Increased Autophagy Markers Are Associated with Ductular Reaction during the Development of Cirrhosis

Increased Autophagy Markers Are Associated with Ductular Reaction during the Development of Cirrhosis

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 ...

7MB Sizes 0 Downloads 65 Views

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

The American Journal of Pathology, Vol. 185, No. 9, September 2015

ajp.amjpathol.org

EPITHELIAL AND MESENCHYMAL CELL BIOLOGY

Increased Autophagy Markers Are Associated with Ductular Reaction during the Development of Cirrhosis Q27

Tzu-Min Hung,*y Ray-Hwang Yuan,*z Wei-Pang Huang,x Yu-Hsuan Chen,{ Yu-Chun Lin,y Chih-Wen Lin,k Hong-Shiee Lai,* and Po-Huang Lee*,** From the Department of Surgery,* National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei; the Departments of Medical Researchy and Surgery,** E-DA Hospital, Kaohsiung; the Department of Integrated Diagnostics and Therapeutics,z National Taiwan University Hospital, Taipei; the Department of Life Science,x National Taiwan University, Taipei; the Institute of Molecular Medicine,{ National Taiwan University College of Medicine, Taipei; and the Division of Gastroenterology and Hepatology,k Department of Medicine, E-DA Hospital/I-Shou University, Kaohsiung, Taiwan Accepted for publication May 12, 2015.

Q4

Q6

Address correspondence to Po-Huang Lee, M.D., Ph.D., Department of Surgery, National Taiwan University Hospital, No. 7 Chung-Shan South Rd, Taipei, Taiwan. E-mail: pohuang1115@ntu. edu.tw.

Autophagy is a regulatory pathway in liver fibrosis. We investigated the roles of autophagy in human cirrhotic livers. Cirrhotic and noncirrhotic liver tissues were obtained from patients with hepatocellular carcinoma, and liver tissues from live donors served as control. Patients with cirrhotic livers had significantly increased levels of various essential autophagy-related genes compared with noncirrhotic livers. In addition, colocalization of autophagy marker microtubule-associated protein 1 light chain 3B (LC3B) with lysosome-associated membrane protein-1, increased levels of lysosome-associated membrane protein-2, and increased maturation of lysosomal cathepsin D were observed in cirrhotic livers. By using dual-immunofluorescence staining, we demonstrated that increased LC3B was located mainly in the cytokeratin 19elabeled ductular reaction (DR) in human cirrhotic livers and in an experimental cirrhosis induced by 2-acetylaminofluorene (AAF) with carbon tetrachloride (CCl4), indicating a conserved response to chronic liver damage. Furthermore, an AAF/CCl4-mediated increase in DR and fibrosis were attenuated after chloroquine treatment, suggesting that the autophagy-lysosome pathway was essential for AAF/CCl4induced DR-fibrosis. In conclusion, we demonstrated that increased autophagy marker positively correlated with DR during the development of cirrhosis. Therefore, targeting autophagy may hold therapeutic value for liver cirrhosis. (Am J Pathol 2015, 185: 1e14; http://dx.doi.org/10.1016/j.ajpath.2015.05.010)

Autophagy is an intracellular degradation process by which protein aggregates, damaged organelles, and invading microbes are delivered to the lysosome to maintain cellular homeostasis. Surrounding the cytosolic constituents, autophagosome is a double-membrane structure that fuses with a lysosome, forming an autolysosome for subsequent degradation in autophagy.1,2 Autophagy was originally discovered from liver experiments.3 Features and functions particular to the liver identify it as an organ in which autophagy potentially plays an important role.4 Furthermore, increasing evidence indicates that alteration of autophagy is the mechanism behind numerous liver diseases.4e6 Cirrhosis, a scarring response that enhances extracellular matrix accumulation after chronic injury, is the last stage of

Q5

liver fibrosis. It is generally believed that the activation of hepatic stellate cells (HSCs) is a key event in liver fibrogenesis.7 Recently, two different groups independently demonstrated that autophagy can regulate lipid droplets in HSCs and then drive HSC activation.8,9 HSC activation, both in vitro and in rodent liver injury models, is associated with features of autophagy induction, including a marked increase in autophagic vacuoles, LC3-II levels, and autophagic flux.8,9 The blocking of autophagy in cultured cells, with either 3-methyladenine or specific siRNAs to Atg5 or Q7 Supported in part by National Science Council research grants NSC-101- Q2 2314-B-650-005 (T.M.H.) and NSC-102-2314-B-650-007-MY3 (P.H.L.). Q3 Disclosures: None declared.

