Sirtuin 1 Retards Hyperphosphatemia-Induced...

31
Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth Muscle Cells Aya Takemura, Katsuya Iijima, Hidetaka Ota, Bo-Kyung Son, Yuki Ito, Sumito Ogawa, Masato Eto, Masahiro Akishita, Yasuyoshi Ouchi Objective—Arterial calcification is associated with cardiovascular disease as a complication of advanced atherosclerosis. Aged vascular cells manifest some morphological features of a senescent phenotype. Recent studies have demonstrated that mammalian sirtuin 1 (SIRT1), a histone deacetylase, is an exciting target for cardiovascular disease management. Here, we investigated the role of SIRT1 in a calcification model of vascular smooth muscle cells (SMCs). Methods and Results—In adenine-induced renal failure rats with hyperphosphatemia, massive calcification was induced in the aortic media. Senescence-associated -galactosidase (SA-gal) activity, a marker of cellular senescence, in medial SMCs was significantly increased, and its induction was positively associated with the degree of calcification. In cultured SMCs, inorganic phosphate (Pi) stimulation dose-dependently increased SA-gal-positive cells, and Pi-induced senescence was associated with downregulation of SIRT1 expression, leading to p21 activation. The activation via SIRT1 downregulation was blunted by inhibition of Pi cotransporter. Activation of SIRT1 by resveratrol significantly reduced the senescence-associated calcification. Conversely, SIRT1 knockdown by small interfering RNA accelerated the Pi-induced SMC senescence and subsequent calcification. In addition, SIRT1 knockdown induced phenotypic change from a differentiated state to osteoblast-like cells. The senescence-related SMC calcification was completely prevented by p21 knockdown. In addition to Pi-induced premature senescence, SMCs with replicative senescence were also more sensitive to Pi-induced calcification compared with young SMCs, and this finding was attributable to augmented p21 expression. Conclusion—SIRT1 plays an essential role in preventing hyperphosphatemia-induced arterial calcification via inhibition of osteoblastic transdifferentiation. In addition, Pi-induced SMC calcification may be associated with both premature and replicative cellular senescence. (Arterioscler Thromb Vasc Biol. 2011;31:2054-2062.) Key Words: cellular senescence hyperphosphatemia longevity gene SIRT1 vascular calcification vascular smooth muscle cell A therosclerotic vascular damage associated with aging manifests several features, namely atherosis, sclerosis, and calcific change, finally leading to cardiovascular events. These pathological changes result in arterial wall thickening (localized morphological changes) and arterial stiffening (functional changes). 1 Arterial calcification makes the man- agement of hemodynamics more difficult in the elderly, because ectopic calcium deposition in the aorta and arteries contributes to vessel wall stiffening and loss of elastic recoil. 2 These pathological conditions result in unstable hemodynamic consequences, finally leading to a decline in end-organ perfusion and subsequent ischemic events. Recently, several reports have demonstrated that aortic calcification detectable on chest X-ray examination is a strong predictor of future cardiovascular events beyond traditional risk factors. 3 Arterial calcification is anatomically separated into two types, intimal and medial calcification. 4 Intimal calcification, which is seen as patchy scattered deposits only occurring within atherosclerotic plaques, is shown to be associated with plaque vulnerability. 5 On the other hand, medial calcification, which is frequently seen in the elderly and in diabetes and chronic renal failure, is observed as continuous linear depos- its along the internal elastic lamina. 6 Advanced atherosclero- sis with both types of calcified lesions is the consequence of overlapping pathological mechanisms. Ectopic calcification in the vasculature has been shown to result from passive precipitation of calcium with aging and osteoporosis, the so-called calcium shift theory, as a previous hypothesis. 7 However, accumulating recent evidence has shown it to be attributable to an active “cell-mediated process” resem- bling osteogenesis in bone rather than passive mineral precipi- tation in vascular smooth muscle cells (SMCs). 8,9 Silent information regulator-2 (Sir2), an NAD -dependent HDAC, is highly conserved in organisms ranging from Archaea Received on: May 12, 2010; final version accepted on: May 25, 2011. From the Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. Correspondence to Katsuya Iijima, MD, PhD, Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan. E-mail [email protected] © 2011 American Heart Association, Inc. Arterioscler Thromb Vasc Biol is available at http://atvb.ahajournals.org DOI: 10.1161/ATVBAHA.110.216739 2054 by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on April 23, 2018 http://atvb.ahajournals.org/ Downloaded from

Transcript of Sirtuin 1 Retards Hyperphosphatemia-Induced...

Page 1: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

Sirtuin 1 Retards Hyperphosphatemia-Induced Calcificationof Vascular Smooth Muscle Cells

Aya Takemura, Katsuya Iijima, Hidetaka Ota, Bo-Kyung Son, Yuki Ito, Sumito Ogawa, Masato Eto,Masahiro Akishita, Yasuyoshi Ouchi

Objective—Arterial calcification is associated with cardiovascular disease as a complication of advanced atherosclerosis.Aged vascular cells manifest some morphological features of a senescent phenotype. Recent studies have demonstratedthat mammalian sirtuin 1 (SIRT1), a histone deacetylase, is an exciting target for cardiovascular disease management.Here, we investigated the role of SIRT1 in a calcification model of vascular smooth muscle cells (SMCs).

Methods and Results—In adenine-induced renal failure rats with hyperphosphatemia, massive calcification was inducedin the aortic media. Senescence-associated �-galactosidase (SA�-gal) activity, a marker of cellular senescence, inmedial SMCs was significantly increased, and its induction was positively associated with the degree of calcification.In cultured SMCs, inorganic phosphate (Pi) stimulation dose-dependently increased SA�-gal-positive cells, andPi-induced senescence was associated with downregulation of SIRT1 expression, leading to p21 activation. Theactivation via SIRT1 downregulation was blunted by inhibition of Pi cotransporter. Activation of SIRT1 by resveratrolsignificantly reduced the senescence-associated calcification. Conversely, SIRT1 knockdown by small interfering RNAaccelerated the Pi-induced SMC senescence and subsequent calcification. In addition, SIRT1 knockdown inducedphenotypic change from a differentiated state to osteoblast-like cells. The senescence-related SMC calcification wascompletely prevented by p21 knockdown. In addition to Pi-induced premature senescence, SMCs with replicativesenescence were also more sensitive to Pi-induced calcification compared with young SMCs, and this finding wasattributable to augmented p21 expression.

Conclusion—SIRT1 plays an essential role in preventing hyperphosphatemia-induced arterial calcification via inhibitionof osteoblastic transdifferentiation. In addition, Pi-induced SMC calcification may be associated with both prematureand replicative cellular senescence. (Arterioscler Thromb Vasc Biol. 2011;31:2054-2062.)

Key Words: cellular senescence � hyperphosphatemia � longevity gene SIRT1 � vascular calcification� vascular smooth muscle cell

Atherosclerotic vascular damage associated with agingmanifests several features, namely atherosis, sclerosis,

and calcific change, finally leading to cardiovascular events.These pathological changes result in arterial wall thickening(localized morphological changes) and arterial stiffening(functional changes).1 Arterial calcification makes the man-agement of hemodynamics more difficult in the elderly,because ectopic calcium deposition in the aorta and arteriescontributes to vessel wall stiffening and loss of elastic recoil.2

These pathological conditions result in unstable hemodynamicconsequences, finally leading to a decline in end-organperfusion and subsequent ischemic events. Recently, severalreports have demonstrated that aortic calcification detectableon chest X-ray examination is a strong predictor of futurecardiovascular events beyond traditional risk factors.3

Arterial calcification is anatomically separated into twotypes, intimal and medial calcification.4 Intimal calcification,

which is seen as patchy scattered deposits only occurringwithin atherosclerotic plaques, is shown to be associated withplaque vulnerability.5 On the other hand, medial calcification,which is frequently seen in the elderly and in diabetes andchronic renal failure, is observed as continuous linear depos-its along the internal elastic lamina.6 Advanced atherosclero-sis with both types of calcified lesions is the consequence ofoverlapping pathological mechanisms.

Ectopic calcification in the vasculature has been shown toresult from passive precipitation of calcium with aging andosteoporosis, the so-called calcium shift theory, as a previoushypothesis.7 However, accumulating recent evidence has shownit to be attributable to an active “cell-mediated process” resem-bling osteogenesis in bone rather than passive mineral precipi-tation in vascular smooth muscle cells (SMCs).8,9

Silent information regulator-2 (Sir2), an NAD�-dependentHDAC, is highly conserved in organisms ranging from Archaea

Received on: May 12, 2010; final version accepted on: May 25, 2011.From the Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.Correspondence to Katsuya Iijima, MD, PhD, Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo,

Bunkyo-ku, Tokyo 113-8655, Japan. E-mail [email protected]© 2011 American Heart Association, Inc.

Arterioscler Thromb Vasc Biol is available at http://atvb.ahajournals.org DOI: 10.1161/ATVBAHA.110.216739

2054

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on A

pril 23, 2018http://atvb.ahajournals.org/

Dow

nloaded from

Page 2: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

to humans.10 In yeast, Sir2 has been shown to play critical rolesin DNA repair, stress resistance, and longevity. Mammaliansirtuin 1 (SIRT1), the closest homolog of Sir2, regulates the cellcycle, apoptosis, and metabolism by interacting with a numberof molecules, including p53, promyelocytic leukemia protein,Foxo, Ku70, and peroxisome proliferator-activated receptor-�.11

A previous study has shown that SIRT1 antagonizes p53-mediated premature senescence in mouse embryo fibroblasts.12

In addition, we have recently demonstrated that SIRT1 inhibitsoxidative stress-induced premature senescence in vascular en-dothelial cells.13 However, the detailed mechanism of howSIRT1 affects vascular SMC senescence and arterial calcifica-tion remains unclear.

In this study, we hypothesized that SIRT1 plays an impor-tant role in preventing arterial calcification due to renalfailure, in association with modulation of cellular senescence.Here, we demonstrated the protective potential of SIRT1against hyperphosphatemia-induced premature and replica-tive senescence and subsequent calcification in SMCs.

MethodsAortic Calcification in Renal Failure RatsRenal failure was induced in rats by a 0.75% adenine-containing dietas previously described.14 All procedures and animal care were inaccordance with the Guide for the Care and Use of Laboratory Animalsof the University of Tokyo. Detailed methods are described in thesupplemental materials, available online at http://atvb.ahajournals.org.

Induction of SMC CalcificationPrimary human aortic SMCs (HASMCs) were treated with a patholog-ical concentration of inorganic phosphate (Pi) up to 3.2 mmol/L inculture medium as previously described.29 To quantitatively measurePi-induced calcification, two distinct experiments were performed aspreviously described14: (1) intracellular calcium deposition as deter-mined by o-cresolphthalein complexone method, and (2) visualizationof mineralization as determined by von Kossa staining, Detailedmethods are described in the supplemental materials.

Senescence-Associated �-Galactosidase StainingTo assess senescent changes in the phenotype of cultured HASMCsor aortic medial cells of rats, staining for senescence-associated�-galactosidase (SA�-gal), a well-established biomarker of cellularsenescence, was performed. Detailed methods are described in thesupplemental materials.

Knockdown of SIRT1 or p21 by SmallInterfering RNAHASMCs were transfected with 200 pmol/L small interfering RNA(siRNA) for SIRT1, p21WAF1/CIP1, or both. Detailed methods aredescribed in the supplemental materials.

Real-Time Polymerase Chain Reaction Analysis:Osteoblastic MarkersTo examine whether Pi stimulation induces change to an osteoblasticphenotype, the expression of Runx-2/Cbfa-1 and alkaline phospha-tase, which are well known to be representative osteoblastic markers,was checked using real time-polymerase chain reaction analysis. Inaddition, the effect of knockdown of SIRT1, p21, or both by siRNAon the osteoblastic phenotypic change in HASMCs was examined.Primer sequences are shown in Supplemental Figure I.

ResultsAssociation of Senescent Vascular Cells WithAortic Medial Calcification in Renal Failure RatsThe adenine-fed rats had severe renal failure, with a hugeincrease in serum creatinine (3.0�0.9 mg/dL in renal failure

rats versus 0.3�0.0 mg/dL in control rats), similar to aprevious report.14 The renal failure rats showed an approxi-mately 2.0-fold increase in serum phosphorus (18.9�4.7mg/dL) compared with control rats (9.8�0.9 mg/dL). Histo-logical assessment using von Kossa staining showed that theaorta in renal failure rats had extensive linear calcification,which was localized in the aortic media, resembling the typicalMonckeberg’s pattern (Figure 1A). Numerous SA�-gal positivecells were found in the aortic media of renal failure rats, whereasthe aortic wall in control rats did not contain senescent cells(Figure 1B). The senescent cells were mainly localized to thecalcified area and its surrounding area, which was defined as thearea not stained black by von Kossa staining. Quantitativeassessment showed that the number of senescent cells with highSA�-gal activity was positively correlated with the calcified areain the aortic media (Figure 1C).

Pi Induces Cellular Senescence in Cultured SMCsOn the basis of our results obtained from animal experiments,we hypothesized that senescent SMCs in the aortic media arestrongly associated with the development of arterial calcifi-cation. Therefore, the effect of excessive Pi stimulation(2.6 mmol/L) on cellular senescence in cultured SMCs wasexamined. SA�-gal-positive senescent HASMCs were signif-icantly induced by not only angiotensin II (Ang II) but also Pi

Figure 1. Presence of senescent vascular cells colocalized withcalcification in aortic media of renal failure rats. A, Rats withsevere renal failure had massive calcification throughout theaorta (right) compared with control rats (left) (n�5). Yellowarrows indicate calcified area. Morphological assessment by vonKossa staining showed extensive calcification in the aorticmedia of renal failure rats. Scale bar�500 �m. B, Senescentvascular cells (senescence-associated �-galactosidase [SA�-gal]-positive: blue) were significantly detected throughout thecalcified area (Calc) in renal failure rats, whereas these senes-cent cells were not present in control rats. Scale bar�100 �m.C, Localized association between calcification and senescentcells is shown in renal failure rats. SA�-gal-positive cells werefrequently found in areas with marked calcification. D, The asso-ciation of the number of SA�-gal-positive cells with the calcifiedarea in each photograph was evaluated. The senescent cellnumber was linearly correlated with the area of calcification inthe aortic media of renal failure rats (calcified area in media:percentage).

Takemura et al SIRT1 Retards Vascular SMC Calcification 2055

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 3: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

stimulation (Figure 2A). Notably, Pi stimulation increasedcalcium deposition; however, Ang II alone did not (Figure2B). It suggests that high-dose Pi condition, but not stress byAng II alone, is indispensable to induce SMC calcification.

These findings also suggest that intracellular Pi influx at leastis essential to induce this SMC calcification model.

In addition, to determine how many days after the initiationof Pi stimulation the cells showed a senescent phenotype andsubsequent calcification, the time-dependent effects of Pistimulation on both SA�-gal activity and calcium depositionwere examined. As shown in Figure 2C, SA�-gal-positivecells were significantly increased by Pi stimulation even onday 1, although calcium deposition was not markedly in-creased at the same time point. A statistically significantincrease in calcium deposition was found from day 3 andlater. Cotreatment with phosphonoformic acid, an inhibitor ofNa-dependent phosphate cotransporter (NPC), showed signif-icant inhibition of Pi-induced senescence (Figure 2D). Ourprevious report showed that treatment with PFA completelyinhibited Pi-induced SMC calcification,15 suggesting theimportance of increased intracellular influx of phosphate inPi-induced SMC senescence.

Downregulation of SIRT1 by PiTreatment of HASMCs with Pi caused downregulation ofSIRT1 expression in a time-dependent manner (Figure 3A).The decline was dependent on Pi concentration (data notshown). An increase in acetylation of both substrates ofSIRT1, histone-3 and p53 (a nonhistone substrate), was foundaccording to the decline in SIRT1 deacetylase activity. Inaddition, expression of p21, a downstream molecule of p53,was significantly induced by Pi as well. Quantitative assess-ment showed that an increase in these expression levels ofacetylated (Ac)-p53 and p21 on day 3 and day 6 wasstatistically significant compared with the pretreatment lev-els, suggesting that downregulation of SIRT1 activity maymediate the subsequent increase in Ac-p53 and p21expression.

To address whether SIRT1 downregulation-related SMCsenescence and calcification are reversible or not, theeffects of continuation or termination of high-dose Pi wereexamined. As shown in Figure 3B, the continuation of Piup to day 10 was associated with SIRT1 downregulationand subsequent upregulation of Ac-p53 and p21, leading toinduction of senescence-related calcification. However,the slight increase in senescent cells was not statisticallysignificant, although calcification was significantly in-duced. Of note, the Pi-induced downregulation of SIRT1was almost completely reversed by withdrawal (termina-tion) of Pi stimulation (exchange of Pi from 2.6 mmol/L to1.4 mmol/L as a normal level on day 6) as shown in Figure3B. According to the restoration of SIRT1, levels of bothAc-p53 and p21 were also decreased without more pro-gression. In addition, termination of Pi showed no progres-sion of senescence-related calcification; however, preex-isting senescent cells and calcification on day 6 continuedwithout regression.

Next, NPC inhibition by PFA completely blunted Pi-induced SIRT1 downregulation and subsequent activation ofits downstream p53/p21 pathway (Figure 3C).

Regulation of SIRT1 Modulates Pi-Induced SMCSenescence and CalcificationThe effects of modulation of SIRT1 activity on Pi-inducedcellular senescence were investigated. First, sirtinol, a chem-

Figure 2. Inorganic phosphate (Pi) stimulation induces cellularsenescence in vascular smooth muscle cells (SMCs) via itscotransporter. A, The effect of Pi on senescent transition inhuman aortic SMCs (HASMCs) was examined. Representative pho-tographs showed that senescence-associated �-galactosidase(SA�-gal) activity (blue) in cells was significantly induced by notonly angiotensin II (Ang II: 10 pmol/L, as a positive control) but alsoPi stimulation (2.6 mmol/L). B, The number of senescent cells wassignificantly increased by not only Ang II but also Pi. Calciumdeposition was significantly increased by Pi; however, calcificationin HASMCs was not induced by Ang II alone in the absence of Pi.C, Senescent cells were significantly increased by Pi stimulationeven on day 1; however, a statistically significant increase in cal-cium deposition was found from day 3 and later. D, Inhibition ofthe phosphate cotransporter Na-dependent phosphate cotrans-porter by the inhibitor phosphonoformic acid (PFA) (100 �mol/L)reduced SA�-gal activity, which was increased by Pi (2.6 mmol/L)in HASMCs. Each experiment was performed al least 3 times.

