1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63

ORIGINAL ARTICLE Q1

Q22

Sirtuin-6 inhibits cardiac fibroblast differentiation into myofibroblasts via inactivation of nuclear factor kB signaling KUNMING TIAN1, ZHIPING LIU1, JIAOJIAO WANG1, SUOWEN XU2, TIANHUI YOU, and PEIQING LIU GUANGZHOU, CHINA

Differentiation of cardiac fibroblasts (CFs) into myofibroblasts represents a key event in cardiac fibrosis that contributes to pathologic cardiac remodeling. However, regulation of this phenotypic transformation remains elusive. Here, we show that sirtuin-6 (SIRT6), a member of the sirtuin family of NAD1-dependent histone deacetylase, plays a role in the regulation of myofibroblast differentiation. SIRT6 expression was upregulated under pathologic conditions in angiotensin II (Ang II)-stimulated CFs and in myocardium of rat subject to abdominal aortic constriction surgery. SIRT6 depletion by RNA interference (small interfering RNA [siRNA]) in CFs resulted in increased cell proliferation and extracellular matrix deposition. Further examination of SIRT6-depleted CFs demonstrated significantly higher expression of a-smooth muscle actin (a-SMA), the classical marker of myofibroblast differentiation, and increased formation of focal adhesions. Notably, SIRT6 depletion further exacerbated Ang II–induced myofibroblast differentiation. Overexpression of SIRT6 restored a-SMA expression in SIRT6-depleted or Ang II–treated CFs. Moreover, SIRT6 depletion induced the DNA binding activity and transcriptional activity of nuclear factor kB (NFkB). Importantly, using an NF-kB p65 siRNA or pyrrolidine dithiocarbamate, a specific inhibitor of NF-kB activity, reversed the expression of phenotypic markers of myofibroblasts. Our findings unravel a novel role of SIRT6 as a key modulator in the phenotypic conversion of CFs to myofibroblasts. (Translational Research 2014;-:1–12)

Q2

Abbreviations: --- ¼ ---

Q3

INTRODUCTION

H

eart failure is a major cause of death in patients with coronary heart disease in the developed world. The development of heart failure is invariably associated with extensive fibrosis, which

1

Kunming Tian, Zhiping Liu, and Jiaojiao Wang contributed equally to this work. 2 Present address: Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY 14642. From the School of Nursing, Guangdong Pharmaceutical University, Guangzhou, China; Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China.

aggravates diastolic dysfunction and predisposes the heart to arrhythmias and sudden cardiac death.1,2 A critical event in the progression of cardiac fibrosis is the differentiation of cardiac fibroblasts (CFs) into myofibroblasts, characterized by the expression of Submitted for publication May 25, 2014; revision submitted August 25, 2014; accepted for publication August 28, 2014. Reprint requests: Peiqing Liu, Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University (Higher Education Mega Center), 132 East Wai-Huan Road, Guangzhou 510006, Guangdong, China; e-mail: [email protected]. 1931-5244/$ - see front matter Ó 2014 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.trsl.2014.08.008

1 FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127

2

128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191

Translational Research - 2014

Tian et al

AT A GLANCE COMMENTARY Tian K, et al. Background

Sirtuin-6 (SIRT6), an NAD1-dependent histone deacetylase, has been implicated in the regulation of genomic stability, cellular metabolism, stress responses, and life-span. However, whether SIRT6 plays a role in the regulation of myofibroblast differentiation and cardiac fibrosis remains largely unknown. Translational Significance

The present study has demonstrated that SIRT6 depletion induces cardiac fibroblast differentiation into myofibroblasts. Previous study has shown that SIRT6 expression was markedly reduced in failing human hearts. Thus, development of specific activators of SIRT6 to increase the levels or activity of this protein may be beneficial for the prevention and/or therapeutic treatment of cardiac fibrosis and heart failure.

Q4

a-smooth muscle actin (a-SMA) and production of extracellular matrix (ECM) components such as fibronectin (FN), collagen type I, and collagen type III.3,4 Myofibroblast differentiation is induced by several profibrotic factors, such as angiotensin II (Ang II), endothelin-1, platelet-derived growth factor, and transforming growth factor b (TGF-b).5-7 Ang II can also stimulate the expression of TGF-b and endothelin-1,8 additionally amplifying in the pathogenesis of cardiac fibrosis. Sirtuins (SIRTs), a family of evolutionary conserved NAD1-dependent histone deacetylases, are implicated in the regulation of multiple pathophysiological processes, including DNA damage repair, metabolic homeostasis, apoptosis, tumorigenesis, neurodegeneration, and aging.9,10 So far, 7 SIRT genes have been found in mammal genomes, named as SIRT1–7. Resveratrol, a well-known SIRT agonist, has been shown to inhibit Ang II and TGF-b–induced CF proliferation and myofibroblast differentiation.11 In the present study, we screened the messenger RNA (mRNA) expressions of all SIRT isoforms in Ang II–induced CFs and found the SIRT1 and SIRT6 were upregulated in response to Ang II stimulation. Importantly, change in the expression of SIRT6 was the most significant, indicating that SIRT6 may play a role in Ang II–treated CFs. Recently, mounting evidence suggests that SIRT6 exerts protective effects in