Copyright ª 2015 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ajpath.2015.05.010

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

63 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

125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186

Hung et al Table 1 Name

Characteristics of the Study Subjects Sex

Age, years

Noncirrhotic group N1 F 59 N2 M 49 Q24 N3 M 78 N4 M 40 N5 M 58 N6 M 63 N7 M 68 N8 M 64 N9 M 79 Cirrhotic groupy Q25 Q26 C1 M 70 C2 F 65 C3 M 53 C4 F 71 C5 F 74 C6 F 61 C7 M 67 C8 F 59 C9 F 55

Hepatitis

Pathological diagnosis*

No No HCV HCV HCV HCV HCV HCV HCV

Donor normal liver Donor normal liver Metavir score, 1 Metavir score, 0 Metavir score, 1 Metavir score, 2 Metavir score, 2 Metavir score, 1 Metavir score, 2

HCV HCV HCV HCV HCV HCV HCV HCV HCV

Grade Grade Grade Grade Grade Grade Grade Grade Grade

A A A A A A B B C

*Normal liver had neither serological nor histopathological evidence of liver disease. Metavir score indicates fibrosis from stages 0 to 4: 0 indicates absent, 1 indicates portal fibrosis, 2 indicates portal fibrosis with few septa, 3 indicates septal fibrosis, and 4 indicates cirrhosis. y Cirrhotic liver specimens were obtained from seven patients with hepatitis Cerelated cirrhosis concurrent with hepatocellular carcinoma (patients C1 to C7). These patients had Child-Pugh grade A, except patient C7. Two other cirrhotic patients were diagnosed with hepatitis Cerelated cirrhosis for liver transplantation and had Child-Pugh grades B and C (patients C8 and C9). F, female; M, male; HCV, hepatitis C virus.

Atg7, leads to attenuated HSC activation and fibrogenesis.8 More important, by using a mouse strain with the HSCspecific deletion of Atg7, Hernández-Gea et al8 demonstrated attenuated fibrosis after sustained liver injury by either carbon tetrachloride (CCl4) or thioacetamide. Although these two studies described the role of autophagy in promoting liver fibrosis, the autophagic function in human cirrhotic livers remains largely unknown and requires to be elucidated. Recently, considerable attention is being directed toward the role of epithelial components (particularly cholangiocytes) in liver fibrosis through the so-called ductular reaction (DR).10 The DR, which occurs at the periphery of portal tracts, is a common typical response to injury observed in human liver diseases.11 The DR can arise from a proliferation of preexisting bile ductular cells, from hepatic progenitor cells, or from the biliary metaplasia of hepatocytes. The term ductular describes that the containing cells exhibit a ductular phenotype. The term reaction is used to recognize that the epithelial component is accompanied by a complex of extracellular matrix, inflammatory cells, endothelial cells, and mesenchymal cells in the reactive lesions.11,12 Increasing evidence indicates a compelling association between the extent of the DR and the severity of fibrosis in patients with chronic

2

hepatitis C (CHC),13,14 alcoholic and nonalcoholic fatty liver diseases,15,16 and genetic hemochromatosis.17 Although the profibrosis effect of autophagy was performed in HSCs, an investigation of autophagy regulation in other liver fibrogenic cells is still lacking. Moreover, clinicopathologic studies to support the link between autophagy and liver fibrosis are insufficient. In this study, both measurements reflecting the number of autophagosomes and measurements of autophagic substrates were applied to patients with and without cirrhosis. We also checked the lysosomal function in these tissue samples. Rats that were treated with 2-acetylaminofluorene (AAF)/ CCL4 were also used as an animal model to confirm the findings in human samples and for a proof-of-concept intervention study.

Materials and Methods Human Liver Specimens The study design was reviewed and approved by the ethics committee of the National Taiwan University Hospital (Taipei, Taiwan; approval number 201212069RIND). Written informed consent was obtained from each subject. Fourteen liver tissues obtained from patients with CHC who underwent curative resection for hepatocellular carcinoma (HCC) were classified into two groups: cirrhotic groups (C-group, C1 to C7; seven cases) and noncirrhotic group (N-group, N3 to N9; seven cases), according to the pathological examination of the nontumorous region of HCC (Table 1). As controls, two normal liver tissues were ½T1 obtained from healthy living liver donors and included into the noncirrhotic group (N1 and N2), and two CHCrelated cirrhotic livers from patients receiving liver transplantation were included into the cirrhotic group (C8 and C9). Detailed pathological diagnoses of all 18 subjects are described in Table 1. All subjects were Q8 selected randomly, and none of them had received any therapy before surgery. For all subjects, a liver specimen was taken after surgery, immediately snap frozen with isopentane cooled by liquid nitrogen, and stored at 80 C until further use.

Animal Model of Liver Cirrhosis Male Sprague-Dawley rats (200 to 250 g body weight) were raised at 20 C to 22 C with a 12-hour light-dark cycle. All animal experimental procedures were approved by the Institute of Animal Care and Use Committees at E-DA Hospital (Kaohsiung, Taiwan). Liver fibrosis was induced by i.p. injection with 50% of CCL4 (diluted 1:1 in Q9 olive oil) twice a week for 6 consecutive weeks. To enhance fibrogenic response and increase DR expansion, AAF (0.02% in pellet form) was administered for 5 continuous days before the first CCL4 injection and every other day during the period of cirrhosis progression. We