2056 Arterioscler Thromb Vasc Biol September 2011

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 4: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

ical inhibitor of SIRT1, induced an increase in SA�-gal-positive cells even under a normal Pi (1.4 mmol/L), and theincreased number of senescent cells induced by Pi wassignificantly augmented by sirtinol (Figure 4A). Sirtinoldose-dependently augmented Pi-induced calcification, al-though no augmentation was found under a normal Pi (Figure4B and 4C). Conversely, treatment with resveratrol, anactivator of SIRT1, significantly reduced both Pi-inducedsenescent transition and calcification in a dose-dependentmanner (Figure 4D to 4F).

Second, complete knockdown of SIRT1 by siRNA causeda significant increase in acetylation of both substrates(histone-3 and p53) and p21 expression (Figure 5A). Simi-larly to sirtinol, SIRT1 inhibition by siRNA also augmentednot only senescent transition (Figure 5A, bottom) but alsocalcium deposition (Figure 5C, top).

Although stimulation with Ang II alone could increase thenumber of SA�-gal-positive cells, it did not increase calcium

deposition. To understand the mechanism of these discrepantphenomena, the effect of Ang II alone on osteoblasticphenotypic change was examined. Ang II alone did notincrease the expression of Runx2 in the absence of Pistimulation, unlike Pi stimulation (Figure 5B).

To understand the detailed mechanism by which SIRT1modulates senescence-related calcification, the effect ofSIRT1 on phenotypic change in HASMCs was examined. Piinhibited the expression of caldesmon, a differentiated SMClineage marker, and complete knockdown of SIRT1 aug-mented the Pi-induced partial downregulation of caldesmon(Figure 5C, middle). In contrast, real-time polymerase chainreaction analysis showed that Pi induced the expression oftwo representative osteoblastic markers, Runx-2/Cbfa-1 andalkaline phosphatase (Figure 5C, bottom) with statisticalsignificance. In addition, complete knockdown of SIRT1using siRNA significantly accelerated the Pi-induced os-

Figure 3. Inorganic phosphate (Pi) stimulation leads to sirtuin 1 (SIRT1) downregulation and subsequent p21 activation. A, The effect ofPi on SIRT1 expression and its downstream pathway was examined. Treatment of human aortic SMCs (HASMCs) with Pi (2.6 mmol/L)showed downregulation of SIRT1 expression, leading to an increase in acetylation of its substrates (acetylated [Ac]-H3 and Ac-p53)and p21 expression. Bottom: Quantitative analysis showed that Pi gradually induced not only SIRT1 downregulation but also upregula-tion of Ac-p53 and p21. B, To address whether SIRT1 downregulation-related senescence and subsequent calcification are reversible,the effects of continuation or termination of high-dose Pi were examined. As shown in 4th lane from left, termination (on day 6) of Pishowed no progression of senescence-related calcification in association with restoration of SIRT1, whereas continuation (up to day10, 3rd lane from left) of Pi stimulation showed further progression of calcification. C, Treatment with phosphonoformic acid (PFA), aNa-dependent phosphate cotransporter inhibitor, completely reversed Pi-induced SIRT1 downregulation. A decline in Ac-H3 andAc-p53 reflected the restoration of SIRT1 deacetylase activity. Pi-induced p21 activation was significantly inhibited by inhibition of Pitransport.

Takemura et al SIRT1 Retards Vascular SMC Calcification 2057

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 5: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

teoblastic phenotypic change, suggesting that modulationof SIRT1 is associated with osteoblastic phenotypicchange in SMCs.

Inhibition of Senescence-Related Calcification inSMCs by p21 KnockdownTo address the association of p21 with senescence-relatedcalcification, knockdown of p21 using siRNA was per-formed. Treatment of p21 siRNA (up to 200 pmol/L) com-pletely inhibited p21 (Figure 5D). p21 knockdown com-pletely inhibited Pi-induced senescence and subsequentcalcification (Figure 5E).

Regulation of NPC-Mediated Runx2 Expression bySIRT1/p21 PathwayAs the next step, the role of SIRT1 in NPC-mediatedRunx2/Cbfa1 expression was examined. First, completeknockdown of SIRT1 did not show any change in bothosteoblastic markers, Runx2 and alkaline phosphatase, in anormal Pi (Supplemental Figure I). As shown in Figure 5F,

Pi-induced Runx2 was significantly blunted by PFA, an NPCinhibitor. SIRT1 activation by resveratrol inhibited Pi-induced Runx2 activation. The Runx2 induction was aug-mented by knockdown of SIRT1 by siRNA, and the activa-tion was completely inhibited by PFA. Surprisingly, Runx2activation was strongly inhibited by knockdown of p21 alone.In addition, the inhibition of Runx2 induction by doubleknockdown of SIRT1 and p21 was less than that by singleknockdown of SIRT1.

To address a difference in senescent induction by Pi or AngII, immunohistological assessment of SIRT1 in HASMCswas examined (Supplemental Figure II). Although SIRT1was predominantly localized in nucleus without Pi, thetranslocation of SIRT1 to cytoplasm was observed after Pistimulation for 24 hours, and its expression disappeared inboth areas on day 6. In contrast, Ang II stimulation did notshow the dynamic translocation.

High Sensitivity of SMCs With ReplicativeSenescence to Pi-Induced CalcificationNot only Pi-induced “premature senescence” in HASMCs butalso the effects of Pi on “replicative senescence” wereevaluated. Senescent cells (passage 18) were more sensitiveto Pi-induced calcification compared with young cells (pas-sage 7) (Figure 6A). SIRT1 expression was downregulated insenescent cells compared with young cells, and the down-regulation was significantly augmented by Pi stimulation(Figure 6B, top). In parallel with this finding, senescent cellsshowed an increase in Ac-p53 and p21 expression. Statisticalanalyses using densitometric measurement showed that (1)downregulation of SIRT1 and upregulation of Ac-53 and p21were augmented by replicative senescence, and (2) Pi inhib-ited the SIRT1-p21 pathway even in cells with replicativesenescence (passage 18) (Figure 6B, bottom).

DiscussionVascular aging, leading to cardiovascular disease, manifestscomplex and diverse vascular changes (eg, impairment ofdistensibility due to loss of arterial elasticity).1,16 Arterial wallstiffness resulting from ectopic calcification is a complicationof advanced atherosclerosis and makes the management ofhemodynamics more difficult in the elderly. Few reports haveaddressed whether cellular senescence is associated withSMC calcification. This study showed the importance ofSIRT1, a longevity gene, in arterial calcification in associa-tion with cellular senescence.

First, our data obtained from animal experiments clearlyshowed the association of senescent SMCs with aortic medialcalcification in the renal failure rats with hyperphosphatemia.Senescent cells showed significant colocalization with cal-cium deposition. Intriguingly, numerous senescent cells couldbe detected before microscopic calcification occurred at 4weeks after the start of renal failure induction (data notshown), suggesting that the transition to a senescent pheno-type in medial SMCs may be associated with the initiationand progression of calcification. Therefore, hyperphos-phatemia, a potent uremic factor, may be a stimulator toinduce senescent phenotypic transition of medial SMCs.

Figure 4. Modulation of sirtuin 1 (SIRT1) affects inorganic phos-phate (Pi)–induced senescent phenotypic change and calcifica-tion in smooth muscle cells (SMCs). The effects of sirtinol (achemical inhibitor of SIRT1 activity; A to C) and resveratrol (anactivator of SIRT1; D to F) on Pi-induced senescent phenotypicchange and calcification were examined (n�6). A, SIRT1 inhibi-tion by sirtinol (10 �mol/L) showed an increase in the number ofsenescence-associated �-galactosidase (SA�-gal)-positive cellseven without Pi stimulation. The increase in Pi-induced senes-cence was significantly augmented by sirtinol. Sirtinol aug-mented Pi-induced calcium deposition in human aortic SMCs(HASMCs) in a time-dependent (B) and dose-dependent manneron day 6 (C). Conversely, treatment with resveratrol (Resv;10 �mol/L) showed a reduction of the Pi-induced senescentphenotype (D) and calcification (E). The inhibitory effect of res-veratrol on calcification was dose dependent (F).

2058 Arterioscler Thromb Vasc Biol September 2011

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 6: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

Figure 5. Augmentation of senescence-related smooth muscle cell (SMC) calcification by sirtuin 1 (SIRT1) knockdown in associationwith osteoblastic phenotypic change and prevention of inorganic phosphate (Pi)–induced changes by p21 knockdown. A, To achieveSIRT1 knockdown in human aortic SMCs (HASMCs), small interfering RNA (siRNA) was simultaneously administered at the start of Pistimulation (2.6 mmol/L). Complete inhibition of SIRT1 showed a significant increase in acetylation of both substrates (acetylated[Ac]-H3 and Ac-p53), p21 expression and senescence-associated �-galactosidase (SA�-gal)-positive cells. B, Angiotensin II (Ang II)alone (10 pmol/L) did not increase the expression of Runx2 in the absence of Pi stimulation, unlike Pi stimulation. C, top: SIRT1 knock-down by siRNA significantly accelerated Pi-induced calcification (n�6), whereas control (Ctrl) siRNA did not. C, middle and bottom:Western blots showed that Pi partially inhibited the expression of a differentiated SMC marker, caldesmon, and complete knockdown ofSIRT1 by siRNA augmented its downregulation. Real-time polymerase chain reaction analysis showed that Pi induced the expression ofRunx-2 and alkaline phosphatase (ALP). Complete knockdown of SIRT1 significantly accelerated the Pi-induced osteoblastic markers.A.U. indicates arbitrary units. *P�0.05. D and E, Knockdown of p21 by siRNA (200 pmol/L) significantly reduced the senescent pheno-typic change and subsequent calcification (n�6). F, The role of SIRT1/p21 axis in Na-dependent phosphate cotransporter-mediatedRunx2 expression was evaluated. Augmentation of Pi-induced Runx2 expression by SIRT1 knockdown was significantly inhibited bydouble knockdown of SIRT1 and p21. *P�0.05 vs control without Pi stimulation (left column), **P�0.05 vs Pi-stimulated cells withSIRT1 siRNA (sixth column from left).

Takemura et al SIRT1 Retards Vascular SMC Calcification 2059

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 7: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

Second, we also confirmed the association of Pi-inducedSMC senescence with calcification in in vitro experiments.Senescent SMCs were significantly increased by Pi even onday 1, although calcium deposition was not markedly in-creased at the same time point. A statistically significantincrease in calcium deposition was found from day 3 and

later. Considering these data, we hypothesize that (1) calciumdeposition may be more readily induced in senescent cellscompared with nonsenescent cells, and (2) Pi-induced senes-cent change is observed earlier than calcium deposition. Inother words, senescent transition associated with Runx2induction may lead to progressive calcification.

Senescent SMCs were associated with the SIRT1-relatedp53/p21 pathway, based on the findings that SIRT1 knockdownaugmented not only cellular senescence but also calcification. Inaddition, p21 knockdown completely inhibited senescence-related calcification induced by Pi. This raises the question ofhow cellular senescence in SMCs is associated with calcifica-tion. Our experiments to understand the detailed mechanisms bywhich SIRT1 modulates senescence-related calcificationshowed that Pi-induced SIRT1 downregulation led to the phe-notypic change from a differentiated state to osteoblast-like cellsin SMCs. It has been reported that Pi induces osteoblasticchange, in which NPC plays a role in inducing Runx2/Cbfa-1expression, in SMCs.17 As the next step, to determine howSIRT1 regulates NPC-mediated Runx2 expression, we exam-ined the effects of knockdown of SIRT1, p21, or both by siRNAon Pi-induced Runx2 expression. Our data shown in Figure 5Fsuggested that (1) NPC plays an essential role in Pi-inducedRunx2 expression, (2) SIRT1 has an inhibitory effect on NPC-mediated Runx2 expression, (3) knockdown of p21 aloneameliorates Runx2 induction, and (4) p21-related osteoblasticchange is at least in part dependent on SIRT1.

There is now the new question of how SIRT1 regulatesRunx2 regulation. A report by Jeon18 has shown that acety-lation of Runx2 itself is important in osteoblast differentia-tion, and it is downregulated by HDAC activities. Based onthis evidence, SIRT1, 1 of the HDACs, may be able todeacetylate Runx2, leading to inhibition of Runx2-relatedosteoblastic transition in SMCs. Therefore, the inhibition ofSIRT1 by hyperphosphatemia may lead to Runx2 activationvia its hyperacetylation. Further investigation of the detailedmechanism of the SIRT1/p21/osteoblastic gene axis isneeded. These data clearly suggest that SIRT1 activation mayinhibit the hyperphosphatemia-induced osteoblastic pheno-typic change of SMCs, and the degree of change may bedependent on SIRT1 expression level. It is possible that theinhibition of SIRT1 expression by Pi alone is “partial,”because complete downregulation of SIRT1 by siRNA wors-ened the dynamic phenotypic change compared with Pi only.We have already shown that tumor necrosis factor-�, a potentatherogenic cytokine, augmented Pi-induced SMC calcifica-tion, as previously described.19 In addition, tumor necrosisfactor-� significantly decreased Pi-induced SIRT1 down-regulation further (data not shown). According to theseresults, we currently hypothesize that hyperphosphatemiainduces SIRT1 downregulation and subsequent osteoblasticphenotypic change in SMCs, leading to calcification, andthese changes are worsened by some harmful atherogenicfactors, which decrease SIRT1 expression/activity further.These results provide a new insight, showing that SIRT1plays an essential role in the prevention of arterial calcifica-tion and that the beneficial effect may be associated with aninhibition in Pi-induced SMC senescent transition.

Figure 6. High sensitivity of smooth muscle cells (SMCs) with rep-licative senescence to inorganic phosphate (Pi)–induced calcifica-tion. The effects of replicative senescence in human aortic SMCs(HASMCs) on Pi-induced calcification (A) and sirtuin 1 (SIRT1)-related molecules (B) were also evaluated. A, Senescent cells (pas-sage 18 [P18]) were more sensitive to Pi-induced calcificationcompared with young cells (passage 7 [P7]) (n�6). Representativephotographs of von Kossa staining (bottom) show strong inductionof calcium deposition by Pi (2.6 mmol/L). B, Senescent HASMCs(P18) showed a decline in SIRT1 expression and an increase inp21 expression compared with young cells (P7). Pi stimulation ofsenescent cells significantly inhibited SIRT1 expression and accel-erated the increase in p21 and acetylated (Ac)-p53. Densitometricanalysis confirmed these more sensitive responses in senescentcells. A.U. indicates arbitrary units. *P�0.05.

2060 Arterioscler Thromb Vasc Biol September 2011

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 8: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

In addition, Ang II did not increase calcium deposition,although the stimulation increased the number of senescentcells. Of note, Ang II alone did not increase Runx2 expres-sion in the absence of Pi (Figure 5B). This result suggests thatSMC senescence shows two different features: one is SA�-gal-positive cells with an increase in Runx2 and the other isSA�-gal-positive cells without. First, it has recently beenreported that SMCs with replicative senescence, rather thanthe cells without senescence, show hypersensitivity in re-sponse to induction of calcification with the more inductionof osteoblastic markers,20 suggesting that the induction ofosteoblastic transdifferentiation is strongly associated withthe senescent change in SMCs. In addition, the translocationof SIRT1 to cytoplasm was observed after Pi stimulation for24 hours, although SIRT1 predominantly localized in nucleuswithout Pi. In contrast, Ang II did not show the dynamictranslocation. Thinking about the mechanism for regulatingthe activity of HDACs, including SIRT1, recent severalreports show the importance of their coordinated shuttlingbetween nucleus and cytoplasm. A report demonstrates thatHDAC7, an HDAC, represses the transcriptional activity ofRunx2 and that osteogenic stimuli induce export of HDAC7from nucleus, leading to a decline in the repressive potentialsof HDAC7 for Runx2.21 On the basis of our findings and aprevious report, the reason that stimulation with Ang II alonedid not induce Runx2 expression and subsequently SMCcalcification may in part depend on the difference of SIRT1translocation after stimulation. Therefore, we strongly hy-pothesize that in the senescent SMCs with upregulation ofp21, Pi stimulation, but not Ang II stimulation, may activateRunx2 via at least two phenomena, the hyperacetylation ofRunx2 by SIRT1 downregulation and the dynamic SIRT1translocation, leading to marked osteoblastic transdifferentia-tion and subsequent calcification. In addition, we haveanother hypothesis. In general, it has been shown thathigh-dose Pi navigates release of matrix vesicles from SMCsin parallel with osteoblastic transdifferentiation. The vesiclesplay an essential role in the initiation of hydroxyapatiteaggregation, so-called nucleation. Accumulating recent re-ports show that the nanocrystal formation as an initial stepunder hyperphosphatemia accelerates the harmful cascade ofosteoblastic transdifferentiation in SMCs via endocytosis.22,23

Maybe Ang II alone does not induce the nanocrystal forma-tion and the cascade of osteoblastic change. Therefore, weexplain that the difference of senescent phenotypic changes inSMCs between both stimulations, Pi and Ang II alone, maydepend on (1) SIRT1 translocation and (2) nanocrystalformation to accelerate calcification. Further investigation toaddress the detailed mechanisms by which SIRT1 regulatesosteoblastic transdifferentiation in SMCs under the cellularsenescence is needed.

Are SIRT1 downregulation-related SMC senescence andsubsequent calcification reversible or not? To answer thisquestion, the effects of continuation or termination of high-dose Pi were examined. As shown in Figure 3B, termination(on day 6) of Pi showed no progression of senescence-relatedcalcification in association with the restoration of SIRT1,whereas continuation (up to day 10) of Pi stimulation showedfurther progression of calcification. It is suggested that a

therapeutic strategy to manage hyperphosphatemia to thenormal range of serum phosphate concentration may lead toat least termination of progressive calcification via reversal ofSIRT1 activity.

Cellular senescence has been shown to have two features:not only stress-induced premature senescence but also repli-cative senescence, indicating a limited number of divisions inculture.24 In fact, both endothelial cells and SMCs derivedfrom human atherosclerotic plaques show a senescent pheno-type earlier than do cells from normal vessels.25 Notably, wefound that senescent HASMCs were significantly more sen-sitive to Pi-induced calcification compared with young cells.These results suggest that calcium deposition may be morereadily induced in arterial medial SMCs with replicativesenescence. This insight may explain the mechanisms bywhich arterial calcification occurs in the elderly more fre-quently than in the young population. Therefore, these obser-vations support our hypothesis that arterial calcification isaccelerated by both senescent types (premature and replica-tive senescence) in SMCs. To explore new therapeuticstrategies against arterial calcification, it is essential toinvestigate how to maintain a higher SIRT1 level in thevasculature, leading to prevention of medial SMC senescenceand which drug is capable of achieving it.