multiple disease models, including obesity,12 liver cancer,13 fatty liver,14 cardiac hypertrophy,15,16 and chronic liver inflammation.17 However, the precise role of SIRT6 in myofibroblast differentiation and cardiac fibrosis remains unknown. This study aims to investigate the possible roles of SIRT6 in the regulation of myofibroblast differentiation and to further explore the potential mechanisms. MATERIALS AND METHODS Chemicals and reagents. Rabbit anti–nuclear factor kB (anti–NF-kB) p65 antibody was purchased from Abcam (Cambridge, Massachusetts). Rabbit anti–IkB-a, a-SMA, paxillin, vinculin, FN, plasminogen activator inhibitor 1 (PAI-1), and collagen type I antibodies were from Santa Cruz Biotechnology (Santa Cruz, California). Rabbit anti-phospho-p65 antibody was Q5 obtained from Cell Signaling Technology (Danvers, Massachusetts). Mouse anti-a-tubulin antibody, rabbit anti-SIRT6, histone H1 antibodies, and pyrrolidine dithiocarbamic acid ammonium salt were purchased from Sigma-Aldrich (St. Louis, Missouri). Other reagents were from Sigma-Aldrich unless otherwise specified. Cell culture. Adult rat left ventricular CFs were isolated as previously described.18 In brief, 8- to 10-week-old (150–200 g) male Sprague-Dawley rats were anaesthetized with an intraperitoneal injection of sodium pentobarbitone (150 mg/kg). Hearts were then excised from anesthetized rats and rinsed in cold phosphate-buffered saline (PBS). The ventricles were Q6 minced and digested in Dulbecco’s modified Eagle’s medium containing 0.1% collagenase type II (Gibco BRL, Grand Island, New York) at 37 C with continuous shaking for 60 minutes. The dissociated cells were collected and plated for 1 hour at 37 C. After preplating, the unattached cells (including myocytes and endothelial cells) were removed and the CFs were cultured in Dulbecco’s modified Eagle’s medium containing 10% new born calf serum (Gibco), 100 U/mL penicillin, and 100 mg/mL streptomycin. The purity of CFs was more than 95% as determined by positive staining for vimentin and negative staining for a-actin and von Willebrand factor. The cells in the second to fourth passage were used in this study. All experimental procedures were conducted as per the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH Publication No. 85-23, revised 1996) and were approved by the Institutional Ethics Review Board of Sun Yat-Sen University. Real-time polymerase chain reaction. RNA isolation Q7 and real-time polymerase chain reaction (PCR) was

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255

Translational Research Volume -, Number 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319

Q8

Q9

Tian et al

conducted as previously described in detail.19 Rat-specific primers for SIRT1–7, a-SMA, paxillin, vinculin, FN, PAI-1, collagen type I, and GAPDH were used (see Supplemental Table 1). Western blot. Western blot analyses were performed as previously described.19 For analysis of p65 protein expression, cytoplasmic and nuclear proteins were extracted with the CelLytic NuCLEAR Extraction Kit (Sigma). Histone H1 and a-tubulin were used to normalize nuclear and cytosolic protein loading. Plasmid transfection. CFs were transiently transfected with SIRT6 (EGFP-SIRT6) or empty vector (EGFP-N3) using Lipofectamine 2000 (Invitrogen, Carlsbad, California) according to the manufacturer’s instructions. After incubation for 24 hours, the cells were treated with or without Ang II for indicated time. RNA interference. Three different duplex small interfering RNAs (siRNAs) for SIRT6, p65, and negative control siRNAs were purchased from GenePharma (Shanghai, China). The sequences of siRNAs are shown in the Supplemental material (Supplemental Tables 2 and 3). Subconfluent CFs (70% confluence) were transfected either with SIRT6 siRNAs or with negative control siRNA using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s instructions. In shortterm silencing experiments, once the transfection complexes were removed, the cultures were maintained in a normal fresh medium for further 48 hours before analysis. In long-term silencing experiments, the cells were harvested by trypsinization after 48 hours of transfection, then seeded at a density of 5 3 106 cells/mL, and maintained in fresh medium for a further 24-hour period before analysis. Real-time PCR and Western blot were used at 48 hours after transfection to compare silencing efficiency of different duplex siRNAs (see Supplemental Figs 1 and 2). In all our siRNA experiments, we did not observe any significant difference between negative siRNA and untreated cells, eliminating the possibility that siRNA has a nonspecific effect on CFs. CFs proliferation assay. Cell Counting Kit-8 (Dojindo, Shanghai, China) was used in CF proliferation assay. Cells were seeded at 5 3 104 cells per well into 96-well plates in a final volume of 100 mL and transfected with negative control siRNA or S6 siRNA. After 72 hours of transfection, cells were subjected to assay by adding 10 mL of Cell Counting Kit-8 solution to each well, and the plate was further incubated for 4 hours at 37 C. The absorbance at 450 nm was measured with a microplate reader. The experiment was performed in 10 duplicates. Hydroxyproline measurement. The content of hydroxyproline in CFs was assayed by a commercial kit (Nanjing Jiancheng Bioengineering Institute,