ajp.amjpathol.org

-

The American Journal of Pathology

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248

print & web 4C=FPO

249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310

Autophagy Link for DR and Cirrhosis Q1

Figure 1

Analysis of autophagy levels in the liver tissues of patients with and without cirrhosis. A: Representative LC3B immunostaining on frozen sections. Note the accumulation of LC3B punctate in cirrhotic liver. B: Fluorescence intensities of LC3B were measured in all 18 subjects and quantified using AxioVision software version 4.8 (Zeiss, Jena, Germany). The numbers 6, 7, and 14 represent outlier subjects. The horizontal line inside the box indicates the median. Significantly increased LC3B levels are present in the cirrhotic group compared with the noncirrhotic group. C: Representative Western blot analysis depicts fibrosis and autophagic markers, including a-smooth muscle actin (a-SMA), LC3B, ATG5-12 complex, and ATG7 (b-actin used as loading control); the intensity of bands was quantitated using the ImageJ software version 1.49v (NIH, Bethesda, MD; http://imagej.nih.gov/ij), and normalization of a-SMA, LC3B, ATG5-12 complex, and ATG7 to b-actin is shown. D: Quantitative RT-PCR analysis of LC3B, ATG5, and ATG7 in liver tissues obtained from the noncirrhotic (N) and cirrhotic (C) patients. E: Representative Western blot analysis of a-SMA, LC3B, and ATG5-12 complex in paired N and C tissues of the same patient. The N specimens were obtained from the first operation for primary hepatocellular carcinoma (HCC) resection. The C specimens were obtained from the second operation for recurrent HCC resection. Relative intensity (RI) shown was calculated by normalization of the intensities of each marker to the loading control. Data are expressed as means  SD from at least three independent experiments (D). n Z 9 for each group (B and D). **P < 0.01, U test; yP < 0.05, yyP < 0.01, Student’s unpaired t-test. GAPDH, glyceraldehyde-3-phosphate dehydrogenase; P1, patient 1; P2, patient 2; P3, patient 3.

used a dose of 2 mL/kg for the initial CCL4 injection and 1 mL/kg for subsequent injection, as described in the study by Chobert et al.18 An additional group of untreated rats was used as a control and fed a standard chow. For measuring autophagic flux, 100 mg/kg chloroquine (CQ; Sigma, St. Louis, MO) was administered to rats via i.p. injection on the end of 6 weeks AAF/CCL4 treatment, and the animals were sacrificed 4 hours thereafter.19 Rats were

The American Journal of Pathology

-

injected with phosphate-buffered saline (PBS) as vehicle control. Two independent experiments were performed.

Dual-Immunofluorescence Staining Dual-immunofluorescence staining was used to determine the localization of LC3B with lysosome-associated membrane protein-1 (LAMP-1), a-smooth muscle actin (a-SMA), and

ajp.amjpathol.org

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

3

Q17

Q18

Q19

311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372

print & web 4C=FPO

373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434

Hung et al

Figure 2 Autophagic functions were assessed in the liver tissues of patients with and without cirrhosis. A: Representative Western blot analysis depicts lysosomal proteins and autophagic substrates, including lysosome-associated membrane protein-1 (LAMP-2), cathepsin D (CathD), p62, and ubiquitinated (Ub) proteins (b-actin used as loading control). Note the antibody of cathepsin D can detect both the precursor form (pre-CathD) and the mature form of CathD. The intensity of bands was quantitated using ImageJ software version 1.49v (NIH, Bethesda, MD; http://imagej.nih.gov/ij), and normalization of LAMP-2, pre-CathD, CathD, p62, and Ub to b-actin is shown. B: Dual-immunofluorescence staining of LC3B and LAMP-1 in cirrhotic livers. Yellow signals in the merged images indicate the colocalization of these two proteins. C: Quantitative RT-PCR analysis of p62 in liver tissues obtained from the noncirrhotic and cirrhotic patients. Data are expressed as the means  SD from at least three independent experiments. n Z 9 for each group. *P < 0.05 for noncirrhotic versus cirrhotic (Student’s unpaired ttest). GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Q10

Q11

cytokeratin (CK) 19. Cryosections from liver tissues (5 mm thick) were fixed in 4% paraformaldehyde in PBS for 10 minutes at 4 C, and subsequently permeabilized in PBS containing 0.5% (v/v) Triton X-100 (Sigma) for 15 minutes at room temperature. After incubation with the Image-iT FX signal enhancer (Life Technologies, Carlsbad, CA) and blocking with 20% normal bovine serum, the sections were incubated with the following primary antibodies: rabbit polyclonal anti-LC3B (dilution, 1:800; Sigma), mouse monoclonal antieLAMP-1 (dilution, 1:200; BD Bioscience, Franklin Lakes, NJ), antiea-SMA (dilution, 1:500; Sigma), or anti-CK19 (dilution, 1:100; Novocastra, Newcastle-upon-Tyne, UK) antibody at 4 C overnight. After washing, the sections were incubated with a cocktail of Alexa Fluor 488econjugated goat anti-rabbit IgG and Alexa Fluor 594econjugated goat anti-mouse IgG (dilution, 1:500; Life Technologies) at room temperature for 1 hour, mounted with Vectashield-DAPI (Vector Laboratories, Burlingame, CA), and observed with a Zeiss (Jena, Germany) LSM780

4

confocal microscope. The negative control condition used nonimmune rabbit IgG or mouse isotype IgG instead of the primary antibody.