How does SIRT1 exert protective effects against SMCcalcification? This study clearly showed that inhibition ofSIRT1 was associated with increases in both Ac-p53 and p21expression. These findings were significantly induced by notonly replicative senescence but also Pi-induced prematuresenescence. SIRT1-mediated deacetylation of p53 inhibitsp53-dependent transactivation of target genes, including p21.A report showed that a decline in cellular deacetylase activityincreases the half-life of endogenous p53,26 suggesting thatp53 acetylation is also associated with p53 stabilization.Therefore, the increased Ac-p53 by Pi-induced SIRT1 down-regulation may induce SMC senescence because of a declinein degradation of p53, leading to calcification. In addition,p53 itself can inhibit SIRT1 transcription because the SIRT1promoter has two response elements to p53.27 Further inves-tigation to address how the SIRT1-p53 negative regulatorypathway is associated with SMC calcification is needed.

On the other hand, regarding p21 activation, it is reportedthat inhibition of p21 expression in the vasculature signifi-cantly attenuates cellular senescence, leading to prevention ofatherosclerosis.28 This evidence suggests a pivotal role of p21in the development of atherosclerosis. p21 activation has beenshown to be regulated by a pathway that is p53 dependent,p53 independent, or both. Okamoto et al have demonstratedthat inhibition of HDAC by trichostatin A showed activationof p21 promoter activity by the Sp1 site even in vascularSMCs, and the induction of p21 was independent of the p53pathway.29 The p21 transcriptional activation in response toHDAC inhibitors was mediated by histone hyperacetylationin its promoter region. Based on these findings, Pi-inducedp21 activation via SIRT1 downregulation may be in partinvolved in a p53-independent pathway, leading to a senes-cent phenotype of SMCs. Further investigation exploringwhich molecule activates the p21 promoter under hyperphos-phatemia is needed.

Takemura et al SIRT1 Retards Vascular SMC Calcification 2061

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 9: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

ConclusionWe showed that SIRT1 exerts a protective role inhyperphosphatemia-based arterial calcification via inhibitionof osteoblastic transdifferentiation, in association with cross-talk between calcification and cellular senescence. Thisability of SIRT1 may orchestrate an analogous protective/longevity paradigm even in vascular SMCs, leading tomaintenance of healthy elasticity of the arterial wall. Strate-gies to maintain a higher level of SIRT1 activity may providenovel therapeutic opportunities for the prevention of arterialcalcification.

AcknowledgmentsThis study was supported by Grants-in-Aid for Scientific Researchfrom the Ministry of Education, Science, Culture and Sports of Japan(No. 19590854, No. 21590947, and No. 20249041), Ono MedicalResearch Foundation, Kanzawa Medical Research Foundation, No-vartis Foundation for Gerontological Research, Takeda ResearchFoundation, and Mitsui-Sumitomo Insurance Welfare Foundation.

Sources of FundingThis manuscript is supported by Grants-in-Aid for Scientific Researchfrom the Ministry of Education, Science, Culture and Sports of Japan(No. 19590854, No. 21590947, No. 20249 041). In particular, this ismainly supported by No. 21590947.

DisclosuresNone.

References1. Safar ME, Levy BI, Struijker-Boudier H. Current perspectives on arterial

stiffness and pulse pressure in hypertension and cardiovascular diseases.Circulation. 2003;107:2864–2869.

2. Abedin M, Tintut Y, Demer LL. Vascular calcification: mechanisms andclinical ramifications. Arterioscler Thromb Vasc Biol. 2004;24:1161–1170.

3. Iijima K, Hashimoto H, Hashimoto M, Son BK, Ota H, Ogawa S, Eto M,Akishita M, Ouchi Y. Aortic arch calcification detectable on chest X-rayis a strong independent predictor of cardiovascular events beyond tradi-tional risk factors. Atherosclerosis. 2010;210:137–144.

4. Demer LL, Tintut Y. Vascular calcification: pathobiology of a multi-faceted disease. Circulation. 2008;117:2938–2948.

5. Ehara S, Kobayashi Y, Yoshiyama M, Shimada K, Shimada Y, Fukuda D,Nakamura Y, Yamashita H, Yamagishi H, Takeuchi K, Naruko T, HazeK, Becker AE, Yoshikawa J, Ueda M. Spotty calcification typifies theculprit plaque in patients with acute myocardial infarction: an intra-vascular ultrasound study. Circulation. 2004;110:3424–3429.

6. Shroff RC, Shanahan CM. The vascular biology of calcification. SeminDial. 2007;20:103–109.

7. Persy V, D’Haese P. Vascular calcification and bone disease: the calci-fication paradox. Trends Mol Med. 2009;15:405–416.

8. Shanahan CM, Cary NR, Salisbury JR, Proudfoot D, Weissberg PL,Edmonds ME. Medial localization of mineralization-regulating proteinsin association with Monckeberg’s sclerosis: evidence for smooth musclecell-mediated vascular calcification. Circulation. 1999;100:2168–2176.

9. Tyson KL, Reynolds JL, McNair R, Zhang Q, Weissberg PL, ShanahanCM. Osteo/chondrocytic transcription factors and their target genesexhibit distinct patterns of expression in human arterial calcification.Arterioscler Thromb Vasc Biol. 2003;23:489–494.

10. Brachmann CB, Sherman JM, Devine SE, Cameron EE, Pillus L, BoekeJD. The SIR2 gene family, conserved from bacteria to humans, functionsin silencing, cell cycle progression, and chromosome stability. GenesDev. 1995;9:2888–2902.

11. Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, Pandita TK,Guarente L, Weinberg RA. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell. 2001;107:149–159.

12. Langley E, Pearson M, Faretta M, Bauer UM, Frye RA, Minucci S,Pelicci PG, Kouzarides T. Human SIR2 deacetylates p53 and antagonizesPML/p53-induced cellular senescence. EMBO J. 2002;21:2383–2396.

13. Ota H, Akishita M, Eto M, Iijima K, Kaneki M, Ouchi Y. Sirt1 modulatespremature senescence-like phenotype in human endothelial cells. J MolCell Cardiol. 2007;43:571–579.

14. Yokozawa T, Zheng PD, Oura H, Koizumi F. Animal model of adenine-induced chronic renal failure in rats. Nephron. 1986;44:230–234.

15. Son BK, Kozaki K, Iijima K, Eto M, Kojima T, Ota H, Senda Y,Maemura K, Nakano T, Akishita M, Ouchi Y. Statins protect humanaortic smooth muscle cells from inorganic phosphate-induced calcifi-cation by restoring Gas6-Axl survival pathway. Circ Res. 2006;98:1024–1031.

16. Ferrari AU, Radaelli A, Centola M. Invited review: aging and the car-diovascular system. J Appl Physiol. 2003;95:2591–2597.

17. Jono S, McKee MD, Murry CE, Shioi A, Nishizawa Y, Mori K, Morii H,Giachelli CM. Phosphate regulation of vascular smooth muscle cell cal-cification. Circ Res. 2000;87:E10–E17.

18. Jeon EJ, Lee KY, Choi NS, Lee MH, Kim HN, Jin YH, Ryoo HM, ChoiJY, Yoshida M, Nishino N, Oh BC, Lee KS, Lee YH, Bae SC. Bonemorphogenetic protein-2 stimulates Runx2 acetylation. J Biol Chem.2006;281:16502–16511.

19. Son BK, Akishita M, Iijima K, Kozaki K, Maemura K, Eto M, Ouchi Y.Adiponectin antagonizes stimulatory effect of tumor necrosis factor-� onvascular smooth muscle cell calcification: regulation of growth arrest-specificgene 6-mediated survival pathway by adenosine 5�-monophosphate-activatedprotein kinase. Endocrinology. 2008;149:1646–1653.

20. Nakano-Kurimoto R, Ikeda K, Uraoka M, Nakagawa Y, Yutaka K, KoideM, Takahashi T, Matoba S, Yamada H, Okigaki M, Matsubara H. Rep-licative senescence of vascular smooth muscle cells enhances the calci-fication through initiating the osteoblastic transition. Am J Physiol HeartCirc Physiol. 2009;297:H1673–H1684.

21. Jensen ED, Gopalakrishnan R, Westendorf JJ. Bone morphogenic protein2 activates protein kinase D to regulate histone deacetylase 7 localizationand repression of Runx2. J Biol Chem. 2009;284:2225–2234.

22. Ewence AE, Bootman M, Roderick HL, Skepper JN, McCarthy G, EppleM, Neumann M, Shanahan CM, Proudfoot D. Calcium phosphate crystalsinduce cell death in human vascular smooth muscle cells: a potentialmechanism in atherosclerotic plaque destabilization. Circ Res. 2008;103:e28–e34.

23. Sage AP, Lu J, Tintut Y, Demer LL. Hyperphosphatemia-induced nano-crystals upregulate the expression of bone morphogenetic protein-2 andosteopontin genes in mouse smooth muscle cells in vitro. Kidney Int.2011;79:414–422.

24. Hayflick L. Current theories of biological aging. Fed Proc. 1975;34:9–13.25. Minamino T, Komuro I. Vascular cell senescence: contribution to ath-

erosclerosis. Circ Res. 2007;100:15–26.26. Ito A, Lai CH, Zhao X, Saito S, Hamilton MH, Appella E, Yao TP.

p300/CBP-mediated p53 acetylation is commonly induced by p53-activating agents and inhibited by MDM2. EMBO J. 2001;20:1331–1340.

27. Nemoto S, Fergusson MM, Finkel T. Nutrient availability regulatesSIRT1 through a forkhead-dependent pathway. Science. 2004;306:2105–2108.

28. Andreassi MG. DNA damage, vascular senescence and atherosclerosis.J Mol Med. 2008;86:1033–1043.

29. Okamoto H, Fujioka Y, Takahashi A, Takahashi T, Taniguchi T, Ishikawa Y,Yokoyama M. Trichostatin A, an inhibitor of histone deacetylase, inhibitssmooth muscle cell proliferation via induction of p21(WAF1). J AtherosclerThromb. 2006;13:183–191.

2062 Arterioscler Thromb Vasc Biol September 2011

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 10: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

Eto, Masahiro Akishita and Yasuyoshi OuchiAya Takemura, Katsuya Iijima, Hidetaka Ota, Bo-Kyung Son, Yuki Ito, Sumito Ogawa, Masato

CellsSirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth Muscle

Print ISSN: 1079-5642. Online ISSN: 1524-4636 Copyright © 2011 American Heart Association, Inc. All rights reserved.

Greenville Avenue, Dallas, TX 75231is published by the American Heart Association, 7272Arteriosclerosis, Thrombosis, and Vascular Biology

doi: 10.1161/ATVBAHA.110.2167392011;31:2054-2062; originally published online June 30, 2011;Arterioscler Thromb Vasc Biol. 

http://atvb.ahajournals.org/content/31/9/2054World Wide Web at:

The online version of this article, along with updated information and services, is located on the

http://atvb.ahajournals.org/content/suppl/2013/10/17/ATVBAHA.110.216739.DC2 http://atvb.ahajournals.org/content/suppl/2011/06/30/ATVBAHA.110.216739.DC1

Data Supplement (unedited) at:

  http://atvb.ahajournals.org//subscriptions/

at: is onlineArteriosclerosis, Thrombosis, and Vascular Biology Information about subscribing to Subscriptions:

  http://www.lww.com/reprints

Information about reprints can be found online at: Reprints: 

document. Question and AnswerPermissions and Rightspage under Services. Further information about this process is available in the

which permission is being requested is located, click Request Permissions in the middle column of the WebCopyright Clearance Center, not the Editorial Office. Once the online version of the published article for

can be obtained via RightsLink, a service of theArteriosclerosis, Thrombosis, and Vascular Biologyin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:

by guest on April 23, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 11: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

Supplement Material

Sirtuin SIRT1 retards hyperphosphatemia-induced calcification of

vascular smooth muscle cells

Methods

Aortic calcification in renal failure rats

Renal failure was induced in rats by a 0.75% adenine-containing diet as previously

described.28

Twelve-week-old male Wistar rats (Nippon Clea Inc., Japan) were pair-fed

standard CE-2 chow (containing 1.2% calcium and 0.6% phosphorus; Nippon Clea Inc.)

in the control group or CE-2 chow containing 0.75% adenine (Sigma) in the renal

failure group for 4 weeks. Then, the diet was returned to normal chow for an additional

4 weeks. After induction of renal failure for 8 weeks in total, the rats were sacrificed to

collect samples. After perfusion with saline at a constant, nonpulsatile pressure of 100

mmHg, the aorta was immediately embedded in OCT compound frozen section and

sequentially cut into cross-sections with 5-μm thickness from each part of the aorta. To

detect calcification in the aortic wall, each cross-section was subjected to von-Kossa

staining to demonstrate mineralization. The calcified area and number of

Page 12: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

SAβ-gal-positive cells in the cross-section were measured by image analysis software

(ImageJ, Scion Image, Maryland, USA). All procedures and animal care were in

accordance with the Guide for the Care and Use of Laboratory Animals of the

University of Tokyo.

Induction of SMC calcification

Primary human aortic SMC (HASMC), derived from the internal thoracic artery

(Clonetics), were treated with a pathological concentration of inorganic phosphate (Pi)

in culture medium. To set up the calcification medium, a mixed solution of Na2HPO4

and NaH2PO4 whose pH was adjusted to 7.4 was added to serum-supplemented DMEM

to final doses of up to 3.2 mmol/L as previously described.29 To quantitatively measure

Pi-induced calcification, two distinct experiments, (1) intracellular calium (Ca)

deposition as determined by o-cresolphthalein complexone method and (2) visualization

of mineralization as determined by von-Kossa staining, were performed as previously

described.28 We have previously confirmed that excessive Pi stimulation dose- and

time-dependently induced calcium deposition in HASMC, whereas a normal Pi dose

(1.4 mmol/L), equivalent to the human physiological level of serum phosphate, did

not.29

Page 13: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

Senescence-associated β-galactosidase (SAβ-gal) staining

To assess senescent changes in the phenotype of cultured HASMC or aortic medial

cells of rats with/without renal failure, staining for senescence-associated

β-galactosidase (SAβ-gal), a well-established biomarker of cellular senescence, was

performed at pH 6.0, as opposed to endogenous lysosomal enzyme detected at pH 4.0 in

normal cells, as previously described.30 Numbers of SAβ-gal-positive cells were

quantitatively counted in the aortic wall or cultured HASMC. As a positive control in in

vitro experiments, angiotensin II (AngII) was used to induce transition to a senescent

phenotype in HASMC.

Knockdown of SIRT1 or p21 by small interfering RNA

HASMC were transfected with 200 pmol/L small interfering RNA (siRNA) for

SIRT1 (GAT GAA GTT GAC CTC CTC A and TGA AGT GCC TCA GAT ATT A,

Santa Cruz Biotechnology) or control (Cntl) using silMPORTER (Upstate). In addition,

knockdown of p21WAF1/CIP1

was performed using 100 to 200 pmol/L siRNA for p21

(CGA CUG UGA UGC GCU AAU G, CCU AAU CCG CCC ACA GGA A, CGU

CAG AAC CCA UGC GGC A, and AGA CCA GCA UGA CAG AUU U) by the same

Page 14: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

method. To inhibit p21 expression effectively and completely, four kinds of sequences

of p21 siRNA were used. HASMC were treated simultaneously with these siRNAs at

the start of Pi stimulation.

Western blot and SDS-PAGE

Protein expression was assessed by Western blot analysis with chemiluminescence

detection. SIRT1 was detected using a rabbit polyclonal anti-SIRT1 antibody (Abcam),

and p53, acetylated

p53 (Ac-p53; Lys-382), acetylated histone-3 (Ac-H3), p21,

caldesmon and -tubulin were detected with monoclonal antibodies (Santa Cruz

Biotechnology). The expression levels of Ac-p53 and Ac-H3 were used to reflect SIRT1

activity as a deacetylase. Caldesmon was used to reflect a lineage marker of SMC

differentiation.

Real-time PCR analysis: Osteoblastic markers

Primer sequences were as follows:

ALP; (forward) ACCATTCCCACGTCTTCACATTTG,

(reverse) AGACATTCTCTCGTTCACCGCC,

Runx-2/Cbfa-1; (forward) TCTGGCCTTCCACTCTCAGT,

Page 15: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

(reverse) GACTGGCGGGGTGTAAGTAA,

SIRT1; (forward) CCTGACTTCAGGTCAAGGGATGGTA,

(reverse) CTGATTAAAAATATCTCCTCGTACAG,

-actin; (forward) CTGGAACGGTGAAGGTGACA,

(reverse) AAGGGACTTCCTGTAACAATGC.

Materials

Angiotensin II (AngII) was used to induce senescent phenotypic change in cultured

HASMC as a positive control. To inhibit activity of Na-dependent phosphate

cotransporter (NPC) stimulated by treatment with exogenous Pi, phosphonoformic acid

(PFA; SIGMA), a chemical inhibitor, was used. Sirtinol (a chemical inhibitor;

Calbiochem) or resveratrol (an activator of SIRT1; WAKO) was used for modulation of

SIRT1 activity. Localization of SIRT1 in HASMC was detected using its antibody

(Santa Cruz: sc-15404).

Immunohistological staining

To address a difference in senescent induction by Pi or AngII, the localization of

SIRT1 in HASMC was compared using immunohistological assessment. SIRT1 specific

Page 16: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

antibody showed localization and its translocation in HSMC before or after stimulation

of Pi alone (2.6 mM) or AngII alone (10 pmol/L). Nucleus was detected by DAPI stain.

Statistical analysis

All results are presented as mean ± standard error (SE). Differences between the

groups were analyzed using ANOVA, followed by Fisher’s PLSD test. A value of P

<0.05 was considered to be significant. All in vitro experiments were performed at least

three times.

Figure legends

Supplemental figure 1. Deterioration of osteoblastic transition by SIRT1

knockdown under high-dose Pi stimulation

The effect of SIRT1 knockdown on osteoblastic markers, Runx2 and ALP, in the

condition of normal Pi or high-dose Pi was examined. Complete knockdown of SIRT1

showed significantly augmented expression of both osteoblastic markers, Runx2 and

ALP, in a high-dose Pi condition; however, augmentation was not found in a normal Pi

condition. These data suggest that intracellular Pi influx by Pi stimulation is essential to

Page 17: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

induce SMC calcification in association with osteoblastic phenotypic change, and the

osteoblastic transition may be correlated with NPC, a cotransporter of Pi.