3

Nanjing, China) according to the manufacturer’s instructions. Immunofluorescence staining. CFs cultured on glass coverslips were fixed in 4% paraformaldehyde for 30 minutes, washed with PBS 3 times, and permeabilized with 0.1% Triton X-100 in PBS for 20 minutes. After rinsing with PBS 3 times, the slides were blocked with 5% goat serum at room temperature for 30 minutes. Next, the cells were incubated with a rabbit polyclonal anti-a-SMA primary antibody (1:100 dilution) overnight at 4 C. The slides were then rinsed and incubated with a goat anti-rabbit secondary antibody conjugated to Alexa Fluor 594 (1:1, 000 dilution; Invitrogen) for 1 hour at room temperature. The slides were counterstained with DAPI (5 mg/mL, Sigma) and mounted in a glycerin jelly medium, Q10 then subjected to confocal microscopy (LSM710; Carl Zeiss). Control coverslips were processed the same way, except for omission of the primary antibody or secondary antibody. Luciferase reporter gene assay. NF-kB–dependent transcriptional activity was conducted as previously described.16 In brief, CFs were cotransfected with NF-kB reporter plasmid and pRL-TK as an internal Q11 control. After 8 hours of incubation, the cells were subjected to Ang II stimulation, RNA interference, or transfection with SIRT6 or empty vector. The luciferase activity was measured using the DualLuciferase Reporter Assay System (Promega, Madison, Wisconsin) according to the manufacturer’s protocol. Relative luciferase activity was calculated as the ratio of firefly luciferase activity to Renilla luciferase activity. Electrophoretic mobility shift assay. Electrophoretic mobility shift assay (EMSA) was performed using a LightShift Chemiluminescent EMSA kit (Pierce) ac- Q12 cording to our established procedure.19,20 Briefly, CFs were washed with ice-cold PBS and scrapped off with a cell scraper. Nuclear extracts were prepared using the Nuclear Extract Kit (Active Motif) according to the manufacturer’s instructions. Two micrograms of nuclear protein was then used to assess DNA binding activity using a 30 -biotin–labeled oligonucleotide probes containing the consensus sequence (underlined) of the NF-kB: 50 -tgg aaa tgg gaa gtc tca tag gac-30 (sense) and 50 -gtc cta tga gac ttc cca ttt cca-30 (antisense). Each sample was electrophoresed in a 6% nondenaturing polyacrylamide gel in 0.53 TBE buffer at 100 V for 60 minutes. For cold competition experiments, 50-fold molar excess unlabeled duplex oligonucleotides containing NF-kB consensus sequence was added to the nuclear extracts before incubation with the biotin-labeled oligonucleotides. Abdominal aortic constriction surgery. SpragueDawley rats (male, weighing 180–200 g, SPF grade,

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383

4

384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447

Translational Research - 2014

Tian et al

certification no. 44008500003151) were supplied by the Experimental Animal Center of Sun Yat-Sen University (Guangzhou, China). Pressure overload was induced by suprarenal abdominal aortic constriction (AAC) as previously described.21 The rats were anaesthetized by an intraperitoneal injection of sodium pentobarbitone (50 mg/kg) before AAC surgery. Loss of reflex was detected by pricking the rats’s feet and legs with forceps. At the end of experiment, rats were sacrificed by CO2 inhalation and then hearts were harvested and stored at 280 C for Western blot analysis. The investigation conformed to the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH Publication No. 85-23, revised 1996). The animal experiments were approved by the Institutional Ethics Review Board of Sun Yat-Sen University. Statistical analysis. Data are presented as the mean 6 standard error of the mean. Statistical analysis was performed with t test or by 1- or 2-way analysis of variance followed by Bonferroni’s post hoc tests as appropriate. In all cases, differences were considered statistically significant with P , 0.05. RESULTS Expression of SIRT6 was upregulated in the in vitro and in vivo model of cardiac fibrosis. To assess whether

SIRT6 might play a relevant role in CFs, we first compared the mRNA expression profile of SIRT1–7 in CFs, using the expression of SIRT1 as a reference. As shown in Supplemental Fig 3, SIRT1, SIRT5, and SIRT6 were highly expressed in CFs, and the mRNA expressions of SIRT1 and SIRT6 were almost equal. Ang II has been well recognized as a potent stimulator of cardiac fibrotic response by promoting CF proliferation and myofibroblast differentiation.5,11 We thus examined the effects of Ang II on mRNA levels of SIRT1–7 in CFs. Fig 1, A shows that the mRNA levels of SIRT1 and SIRT6 were increased in response to Ang II stimulation, and the increase in SIRT6 was the most prominent. However, we did not observe significant alterations in the mRNA expression of other SIRTs. Moreover, Western blot analysis revealed that Ang II in a time- and concentration-dependent manner increased the protein expression of SIRT6 (Fig 1, B and C). Comparing with the untreated group, treatment with 100 nM of Ang II for 24 hours resulted in the increase in protein level of SIRT6 by 3.05 6 0.36 fold (P , 0.05). To further examine whether there was a similar alteration of SIRT6 in pressure overload-induced cardiac fibrosis of rats, we analyzed SIRT6 expression in the hearts from AAC or sham-operated rats at the age of 8 weeks. Collagen deposition and the expression of

fibrotic markers including a-SMA, collagen type I, and FN were significantly increased in AAC rats (Supplemental Fig 4), suggesting the successful induction of cardiac fibrosis by AAC surgery. In addition, the protein expression of SIRT6 was markedly upregulated in pressure-overloaded hearts induced by AAC (Fig 1, D) and in CFs isolated from the hearts of AAC rat (Supplemental Fig 5). These results raised the possibility that SIRT6 may play a critical role in the development of cardiac fibrosis. Silencing of SIRT6-induced CF proliferation and ECM deposition. To explore the potential role of SIRT6 in