Western Blot Analysis Frozen liver tissues were homogenized, and the total protein extracts were prepared as previously described.20 The membranes were probed with the following antibodies: polyclonal anti-LC3B (dilution, 1:4000; Sigma), monoclonal anti-ATG5 (dilution, 1:1000; Sigma), polyclonal antiATG7 (dilution, 1:4000; Sigma), polyclonal antieLAMP-2 (dilution, 1:2000; GeneTex, Irvine, CA), polyclonal anticathepsin D (dilution, 1:2000; GeneTex), polyclonal antip62/SQSTM1 (dilution, 1:4000; Sigma), monoclonal anti-ubiquitin (dilution, 1:1000; Sigma), and monoclonal antieb-actin (dilution, 1:10,000; Novus, Littleton, CO). After overnight incubation with each primary antibody, the

ajp.amjpathol.org

-

The American Journal of Pathology

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

Q20

Q21

435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496

in 0.1 mol/L PBS and were stored at 4 C until embedding. Tissue samples were then postfixed in 1% phosphatebuffered osmium tetroxide and embedded in Spurr’s resin. Ultrathin sections were stained with 0.2% lead citrate and 1% uranyl acetate. Images were acquired using a transmission electron microscope (model TEM-1400; Jeol, Tokyo, Japan).

print & web 4C=FPO

497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558

Autophagy Link for DR and Cirrhosis

Q22

Figure 3 LC3B does not co-express in a-smooth muscle actin (a-SMA)e positive myofibroblasts. Dual-immunofluorescence staining for LC3B (A) and aSMA (B) in a chronic hepatitis C patient with cirrhosis. Liver tissues were stained with a-SMA to identify myofibroblasts (including portal fibroblasts and activated hepatic stellate cells). C: Merged images show no colocalization of LC3B and a-SMA. D: Boxed areas in C. A closer look at the ductule structures.

membranes were incubated with horseradish peroxidasee conjugated secondary antibody. The proteins were then detected using an enhanced chemiluminescence detection system (Thermo Fisher Scientific Inc., Waltham, MA).

Isolation of Total RNA and Quantitative Real-Time PCR Analysis

Q12

The total RNA from specimens was extracted using TRIzol reagent (Life Technologies). Total RNA (1 mg) was reverse transcribed using random hexamer and Moloney Murine Leukemia Virus Reverse Transcriptase (Thermo Fisher Scientific Inc.). Subsequently, quantitative RT-PCR was performed using TaqMan Gene Expression Assays (Life Technologies). The assay identification numbers of the validated genes are as follows: Hs00797944 for LC3B, Hs00169468 for ATG5, Hs00197348 for ATG7, Hs00177654 for p62/SQSTM1, and Hs99999905 for glyceraldehyde-3-phosphate dehydrogenase. mRNA transcript levels were normalized to glyceraldehyde-3phosphate dehydrogenase mRNA levels (DCT). For profibrogenic gene expression, quantitative RT-PCR was performed using gene-specific primers for collagen type 1 a 1, a-SMA, and transforming growth factor-b1. The primer sequence was the same as our previous study.21

Transmission Electron Microscopy The specimens were excised and fixed with fixative buffer containing 2% paraformaldehyde and 2.5% glutaraldehyde

The American Journal of Pathology

-

559 560 561 562 563 564 565 566 567 568 Statistical Analysis 569 570 All statistical analyses were performed using SPSS version 571 Q13 16.0 statistical software package (SPSS, Chicago, IL). 572 Quantitative variables were presented as the means  SD. 573 Between-group comparisons between different groups were 574 Q14 performed using the U test or t-test. Pearson’s correlation 575 was used to determine the correlation coefficients between 576 the expression levels of LC3B and CK19 of liver tissues 577 in 18 patients. All tests were two sided, with a significant 578 P < 0.05. 579 580 581 Results 582 583 Increased Autophagy Markers in Human Cirrhotic 584 Livers 585 586 Immunofluorescence analysis showed a markedly increased 587 punctate LC3B staining in the cirrhotic livers when 588 compared with noncirrhotic livers (Figure 1A). After ½F1 589 quantification, we observed that the intensity of LC3B was 590 significantly higher in the C-group than in the N-group 591 (P < 0.01) (Figure 1B). 592 593 LC3B-II is a reliable protein marker for autophagosomes, 594 and its levels correlate with the number of autophagic 595 vesicles. To confirm that LC3B staining in cirrhotic livers 596 reflects an increase in the autophagosome-bound LC3B-II 597 rather than the cytoplasmic LC3B-I, we used Western blot 598 analysis to investigate the protein expression of LC3B. The 599 LC3B-II protein was expressed abundantly in the cirrhotic 600 livers and only moderately in noncirrhotic livers 601 (Figure 1C). Western blot analysis for other autophagic602 related genes was performed in parallel on all samples in 603 the two groups. The ATG5-12 complex and ATG7 protein 604 605 levels were significantly higher in the subjects from the C606 group than in the subjects from the N-group (Figure 1C). 607 The expression of a-SMA is used as a fibrogenesis marker. 608 To establish an overview of the autophagic process, 609 mRNA levels of the autophagy genes were also assessed. In 610 alignment with the protein results, the LC3B and ATG7 611 mRNA levels were significantly higher in the cirrhotic livers 612 than in the noncirrhotic controls (Figure 1D). To decrease the 613 variability between individual patients, three sets of surgical 614 specimens taken from first operation for the primary HCC 615 and secondary operation for recurrent HCC in the same pa616 617 tients were used to examine the association between auto618 phagy and cirrhosis. The LC3B-II and ATG5-12 complex 619 protein levels were greater in the cirrhotic tissues compared 620