Supplemental figure 2. Translocation of SIRT1 in HASMC is induced by Pi, but

not AngII.

To address a difference in senescent induction by Pi or AngII, immunohistological

assessment of SIRT1 in HASMC was examined. SIRT1 was predominantly localized in

nucleus without Pi. Dynamic translocation of SIRT1 to cytoplasm was observed after Pi

stimulation (2.6 mM) for 24 hr and its expression disappeared in both areas on day 6. In

contrast, AngII alone (10 pmol/L) did not show the dynamic translocation. SIRT1

siRNA shows complete knockdown of SIRT1 expression. DAPI shows nuclear stain.

Page 18: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

Supplement Material Figure Ⅰ

Ctrl siRNA

SIRT1 siRNA

-

+

Pi

- -

-

- + - +

-

- + -

+

- +

+

-

0

2

4

6

8

10

0

2

4

6

8

10

Ru

nx2

(

A.U

.)

AL

P

(A

.U.)

* *

*

* *

*

** ** n.s.

** ** n.s.

n.s. n.s. n.s.

n.s. n.s. n.s.

Page 19: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

-

SIRT1 DAPI

Pi (day 1)

Pi (day 6)

SIRT1 siRNA

Ang II (day 1)

Supplement Material Figure Ⅱ

Page 20: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

45

19. Wasselius JA, Larsson SA, Jacobsson H. FDG-accumulating athero-sclerotic plaques identified with 18F-FDG-PET/CT in 141 patients. Molimaging Biol. 2009;11:455–459.

20. Rudd JH, Warburton EA, Fryer TD, Jones HA, Clark JC, Antoun N,Johnstrom P, Davenport AP, Kirkpatrick PJ, Arch BN, Pickard JD,Weissberg PL. Imaging atherosclerotic plaque inflammation with [18F]-fluorodeoxyglucose positron emission tomography. Circulation. 2002;105:2708–2711.

21. Hyafil F, Cornily JC, Rudd JH, Machac J, Feldman LJ, Fayad ZA.Quantification of inflammation within rabbit atherosclerotic plaquesusing the macrophage-specific CT contrast agent N1177: a comparisonwith 18F-FDG PET/CT and histology. J Nucl Med. 2009;50:959–965.

22. Rudd JH, Narula J, Strauss HW, Virmani R, Machac J, Klimas M, TaharaN, Fuster V, Warburton EA, Fayad ZA, Tawakol AA. Imaging athero-sclerotic plaque inflammation by fluorodeoxyglucose with positronemission tomography: ready for prime time? J Am Coll Cardiol. 2010;55:2527–2535.

23. Avart SJ, Bernard DW, Jerome WG, Glick JM. Cholesteryl esterhydrolysis in J774 macrophages occurs in the cytoplasm and lysosomes.J Lipid Res. 1999;405:405–414.

24. Tangirala RK, Jerome WG, Jones NL, Small DM, Johnson WJ, Glick JM,Mahlberg FH, Rothblat GH. Formation of cholesterol monotydratecrystals in macrophage-derived foam cells. J Lipid Res. 1994;35:93–104.

25. Calcagno C, Cornily J-C, Hyafil F, Rudd JHF, Briley-Saebo KC, Mani V,Goldschlager G, Machac J, Fuster V, Fayad ZA. Detection of neovesselsin atherosclerotic plaques of rabbits using dynamic contrast enhancedMRI and 18F-FDG PET. Arterio Thromb Vasc Biol. 2008;28:1311–1317.

26. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG,Abela GS, Franchi L, Nunuez G, Schnurr M, Espevik T, Lien E,Fitzgerald KA, Rock KL, Moore KJ, Wright SD, Hornung V, Latz E.NLRP3 inflammasomes are required for atherogenesis and activated bycholesterol crystals. Nature. 2010;464:1357–1362.

27. Geng Y-J, Phillips JE, Mason RP, Casscells SW. Cholesterol crystalli-zation and macrophage apoptosis: implication for atherosclerotic plaqueinstability and rupture. Biochem Pharm. 2003;66:1485–1492.

28. Tabas I. Consequences and therapeutic implications of macrophage apo-ptosis in atherosclerosis: the importance of lesion stage and phagocyticactivity. Arterioscler Thromb Vasc Biol. 2005;25:2255–2264.

29. Abela GS. Cholesterol crystal piercing the arterial plaque and intimatriggers local and systemic inflammation. J Clin Lipidol. 2010;4:156–164.

30. LaRosa JC, Grundy SM, Waters DD, Shear C, Barter P, Fruchart JC,Gotto AM, Greten H, Kastelein JJ, Shepherd J, Wenger NK; Treating toNew Targets (TNT) Investigators. Intensive lipid lowering with atorva-statin in patients with stable coronary disease. N Engl J Med. 2005;352:1425–1435.

2014 Arterioscler Thromb Vasc Biol September 2011

by IMED Korea on October 9, 2011http://atvb.ahajournals.org/Downloaded from

Summary

목적

동맥의 석회화는 진행된 죽상동맥경화의 합병증

으로 심혈관질환과 관련되어 있다. 노인의 혈관세

포는 노화에 의한 특징적인 구조적인 특성을 보인

다. 최근 연구는 포유류의 histone deacetylase인

SIRT1 (sirtuin 1)이 심혈관질환의 새로운 치료제

를 개발하는데 이용될 수 있는 표적임을 보여주고

있다. 우리는 혈관 평활근세포를 이용한 혈관석회

화 모델에서 SIRT1의 역할에 대해 알아보고자 하

였다.

방법 및 결과

아데닌에 의한 신부전, 고인산혈증 백서 모델에서

대동맥의 중막(media)에서 광범위한 혈관석회화

가 관찰되었다. 세포노화의 지표인 노화관련

ß-galactosidase(SA ß-gal)의 활성도는 중막의

혈관 평활근세포에서 유의하게 증가하였으며

이는 혈관석회화 정도와 양의 상관관계를 보였다.

배양된 혈관 평활근세포에서 무기인산은 SA ß-gal

양성인 세포를 용량의존적으로 증가시켰다.

인산에 의해 유도된 세포노화는 SIRT1 발현 감소

에 의한 p21 활성화와 관련되었다. 인산의 공동전

달체를 차단함으로써 SIRT1 저하에 의한 활성화

는 소실되었다. 레스베라트롤(resveratrol)에 의한

SIRT1의 활성화는 세포노화에 의한 혈관석회화

를 유의하게 감소시켰다. 반면 small interfering

RNA로 SIRT1의 발현을 억제하면 인산에 의한

혈관 평활근세포의 노화 및 혈관석회화가 급속

하게 진행하였다. 또한 SIRT1 발현 억제는 혈관

평활근세포가 조골세포 유사세포(osteoblast-

like cells)의 형태로 변화되는 것을 유도하였다.

세포노화와 관련된 혈관 평활근세포의 석회화는

p21의 발현억제에 의해 완전하게 예방되었다.

인산에 의해 유도된 조기 세포노화와 같이 세포

복제에 의해 노화(replicative senescence)된 혈관

평활근세포가 젊은 혈관 평활근세포에 비해

인산에 의한 혈관석회화에 보다 취약하였고

이러한 결과는 항진된 p21 발현과 관련되었다.

결론

SIRT1은 혈관 평활근세포가 조골세포 (osteoblast)

로의 transdifferentiation을 억제함 으로써 고인

산혈증에 의한 혈관석회화를 예방하는데 중요

한 역할을 한다. 또한 인산에 의해 유도된 혈관

평활근세포의 석회화는 조기 세포노화 및 세포

복제에 의한 노화 모두와 관련이 있는 것으로

생각된다.

SIRT1의 새로운 역할: SIRT1은 고인산혈증에 의한 혈관석회화를 억제한다.

김 광 일 교수분당서울대학교병원 노인병내과

Page 21: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

46

Commentary

혈관석회화는 혈관에 무기질이 침착됨으로써 혈

관의 경직도가 증가하고 죽상동맥경화반의 파열

을 초래하여 심혈관질환의 발생 및 합병증의 위험

성을 증가시킨다. 특히 고령 환자에게는 동반 질환

이 없이도 흔히 관찰되는 대표적인 혈관 노화 현

상이나 아직까지 원인 및 기전에 관한 연구는 초

보적인 단계에 머물러 있는 상태이다.

혈관석회화는 노화, 당뇨병, 만성 신부전, 죽상동

맥경화 등에 따른 이차적인 현상이라고 생각되어

왔으나 최근 혈관석회화의 병태 생리에 대한 연구

가 진행되면서 혈관석회화는 독립적인 기전에 의

해 이루어지는 질병 과정이며 흥미롭게도 골형성

과정과 유사한 기전에 의해 조절된다는 사실이 알

려지고 있다. 혈관석회화는 이전에는 노화에 따른

당연한 결과이며 다른 원인에 의한 이차적인 현상

으로 생각되어 크게 주목받지 못했으나 최근 노

인 인구의 증가로 혈관석회화를 동반한 노인 환자

가 증가하게 되었고 혈관석회화가 관찰되는 환자

에서 향후 심혈관질환의 발생 및 합병증의 위험이

증가하는 것으로 밝혀짐에 따라 그 기전 및 조기

진단, 그리고 치료에 대한 관심이 증가하고 있다.

특히, 미국 UCLA의 Demer 그룹에 의해 혈관내

부에도 석회화 능력을 가진 전구세포로 조골세포

와 유사한 역할을 하는 CVC (Calcifying vascular

cell)가 존재한다는 사실이 밝혀지고 난 후, 이 세

포의 석회화 기전에 영향을 미치는 인자 및 그 기

전에 관한 연구가 활발히 진행되고 있다.1 하지만

아직까지 혈관석회화를 일으키는 근원세포의 기

원과 그 특성을 명확하게 규명한 연구 결과는 많

지 않으며 혈관석회화 과정에 영향을 미치는 여러

인자의 상호 관계 및 기전에 관한 연구는 초기 단

계에 머물러 있는 실정이다.

Sirtuin (SIRTs)은 항상성 유지 및 외부 스트레스

에 대한 반응을 조절하는 물질로 대부분의 sirtuin

(SIRT1, SIRT2, SIRT3, SIRT5)은 NAD+-dependent

deacetylation 과정을 촉매하며 SIRT4, SIRT6은

ADP-ribosylation을 매개한다.2 효모균에서 Sir2

(silent information regulator 2)는 budding 및

수명을 조절하는 역할을 담당하며 외부 스트레

스나 식이 제한으로부터 개체의 생존을 유지하

는데 중요한 역할을 한다. 포유류에서는 Sir2의

homolog인 SIRT1이 세포노화, 세포고사, 인슐린

감수성, 염증반응, 유전자 안정성, 산화 스트레스

등 여러 기전에 관여하는 것으로 알려져 있다.

비록 효모균에서 Sir2가 외부 스트레스에 대한 개

체 보호효과를 가짐으로써 수명을 연장시킨다는

사실이 잘 알려져 있지만 포유류에서도 SIRT1이

노화를 억제하고 수명을 연장시키는 가에 대해서

는 아직까지는 명확하게 밝혀져 있지는 않다.3 그

러나 포유류에서도 수명 연장 효과가 관찰된 식

이 제한 모델에서 식이 제한에 의해 SIRT1의 발현

이 증가된다는 사실이 알려짐으로써 포유류에서

도 수명을 연장시키고 노화와 관련된 여러 장기

의 변화를 조절하고 노화 관련 질병을 예방하는

데 관여할 것으로 기대되고 있다. 따라서 최근에

는 SIRT1 activator의 임상적 효능에 대해 많은 관

심을 가지고 연구가 진행 중에 있다.

혈관 노화에 있어서 SIRT1의 역할에 대해서도 많

은 관심을 받고 있다. SIRT1은 p53에 의해 유도된

세포노화를 억제하고 혈관내피세포에서 산화 스

트레스에 의한 세포조기노화를 차단한다는 사실

이 보고된 바 있다. 하지만 아직까지 SIRT1이 혈관

의 노화를 억제하는 기전에 대해서는 잘 밝혀져

있지 못한 상태이다.

본 논문에서 동경대학교 노년내과학 교실의 연

구자들은 신부전에 의한 고인산혈증 모델에서

Page 22: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

47

SIRT1이 혈관석회화 과정을 조절하는 기전에 대

해 밝히고자 하였다. 신부전 환자에서는 혈관석회

화가 흔히 관찰되며 만성 신부전 환자의 주된 사

망원인이 심혈관질환이라는 점을 고려하면 연구

자들의 실험 모델은 임상적으로도 혈관석회화 연

구를 수행하는데 적절한 모델이라고 생각된다. 연

구자들이 새롭게 밝혀낸 사실은 SIRT1이 고인산

혈증에 의해 유도된 혈관석회화 과정을 차단하는

보호작용을 하며 특히 혈관석회화 과정은 노화

된 혈관 평활근세포에서 보다 쉽게 유도되기 때문

에 SIRT1 활성도를 혈관석회화에 보다 취약한 것

으로 생각되는 노인에서 어떻게 하면 높게 유지할

수 있는가를 찾아내는 것이 향후 혈관석회화를

억제할 수 있는 새로운 치료법 개발을 위해 필수

적일 것으로 제시하였다. 또한 SIRT1의 혈관석회

화 억제 기전이 p53/p21 경로를 통한다는 사실을

밝힘으로써 혈관 노화/석회화의 상호 관련성을

이해하는데 도움이 되게 하였다. 즉, 혈관세포의

노화는 석회화에 보다 취약해지는데 이러한 기전

이 같은 경로에 의해 발생한다는 사실을 밝혔다.

이러한 일련의 연구를 통해 이전까지 수동적인 과

정이라고 생각되었던 혈관석회화가 능동적인 자

체 기전에 의해 발생한다는 사실을 알게 되었고

또한 혈관석회화를 촉진 또는 억제하는 다양한

기전을 밝혀냄으로써 향후 인구의 고령화로 인해

임상에서 더욱 문제가 될 것으로 생각되는 혈관석

회화의 근본적인 치료방법을 개발하는데 한걸음

나아갈 수 있을 것으로 기대된다.

REFERENCESShao JS, Cai J, Towler DA. Molecular mechanisms of vascular 1. calcif ication: Lessons learned from the aorta. Arterioscler Thromb Vasc Biol. 2006;26:1423-1430.Potente M, Dimmeler S. No targets sirt1: A novel signaling 2. network in endothelial senescence. Arterioscler Thromb Vasc Biol. 2008;28:1577-1579.Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu 3. S, Wood JG, Zipkin RE, Chung P, Kisielewski A, Zhang LL, Scherer B, Sinclair DA. Small molecule activators of sirtuins extend saccharomyces cerevisiae lifespan. Nature. 2003;425:191-196.

Page 23: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

to humans.10 In yeast, Sir2 has been shown to play critical rolesin DNA repair, stress resistance, and longevity. Mammaliansirtuin 1 (SIRT1), the closest homolog of Sir2, regulates the cellcycle, apoptosis, and metabolism by interacting with a numberof molecules, including p53, promyelocytic leukemia protein,Foxo, Ku70, and peroxisome proliferator-activated receptor-�.11

A previous study has shown that SIRT1 antagonizes p53-mediated premature senescence in mouse embryo fibroblasts.12

In addition, we have recently demonstrated that SIRT1 inhibitsoxidative stress-induced premature senescence in vascular en-dothelial cells.13 However, the detailed mechanism of howSIRT1 affects vascular SMC senescence and arterial calcifica-tion remains unclear.

In this study, we hypothesized that SIRT1 plays an impor-tant role in preventing arterial calcification due to renalfailure, in association with modulation of cellular senescence.Here, we demonstrated the protective potential of SIRT1against hyperphosphatemia-induced premature and replica-tive senescence and subsequent calcification in SMCs.

MethodsAortic Calcification in Renal Failure RatsRenal failure was induced in rats by a 0.75% adenine-containing dietas previously described.14 All procedures and animal care were inaccordance with the Guide for the Care and Use of Laboratory Animalsof the University of Tokyo. Detailed methods are described in thesupplemental materials, available online at http://atvb.ahajournals.org.

Induction of SMC CalcificationPrimary human aortic SMCs (HASMCs) were treated with a patholog-ical concentration of inorganic phosphate (Pi) up to 3.2 mmol/L inculture medium as previously described.29 To quantitatively measurePi-induced calcification, two distinct experiments were performed aspreviously described14: (1) intracellular calcium deposition as deter-mined by o-cresolphthalein complexone method, and (2) visualizationof mineralization as determined by von Kossa staining, Detailedmethods are described in the supplemental materials.

Senescence-Associated �-Galactosidase StainingTo assess senescent changes in the phenotype of cultured HASMCsor aortic medial cells of rats, staining for senescence-associated�-galactosidase (SA�-gal), a well-established biomarker of cellularsenescence, was performed. Detailed methods are described in thesupplemental materials.

Knockdown of SIRT1 or p21 by SmallInterfering RNAHASMCs were transfected with 200 pmol/L small interfering RNA(siRNA) for SIRT1, p21WAF1/CIP1, or both. Detailed methods aredescribed in the supplemental materials.

Real-Time Polymerase Chain Reaction Analysis:Osteoblastic MarkersTo examine whether Pi stimulation induces change to an osteoblasticphenotype, the expression of Runx-2/Cbfa-1 and alkaline phospha-tase, which are well known to be representative osteoblastic markers,was checked using real time-polymerase chain reaction analysis. Inaddition, the effect of knockdown of SIRT1, p21, or both by siRNAon the osteoblastic phenotypic change in HASMCs was examined.Primer sequences are shown in Supplemental Figure I.

ResultsAssociation of Senescent Vascular Cells WithAortic Medial Calcification in Renal Failure RatsThe adenine-fed rats had severe renal failure, with a hugeincrease in serum creatinine (3.0�0.9 mg/dL in renal failure

rats versus 0.3�0.0 mg/dL in control rats), similar to aprevious report.14 The renal failure rats showed an approxi-mately 2.0-fold increase in serum phosphorus (18.9�4.7mg/dL) compared with control rats (9.8�0.9 mg/dL). Histo-logical assessment using von Kossa staining showed that theaorta in renal failure rats had extensive linear calcification,which was localized in the aortic media, resembling the typicalMonckeberg’s pattern (Figure 1A). Numerous SA�-gal positivecells were found in the aortic media of renal failure rats, whereasthe aortic wall in control rats did not contain senescent cells(Figure 1B). The senescent cells were mainly localized to thecalcified area and its surrounding area, which was defined as thearea not stained black by von Kossa staining. Quantitativeassessment showed that the number of senescent cells with highSA�-gal activity was positively correlated with the calcified areain the aortic media (Figure 1C).