cardiac fibrosis, we first knocked down SIRT6 in CFs using siRNA. Real-time PCR and Western blot were performed to evaluate the effect of 3 independent siRNAs, marked si1, si2, and si3, respectively. As shown in Supplemental Fig 1, si3 reduced the mRNA and protein expressions of SIRT6 by 85% and 92%, respectively, without affecting other SIRT isoforms (Supplemental Fig 6). Therefore, si3 was used in the following experiments. CFs transfected with the negative control siRNA were well spread and displayed few plasma membrane protrusions (Fig 2, A). In contrast, SIRT6-silenced CFs exhibited an irregular cell shape, which was characterized by numerous protrusions of the cytoplasm (Fig 2, B). SIRT6-silenced cells showed increased rates of proliferation (Fig 2, C), suggesting a proproliferative phenotype of SIRT6 depletion. Cardiac fibrosis leads to ECM deposition and changes in gene expression reminiscent of a pathologic phenotype, such as FN, collagen type I, and PAI-1. Consistent with this notion, Fig 2, D shows that in SIRT6-depleted CFs there is a significant increase in hydroxyproline content. In addition, real-time PCR analysis showed that SIRT6-silenced CFs displayed higher mRNA levels of FN, collagen type I, and PAI-1 (Fig 2, E). Similarly, testing for these fibrosis-related genes by Western blot showed paralleling changes at the protein level (Fig 2, F). Silencing of SIRT6 induced CF differentiation into myofibroblasts. To examine the potential effect of

SIRT6 on myofibroblast differentiation, CFs were transfected with SIRT6 siRNA or/and treated with Ang II. Q13 SIRT6 depletion could mimic the effects of Ang II to markedly induce mRNA (Supplemental Fig 7) and protein (Fig 3, A) levels of a-SMA. Importantly, SIRT6 siRNA transfection exacerbated the effect of Ang II-mediated upregulation of a-SMA mRNA and protein expression. This effect was confirmed by immunofluorescence analysis (Fig 3, B). Furthermore, immunofluorescent staining assay also revealed increased cell size of CFs and a-SMA containing stress-fibers indicative of myofibroblasts formation in

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511

Translational Research Volume -, Number 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575

Tian et al

5

Fig 1. SIRT6 was upregulated in Ang II–treated CFs and in AAC rats. (A) Adult rat left ventricular CFs were treated with 100 nM of Ang II for 24 hours, and real-time polymerase chain reaction was performed to screen the mRNA expression profile of SIRT1–7. Data are presented as the mean 6 SEM (*P , 0.05 vs control, n 5 5). (B and C) CFs were treated with 100 nM of Ang II for indicated time (left panel) or with indicated concentrations of Ang II for 24 hours (right panel). The levels of mRNA (B) and protein (C) of SIRT6 were detected and normalized (mRNA by GAPDH and protein by a-tubulin) and then presented as fold of control level. Data are presented as the mean 6 SEM (*P , 0.05 vs control, n 5 6). (D) Sprague-Dawley rats were subjected to AAC surgery for 8 weeks. Hearts were collected from sham rats or AAC rats, and subjected to Western blot to examine the expression of SIRT6 and a-tubulin. Data are presented as the mean 6 SEM (*P , 0.05 vs sham group, n 5 8). AAC, abdominal aortic constriction; CF, cardiac fibroblast; Ang II, angiotensin II; SIRT, sirtuin; SEM, standard error of the mean; mRNA, messenger RNA.

SIRT6-silenced cells. In contrast, overexpression of SIRT6 partially reversed the increase in a-SMA protein expression in cells treated with Ang II (Fig 3, C) or SIRT6 siRNA (Fig 3, D). Silencing of SIRT6 increased focal adhesions in CFs. Another morphologic hallmark of myofibroblasts

is focal adhesions. We therefore examined the impact of depletion of SIRT6 on paxillin and vinculin. Realtime PCR (Fig 4, A) and Western blot (Fig 4, B) analysis showed that SIRT6-silenced CFs displayed higher levels of mRNA and protein expressions of both genes, mimicking the effects of Ang II stimulation. Furthermore, our results also demonstrated a significant trend toward the increase in protein expression of both genes in the SIRT6 siRNA

plus Ang II-treated cells compared with SIRT6 siRNA and Ang II alone, suggesting SIRT6 depletion aggravates Ang II–induced CF phenotypic conversion. Silencing of SIRT6 activated NF-kB signaling. Previous studies have demonstrated that NF-kB activity is increased in multiple cardiovascular diseases and its signaling is critically implicated in the development of cardiac fibrosis.22-24 Kawahara et al25 found that SIRT6 deacetylates histone H3 lysine 9 (H3K9) and acts as a transcriptional corepressor of NF-kB. Therefore, we supposed that NF-kB signaling might be involved in SIRT6-mediated protection against cardiac fibrosis in CFs. To test this hypothesis, we first examined the effects of SIRT6 depletion on the activation of NF-kB. EMSA assay demonstrated that

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639

6

640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703

Translational Research - 2014

Tian et al

Fig 2. Depletion of SIRT6 induced CF proliferation and ECM deposition. (A and B) Morphology of CFs transfected with NC siRNA (A) or S6 siRNA (B), 3200 magnification. (C) Cell proliferation was measured by CCK-8 assay after 72 hours of transfection with NC siRNA or S6 siRNA under standard culture conditions (10% FCS). *P , 0.05 vs NC siRNA, n 5 5. (D) Collagen synthesis was determined by hydroxyproline assay in CFs after 72 hours of transfection with NC siRNA or S6 siRNA, *P , 0.05 vs NC siRNA, n 5 6. (E and F) Real-time polymerase chain reaction (E) and Western blot (F) analysis of mRNA and protein levels of PAI-1, collagen type I, and FN in CFs transfected with NC or S6 siRNA. Data are expressed as the mean 6 SEM, *P , 0.05 vs NC siRNA, n 5 6. CF, cardiac fibroblast; SIRT, sirtuin; SEM, standard error of the mean; NC, negative control; siRNA, small interfering RNA; ECM, extracellular matrix; CCK-8, Cell Counting Kit-8; PAI-1, plasminogen activator inhibitor 1; FN, fibronectin.