ajp.amjpathol.org

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

5

print & web 4C=FPO

621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682

Hung et al

Figure 4

Increased autophagy markers in bile ductular lineage cells. A: LC3B expression in ductular reaction cells. Dual-immunofluorescence images for LC3B (green) and cytokeratin 19 (CK19; red) in three subjects show that CK19-labeled ductular cells express LC3B (merged images). BeD: Ultrastructural assessment of autophagic changes in the bile ductules of livers from a donor and a recipient. Electron micrographs illustrate autophagic vacuoles in the bile ductules from a healthy donor (B) and a cirrhotic recipient (C and D). Higher magnifications of the individual cholangiocyte shown below are enlarged micrographs from the respective boxed area. Arrows point to double-membrane autophagosomes. AVd, degradative autophagic vacuoles.

6

ajp.amjpathol.org

-

The American Journal of Pathology

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744

745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806

Autophagy Link for DR and Cirrhosis with the noncirrhotic tissues in patients 1 and 3 (Figure 1E). Together, these results demonstrated that increased autophagy markers were present in human cirrhotic livers.

Increased Autophagic Process Could Be Functional in Human Cirrhotic Livers Because autophagy is a lysosomal degradation mechanism, lysosomal functions were assessed in parallel in human samples. In cirrhotic livers, the protein levels of LAMP-2 were ½F2 greater than those in noncirrhotic livers (Figure 2A), suggesting an increase of lysosome number. We next investigated the maturation of the lysosomal hydrolase, cathepsin D. Synthesized as 46-kDa procathepsin D, cathepsin D is targeted to lysosomes and further cleaved into a mature enzyme comprising noncovalent 28- and 15-kDa polypeptides. By using an antibody that detects both 28-kDa and the precursor forms of cathepsin D, a decrease in the precursor form and an accompanying increase in the mature 28-kDa form of cathepsin D were observed in the cirrhotic livers, indicating increased lysosomal activity (Figure 2A). The clearance of autophagosomes occurs via fusion with lysosomes. We analyzed the subcellular localization of the lysosomal marker LAMP-1 and the autophagosomal marker LC3B using confocal microscopy. The punctate LC3B staining of cirrhotic livers was colocalized with LAMP-1, thereby providing evidence of the fusion of autophagosomal compartments with lysosome (Figure 2B). To examine the autophagic flux in another manner, we analyzed the levels of the LC3-binding protein p62, which is known to be degraded by autophagy and to act as an adaptor to convey ubiquitinated proteins to autophagosomes for lysosomal degradation.22 There was no significant difference in the protein levels of p62 between the noncirrhotic and cirrhotic livers (Figure 2A). However, a decrease in the ubiquitinated proteins was observed in the cirrhotic livers (Figure 2A). It has been reported that increases in the amount of p62 are seen in some situations where there is an increase in autophagic flux, and they might be explained by simultaneous transcriptional induction of the gene encoding p62.23,24 Quantitative RT-PCR analysis showed the p62 mRNA levels to be significantly higher in the cirrhotic livers than in the noncirrhotic controls (Figure 2C), further supporting this possibility.

Increased Autophagy Marker Correlates the Degree of DR and Fibrosis Severity We performed confocal colocalization studies to further identify which cells express abundant LC3B in cirrhotic livers. We initially speculated that the previously reported release of lipids through autophagy could be the mechanism underlying the association between the activation of HSCs and liver fibrogenesis.8 However, we did not observe LC3B to be colocalized with a-SMA, a marker of activated HSCs ½F3 (Figure 3). In contrast, LC3B staining was strongly positive