Pi Induces Cellular Senescence in Cultured SMCsOn the basis of our results obtained from animal experiments,we hypothesized that senescent SMCs in the aortic media arestrongly associated with the development of arterial calcifi-cation. Therefore, the effect of excessive Pi stimulation(2.6 mmol/L) on cellular senescence in cultured SMCs wasexamined. SA�-gal-positive senescent HASMCs were signif-icantly induced by not only angiotensin II (Ang II) but also Pi

Figure 1. Presence of senescent vascular cells colocalized withcalcification in aortic media of renal failure rats. A, Rats withsevere renal failure had massive calcification throughout theaorta (right) compared with control rats (left) (n�5). Yellowarrows indicate calcified area. Morphological assessment by vonKossa staining showed extensive calcification in the aorticmedia of renal failure rats. Scale bar�500 �m. B, Senescentvascular cells (senescence-associated �-galactosidase [SA�-gal]-positive: blue) were significantly detected throughout thecalcified area (Calc) in renal failure rats, whereas these senes-cent cells were not present in control rats. Scale bar�100 �m.C, Localized association between calcification and senescentcells is shown in renal failure rats. SA�-gal-positive cells werefrequently found in areas with marked calcification. D, The asso-ciation of the number of SA�-gal-positive cells with the calcifiedarea in each photograph was evaluated. The senescent cellnumber was linearly correlated with the area of calcification inthe aortic media of renal failure rats (calcified area in media:percentage).

Takemura et al SIRT1 Retards Vascular SMC Calcification 2055

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from 48

Sirtuin 1 Retards Hyperphosphatemia-Induced Calcificationof Vascular Smooth Muscle Cells

Aya Takemura, Katsuya Iijima, Hidetaka Ota, Bo-Kyung Son, Yuki Ito, Sumito Ogawa, Masato Eto,Masahiro Akishita, Yasuyoshi Ouchi

Objective—Arterial calcification is associated with cardiovascular disease as a complication of advanced atherosclerosis.Aged vascular cells manifest some morphological features of a senescent phenotype. Recent studies have demonstratedthat mammalian sirtuin 1 (SIRT1), a histone deacetylase, is an exciting target for cardiovascular disease management.Here, we investigated the role of SIRT1 in a calcification model of vascular smooth muscle cells (SMCs).

Methods and Results—In adenine-induced renal failure rats with hyperphosphatemia, massive calcification was inducedin the aortic media. Senescence-associated �-galactosidase (SA�-gal) activity, a marker of cellular senescence, inmedial SMCs was significantly increased, and its induction was positively associated with the degree of calcification.In cultured SMCs, inorganic phosphate (Pi) stimulation dose-dependently increased SA�-gal-positive cells, andPi-induced senescence was associated with downregulation of SIRT1 expression, leading to p21 activation. Theactivation via SIRT1 downregulation was blunted by inhibition of Pi cotransporter. Activation of SIRT1 by resveratrolsignificantly reduced the senescence-associated calcification. Conversely, SIRT1 knockdown by small interfering RNAaccelerated the Pi-induced SMC senescence and subsequent calcification. In addition, SIRT1 knockdown inducedphenotypic change from a differentiated state to osteoblast-like cells. The senescence-related SMC calcification wascompletely prevented by p21 knockdown. In addition to Pi-induced premature senescence, SMCs with replicativesenescence were also more sensitive to Pi-induced calcification compared with young SMCs, and this finding wasattributable to augmented p21 expression.

Conclusion—SIRT1 plays an essential role in preventing hyperphosphatemia-induced arterial calcification via inhibitionof osteoblastic transdifferentiation. In addition, Pi-induced SMC calcification may be associated with both prematureand replicative cellular senescence. (Arterioscler Thromb Vasc Biol. 2011;31:2054-2062.)

Key Words: cellular senescence � hyperphosphatemia � longevity gene SIRT1 � vascular calcification� vascular smooth muscle cell

Atherosclerotic vascular damage associated with agingmanifests several features, namely atherosis, sclerosis,

and calcific change, finally leading to cardiovascular events.These pathological changes result in arterial wall thickening(localized morphological changes) and arterial stiffening(functional changes).1 Arterial calcification makes the man-agement of hemodynamics more difficult in the elderly,because ectopic calcium deposition in the aorta and arteriescontributes to vessel wall stiffening and loss of elastic recoil.2

These pathological conditions result in unstable hemodynamicconsequences, finally leading to a decline in end-organperfusion and subsequent ischemic events. Recently, severalreports have demonstrated that aortic calcification detectableon chest X-ray examination is a strong predictor of futurecardiovascular events beyond traditional risk factors.3

Arterial calcification is anatomically separated into twotypes, intimal and medial calcification.4 Intimal calcification,

which is seen as patchy scattered deposits only occurringwithin atherosclerotic plaques, is shown to be associated withplaque vulnerability.5 On the other hand, medial calcification,which is frequently seen in the elderly and in diabetes andchronic renal failure, is observed as continuous linear depos-its along the internal elastic lamina.6 Advanced atherosclero-sis with both types of calcified lesions is the consequence ofoverlapping pathological mechanisms.

Ectopic calcification in the vasculature has been shown toresult from passive precipitation of calcium with aging andosteoporosis, the so-called calcium shift theory, as a previoushypothesis.7 However, accumulating recent evidence has shownit to be attributable to an active “cell-mediated process” resem-bling osteogenesis in bone rather than passive mineral precipi-tation in vascular smooth muscle cells (SMCs).8,9

Silent information regulator-2 (Sir2), an NAD�-dependentHDAC, is highly conserved in organisms ranging from Archaea

Received on: May 12, 2010; final version accepted on: May 25, 2011.From the Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.Correspondence to Katsuya Iijima, MD, PhD, Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo,

Bunkyo-ku, Tokyo 113-8655, Japan. E-mail [email protected]© 2011 American Heart Association, Inc.

Arterioscler Thromb Vasc Biol is available at http://atvb.ahajournals.org DOI: 10.1161/ATVBAHA.110.216739

2054 by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Page 24: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

49

to humans.10 In yeast, Sir2 has been shown to play critical rolesin DNA repair, stress resistance, and longevity. Mammaliansirtuin 1 (SIRT1), the closest homolog of Sir2, regulates the cellcycle, apoptosis, and metabolism by interacting with a numberof molecules, including p53, promyelocytic leukemia protein,Foxo, Ku70, and peroxisome proliferator-activated receptor-�.11

A previous study has shown that SIRT1 antagonizes p53-mediated premature senescence in mouse embryo fibroblasts.12

In addition, we have recently demonstrated that SIRT1 inhibitsoxidative stress-induced premature senescence in vascular en-dothelial cells.13 However, the detailed mechanism of howSIRT1 affects vascular SMC senescence and arterial calcifica-tion remains unclear.

In this study, we hypothesized that SIRT1 plays an impor-tant role in preventing arterial calcification due to renalfailure, in association with modulation of cellular senescence.Here, we demonstrated the protective potential of SIRT1against hyperphosphatemia-induced premature and replica-tive senescence and subsequent calcification in SMCs.

MethodsAortic Calcification in Renal Failure RatsRenal failure was induced in rats by a 0.75% adenine-containing dietas previously described.14 All procedures and animal care were inaccordance with the Guide for the Care and Use of Laboratory Animalsof the University of Tokyo. Detailed methods are described in thesupplemental materials, available online at http://atvb.ahajournals.org.

Induction of SMC CalcificationPrimary human aortic SMCs (HASMCs) were treated with a patholog-ical concentration of inorganic phosphate (Pi) up to 3.2 mmol/L inculture medium as previously described.29 To quantitatively measurePi-induced calcification, two distinct experiments were performed aspreviously described14: (1) intracellular calcium deposition as deter-mined by o-cresolphthalein complexone method, and (2) visualizationof mineralization as determined by von Kossa staining, Detailedmethods are described in the supplemental materials.

Senescence-Associated �-Galactosidase StainingTo assess senescent changes in the phenotype of cultured HASMCsor aortic medial cells of rats, staining for senescence-associated�-galactosidase (SA�-gal), a well-established biomarker of cellularsenescence, was performed. Detailed methods are described in thesupplemental materials.

Knockdown of SIRT1 or p21 by SmallInterfering RNAHASMCs were transfected with 200 pmol/L small interfering RNA(siRNA) for SIRT1, p21WAF1/CIP1, or both. Detailed methods aredescribed in the supplemental materials.

Real-Time Polymerase Chain Reaction Analysis:Osteoblastic MarkersTo examine whether Pi stimulation induces change to an osteoblasticphenotype, the expression of Runx-2/Cbfa-1 and alkaline phospha-tase, which are well known to be representative osteoblastic markers,was checked using real time-polymerase chain reaction analysis. Inaddition, the effect of knockdown of SIRT1, p21, or both by siRNAon the osteoblastic phenotypic change in HASMCs was examined.Primer sequences are shown in Supplemental Figure I.

ResultsAssociation of Senescent Vascular Cells WithAortic Medial Calcification in Renal Failure RatsThe adenine-fed rats had severe renal failure, with a hugeincrease in serum creatinine (3.0�0.9 mg/dL in renal failure

rats versus 0.3�0.0 mg/dL in control rats), similar to aprevious report.14 The renal failure rats showed an approxi-mately 2.0-fold increase in serum phosphorus (18.9�4.7mg/dL) compared with control rats (9.8�0.9 mg/dL). Histo-logical assessment using von Kossa staining showed that theaorta in renal failure rats had extensive linear calcification,which was localized in the aortic media, resembling the typicalMonckeberg’s pattern (Figure 1A). Numerous SA�-gal positivecells were found in the aortic media of renal failure rats, whereasthe aortic wall in control rats did not contain senescent cells(Figure 1B). The senescent cells were mainly localized to thecalcified area and its surrounding area, which was defined as thearea not stained black by von Kossa staining. Quantitativeassessment showed that the number of senescent cells with highSA�-gal activity was positively correlated with the calcified areain the aortic media (Figure 1C).

Pi Induces Cellular Senescence in Cultured SMCsOn the basis of our results obtained from animal experiments,we hypothesized that senescent SMCs in the aortic media arestrongly associated with the development of arterial calcifi-cation. Therefore, the effect of excessive Pi stimulation(2.6 mmol/L) on cellular senescence in cultured SMCs wasexamined. SA�-gal-positive senescent HASMCs were signif-icantly induced by not only angiotensin II (Ang II) but also Pi

Figure 1. Presence of senescent vascular cells colocalized withcalcification in aortic media of renal failure rats. A, Rats withsevere renal failure had massive calcification throughout theaorta (right) compared with control rats (left) (n�5). Yellowarrows indicate calcified area. Morphological assessment by vonKossa staining showed extensive calcification in the aorticmedia of renal failure rats. Scale bar�500 �m. B, Senescentvascular cells (senescence-associated �-galactosidase [SA�-gal]-positive: blue) were significantly detected throughout thecalcified area (Calc) in renal failure rats, whereas these senes-cent cells were not present in control rats. Scale bar�100 �m.C, Localized association between calcification and senescentcells is shown in renal failure rats. SA�-gal-positive cells werefrequently found in areas with marked calcification. D, The asso-ciation of the number of SA�-gal-positive cells with the calcifiedarea in each photograph was evaluated. The senescent cellnumber was linearly correlated with the area of calcification inthe aortic media of renal failure rats (calcified area in media:percentage).

Takemura et al SIRT1 Retards Vascular SMC Calcification 2055

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Sirtuin 1 Retards Hyperphosphatemia-Induced Calcificationof Vascular Smooth Muscle Cells

Aya Takemura, Katsuya Iijima, Hidetaka Ota, Bo-Kyung Son, Yuki Ito, Sumito Ogawa, Masato Eto,Masahiro Akishita, Yasuyoshi Ouchi

Objective—Arterial calcification is associated with cardiovascular disease as a complication of advanced atherosclerosis.Aged vascular cells manifest some morphological features of a senescent phenotype. Recent studies have demonstratedthat mammalian sirtuin 1 (SIRT1), a histone deacetylase, is an exciting target for cardiovascular disease management.Here, we investigated the role of SIRT1 in a calcification model of vascular smooth muscle cells (SMCs).

Methods and Results—In adenine-induced renal failure rats with hyperphosphatemia, massive calcification was inducedin the aortic media. Senescence-associated �-galactosidase (SA�-gal) activity, a marker of cellular senescence, inmedial SMCs was significantly increased, and its induction was positively associated with the degree of calcification.In cultured SMCs, inorganic phosphate (Pi) stimulation dose-dependently increased SA�-gal-positive cells, andPi-induced senescence was associated with downregulation of SIRT1 expression, leading to p21 activation. Theactivation via SIRT1 downregulation was blunted by inhibition of Pi cotransporter. Activation of SIRT1 by resveratrolsignificantly reduced the senescence-associated calcification. Conversely, SIRT1 knockdown by small interfering RNAaccelerated the Pi-induced SMC senescence and subsequent calcification. In addition, SIRT1 knockdown inducedphenotypic change from a differentiated state to osteoblast-like cells. The senescence-related SMC calcification wascompletely prevented by p21 knockdown. In addition to Pi-induced premature senescence, SMCs with replicativesenescence were also more sensitive to Pi-induced calcification compared with young SMCs, and this finding wasattributable to augmented p21 expression.

Conclusion—SIRT1 plays an essential role in preventing hyperphosphatemia-induced arterial calcification via inhibitionof osteoblastic transdifferentiation. In addition, Pi-induced SMC calcification may be associated with both prematureand replicative cellular senescence. (Arterioscler Thromb Vasc Biol. 2011;31:2054-2062.)

Key Words: cellular senescence � hyperphosphatemia � longevity gene SIRT1 � vascular calcification� vascular smooth muscle cell

Atherosclerotic vascular damage associated with agingmanifests several features, namely atherosis, sclerosis,

and calcific change, finally leading to cardiovascular events.These pathological changes result in arterial wall thickening(localized morphological changes) and arterial stiffening(functional changes).1 Arterial calcification makes the man-agement of hemodynamics more difficult in the elderly,because ectopic calcium deposition in the aorta and arteriescontributes to vessel wall stiffening and loss of elastic recoil.2

These pathological conditions result in unstable hemodynamicconsequences, finally leading to a decline in end-organperfusion and subsequent ischemic events. Recently, severalreports have demonstrated that aortic calcification detectableon chest X-ray examination is a strong predictor of futurecardiovascular events beyond traditional risk factors.3

Arterial calcification is anatomically separated into twotypes, intimal and medial calcification.4 Intimal calcification,

which is seen as patchy scattered deposits only occurringwithin atherosclerotic plaques, is shown to be associated withplaque vulnerability.5 On the other hand, medial calcification,which is frequently seen in the elderly and in diabetes andchronic renal failure, is observed as continuous linear depos-its along the internal elastic lamina.6 Advanced atherosclero-sis with both types of calcified lesions is the consequence ofoverlapping pathological mechanisms.

Ectopic calcification in the vasculature has been shown toresult from passive precipitation of calcium with aging andosteoporosis, the so-called calcium shift theory, as a previoushypothesis.7 However, accumulating recent evidence has shownit to be attributable to an active “cell-mediated process” resem-bling osteogenesis in bone rather than passive mineral precipi-tation in vascular smooth muscle cells (SMCs).8,9

Silent information regulator-2 (Sir2), an NAD�-dependentHDAC, is highly conserved in organisms ranging from Archaea

Received on: May 12, 2010; final version accepted on: May 25, 2011.From the Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.Correspondence to Katsuya Iijima, MD, PhD, Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo,

Bunkyo-ku, Tokyo 113-8655, Japan. E-mail [email protected]© 2011 American Heart Association, Inc.

Arterioscler Thromb Vasc Biol is available at http://atvb.ahajournals.org DOI: 10.1161/ATVBAHA.110.216739

2054 by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Page 25: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

50

ical inhibitor of SIRT1, induced an increase in SA�-gal-positive cells even under a normal Pi (1.4 mmol/L), and theincreased number of senescent cells induced by Pi wassignificantly augmented by sirtinol (Figure 4A). Sirtinoldose-dependently augmented Pi-induced calcification, al-though no augmentation was found under a normal Pi (Figure4B and 4C). Conversely, treatment with resveratrol, anactivator of SIRT1, significantly reduced both Pi-inducedsenescent transition and calcification in a dose-dependentmanner (Figure 4D to 4F).

Second, complete knockdown of SIRT1 by siRNA causeda significant increase in acetylation of both substrates(histone-3 and p53) and p21 expression (Figure 5A). Simi-larly to sirtinol, SIRT1 inhibition by siRNA also augmentednot only senescent transition (Figure 5A, bottom) but alsocalcium deposition (Figure 5C, top).

Although stimulation with Ang II alone could increase thenumber of SA�-gal-positive cells, it did not increase calcium

deposition. To understand the mechanism of these discrepantphenomena, the effect of Ang II alone on osteoblasticphenotypic change was examined. Ang II alone did notincrease the expression of Runx2 in the absence of Pistimulation, unlike Pi stimulation (Figure 5B).

To understand the detailed mechanism by which SIRT1modulates senescence-related calcification, the effect ofSIRT1 on phenotypic change in HASMCs was examined. Piinhibited the expression of caldesmon, a differentiated SMClineage marker, and complete knockdown of SIRT1 aug-mented the Pi-induced partial downregulation of caldesmon(Figure 5C, middle). In contrast, real-time polymerase chainreaction analysis showed that Pi induced the expression oftwo representative osteoblastic markers, Runx-2/Cbfa-1 andalkaline phosphatase (Figure 5C, bottom) with statisticalsignificance. In addition, complete knockdown of SIRT1using siRNA significantly accelerated the Pi-induced os-

Figure 3. Inorganic phosphate (Pi) stimulation leads to sirtuin 1 (SIRT1) downregulation and subsequent p21 activation. A, The effect ofPi on SIRT1 expression and its downstream pathway was examined. Treatment of human aortic SMCs (HASMCs) with Pi (2.6 mmol/L)showed downregulation of SIRT1 expression, leading to an increase in acetylation of its substrates (acetylated [Ac]-H3 and Ac-p53)and p21 expression. Bottom: Quantitative analysis showed that Pi gradually induced not only SIRT1 downregulation but also upregula-tion of Ac-p53 and p21. B, To address whether SIRT1 downregulation-related senescence and subsequent calcification are reversible,the effects of continuation or termination of high-dose Pi were examined. As shown in 4th lane from left, termination (on day 6) of Pishowed no progression of senescence-related calcification in association with restoration of SIRT1, whereas continuation (up to day10, 3rd lane from left) of Pi stimulation showed further progression of calcification. C, Treatment with phosphonoformic acid (PFA), aNa-dependent phosphate cotransporter inhibitor, completely reversed Pi-induced SIRT1 downregulation. A decline in Ac-H3 andAc-p53 reflected the restoration of SIRT1 deacetylase activity. Pi-induced p21 activation was significantly inhibited by inhibition of Pitransport.