silencing of SIRT6 induced a significant increase in NF-kB DNA binding activity (Fig 5, A). We also examined the effect of SIRT6 on NF-kB–dependent

transcriptional activity by using dual-luciferase reporter gene assay. As shown in Fig 5, B, SIRT6 depletion led to increased NF-kB–luciferase reporter gene

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

Q17

704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767

Translational Research Volume -, Number -

7

web 4C=FPO

768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831

Tian et al

Fig 3. Depletion of SIRT6 induced CF differentiation into myofibroblasts. (A) Representative Western blot (upper panel) and quantitative analysis (lower panel) of a-SMA and SIRT6 protein expression in CFs transfected with NC siRNA or S6 siRNA for 48 hours in the presence or absence of Ang II (100 nM). Data are presented as the mean 6 SEM (*P , 0.05 vs NC siRNA, n 5 5). (B) Representative images of immunofluorescence staining for a-SMA (red) in CFs treated as mentioned previously. Nuclei were stained with DAPI (blue). Scale bar, 20 mm. (C) Representative Western blot (upper panel) and quantitative analysis (lower panel) of the protein levels of a-SMA and SIRT6 in CFs transfected with empty vector (EGFP-N3) or SIRT6 plasmid for 48 hours in the presence or absence of Ang II (100 nM). Data are presented as the mean 6 SEM (n 5 6). ***P , 0.001; #P , 0.05. (D) Representative Western blot (upper panel) and quantitative analysis (lower panel) of the protein levels of a-SMA and SIRT6 in CFs treated with S6 siRNA, with or without SIRT6 overexpression. Data are presented as the mean 6 SEM (n 5 6). ***P , 0.001; #P , 0.05. CF, cardiac fibroblast; SIRT, sirtuin; SEM, standard error of the mean; NC, negative control; a-SMA, a-smooth muscle actin; siRNA, small interfering RNA; Ang II, angiotensin II. For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.

activity, mimicking the effect of Ang II stimulation. On the contrary, SIRT6 overexpression repressed both basal and Ang II–induced NF-kB transcriptional activity. SIRT6 inhibited myofibroblast differentiation via inactivation of NF-kB–dependent transcriptional activity. To further investigate the potential role of

NF-kB in SIRT6-depleted CFs during myofibroblast transformation, CFs were transfected with p65 siRNA or treated with pyrrolidine dithiocarbamate (PDTC), a specific inhibitor of NF-kB activity.26 As shown in Fig 6, A and B, SIRT6 depletion increased the mRNA

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

Q18

832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895

8

896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959

Translational Research - 2014

Tian et al

Fig 4. Depletion of SIRT6 increased focal adhesions in CFs. (A and B) Real-time polymerase chain reaction (A) and Western blot (B) analysis of mRNA and protein levels of paxillin and vinculin in CFs transfected with NC siRNA or S6 siRNA for 48 hours in the presence or absence of Ang II (100 nM). Data are presented as the mean 6 SEM (*P , 0.05 vs NC siRNA, n 5 6). CF, cardiac fibroblast; SEM, standard error of the mean; NC, negative control; Ang II, angiotensin II; siRNA, small interfering RNA.

and protein expressions of a-SMA, which can be abrogated by p65 siRNA or PDTC. This finding was also confirmed by confocal microscopy (Fig 6, C). Similarly, the effects of SIRT6 depletion on paxillin (Fig 7, A) and vinculin (Fig 7, B) were also abolished by PDTC. Taken together, these results indicate that the effect of SIRT6 on myofibroblast transformation required the participation of NF-kB. DISCUSSION

Emerging evidence suggests that SIRTs are involved in the onset and progression of cardiovascular diseases.

For example, SIRT1 protects the heart from aging, stress, inflammation, and hypertrophy27,28; SIRT3 and SIRT7 also play important roles in the regulation of stress responses in the heart.29,30 We have previously showed that SIRT6 acts as a negative regulator of cardiac hypertrophy,16 indicating its potential cardioprotective role; however, whether SIRT6 plays a role in myofibroblast transformation and cardiac fibrosis is not clear. Here, we have provided the first evidence that SIRT6 negatively regulates CF differentiation into myofibroblasts. Additionally, our data show that SIRT6 depletion increases CF proliferation, ECM deposition, and upregulates focal adhesion-related genes

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023

Translational Research Volume -, Number 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087

Tian et al

9

Fig 5. Depletion of SIRT6 activated NF-kB signaling in CFs. (A) CFs were transfected with NC siRNA or S6 siRNA, or treated with Ang II (100 nM) or pyrrolidine dithiocarbamate (PDTC). ESMA assays were performed to evaluate the influence of SIRT6 depletion on DNA binding activity. (B) Dual-luciferase reporter assays were performed to evaluate the influence of SIRT6 depletion, overexpression, and/or Ang II (100 nM) treatment on NF-kB–dependent transcriptional activity. Data are presented as the mean 6 SEM (*P , 0.05 vs control, # P , 0.05 vs Ang II treatment, n 5 6). CF, cardiac fibroblast; SIRT, sirtuin; SEM, standard error of the mean; NF-kB, nuclear factor kB; NC, negative control; siRNA, small interfering RNA; Ang II, angiotensin II.