The American Journal of Pathology

-

807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 Correlation between Increased Autophagy Marker and 848 DR 849 850 In addition to CHC-related cirrhosis, other types of 851 cirrhosis, including chronic hepatitis B, alcoholic liver dis852 853 ease, and primary biliary cirrhosis, were also assessed 854 whether a similar pattern of protein expression would be 855 seen across a spectrum of liver disease. LC3B and CK19 856 colocalization was associated with bile ductular proliferation 857 in all other liver diseases (Figure 6). ½F6 858 Similar CK19-positive DR has also been described in the 859 18 rodent model ; we, therefore, studied whether increased DR 860 was accompanied by autophagic changes during the progres861 sion of experimental cirrhosis in rat. Sprague-Dawley rats 862 were fed with chronic AAF in combination with CCL4 863 administration for 6 weeks. At the end of the treatment, the 864 865 ratio of the liver weight/body weight, serum AST, and ALT 866 significantly increased in AAF/CCL4 rats compared with untreated rats (Figure 7A). On histological examination, the ½F7 867 868

in glandular, tubular structures that lack clear lumens (Figure 3); these structures were similar to the characteristics of DR.12 CK19 is a well-known marker for bile ductular lineage and can label the process of DR.25 Therefore, double staining with LC3B and CK19 was performed to determine whether the cells were positive for both markers. LC3B coexpressed with CK19 in linear, circular, and tubular structures, and the pattern of LC3B and CK19 staining had good consistency (Figure 4A). ½F4 In the next step, transmission electron microscopy was used to detect autophagy features specifically in proliferating bile ductules. Ultrastructurally, several autophagic vacuoles were observed in damaged small bile ducts (composed of four to eight cells) in the cirrhotic liver, whereas little vacuole formation was observed in the normal liver (Figure 4, BeD). Intact mitochondria can be observed in the cytoplasm of control cells; however, in cirrhotic cells, degradative autophagic vacuoles, which usually have only one limiting membrane and contain electron-dense amorphous material, were observed in the cytoplasm (Figure 4, BeD). These data demonstrated that bile ductular cells in cirrhosis sections clearly showed autophagic response elevation. Several studies indicate that the extent of DR closely correlates with the severity of fibrosis.13e17 Hence, we next compared the correlation between autophagy and the status of fibrosis. In noncirrhotic livers, LC3B plus CK19 dual staining was primarily expressed in the mature bile ducts (Figure 5, A and B), whereas in cirrhotic livers, reaction ½F5 products localized in the DR of portal areas, including bile duct, ductule structures, and cell clusters (Figure 5C). Moreover, a progressive increase in LC3B and CK19 expression was observed as the severity of fibrosis increased from the F1 stage to the F2 and F4 stages (Figure 5); the correlation between the expression levels of LC3B and CK19 was statistically significant (P < 0.001) (Figure 5E). Q15

ajp.amjpathol.org

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

7

print & web 4C=FPO

869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930

Hung et al

Increased autophagy marker correlates the degree of ductular reaction and fibrosis severity. AeC: Dual-immunofluorescence analysis showing the colocalization of LC3B (green) staining with cytokeratin 19 (CK19; red). The expression of both molecules increased from mild to severe fibrosis. Images are from representative patients with fibrosis stage F1 (A), stage F2 (B), and stage F4 (C). Insets: Higher magnifications of the bile duct (asterisks). D: Negative control counterstain (a consecutive section of the same cirrhotic tissues in which the primary antibodies were replaced with mouse and rabbit normal IgG). E: The fluorescence intensities of the LC3B and CK19 of 18 subjects were measured and used for a correlation analysis with one another. The correlation coefficients (R) and their respective significance levels were calculated using Pearson’s correlation analysis. P < 0.001.

Figure 5

livers of AAF/CCL4 rats exhibited marked fibrosis with portalcentral bridging (Figure 7B). In addition, CK19-positive ductular cells extended out into lobules, forming bridges that separate islands of parenchyma, in the livers of AAF/CCL4 rats (Figure 7C), supporting the clinical relevance of this animal model. By using dual-immunofluorescence staining, we observed that the CK19-positive cells were also positive for LC3B staining (Figure 7D), which was similar to that observed in humans.

8

In addition, we monitored the autophagic flux analyzing LC3B turnover assay using CQ, which can block autophagy degradation in the lysosome, thereby triggering the accumulation of autophagosome. We observed that in the presence of CQ, the LC3B-II levels induced by AAF/CCL4 were further increased compared with either AAF/CCL4 or CQ treatment alone (Figure 7E). Moreover, p62 levels were increased by CQ, suggesting that autophagic flux was increased during fibrogenesis in the AAF/CCL4 model.

ajp.amjpathol.org

-

The American Journal of Pathology

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992

print & web 4C=FPO

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 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

Autophagy Link for DR and Cirrhosis

Figure 6

Evidence for increased autophagy marker in ductular reaction in cirrhotic patients with different etiology. Liver sections were obtained from individuals with chronic hepatitis C (CHC), chronic hepatitis B (CHB), alcoholic liver disease (ALD), and primary biliary cirrhosis (PBC), and stained for autophagy marker LC3B and the ductular reaction marker cytokeratin 19 (CK19). All subjects were diagnosed as having liver cirrhosis for liver transplantation, and the cirrhotic specimens were taken during the operation. Dual immunofluorescence for LC3B (green) and CK19 (red) showed that CK19-labeled ductular cells co-express LC3B (yellow in merged images). Representative images of two experiments on liver sections from two patients in each disease etiology.