Takemura et al SIRT1 Retards Vascular SMC Calcification 2057

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

stimulation (Figure 2A). Notably, Pi stimulation increasedcalcium deposition; however, Ang II alone did not (Figure2B). It suggests that high-dose Pi condition, but not stress byAng II alone, is indispensable to induce SMC calcification.

These findings also suggest that intracellular Pi influx at leastis essential to induce this SMC calcification model.

In addition, to determine how many days after the initiationof Pi stimulation the cells showed a senescent phenotype andsubsequent calcification, the time-dependent effects of Pistimulation on both SA�-gal activity and calcium depositionwere examined. As shown in Figure 2C, SA�-gal-positivecells were significantly increased by Pi stimulation even onday 1, although calcium deposition was not markedly in-creased at the same time point. A statistically significantincrease in calcium deposition was found from day 3 andlater. Cotreatment with phosphonoformic acid, an inhibitor ofNa-dependent phosphate cotransporter (NPC), showed signif-icant inhibition of Pi-induced senescence (Figure 2D). Ourprevious report showed that treatment with PFA completelyinhibited Pi-induced SMC calcification,15 suggesting theimportance of increased intracellular influx of phosphate inPi-induced SMC senescence.

Downregulation of SIRT1 by PiTreatment of HASMCs with Pi caused downregulation ofSIRT1 expression in a time-dependent manner (Figure 3A).The decline was dependent on Pi concentration (data notshown). An increase in acetylation of both substrates ofSIRT1, histone-3 and p53 (a nonhistone substrate), was foundaccording to the decline in SIRT1 deacetylase activity. Inaddition, expression of p21, a downstream molecule of p53,was significantly induced by Pi as well. Quantitative assess-ment showed that an increase in these expression levels ofacetylated (Ac)-p53 and p21 on day 3 and day 6 wasstatistically significant compared with the pretreatment lev-els, suggesting that downregulation of SIRT1 activity maymediate the subsequent increase in Ac-p53 and p21expression.

To address whether SIRT1 downregulation-related SMCsenescence and calcification are reversible or not, theeffects of continuation or termination of high-dose Pi wereexamined. As shown in Figure 3B, the continuation of Piup to day 10 was associated with SIRT1 downregulationand subsequent upregulation of Ac-p53 and p21, leading toinduction of senescence-related calcification. However,the slight increase in senescent cells was not statisticallysignificant, although calcification was significantly in-duced. Of note, the Pi-induced downregulation of SIRT1was almost completely reversed by withdrawal (termina-tion) of Pi stimulation (exchange of Pi from 2.6 mmol/L to1.4 mmol/L as a normal level on day 6) as shown in Figure3B. According to the restoration of SIRT1, levels of bothAc-p53 and p21 were also decreased without more pro-gression. In addition, termination of Pi showed no progres-sion of senescence-related calcification; however, preex-isting senescent cells and calcification on day 6 continuedwithout regression.

Next, NPC inhibition by PFA completely blunted Pi-induced SIRT1 downregulation and subsequent activation ofits downstream p53/p21 pathway (Figure 3C).

Regulation of SIRT1 Modulates Pi-Induced SMCSenescence and CalcificationThe effects of modulation of SIRT1 activity on Pi-inducedcellular senescence were investigated. First, sirtinol, a chem-

Figure 2. Inorganic phosphate (Pi) stimulation induces cellularsenescence in vascular smooth muscle cells (SMCs) via itscotransporter. A, The effect of Pi on senescent transition inhuman aortic SMCs (HASMCs) was examined. Representative pho-tographs showed that senescence-associated �-galactosidase(SA�-gal) activity (blue) in cells was significantly induced by notonly angiotensin II (Ang II: 10 pmol/L, as a positive control) but alsoPi stimulation (2.6 mmol/L). B, The number of senescent cells wassignificantly increased by not only Ang II but also Pi. Calciumdeposition was significantly increased by Pi; however, calcificationin HASMCs was not induced by Ang II alone in the absence of Pi.C, Senescent cells were significantly increased by Pi stimulationeven on day 1; however, a statistically significant increase in cal-cium deposition was found from day 3 and later. D, Inhibition ofthe phosphate cotransporter Na-dependent phosphate cotrans-porter by the inhibitor phosphonoformic acid (PFA) (100 �mol/L)reduced SA�-gal activity, which was increased by Pi (2.6 mmol/L)in HASMCs. Each experiment was performed al least 3 times.

2056 Arterioscler Thromb Vasc Biol September 2011

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Page 26: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

51

ical inhibitor of SIRT1, induced an increase in SA�-gal-positive cells even under a normal Pi (1.4 mmol/L), and theincreased number of senescent cells induced by Pi wassignificantly augmented by sirtinol (Figure 4A). Sirtinoldose-dependently augmented Pi-induced calcification, al-though no augmentation was found under a normal Pi (Figure4B and 4C). Conversely, treatment with resveratrol, anactivator of SIRT1, significantly reduced both Pi-inducedsenescent transition and calcification in a dose-dependentmanner (Figure 4D to 4F).

Second, complete knockdown of SIRT1 by siRNA causeda significant increase in acetylation of both substrates(histone-3 and p53) and p21 expression (Figure 5A). Simi-larly to sirtinol, SIRT1 inhibition by siRNA also augmentednot only senescent transition (Figure 5A, bottom) but alsocalcium deposition (Figure 5C, top).

Although stimulation with Ang II alone could increase thenumber of SA�-gal-positive cells, it did not increase calcium

deposition. To understand the mechanism of these discrepantphenomena, the effect of Ang II alone on osteoblasticphenotypic change was examined. Ang II alone did notincrease the expression of Runx2 in the absence of Pistimulation, unlike Pi stimulation (Figure 5B).

To understand the detailed mechanism by which SIRT1modulates senescence-related calcification, the effect ofSIRT1 on phenotypic change in HASMCs was examined. Piinhibited the expression of caldesmon, a differentiated SMClineage marker, and complete knockdown of SIRT1 aug-mented the Pi-induced partial downregulation of caldesmon(Figure 5C, middle). In contrast, real-time polymerase chainreaction analysis showed that Pi induced the expression oftwo representative osteoblastic markers, Runx-2/Cbfa-1 andalkaline phosphatase (Figure 5C, bottom) with statisticalsignificance. In addition, complete knockdown of SIRT1using siRNA significantly accelerated the Pi-induced os-

Figure 3. Inorganic phosphate (Pi) stimulation leads to sirtuin 1 (SIRT1) downregulation and subsequent p21 activation. A, The effect ofPi on SIRT1 expression and its downstream pathway was examined. Treatment of human aortic SMCs (HASMCs) with Pi (2.6 mmol/L)showed downregulation of SIRT1 expression, leading to an increase in acetylation of its substrates (acetylated [Ac]-H3 and Ac-p53)and p21 expression. Bottom: Quantitative analysis showed that Pi gradually induced not only SIRT1 downregulation but also upregula-tion of Ac-p53 and p21. B, To address whether SIRT1 downregulation-related senescence and subsequent calcification are reversible,the effects of continuation or termination of high-dose Pi were examined. As shown in 4th lane from left, termination (on day 6) of Pishowed no progression of senescence-related calcification in association with restoration of SIRT1, whereas continuation (up to day10, 3rd lane from left) of Pi stimulation showed further progression of calcification. C, Treatment with phosphonoformic acid (PFA), aNa-dependent phosphate cotransporter inhibitor, completely reversed Pi-induced SIRT1 downregulation. A decline in Ac-H3 andAc-p53 reflected the restoration of SIRT1 deacetylase activity. Pi-induced p21 activation was significantly inhibited by inhibition of Pitransport.

Takemura et al SIRT1 Retards Vascular SMC Calcification 2057

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

stimulation (Figure 2A). Notably, Pi stimulation increasedcalcium deposition; however, Ang II alone did not (Figure2B). It suggests that high-dose Pi condition, but not stress byAng II alone, is indispensable to induce SMC calcification.

These findings also suggest that intracellular Pi influx at leastis essential to induce this SMC calcification model.

In addition, to determine how many days after the initiationof Pi stimulation the cells showed a senescent phenotype andsubsequent calcification, the time-dependent effects of Pistimulation on both SA�-gal activity and calcium depositionwere examined. As shown in Figure 2C, SA�-gal-positivecells were significantly increased by Pi stimulation even onday 1, although calcium deposition was not markedly in-creased at the same time point. A statistically significantincrease in calcium deposition was found from day 3 andlater. Cotreatment with phosphonoformic acid, an inhibitor ofNa-dependent phosphate cotransporter (NPC), showed signif-icant inhibition of Pi-induced senescence (Figure 2D). Ourprevious report showed that treatment with PFA completelyinhibited Pi-induced SMC calcification,15 suggesting theimportance of increased intracellular influx of phosphate inPi-induced SMC senescence.

Downregulation of SIRT1 by PiTreatment of HASMCs with Pi caused downregulation ofSIRT1 expression in a time-dependent manner (Figure 3A).The decline was dependent on Pi concentration (data notshown). An increase in acetylation of both substrates ofSIRT1, histone-3 and p53 (a nonhistone substrate), was foundaccording to the decline in SIRT1 deacetylase activity. Inaddition, expression of p21, a downstream molecule of p53,was significantly induced by Pi as well. Quantitative assess-ment showed that an increase in these expression levels ofacetylated (Ac)-p53 and p21 on day 3 and day 6 wasstatistically significant compared with the pretreatment lev-els, suggesting that downregulation of SIRT1 activity maymediate the subsequent increase in Ac-p53 and p21expression.

To address whether SIRT1 downregulation-related SMCsenescence and calcification are reversible or not, theeffects of continuation or termination of high-dose Pi wereexamined. As shown in Figure 3B, the continuation of Piup to day 10 was associated with SIRT1 downregulationand subsequent upregulation of Ac-p53 and p21, leading toinduction of senescence-related calcification. However,the slight increase in senescent cells was not statisticallysignificant, although calcification was significantly in-duced. Of note, the Pi-induced downregulation of SIRT1was almost completely reversed by withdrawal (termina-tion) of Pi stimulation (exchange of Pi from 2.6 mmol/L to1.4 mmol/L as a normal level on day 6) as shown in Figure3B. According to the restoration of SIRT1, levels of bothAc-p53 and p21 were also decreased without more pro-gression. In addition, termination of Pi showed no progres-sion of senescence-related calcification; however, preex-isting senescent cells and calcification on day 6 continuedwithout regression.

Next, NPC inhibition by PFA completely blunted Pi-induced SIRT1 downregulation and subsequent activation ofits downstream p53/p21 pathway (Figure 3C).

Regulation of SIRT1 Modulates Pi-Induced SMCSenescence and CalcificationThe effects of modulation of SIRT1 activity on Pi-inducedcellular senescence were investigated. First, sirtinol, a chem-

Figure 2. Inorganic phosphate (Pi) stimulation induces cellularsenescence in vascular smooth muscle cells (SMCs) via itscotransporter. A, The effect of Pi on senescent transition inhuman aortic SMCs (HASMCs) was examined. Representative pho-tographs showed that senescence-associated �-galactosidase(SA�-gal) activity (blue) in cells was significantly induced by notonly angiotensin II (Ang II: 10 pmol/L, as a positive control) but alsoPi stimulation (2.6 mmol/L). B, The number of senescent cells wassignificantly increased by not only Ang II but also Pi. Calciumdeposition was significantly increased by Pi; however, calcificationin HASMCs was not induced by Ang II alone in the absence of Pi.C, Senescent cells were significantly increased by Pi stimulationeven on day 1; however, a statistically significant increase in cal-cium deposition was found from day 3 and later. D, Inhibition ofthe phosphate cotransporter Na-dependent phosphate cotrans-porter by the inhibitor phosphonoformic acid (PFA) (100 �mol/L)reduced SA�-gal activity, which was increased by Pi (2.6 mmol/L)in HASMCs. Each experiment was performed al least 3 times.

2056 Arterioscler Thromb Vasc Biol September 2011

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Page 27: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

52

Figure 5. Augmentation of senescence-related smooth muscle cell (SMC) calcification by sirtuin 1 (SIRT1) knockdown in associationwith osteoblastic phenotypic change and prevention of inorganic phosphate (Pi)–induced changes by p21 knockdown. A, To achieveSIRT1 knockdown in human aortic SMCs (HASMCs), small interfering RNA (siRNA) was simultaneously administered at the start of Pistimulation (2.6 mmol/L). Complete inhibition of SIRT1 showed a significant increase in acetylation of both substrates (acetylated[Ac]-H3 and Ac-p53), p21 expression and senescence-associated �-galactosidase (SA�-gal)-positive cells. B, Angiotensin II (Ang II)alone (10 pmol/L) did not increase the expression of Runx2 in the absence of Pi stimulation, unlike Pi stimulation. C, top: SIRT1 knock-down by siRNA significantly accelerated Pi-induced calcification (n�6), whereas control (Ctrl) siRNA did not. C, middle and bottom:Western blots showed that Pi partially inhibited the expression of a differentiated SMC marker, caldesmon, and complete knockdown ofSIRT1 by siRNA augmented its downregulation. Real-time polymerase chain reaction analysis showed that Pi induced the expression ofRunx-2 and alkaline phosphatase (ALP). Complete knockdown of SIRT1 significantly accelerated the Pi-induced osteoblastic markers.A.U. indicates arbitrary units. *P�0.05. D and E, Knockdown of p21 by siRNA (200 pmol/L) significantly reduced the senescent pheno-typic change and subsequent calcification (n�6). F, The role of SIRT1/p21 axis in Na-dependent phosphate cotransporter-mediatedRunx2 expression was evaluated. Augmentation of Pi-induced Runx2 expression by SIRT1 knockdown was significantly inhibited bydouble knockdown of SIRT1 and p21. *P�0.05 vs control without Pi stimulation (left column), **P�0.05 vs Pi-stimulated cells withSIRT1 siRNA (sixth column from left).

Takemura et al SIRT1 Retards Vascular SMC Calcification 2059

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

teoblastic phenotypic change, suggesting that modulationof SIRT1 is associated with osteoblastic phenotypicchange in SMCs.

Inhibition of Senescence-Related Calcification inSMCs by p21 KnockdownTo address the association of p21 with senescence-relatedcalcification, knockdown of p21 using siRNA was per-formed. Treatment of p21 siRNA (up to 200 pmol/L) com-pletely inhibited p21 (Figure 5D). p21 knockdown com-pletely inhibited Pi-induced senescence and subsequentcalcification (Figure 5E).

Regulation of NPC-Mediated Runx2 Expression bySIRT1/p21 PathwayAs the next step, the role of SIRT1 in NPC-mediatedRunx2/Cbfa1 expression was examined. First, completeknockdown of SIRT1 did not show any change in bothosteoblastic markers, Runx2 and alkaline phosphatase, in anormal Pi (Supplemental Figure I). As shown in Figure 5F,

Pi-induced Runx2 was significantly blunted by PFA, an NPCinhibitor. SIRT1 activation by resveratrol inhibited Pi-induced Runx2 activation. The Runx2 induction was aug-mented by knockdown of SIRT1 by siRNA, and the activa-tion was completely inhibited by PFA. Surprisingly, Runx2activation was strongly inhibited by knockdown of p21 alone.In addition, the inhibition of Runx2 induction by doubleknockdown of SIRT1 and p21 was less than that by singleknockdown of SIRT1.

To address a difference in senescent induction by Pi or AngII, immunohistological assessment of SIRT1 in HASMCswas examined (Supplemental Figure II). Although SIRT1was predominantly localized in nucleus without Pi, thetranslocation of SIRT1 to cytoplasm was observed after Pistimulation for 24 hours, and its expression disappeared inboth areas on day 6. In contrast, Ang II stimulation did notshow the dynamic translocation.

High Sensitivity of SMCs With ReplicativeSenescence to Pi-Induced CalcificationNot only Pi-induced “premature senescence” in HASMCs butalso the effects of Pi on “replicative senescence” wereevaluated. Senescent cells (passage 18) were more sensitiveto Pi-induced calcification compared with young cells (pas-sage 7) (Figure 6A). SIRT1 expression was downregulated insenescent cells compared with young cells, and the down-regulation was significantly augmented by Pi stimulation(Figure 6B, top). In parallel with this finding, senescent cellsshowed an increase in Ac-p53 and p21 expression. Statisticalanalyses using densitometric measurement showed that (1)downregulation of SIRT1 and upregulation of Ac-53 and p21were augmented by replicative senescence, and (2) Pi inhib-ited the SIRT1-p21 pathway even in cells with replicativesenescence (passage 18) (Figure 6B, bottom).

DiscussionVascular aging, leading to cardiovascular disease, manifestscomplex and diverse vascular changes (eg, impairment ofdistensibility due to loss of arterial elasticity).1,16 Arterial wallstiffness resulting from ectopic calcification is a complicationof advanced atherosclerosis and makes the management ofhemodynamics more difficult in the elderly. Few reports haveaddressed whether cellular senescence is associated withSMC calcification. This study showed the importance ofSIRT1, a longevity gene, in arterial calcification in associa-tion with cellular senescence.

First, our data obtained from animal experiments clearlyshowed the association of senescent SMCs with aortic medialcalcification in the renal failure rats with hyperphosphatemia.Senescent cells showed significant colocalization with cal-cium deposition. Intriguingly, numerous senescent cells couldbe detected before microscopic calcification occurred at 4weeks after the start of renal failure induction (data notshown), suggesting that the transition to a senescent pheno-type in medial SMCs may be associated with the initiationand progression of calcification. Therefore, hyperphos-phatemia, a potent uremic factor, may be a stimulator toinduce senescent phenotypic transition of medial SMCs.