Q14

(paxillin and vinculin) and fibrosis-related genes (FN, collagen type I, and PAI-1). Mechanistically, we have documented a strong correlation between SIRT6 and NF-kB signaling in cardiac fibrotic response. In the first set of the experiments, we have revealed that SIRT6 is upregulated in Ang II–stimulated CFs and pressure overload–induced cardiac fibrosis. It seems paradoxical that SIRT6 is upregulated in the pressure-overloaded myocardium and inhibits fibroblast activation. In previous studies, Mao et al31 found that SIRT6 is upregulated in response to oxidative stress and promotes DNA repair; Minagawa et al32 also showed that SIRT6 expression is increased in idiopathic pulmonary fibrosis lung and inhibits senescence of human bronchial epithelial cells. They speculated that increased expression of SIRT6 might be an insufficient compensatory mechanism against stress-induced cellular senescence. Similarly, upregulation of SIRT6 in the pressure-overloaded myocardium might also be a compensatory mechanism against stress-induced fibroblast activation. Another potential explanation is that SIRT6 upregulation in the pressure-overloaded heart may act as a STOP signal limiting the fibrotic process. CF proliferation is vital for pathologic cardiac remodeling. In the present study, we showed a proproliferative phenotype of SIRT6-silenced CFs. Previous study has shown that depletion of SIRT6 reduces endothelial cell proliferation33; it was also demonstrated that depletion of SIRT6 had no significant effect on U2OS cell proliferation.34 Therefore, it appears that the effect of STRT6 on proliferation may be cell type-specific. One of the key features of cardiac fibrosis is the increased deposition of the ECM. The expression of

FN and collagen in CFs has been well established in de novo biosynthesis of ECM components and the fibrotic response. Consistent with this notion, our work showed that depletion of SIRT6 led to an increase in the mRNA and protein levels of the factor FN, collagen type I, and PAI-1. Furthermore, depletion of SIRT6 resulted in an increase in the content of hydroxyproline, the most specific amino acid in collagen and its change in concentration indicates a change in collagen levels.35 Taken together, these findings suggest that SIRT6 plays a prominent role in regulating collagen synthesis during pathologic cardiac remodeling. A major finding of this study is that SIRT6 is a negative regulator of myofibroblast differentiation, another key event in the cardiac remodeling processes. Myofibroblasts are highly active cells characterized by augmented expression of a-SMA, production of ECM components, and formation of focal adhesions. Upon injury, myofibroblast differentiation is induced by profibrotic factors such as Ang II/TGF-b and plays a key role in wound healing and remodeling processes.36,37 However, persistent myofibroblasts expedite scar formation and fibrosis, eventually leading to myocardial stiffness and impairment of cardiac function.38,39 Prevention of myofibroblast differentiation might, therefore, be a relevant approach to delay fibrosis in the heart. In the present study, we found that depletion of SIRT6 led to a significant increase in the mRNA and protein levels of a-SMA. Furthermore, overexpression of SIRT6 partially reversed the increase in a-SMA protein expression, in SIRT6-depleted or Ang II–treated CFs. In line with these findings, increased formation of focal adhesions occurred in SIRT6-depleted

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

Q19

Q20

1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151

1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215

Translational Research - 2014

Tian et al

web 4C=FPO

10

Fig 6. Depletion of SIRT6 induced myofibroblast differentiation by activating NF-kB. (A and B) Real-time polymerase chain reaction (A) and Western blot (B) analysis of mRNA and protein levels of a-SMA in SIRT6-depleted CFs treated with or without PDTC, or transfected with or without p65 siRNA. Data are presented as the mean 6 SEM (*P , 0.05 vs NC siRNA, #P , 0.05 vs S6 siRNA, n 5 6). (C) Representative images of immunofluorescent staining for a-SMA (red) in CFs treated as mentioned previously. Nuclei were stained with DAPI (blue). Scale bar, 20 mm. CF, cardiac fibroblast; SIRT, sirtuin; SEM, standard error of the mean; NF-kB, nuclear factor kB; a-SMA, a-smooth muscle actin; NC, negative control; siRNA, small interfering RNA; PDTC, pyrrolidine dithiocarbamate. For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.

CFs. Taken together, these data strongly suggested that SIRT6 plays a crucial role in the phenotypic transformation of CFs to myofibroblasts. Mechanistically, SIRT6 inhibits myofibroblast differentiation through its ability to suppress the transcriptional activity of NF-kB. NF-kB is a key regulator of inflammatory reaction. Activation of NF-KB signaling results in the synthesis and release of numbers of cytokine and chemotactic factors, which can induce the expression of a-SMA and other fibrosis-related genes, eventually contribute to myofibroblast differentiation and cardiac fibrosis.40 Accumulating evidence suggests that NF-kB is involved in the development of pathologic

cardiac remodeling. NF-kB activation leads to myocardial hypertrophy, myocardial fibrosis, and subsequent heart failure.22,23,41,42 In this study, we revealed that NF-kB DNA binding activity and transcriptional activity were increased in SIRT6-depleted CFs. In addition, overexpression of SIRT6 repressed both basal and Ang II–induced NF-kB transcriptional activity. Most importantly, inhibition of NF-kB signaling was sufficient to block myofibroblast differentiation in SIRT6-depleted CFs, suggesting that NF-kB signaling has a major role in this phenotypic transformation. SIRT6 is a nuclear histone deacetylase, and it is generally accepted that the biological functions of SIRT6

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

Q21

1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279

Translational Research Volume -, Number 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343

Tian et al

11

Fig 7. Depletion of SIRT6 increased focal adhesions in CFs by activating NF-kB. (A and B) Representative Western blot (upper panel) and quantitative analysis (lower panel) of protein expression of paxillin (A) and vinculin (B) in CFs treated with or without PDTC. Data are presented as the mean 6 SEM (*P , 0.05 vs NC siRNA, #P , 0.05 vs S6 siRNA, n 5 5). CF, cardiac fibroblast; SIRT, sirtuin; SEM, standard error of the mean; NF-kB, nuclear factor kB; NC, negative control; siRNA, small interfering RNA; PDTC, pyrrolidine dithiocarbamate.