Our data showed that p62 levels did not decrease in the AAF/CCL4 rats despite autophagic degradation; this finding might be explained by aberrant accumulation of p62 in a detergent-insoluble fraction.23,24 Consistently, we observed that a substantial amount of p62 in a Triton X100einsoluble fraction accounts for the increase of p62 protein in AAF/CCL4 rats (Supplemental Figure S1). In contrast, AAF/CCL4-induced degradation of p62 was observed in a Triton X-100esoluble fraction, and this was rescued when autophagic degradation is inhibited by CQ.

Inhibited Autophagy Alleviated AAF/CCL4-Induced Liver Fibrosis To prove that autophagy is required for progression of DRfibrosis, we co-administered CQ in AAF/CCL4 rats. Treatment with 50 mg/kg per day CQ was initiated when rats were pretreated with AAF/CCL4 for 2 weeks, followed by 4 weeks of cotreatment.

The American Journal of Pathology

-

CQ treatment showed a decline of DR, as evidenced by immunohistochemistry and Western blot analysis for CK19 expression (Figure 8). Moreover, autophagy suppression ameliorated liver injury and decreased fibrosis induced by AAF/CCL4, as measured by Sirius red (Figure 8A) and by the levels of profibrogenic gene expression (Figure 8B) in the liver. However, combined CQ and AAF/CCL4 treatment did not further increase the endogenous LC3B-II levels (Figure 8C), although the lysosomal activity was suppressed, as evidenced by decreased expression levels and activity of cathepsin D (Supplemental Figure S2). Because our CQ was co-administered with injury in the early stage of liver fibrosis, we hypothesized that the ability of CQ to neutralize inflammation, which is a well-known indication used in a clinic,26 might counteract the AAF/CCL4-induced toxicity and, as a result, hinder the induction of autophagy. The mechanism by which CQ suppresses AAF/CCL4induced fibrosis remains to be further clarified.

ajp.amjpathol.org

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

9

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 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 ½F8 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116

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 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

print & web 4C/FPO

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

Hung et al

10

ajp.amjpathol.org

-

The American Journal of Pathology

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

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 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302

Autophagy Link for DR and Cirrhosis

Discussion The autophagy function in hepatic fibrosis has only been recently described,8,9 and whether increased autophagy results in human cirrhosis remains to be investigated. The research conducted herein is directed at understanding the autophagic functioning of the human cirrhotic liver. We demonstrated that autophagy markers were strongly up-regulated (on the basis of LC3B immunostaining, Western blot analysis, and transmission electron microscopy) in cirrhotic tissues compared with noncirrhotic tissues. The increased level of LC3B mRNA in cirrhotic livers further provided correlative data related to the induction of autophagy. We also verified that the increased number of autophagosomes in cirrhotic tissues was caused by an enhanced autophagic flux rather than by impaired autophagosome maturation. First, we verified the occurrence of the late steps of autophagolysosome formation in cirrhotic tissues by monitoring the colocalization of LC3B with LAMP-1. Second, we confirmed an increased lysosomal activity by examining the maturation of cathepsin D. Furthermore, apart from static measurements in patient samples, autophagic flux was also monitored by treating AAF/CCL4 rats with CQ. Increased levels of LC3B and p62 by CQ suggested that autophagic flux increased during fibrogenesis in the AAF/ CCL4-treated model. An increased number of autophagosomes in hepatic C virus (HCV)einfected cells and/or livers had been described previously.27,28 Autophagy was proposed to be nonfunctional in these reports because no decreases in the p62 protein were observed.29 However, another study demonstrated that a selective autophagy for lipids rather than a defective process is induced by HCV infection.30 By using lysosomal inhibitor approaches, the colocalization of LC3 and LAMP-1 and a tandem fluorescent-tagged LC3 to determine autophagic flux, Vescovo et al30 demonstrated that an HCV-induced autophagic process can advance to completion. It is now evident that autophagosomes can engulf a substrate selectively rather stochastically and that ubiquitinated proteins, ribosomes, mitochondria, peroxisomes, and intracellular bacteria may all be selectively targeted for autophagic degradation.31 Therefore, consistent with the previous reports,30 our results showed an absence of decreased level in p62 protein and increased autophagic markers in cirrhotic patients with HCV infection.