Figure 4. Modulation of sirtuin 1 (SIRT1) affects inorganic phos-phate (Pi)–induced senescent phenotypic change and calcifica-tion in smooth muscle cells (SMCs). The effects of sirtinol (achemical inhibitor of SIRT1 activity; A to C) and resveratrol (anactivator of SIRT1; D to F) on Pi-induced senescent phenotypicchange and calcification were examined (n�6). A, SIRT1 inhibi-tion by sirtinol (10 �mol/L) showed an increase in the number ofsenescence-associated �-galactosidase (SA�-gal)-positive cellseven without Pi stimulation. The increase in Pi-induced senes-cence was significantly augmented by sirtinol. Sirtinol aug-mented Pi-induced calcium deposition in human aortic SMCs(HASMCs) in a time-dependent (B) and dose-dependent manneron day 6 (C). Conversely, treatment with resveratrol (Resv;10 �mol/L) showed a reduction of the Pi-induced senescentphenotype (D) and calcification (E). The inhibitory effect of res-veratrol on calcification was dose dependent (F).

2058 Arterioscler Thromb Vasc Biol September 2011

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Page 28: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

53

Figure 5. Augmentation of senescence-related smooth muscle cell (SMC) calcification by sirtuin 1 (SIRT1) knockdown in associationwith osteoblastic phenotypic change and prevention of inorganic phosphate (Pi)–induced changes by p21 knockdown. A, To achieveSIRT1 knockdown in human aortic SMCs (HASMCs), small interfering RNA (siRNA) was simultaneously administered at the start of Pistimulation (2.6 mmol/L). Complete inhibition of SIRT1 showed a significant increase in acetylation of both substrates (acetylated[Ac]-H3 and Ac-p53), p21 expression and senescence-associated �-galactosidase (SA�-gal)-positive cells. B, Angiotensin II (Ang II)alone (10 pmol/L) did not increase the expression of Runx2 in the absence of Pi stimulation, unlike Pi stimulation. C, top: SIRT1 knock-down by siRNA significantly accelerated Pi-induced calcification (n�6), whereas control (Ctrl) siRNA did not. C, middle and bottom:Western blots showed that Pi partially inhibited the expression of a differentiated SMC marker, caldesmon, and complete knockdown ofSIRT1 by siRNA augmented its downregulation. Real-time polymerase chain reaction analysis showed that Pi induced the expression ofRunx-2 and alkaline phosphatase (ALP). Complete knockdown of SIRT1 significantly accelerated the Pi-induced osteoblastic markers.A.U. indicates arbitrary units. *P�0.05. D and E, Knockdown of p21 by siRNA (200 pmol/L) significantly reduced the senescent pheno-typic change and subsequent calcification (n�6). F, The role of SIRT1/p21 axis in Na-dependent phosphate cotransporter-mediatedRunx2 expression was evaluated. Augmentation of Pi-induced Runx2 expression by SIRT1 knockdown was significantly inhibited bydouble knockdown of SIRT1 and p21. *P�0.05 vs control without Pi stimulation (left column), **P�0.05 vs Pi-stimulated cells withSIRT1 siRNA (sixth column from left).

Takemura et al SIRT1 Retards Vascular SMC Calcification 2059

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

teoblastic phenotypic change, suggesting that modulationof SIRT1 is associated with osteoblastic phenotypicchange in SMCs.

Inhibition of Senescence-Related Calcification inSMCs by p21 KnockdownTo address the association of p21 with senescence-relatedcalcification, knockdown of p21 using siRNA was per-formed. Treatment of p21 siRNA (up to 200 pmol/L) com-pletely inhibited p21 (Figure 5D). p21 knockdown com-pletely inhibited Pi-induced senescence and subsequentcalcification (Figure 5E).

Regulation of NPC-Mediated Runx2 Expression bySIRT1/p21 PathwayAs the next step, the role of SIRT1 in NPC-mediatedRunx2/Cbfa1 expression was examined. First, completeknockdown of SIRT1 did not show any change in bothosteoblastic markers, Runx2 and alkaline phosphatase, in anormal Pi (Supplemental Figure I). As shown in Figure 5F,

Pi-induced Runx2 was significantly blunted by PFA, an NPCinhibitor. SIRT1 activation by resveratrol inhibited Pi-induced Runx2 activation. The Runx2 induction was aug-mented by knockdown of SIRT1 by siRNA, and the activa-tion was completely inhibited by PFA. Surprisingly, Runx2activation was strongly inhibited by knockdown of p21 alone.In addition, the inhibition of Runx2 induction by doubleknockdown of SIRT1 and p21 was less than that by singleknockdown of SIRT1.

To address a difference in senescent induction by Pi or AngII, immunohistological assessment of SIRT1 in HASMCswas examined (Supplemental Figure II). Although SIRT1was predominantly localized in nucleus without Pi, thetranslocation of SIRT1 to cytoplasm was observed after Pistimulation for 24 hours, and its expression disappeared inboth areas on day 6. In contrast, Ang II stimulation did notshow the dynamic translocation.

High Sensitivity of SMCs With ReplicativeSenescence to Pi-Induced CalcificationNot only Pi-induced “premature senescence” in HASMCs butalso the effects of Pi on “replicative senescence” wereevaluated. Senescent cells (passage 18) were more sensitiveto Pi-induced calcification compared with young cells (pas-sage 7) (Figure 6A). SIRT1 expression was downregulated insenescent cells compared with young cells, and the down-regulation was significantly augmented by Pi stimulation(Figure 6B, top). In parallel with this finding, senescent cellsshowed an increase in Ac-p53 and p21 expression. Statisticalanalyses using densitometric measurement showed that (1)downregulation of SIRT1 and upregulation of Ac-53 and p21were augmented by replicative senescence, and (2) Pi inhib-ited the SIRT1-p21 pathway even in cells with replicativesenescence (passage 18) (Figure 6B, bottom).

DiscussionVascular aging, leading to cardiovascular disease, manifestscomplex and diverse vascular changes (eg, impairment ofdistensibility due to loss of arterial elasticity).1,16 Arterial wallstiffness resulting from ectopic calcification is a complicationof advanced atherosclerosis and makes the management ofhemodynamics more difficult in the elderly. Few reports haveaddressed whether cellular senescence is associated withSMC calcification. This study showed the importance ofSIRT1, a longevity gene, in arterial calcification in associa-tion with cellular senescence.

First, our data obtained from animal experiments clearlyshowed the association of senescent SMCs with aortic medialcalcification in the renal failure rats with hyperphosphatemia.Senescent cells showed significant colocalization with cal-cium deposition. Intriguingly, numerous senescent cells couldbe detected before microscopic calcification occurred at 4weeks after the start of renal failure induction (data notshown), suggesting that the transition to a senescent pheno-type in medial SMCs may be associated with the initiationand progression of calcification. Therefore, hyperphos-phatemia, a potent uremic factor, may be a stimulator toinduce senescent phenotypic transition of medial SMCs.

Figure 4. Modulation of sirtuin 1 (SIRT1) affects inorganic phos-phate (Pi)–induced senescent phenotypic change and calcifica-tion in smooth muscle cells (SMCs). The effects of sirtinol (achemical inhibitor of SIRT1 activity; A to C) and resveratrol (anactivator of SIRT1; D to F) on Pi-induced senescent phenotypicchange and calcification were examined (n�6). A, SIRT1 inhibi-tion by sirtinol (10 �mol/L) showed an increase in the number ofsenescence-associated �-galactosidase (SA�-gal)-positive cellseven without Pi stimulation. The increase in Pi-induced senes-cence was significantly augmented by sirtinol. Sirtinol aug-mented Pi-induced calcium deposition in human aortic SMCs(HASMCs) in a time-dependent (B) and dose-dependent manneron day 6 (C). Conversely, treatment with resveratrol (Resv;10 �mol/L) showed a reduction of the Pi-induced senescentphenotype (D) and calcification (E). The inhibitory effect of res-veratrol on calcification was dose dependent (F).

2058 Arterioscler Thromb Vasc Biol September 2011

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Page 29: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

54

In addition, Ang II did not increase calcium deposition,although the stimulation increased the number of senescentcells. Of note, Ang II alone did not increase Runx2 expres-sion in the absence of Pi (Figure 5B). This result suggests thatSMC senescence shows two different features: one is SA�-gal-positive cells with an increase in Runx2 and the other isSA�-gal-positive cells without. First, it has recently beenreported that SMCs with replicative senescence, rather thanthe cells without senescence, show hypersensitivity in re-sponse to induction of calcification with the more inductionof osteoblastic markers,20 suggesting that the induction ofosteoblastic transdifferentiation is strongly associated withthe senescent change in SMCs. In addition, the translocationof SIRT1 to cytoplasm was observed after Pi stimulation for24 hours, although SIRT1 predominantly localized in nucleuswithout Pi. In contrast, Ang II did not show the dynamictranslocation. Thinking about the mechanism for regulatingthe activity of HDACs, including SIRT1, recent severalreports show the importance of their coordinated shuttlingbetween nucleus and cytoplasm. A report demonstrates thatHDAC7, an HDAC, represses the transcriptional activity ofRunx2 and that osteogenic stimuli induce export of HDAC7from nucleus, leading to a decline in the repressive potentialsof HDAC7 for Runx2.21 On the basis of our findings and aprevious report, the reason that stimulation with Ang II alonedid not induce Runx2 expression and subsequently SMCcalcification may in part depend on the difference of SIRT1translocation after stimulation. Therefore, we strongly hy-pothesize that in the senescent SMCs with upregulation ofp21, Pi stimulation, but not Ang II stimulation, may activateRunx2 via at least two phenomena, the hyperacetylation ofRunx2 by SIRT1 downregulation and the dynamic SIRT1translocation, leading to marked osteoblastic transdifferentia-tion and subsequent calcification. In addition, we haveanother hypothesis. In general, it has been shown thathigh-dose Pi navigates release of matrix vesicles from SMCsin parallel with osteoblastic transdifferentiation. The vesiclesplay an essential role in the initiation of hydroxyapatiteaggregation, so-called nucleation. Accumulating recent re-ports show that the nanocrystal formation as an initial stepunder hyperphosphatemia accelerates the harmful cascade ofosteoblastic transdifferentiation in SMCs via endocytosis.22,23

Maybe Ang II alone does not induce the nanocrystal forma-tion and the cascade of osteoblastic change. Therefore, weexplain that the difference of senescent phenotypic changes inSMCs between both stimulations, Pi and Ang II alone, maydepend on (1) SIRT1 translocation and (2) nanocrystalformation to accelerate calcification. Further investigation toaddress the detailed mechanisms by which SIRT1 regulatesosteoblastic transdifferentiation in SMCs under the cellularsenescence is needed.

Are SIRT1 downregulation-related SMC senescence andsubsequent calcification reversible or not? To answer thisquestion, the effects of continuation or termination of high-dose Pi were examined. As shown in Figure 3B, termination(on day 6) of Pi showed no progression of senescence-relatedcalcification in association with the restoration of SIRT1,whereas continuation (up to day 10) of Pi stimulation showedfurther progression of calcification. It is suggested that a

therapeutic strategy to manage hyperphosphatemia to thenormal range of serum phosphate concentration may lead toat least termination of progressive calcification via reversal ofSIRT1 activity.

Cellular senescence has been shown to have two features:not only stress-induced premature senescence but also repli-cative senescence, indicating a limited number of divisions inculture.24 In fact, both endothelial cells and SMCs derivedfrom human atherosclerotic plaques show a senescent pheno-type earlier than do cells from normal vessels.25 Notably, wefound that senescent HASMCs were significantly more sen-sitive to Pi-induced calcification compared with young cells.These results suggest that calcium deposition may be morereadily induced in arterial medial SMCs with replicativesenescence. This insight may explain the mechanisms bywhich arterial calcification occurs in the elderly more fre-quently than in the young population. Therefore, these obser-vations support our hypothesis that arterial calcification isaccelerated by both senescent types (premature and replica-tive senescence) in SMCs. To explore new therapeuticstrategies against arterial calcification, it is essential toinvestigate how to maintain a higher SIRT1 level in thevasculature, leading to prevention of medial SMC senescenceand which drug is capable of achieving it.

How does SIRT1 exert protective effects against SMCcalcification? This study clearly showed that inhibition ofSIRT1 was associated with increases in both Ac-p53 and p21expression. These findings were significantly induced by notonly replicative senescence but also Pi-induced prematuresenescence. SIRT1-mediated deacetylation of p53 inhibitsp53-dependent transactivation of target genes, including p21.A report showed that a decline in cellular deacetylase activityincreases the half-life of endogenous p53,26 suggesting thatp53 acetylation is also associated with p53 stabilization.Therefore, the increased Ac-p53 by Pi-induced SIRT1 down-regulation may induce SMC senescence because of a declinein degradation of p53, leading to calcification. In addition,p53 itself can inhibit SIRT1 transcription because the SIRT1promoter has two response elements to p53.27 Further inves-tigation to address how the SIRT1-p53 negative regulatorypathway is associated with SMC calcification is needed.

On the other hand, regarding p21 activation, it is reportedthat inhibition of p21 expression in the vasculature signifi-cantly attenuates cellular senescence, leading to prevention ofatherosclerosis.28 This evidence suggests a pivotal role of p21in the development of atherosclerosis. p21 activation has beenshown to be regulated by a pathway that is p53 dependent,p53 independent, or both. Okamoto et al have demonstratedthat inhibition of HDAC by trichostatin A showed activationof p21 promoter activity by the Sp1 site even in vascularSMCs, and the induction of p21 was independent of the p53pathway.29 The p21 transcriptional activation in response toHDAC inhibitors was mediated by histone hyperacetylationin its promoter region. Based on these findings, Pi-inducedp21 activation via SIRT1 downregulation may be in partinvolved in a p53-independent pathway, leading to a senes-cent phenotype of SMCs. Further investigation exploringwhich molecule activates the p21 promoter under hyperphos-phatemia is needed.

Takemura et al SIRT1 Retards Vascular SMC Calcification 2061

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Second, we also confirmed the association of Pi-inducedSMC senescence with calcification in in vitro experiments.Senescent SMCs were significantly increased by Pi even onday 1, although calcium deposition was not markedly in-creased at the same time point. A statistically significantincrease in calcium deposition was found from day 3 and

later. Considering these data, we hypothesize that (1) calciumdeposition may be more readily induced in senescent cellscompared with nonsenescent cells, and (2) Pi-induced senes-cent change is observed earlier than calcium deposition. Inother words, senescent transition associated with Runx2induction may lead to progressive calcification.

Senescent SMCs were associated with the SIRT1-relatedp53/p21 pathway, based on the findings that SIRT1 knockdownaugmented not only cellular senescence but also calcification. Inaddition, p21 knockdown completely inhibited senescence-related calcification induced by Pi. This raises the question ofhow cellular senescence in SMCs is associated with calcifica-tion. Our experiments to understand the detailed mechanisms bywhich SIRT1 modulates senescence-related calcificationshowed that Pi-induced SIRT1 downregulation led to the phe-notypic change from a differentiated state to osteoblast-like cellsin SMCs. It has been reported that Pi induces osteoblasticchange, in which NPC plays a role in inducing Runx2/Cbfa-1expression, in SMCs.17 As the next step, to determine howSIRT1 regulates NPC-mediated Runx2 expression, we exam-ined the effects of knockdown of SIRT1, p21, or both by siRNAon Pi-induced Runx2 expression. Our data shown in Figure 5Fsuggested that (1) NPC plays an essential role in Pi-inducedRunx2 expression, (2) SIRT1 has an inhibitory effect on NPC-mediated Runx2 expression, (3) knockdown of p21 aloneameliorates Runx2 induction, and (4) p21-related osteoblasticchange is at least in part dependent on SIRT1.

There is now the new question of how SIRT1 regulatesRunx2 regulation. A report by Jeon18 has shown that acety-lation of Runx2 itself is important in osteoblast differentia-tion, and it is downregulated by HDAC activities. Based onthis evidence, SIRT1, 1 of the HDACs, may be able todeacetylate Runx2, leading to inhibition of Runx2-relatedosteoblastic transition in SMCs. Therefore, the inhibition ofSIRT1 by hyperphosphatemia may lead to Runx2 activationvia its hyperacetylation. Further investigation of the detailedmechanism of the SIRT1/p21/osteoblastic gene axis isneeded. These data clearly suggest that SIRT1 activation mayinhibit the hyperphosphatemia-induced osteoblastic pheno-typic change of SMCs, and the degree of change may bedependent on SIRT1 expression level. It is possible that theinhibition of SIRT1 expression by Pi alone is “partial,”because complete downregulation of SIRT1 by siRNA wors-ened the dynamic phenotypic change compared with Pi only.We have already shown that tumor necrosis factor-�, a potentatherogenic cytokine, augmented Pi-induced SMC calcifica-tion, as previously described.19 In addition, tumor necrosisfactor-� significantly decreased Pi-induced SIRT1 down-regulation further (data not shown). According to theseresults, we currently hypothesize that hyperphosphatemiainduces SIRT1 downregulation and subsequent osteoblasticphenotypic change in SMCs, leading to calcification, andthese changes are worsened by some harmful atherogenicfactors, which decrease SIRT1 expression/activity further.These results provide a new insight, showing that SIRT1plays an essential role in the prevention of arterial calcifica-tion and that the beneficial effect may be associated with aninhibition in Pi-induced SMC senescent transition.

Figure 6. High sensitivity of smooth muscle cells (SMCs) with rep-licative senescence to inorganic phosphate (Pi)–induced calcifica-tion. The effects of replicative senescence in human aortic SMCs(HASMCs) on Pi-induced calcification (A) and sirtuin 1 (SIRT1)-related molecules (B) were also evaluated. A, Senescent cells (pas-sage 18 [P18]) were more sensitive to Pi-induced calcificationcompared with young cells (passage 7 [P7]) (n�6). Representativephotographs of von Kossa staining (bottom) show strong inductionof calcium deposition by Pi (2.6 mmol/L). B, Senescent HASMCs(P18) showed a decline in SIRT1 expression and an increase inp21 expression compared with young cells (P7). Pi stimulation ofsenescent cells significantly inhibited SIRT1 expression and accel-erated the increase in p21 and acetylated (Ac)-p53. Densitometricanalysis confirmed these more sensitive responses in senescentcells. A.U. indicates arbitrary units. *P�0.05.