have been linked to its deacetylase activity on H3K9 or H3K56.43,44 Therefore, SIRT6 might negatively regulate NF-kB–dependent transcription via histone deacetylation. In fact, Kawahara et al25 have shown a model in which SIRT6 was recruited by transcription factor NF-kB to the promoter regions of target genes and deacetylates H3K9, thereby suppressing the target gene expression and leading to NF-kB signal termination. Q15

Q16

CONCLUSIONS

Our study suggests that SIRT6 protects CFs from differentiation into myofibroblasts via inhibition of NF-kB–dependent transcriptional activation. Previous study has shown that SIRT6 is deficient in failing human hearts and in mouse hearts undergoing pressure overload.15 In this context, our findings that depletion of SIRT6 in CFs induces a pathologic phenotype (CF proliferation, ECM deposition, and myofibroblast differentiation) indicate that therapeutic approaches by increasing SIRT6 expression/activity may be a promising strategy for the prevention/treatment of cardiac fibrosis. ACKNOWLEDGMENTS

Conflicts of Interest: The authors declare no conflict of interest. All authors have read the journal’s policy on disclosure of potential conflicts of interest and the journal’s authorship agreement. The authors gratefully acknowledge financial support from the National Natural Science Foundation of China

(Nos. 81072641 and 81273499), the National Science and Technology Major Project of China ‘‘Key New Drug Creation and Manufacturing Program’’ (No. 2011ZX09401-307), Team Item of the Natural Science Foundation of Guangdong Province (No. S201103 0003190), Natural Science Foundation of Guangdong Province (No. S2013010016210), Major Project of Guangdong Province (Nos. 2008A030201013 and 2012A080201007), Major Project of the Department of Education of Guangdong Province (No. CXZ D1006), and the fostering discipline construction projects of Guangdong Pharmaceutical University. Supplementary Data

Supplementary data related to this article can be found at http://dx.doi.org/10.1016/j.trsl.2014.08.008.

REFERENCES

1. Swynghedauw B. Molecular mechanisms of myocardial remodeling. Physiol Rev 1999;79:215–62. 2. Weber KT. Are myocardial fibrosis and diastolic dysfunction reversible in hypertensive heart disease? Congest Heart Fail 2005;11:322–4. 3. Weber KT. Fibrosis in hypertensive heart disease: focus on cardiac fibroblasts. J Hypertens 2004;22:47–50. 4. Petrov VV, Fagard RH, Lijnen PJ. Stimulation of collagen production by transforming growth factor-beta1 during differentiation of cardiac fibroblasts to myofibroblasts. Hypertension 2002;39: 258–63. 5. Kawano H, Do YS, Kawano Y, et al. Angiotensin II has multiple profibrotic effects in human cardiac fibroblasts. Circulation 2000; 101:1130–7.

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402 1403 1404 1405 1406 1407

12

1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471

Translational Research - 2014

Tian et al

6. Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol 2002;3:349–63. 7. Powell DW, Mifflin RC, Valentich JD, Crowe SE, Saada JI, West AB. Myofibroblasts. I. Paracrine cells important in health and disease. Am J Physiol 1999;277:C1–9. 8. Manabe I, Shindo T, Nagai R. Gene expression in fibroblasts and fibrosis: involvement in cardiac hypertrophy. Circ Res 2002;91: 1103–13. 9. Finkel T, Deng CX, Mostoslavsky R. Recent progress in the biology and physiology of sirtuins. Nature 2009;460:587–91. 10. Nakagawa T, Guarente L. Sirtuins at a glance. J Cell Sci 2011; 124:833–8. 11. Olson ER, Naugle JE, Zhang X, Bomser JA, Meszaros JG. Inhibition of cardiac fibroblast proliferation and myofibroblast differentiation by resveratrol. Am J Physiol Heart Circ Physiol 2005;288: H1131–8. 12. Kanfi Y, Peshti V, Gil R, et al. SIRT6 protects against pathological damage caused by diet-induced obesity. Aging Cell 2010;9: 162–73. 13. Min L, Ji Y, Bakiri L, et al. Liver cancer initiation is controlled by AP-1 through SIRT6-dependent inhibition of survivin. Nat Cell Biol 2012;14:1203–11. 14. Kim HS, Xiao C, Wang RH, et al. Hepatic-specific disruption of SIRT6 in mice results in fatty liver formation due to enhanced glycolysis and triglyceride synthesis. Cell Metab 2010;12: 224–36. 15. Sundaresan NR, Vasudevan P, Zhong L, et al. The sirtuin SIRT6 blocks IGF-Akt signaling and development of cardiac hypertrophy by targeting c-Jun. Nat Med 2012;18:1643–50. 16. Yu SS, Cai Y, Ye JT, et al. Sirtuin 6 protects cardiomyocytes from hypertrophy in vitro via inhibition of NF-kappaB-dependent transcriptional activity. Br J Pharmacol 2013;168:117–28. 17. Xiao C, Wang RH, Lahusen TJ, et al. Progression of chronic liver inflammation and fibrosis driven by activation of c-JUN signaling in Sirt6 mutant mice. J Biol Chem 2012;287: 41903–13. 18. Teunissen BE, Smeets PJ, Willemsen PH, De Windt LJ, Van der Vusse GJ, Van Bilsen M. Activation of PPARdelta inhibits cardiac fibroblast proliferation and the transdifferentiation into myofibroblasts. Cardiovasc Res 2007;75:519–29. 19. Xu S, Liu Z, Huang Y, et al. Tanshinone II-A inhibits oxidized LDL-induced LOX-1 expression in macrophages by reducing intracellular superoxide radical generation and NF-kappaB activation. Transl Res 2012;160:114–24. 20. Wu X, Huang H, Tang F, Le K, Xu S, Liu P. Regulated expression of endothelial lipase in atherosclerosis. Mol Cell Endocrinol 2010;315:233–8. 21. Phrommintikul A, Tran L, Kompa A, et al. Effects of a Rho kinase inhibitor on pressure overload induced cardiac hypertrophy and associated diastolic dysfunction. Am J Physiol Heart Circ Physiol 2008;294:H1804–14. 22. Van der Heiden K, Cuhlmann S, Luong le A, Zakkar M, Evans PC. Role of nuclear factor kappaB in cardiovascular health and disease. Clin Sci (Lond) 2010;118:593–605. 23. Gordon JW, Shaw JA, Kirshenbaum LA. Multiple facets of NFkappaB in the heart: to be or not to NF-kappaB. Circ Res 2011; 108:1122–32. 24. Xu S, Zhi H, Hou X, Cohen RA, Jiang B. IkappaBbeta attenuates angiotensin II-induced cardiovascular inflammation and fibrosis in mice. Hypertension 2011;58:310–6.