2060 Arterioscler Thromb Vasc Biol September 2011

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Page 30: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

55

In addition, Ang II did not increase calcium deposition,although the stimulation increased the number of senescentcells. Of note, Ang II alone did not increase Runx2 expres-sion in the absence of Pi (Figure 5B). This result suggests thatSMC senescence shows two different features: one is SA�-gal-positive cells with an increase in Runx2 and the other isSA�-gal-positive cells without. First, it has recently beenreported that SMCs with replicative senescence, rather thanthe cells without senescence, show hypersensitivity in re-sponse to induction of calcification with the more inductionof osteoblastic markers,20 suggesting that the induction ofosteoblastic transdifferentiation is strongly associated withthe senescent change in SMCs. In addition, the translocationof SIRT1 to cytoplasm was observed after Pi stimulation for24 hours, although SIRT1 predominantly localized in nucleuswithout Pi. In contrast, Ang II did not show the dynamictranslocation. Thinking about the mechanism for regulatingthe activity of HDACs, including SIRT1, recent severalreports show the importance of their coordinated shuttlingbetween nucleus and cytoplasm. A report demonstrates thatHDAC7, an HDAC, represses the transcriptional activity ofRunx2 and that osteogenic stimuli induce export of HDAC7from nucleus, leading to a decline in the repressive potentialsof HDAC7 for Runx2.21 On the basis of our findings and aprevious report, the reason that stimulation with Ang II alonedid not induce Runx2 expression and subsequently SMCcalcification may in part depend on the difference of SIRT1translocation after stimulation. Therefore, we strongly hy-pothesize that in the senescent SMCs with upregulation ofp21, Pi stimulation, but not Ang II stimulation, may activateRunx2 via at least two phenomena, the hyperacetylation ofRunx2 by SIRT1 downregulation and the dynamic SIRT1translocation, leading to marked osteoblastic transdifferentia-tion and subsequent calcification. In addition, we haveanother hypothesis. In general, it has been shown thathigh-dose Pi navigates release of matrix vesicles from SMCsin parallel with osteoblastic transdifferentiation. The vesiclesplay an essential role in the initiation of hydroxyapatiteaggregation, so-called nucleation. Accumulating recent re-ports show that the nanocrystal formation as an initial stepunder hyperphosphatemia accelerates the harmful cascade ofosteoblastic transdifferentiation in SMCs via endocytosis.22,23

Maybe Ang II alone does not induce the nanocrystal forma-tion and the cascade of osteoblastic change. Therefore, weexplain that the difference of senescent phenotypic changes inSMCs between both stimulations, Pi and Ang II alone, maydepend on (1) SIRT1 translocation and (2) nanocrystalformation to accelerate calcification. Further investigation toaddress the detailed mechanisms by which SIRT1 regulatesosteoblastic transdifferentiation in SMCs under the cellularsenescence is needed.

Are SIRT1 downregulation-related SMC senescence andsubsequent calcification reversible or not? To answer thisquestion, the effects of continuation or termination of high-dose Pi were examined. As shown in Figure 3B, termination(on day 6) of Pi showed no progression of senescence-relatedcalcification in association with the restoration of SIRT1,whereas continuation (up to day 10) of Pi stimulation showedfurther progression of calcification. It is suggested that a

therapeutic strategy to manage hyperphosphatemia to thenormal range of serum phosphate concentration may lead toat least termination of progressive calcification via reversal ofSIRT1 activity.

Cellular senescence has been shown to have two features:not only stress-induced premature senescence but also repli-cative senescence, indicating a limited number of divisions inculture.24 In fact, both endothelial cells and SMCs derivedfrom human atherosclerotic plaques show a senescent pheno-type earlier than do cells from normal vessels.25 Notably, wefound that senescent HASMCs were significantly more sen-sitive to Pi-induced calcification compared with young cells.These results suggest that calcium deposition may be morereadily induced in arterial medial SMCs with replicativesenescence. This insight may explain the mechanisms bywhich arterial calcification occurs in the elderly more fre-quently than in the young population. Therefore, these obser-vations support our hypothesis that arterial calcification isaccelerated by both senescent types (premature and replica-tive senescence) in SMCs. To explore new therapeuticstrategies against arterial calcification, it is essential toinvestigate how to maintain a higher SIRT1 level in thevasculature, leading to prevention of medial SMC senescenceand which drug is capable of achieving it.

How does SIRT1 exert protective effects against SMCcalcification? This study clearly showed that inhibition ofSIRT1 was associated with increases in both Ac-p53 and p21expression. These findings were significantly induced by notonly replicative senescence but also Pi-induced prematuresenescence. SIRT1-mediated deacetylation of p53 inhibitsp53-dependent transactivation of target genes, including p21.A report showed that a decline in cellular deacetylase activityincreases the half-life of endogenous p53,26 suggesting thatp53 acetylation is also associated with p53 stabilization.Therefore, the increased Ac-p53 by Pi-induced SIRT1 down-regulation may induce SMC senescence because of a declinein degradation of p53, leading to calcification. In addition,p53 itself can inhibit SIRT1 transcription because the SIRT1promoter has two response elements to p53.27 Further inves-tigation to address how the SIRT1-p53 negative regulatorypathway is associated with SMC calcification is needed.

On the other hand, regarding p21 activation, it is reportedthat inhibition of p21 expression in the vasculature signifi-cantly attenuates cellular senescence, leading to prevention ofatherosclerosis.28 This evidence suggests a pivotal role of p21in the development of atherosclerosis. p21 activation has beenshown to be regulated by a pathway that is p53 dependent,p53 independent, or both. Okamoto et al have demonstratedthat inhibition of HDAC by trichostatin A showed activationof p21 promoter activity by the Sp1 site even in vascularSMCs, and the induction of p21 was independent of the p53pathway.29 The p21 transcriptional activation in response toHDAC inhibitors was mediated by histone hyperacetylationin its promoter region. Based on these findings, Pi-inducedp21 activation via SIRT1 downregulation may be in partinvolved in a p53-independent pathway, leading to a senes-cent phenotype of SMCs. Further investigation exploringwhich molecule activates the p21 promoter under hyperphos-phatemia is needed.

Takemura et al SIRT1 Retards Vascular SMC Calcification 2061

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Second, we also confirmed the association of Pi-inducedSMC senescence with calcification in in vitro experiments.Senescent SMCs were significantly increased by Pi even onday 1, although calcium deposition was not markedly in-creased at the same time point. A statistically significantincrease in calcium deposition was found from day 3 and

later. Considering these data, we hypothesize that (1) calciumdeposition may be more readily induced in senescent cellscompared with nonsenescent cells, and (2) Pi-induced senes-cent change is observed earlier than calcium deposition. Inother words, senescent transition associated with Runx2induction may lead to progressive calcification.

Senescent SMCs were associated with the SIRT1-relatedp53/p21 pathway, based on the findings that SIRT1 knockdownaugmented not only cellular senescence but also calcification. Inaddition, p21 knockdown completely inhibited senescence-related calcification induced by Pi. This raises the question ofhow cellular senescence in SMCs is associated with calcifica-tion. Our experiments to understand the detailed mechanisms bywhich SIRT1 modulates senescence-related calcificationshowed that Pi-induced SIRT1 downregulation led to the phe-notypic change from a differentiated state to osteoblast-like cellsin SMCs. It has been reported that Pi induces osteoblasticchange, in which NPC plays a role in inducing Runx2/Cbfa-1expression, in SMCs.17 As the next step, to determine howSIRT1 regulates NPC-mediated Runx2 expression, we exam-ined the effects of knockdown of SIRT1, p21, or both by siRNAon Pi-induced Runx2 expression. Our data shown in Figure 5Fsuggested that (1) NPC plays an essential role in Pi-inducedRunx2 expression, (2) SIRT1 has an inhibitory effect on NPC-mediated Runx2 expression, (3) knockdown of p21 aloneameliorates Runx2 induction, and (4) p21-related osteoblasticchange is at least in part dependent on SIRT1.

There is now the new question of how SIRT1 regulatesRunx2 regulation. A report by Jeon18 has shown that acety-lation of Runx2 itself is important in osteoblast differentia-tion, and it is downregulated by HDAC activities. Based onthis evidence, SIRT1, 1 of the HDACs, may be able todeacetylate Runx2, leading to inhibition of Runx2-relatedosteoblastic transition in SMCs. Therefore, the inhibition ofSIRT1 by hyperphosphatemia may lead to Runx2 activationvia its hyperacetylation. Further investigation of the detailedmechanism of the SIRT1/p21/osteoblastic gene axis isneeded. These data clearly suggest that SIRT1 activation mayinhibit the hyperphosphatemia-induced osteoblastic pheno-typic change of SMCs, and the degree of change may bedependent on SIRT1 expression level. It is possible that theinhibition of SIRT1 expression by Pi alone is “partial,”because complete downregulation of SIRT1 by siRNA wors-ened the dynamic phenotypic change compared with Pi only.We have already shown that tumor necrosis factor-�, a potentatherogenic cytokine, augmented Pi-induced SMC calcifica-tion, as previously described.19 In addition, tumor necrosisfactor-� significantly decreased Pi-induced SIRT1 down-regulation further (data not shown). According to theseresults, we currently hypothesize that hyperphosphatemiainduces SIRT1 downregulation and subsequent osteoblasticphenotypic change in SMCs, leading to calcification, andthese changes are worsened by some harmful atherogenicfactors, which decrease SIRT1 expression/activity further.These results provide a new insight, showing that SIRT1plays an essential role in the prevention of arterial calcifica-tion and that the beneficial effect may be associated with aninhibition in Pi-induced SMC senescent transition.

Figure 6. High sensitivity of smooth muscle cells (SMCs) with rep-licative senescence to inorganic phosphate (Pi)–induced calcifica-tion. The effects of replicative senescence in human aortic SMCs(HASMCs) on Pi-induced calcification (A) and sirtuin 1 (SIRT1)-related molecules (B) were also evaluated. A, Senescent cells (pas-sage 18 [P18]) were more sensitive to Pi-induced calcificationcompared with young cells (passage 7 [P7]) (n�6). Representativephotographs of von Kossa staining (bottom) show strong inductionof calcium deposition by Pi (2.6 mmol/L). B, Senescent HASMCs(P18) showed a decline in SIRT1 expression and an increase inp21 expression compared with young cells (P7). Pi stimulation ofsenescent cells significantly inhibited SIRT1 expression and accel-erated the increase in p21 and acetylated (Ac)-p53. Densitometricanalysis confirmed these more sensitive responses in senescentcells. A.U. indicates arbitrary units. *P�0.05.

2060 Arterioscler Thromb Vasc Biol September 2011

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from

Page 31: Sirtuin 1 Retards Hyperphosphatemia-Induced …atvb.ahajournals.org/content/atvbaha/31/9/2054.full.pdf · Sirtuin 1 Retards Hyperphosphatemia-Induced Calcification of Vascular Smooth

56

ConclusionWe showed that SIRT1 exerts a protective role inhyperphosphatemia-based arterial calcification via inhibitionof osteoblastic transdifferentiation, in association with cross-talk between calcification and cellular senescence. Thisability of SIRT1 may orchestrate an analogous protective/longevity paradigm even in vascular SMCs, leading tomaintenance of healthy elasticity of the arterial wall. Strate-gies to maintain a higher level of SIRT1 activity may providenovel therapeutic opportunities for the prevention of arterialcalcification.

AcknowledgmentsThis study was supported by Grants-in-Aid for Scientific Researchfrom the Ministry of Education, Science, Culture and Sports of Japan(No. 19590854, No. 21590947, and No. 20249041), Ono MedicalResearch Foundation, Kanzawa Medical Research Foundation, No-vartis Foundation for Gerontological Research, Takeda ResearchFoundation, and Mitsui-Sumitomo Insurance Welfare Foundation.

Sources of FundingThis manuscript is supported by Grants-in-Aid for Scientific Researchfrom the Ministry of Education, Science, Culture and Sports of Japan(No. 19590854, No. 21590947, No. 20249 041). In particular, this ismainly supported by No. 21590947.

DisclosuresNone.

References1. Safar ME, Levy BI, Struijker-Boudier H. Current perspectives on arterial

stiffness and pulse pressure in hypertension and cardiovascular diseases.Circulation. 2003;107:2864–2869.

2. Abedin M, Tintut Y, Demer LL. Vascular calcification: mechanisms andclinical ramifications. Arterioscler Thromb Vasc Biol. 2004;24:1161–1170.

3. Iijima K, Hashimoto H, Hashimoto M, Son BK, Ota H, Ogawa S, Eto M,Akishita M, Ouchi Y. Aortic arch calcification detectable on chest X-rayis a strong independent predictor of cardiovascular events beyond tradi-tional risk factors. Atherosclerosis. 2010;210:137–144.

4. Demer LL, Tintut Y. Vascular calcification: pathobiology of a multi-faceted disease. Circulation. 2008;117:2938–2948.

5. Ehara S, Kobayashi Y, Yoshiyama M, Shimada K, Shimada Y, Fukuda D,Nakamura Y, Yamashita H, Yamagishi H, Takeuchi K, Naruko T, HazeK, Becker AE, Yoshikawa J, Ueda M. Spotty calcification typifies theculprit plaque in patients with acute myocardial infarction: an intra-vascular ultrasound study. Circulation. 2004;110:3424–3429.

6. Shroff RC, Shanahan CM. The vascular biology of calcification. SeminDial. 2007;20:103–109.

7. Persy V, D’Haese P. Vascular calcification and bone disease: the calci-fication paradox. Trends Mol Med. 2009;15:405–416.

8. Shanahan CM, Cary NR, Salisbury JR, Proudfoot D, Weissberg PL,Edmonds ME. Medial localization of mineralization-regulating proteinsin association with Monckeberg’s sclerosis: evidence for smooth musclecell-mediated vascular calcification. Circulation. 1999;100:2168–2176.

9. Tyson KL, Reynolds JL, McNair R, Zhang Q, Weissberg PL, ShanahanCM. Osteo/chondrocytic transcription factors and their target genesexhibit distinct patterns of expression in human arterial calcification.Arterioscler Thromb Vasc Biol. 2003;23:489–494.

10. Brachmann CB, Sherman JM, Devine SE, Cameron EE, Pillus L, BoekeJD. The SIR2 gene family, conserved from bacteria to humans, functionsin silencing, cell cycle progression, and chromosome stability. GenesDev. 1995;9:2888–2902.

11. Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, Pandita TK,Guarente L, Weinberg RA. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell. 2001;107:149–159.

12. Langley E, Pearson M, Faretta M, Bauer UM, Frye RA, Minucci S,Pelicci PG, Kouzarides T. Human SIR2 deacetylates p53 and antagonizesPML/p53-induced cellular senescence. EMBO J. 2002;21:2383–2396.

13. Ota H, Akishita M, Eto M, Iijima K, Kaneki M, Ouchi Y. Sirt1 modulatespremature senescence-like phenotype in human endothelial cells. J MolCell Cardiol. 2007;43:571–579.

14. Yokozawa T, Zheng PD, Oura H, Koizumi F. Animal model of adenine-induced chronic renal failure in rats. Nephron. 1986;44:230–234.

15. Son BK, Kozaki K, Iijima K, Eto M, Kojima T, Ota H, Senda Y,Maemura K, Nakano T, Akishita M, Ouchi Y. Statins protect humanaortic smooth muscle cells from inorganic phosphate-induced calcifi-cation by restoring Gas6-Axl survival pathway. Circ Res. 2006;98:1024–1031.

16. Ferrari AU, Radaelli A, Centola M. Invited review: aging and the car-diovascular system. J Appl Physiol. 2003;95:2591–2597.

17. Jono S, McKee MD, Murry CE, Shioi A, Nishizawa Y, Mori K, Morii H,Giachelli CM. Phosphate regulation of vascular smooth muscle cell cal-cification. Circ Res. 2000;87:E10–E17.

18. Jeon EJ, Lee KY, Choi NS, Lee MH, Kim HN, Jin YH, Ryoo HM, ChoiJY, Yoshida M, Nishino N, Oh BC, Lee KS, Lee YH, Bae SC. Bonemorphogenetic protein-2 stimulates Runx2 acetylation. J Biol Chem.2006;281:16502–16511.

19. Son BK, Akishita M, Iijima K, Kozaki K, Maemura K, Eto M, Ouchi Y.Adiponectin antagonizes stimulatory effect of tumor necrosis factor-� onvascular smooth muscle cell calcification: regulation of growth arrest-specificgene 6-mediated survival pathway by adenosine 5�-monophosphate-activatedprotein kinase. Endocrinology. 2008;149:1646–1653.

20. Nakano-Kurimoto R, Ikeda K, Uraoka M, Nakagawa Y, Yutaka K, KoideM, Takahashi T, Matoba S, Yamada H, Okigaki M, Matsubara H. Rep-licative senescence of vascular smooth muscle cells enhances the calci-fication through initiating the osteoblastic transition. Am J Physiol HeartCirc Physiol. 2009;297:H1673–H1684.

21. Jensen ED, Gopalakrishnan R, Westendorf JJ. Bone morphogenic protein2 activates protein kinase D to regulate histone deacetylase 7 localizationand repression of Runx2. J Biol Chem. 2009;284:2225–2234.

22. Ewence AE, Bootman M, Roderick HL, Skepper JN, McCarthy G, EppleM, Neumann M, Shanahan CM, Proudfoot D. Calcium phosphate crystalsinduce cell death in human vascular smooth muscle cells: a potentialmechanism in atherosclerotic plaque destabilization. Circ Res. 2008;103:e28–e34.

23. Sage AP, Lu J, Tintut Y, Demer LL. Hyperphosphatemia-induced nano-crystals upregulate the expression of bone morphogenetic protein-2 andosteopontin genes in mouse smooth muscle cells in vitro. Kidney Int.2011;79:414–422.

24. Hayflick L. Current theories of biological aging. Fed Proc. 1975;34:9–13.25. Minamino T, Komuro I. Vascular cell senescence: contribution to ath-

erosclerosis. Circ Res. 2007;100:15–26.26. Ito A, Lai CH, Zhao X, Saito S, Hamilton MH, Appella E, Yao TP.

p300/CBP-mediated p53 acetylation is commonly induced by p53-activating agents and inhibited by MDM2. EMBO J. 2001;20:1331–1340.

27. Nemoto S, Fergusson MM, Finkel T. Nutrient availability regulatesSIRT1 through a forkhead-dependent pathway. Science. 2004;306:2105–2108.

28. Andreassi MG. DNA damage, vascular senescence and atherosclerosis.J Mol Med. 2008;86:1033–1043.

29. Okamoto H, Fujioka Y, Takahashi A, Takahashi T, Taniguchi T, Ishikawa Y,Yokoyama M. Trichostatin A, an inhibitor of histone deacetylase, inhibitssmooth muscle cell proliferation via induction of p21(WAF1). J AtherosclerThromb. 2006;13:183–191.

2062 Arterioscler Thromb Vasc Biol September 2011

by IMED Korea on October 5, 2011http://atvb.ahajournals.org/Downloaded from