25. Kawahara TL, Michishita E, Adler AS, et al. SIRT6 links histone H3 lysine 9 deacetylation to NF-kappaB-dependent gene expression and organismal life span. Cell 2009;136:62–74. 26. Mahmoodzadeh S, Fritschka S, Dworatzek E, et al. Nuclear factor-kappaB regulates estrogen receptor-alpha transcription in the human heart. J Biol Chem 2009;284:24705–14. 27. Alcendor RR, Gao S, Zhai P, et al. Sirt1 regulates aging and resistance to oxidative stress in the heart. Circ Res 2007;100: 1512–21. 28. Planavila A, Iglesias R, Giralt M, Villarroya F. Sirt1 acts in association with PPARalpha to protect the heart from hypertrophy, metabolic dysregulation, and inflammation. Cardiovasc Res 2011;90:276–84. 29. Sundaresan NR, Gupta M, Kim G, Rajamohan SB, Isbatan A, Gupta MP. Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J Clin Invest 2009;119:2758–71. 30. Vakhrusheva O, Smolka C, Gajawada P, et al. Sirt7 increases stress resistance of cardiomyocytes and prevents apoptosis and inflammatory cardiomyopathy in mice. Circ Res 2008;102: 703–10. 31. Mao Z, Hine C, Tian X, et al. SIRT6 promotes DNA repair under stress by activating PARP1. Science 2011;332:1443–6. 32. Minagawa S, Araya J, Numata T, et al. Accelerated epithelial cell senescence in IPF and the inhibitory role of SIRT6 in TGF-betainduced senescence of human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 2011;300:L391–401. 33. Cardus A, Uryga AK, Walters G, Erusalimsky JD. SIRT6 protects human endothelial cells from DNA damage, telomere dysfunction, and senescence. Cardiovasc Res 2013;97:571–9. 34. Kaidi A, Weinert BT, Choudhary C, Jackson SP. Human SIRT6 promotes DNA end resection through CtIP deacetylation. Science 2010;329:1348–53. 35. Wu G, Bazer FW, Burghardt RC, et al. Proline and hydroxyproline metabolism: implications for animal and human nutrition. Amino Acids 2011;40:1053–63. 36. Sun Y, Weber KT. Angiotensin converting enzyme and myofibroblasts during tissue repair in the rat heart. J Mol Cell Cardiol 1996; 28:851–8. 37. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature 2008;453:314–21. 38. Frangogiannis NG. Targeting the inflammatory response in healing myocardial infarcts. Curr Med Chem 2006;13:1877–93. 39. Sun Y, Weber KT. Infarct scar: a dynamic tissue. Cardiovasc Res 2000;46:250–6. 40. Hall G, Hasday JD, Rogers TB. Regulating the regulator: NF-kappaB signaling in heart. J Mol Cell Cardiol 2006;41: 580–91. 41. Purcell NH, Molkentin JD. Is nuclear factor kappaB an attractive therapeutic target for treating cardiac hypertrophy? Circulation 2003;108:638–40. 42. Zou J, Le K, Xu S, et al. Fenofibrate ameliorates cardiac hypertrophy by activation of peroxisome proliferator-activated receptoralpha partly via preventing p65-NFkappaB binding to NFATc4. Mol Cell Endocrinol 2013;370:103–12. 43. Michishita E, McCord RA, Berber E, et al. SIRT6 is a histone H3 lysine 9 deacetylase that modulates telomeric chromatin. Nature 2008;452:492–6. 44. Michishita E, McCord RA, Boxer LD, et al. Cell cycle-dependent deacetylation of telomeric histone H3 lysine K56 by human SIRT6. Cell Cycle 2009;8:2664–6.

FLA 5.2.0 DTD  TRSL851_proof  1 December 2014  5:36 pm  ce

1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500 1501 1502 1503 1504 1505 1506 1507 1508 1509 1510 1511 1512 1513 1514 1515 1516 1517 1518 1519 1520 1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535

Sirtuin-6 inhibits cardiac fibroblasts differentiation into myofibroblasts via inactivation of nuclear factor κB signaling.

Differentiation of cardiac fibroblasts (CFs) into myofibroblasts represents a key event in cardiac fibrosis that contributes to pathologic cardiac rem...
2MB Sizes 1 Downloads 10 Views