To further exclude the possibility that the observed autophagic changes were HCV specific, experiments were repeated by analyzing cirrhotic livers from patients with chronic hepatitis B, alcoholic liver disease, and primary biliary cirrhosis and from a rodent model of AAF/CCL4. Our dual-immunofluorescence analysis of cirrhotic livers localized LC3B to CK19-positive ductular cells, providing the first in vivo evidence, to our knowledge, that autophagy regulation could occur in other fibrogenic cells apart from HSCs. Consistently, Sasaki et al32 also reported recently that autophagy is frequently seen in bile ductular cells in primary biliary cirrhosis. Although our work fails to demonstrate increased LC3B punctate in a-SMAepositive HSCs (Figure 3 and Supplemental Figure S3), it is possible that a-SMAepositive HSCs undergoing autophagy activation were not detected in our experimental condition or that a-SMAenegative HSCs underwent autophagy activation. Further studies are required to clarify this point by tracking the behavior of both types of HSCs in the liver. Nevertheless, our data showed that CQ treatment, leading to decreased expression of a-SMA, supported a role for autophagy in HSC activation (Figure 8, B and C). There are two mechanisms proposed to demonstrate how DR promotes liver fibrosis.33 First, reactive ductular cells can secrete profibrogenic factors, which then activate matrix-producing cells. Numerous studies have demonstrated an intimate cross talk between ductular cells and a-SMAepositive myofibroblasts.18,34,35 In this regard, our data showed enrichment of a-SMAepositive cells in close proximity to the LC3B-positive ductule structures (Supplemental Figure S3), providing evidence that autophagy has a role in this paracrine interaction between DR and myofibroblast. The other mechanism is that cells that are involved in DR during chronic liver injury are capable of epithelial-mesenchymal transition (EMT),14,36e38 which is a novel mechanism considered to contribute to organ fibrosis.39 Svegliati-Baroni et al14 have demonstrated that the nuclear expression of Snail, down-regulation of E-cadherin, and expression of fibroblast-specific protein-1 by CK7-positive cells in HCV liver biopsy specimens provide clear evidence that these ductular cells actively engage in EMT. Rygiel et al37 also documented co-expression of epithelial and mesenchymal markers in DR cells in human tissues. Recently, a study found that starvation-induced autophagy could induce the expression of EMT markers

Figure 7 Increased autophagy marker correlates with ductular reaction in the rats with 2-acetylaminofluorene (AAF)/carbon tetrachloride (CCL4) administration. A: After 6 weeks of treatment, rats were sacrificed and the livers were removed to weight. Blood samples were collected to measure serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels by using enzyme-linked immunosorbent assay. B: Formalin-fixed, paraffinembedded liver tissues were divided into sections and followed by Sirius red staining. Extensive fibrous septae that separate hepatocyte into nodules are present in AAF/CCL4 liver. C: Cytokeratin 19 (CK19) immunofluorescence detection (red) on frozen liver sections. Positive signal was observed in bile duct of normal liver and ductular cells along with bridging fibrous septa of AAF/CCL4 livers. D: Dual-immunofluorescence staining of autophagy marker LC3B (green) combined with CK19 (red). Merged images reveal that LC3B is expressed in the bile duct of normal liver and the ductular cells of AAF/CCL4 livers. E: Evaluation of autophagic flux using lysosomal inhibitor chloroquine (CQ). Normal and AAF/CCL4-treated rats were given phosphate-buffered saline or CQ 4 hours before sacrifice. Levels of LC3B-II and p62 quantified by densitometry (normalized to the loading control and to the normal without CQ) are shown below the blot. Representative Western blot analysis is from three independent experiments. Data are expressed as the means  SEM from at least three independent experiments (A). n Z 3 rats in each group (A); n Z 2 to 4 rats for each experimental condition (E). *P < 0.05, Student’s unpaired t-test.

The American Journal of Pathology

-

ajp.amjpathol.org

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

11

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 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364

print & web 4C=FPO

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 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426

Hung et al

Figure 8 Chloroquine (CQ) treatment attenuates 2-acetylaminofluorene (AAF)/carbon tetrachloride (CCL4)-induced liver fibrosis. Treatment with 50 mg/kg per day CQ was initiated when rats were pretreated with AAF/CCL4 for 2 weeks, followed by 4 weeks of cotreatment. A: Normal, AAF/CCL4, and AAF/CCL4 þ CQ liver were stained with hematoxylin and eosin (H&E), Sirius red, and immunohistochemistry for cytokeratin 19 (CK19). Quantification of areas stained for Sirius red and serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels by using enzyme-linked immunosorbent assay. B: Expression of collagen type 1 a 1 (Col1A1), a-smooth muscle actin (a-SMA), and transforming growth factor (TGF)-b1 mRNA in liver tissues of normal, AAF/CCL4, and AAF/ CCL4 þ CQ rats. Statistical analysis was analyzed by Student’s unpaired t-test. C: Representative Western blot analysis depicts levels of CK19, a-SMA, LC3B, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in liver tissues of normal, AAF/CCL4, and AAF/CCL4 þ CQ rats. Relative intensity (RI) shown was calculated by normalization of the intensities of each marker to the loading control and to the number 1 of normal group. Each lane represents one rat. Data are means  SEM (A and B). n Z 4 to 6 rats per group (A and B).*P < 0.05 versus normal group; yP < 0.05 versus AAF/CCL4 group.

and invasion in hepatic carcinoma cells through a transforming growth factor-b/Smad3 signaling-dependent manner.40 Whether activation of autophagy may precede EMT of bile ductular cells in DR remains to be investigated. Herein, we found that LC3B staining was strongly positive in mature bile ducts in noncirrhotic human livers and normal rat liver (Figure 5, A and B, and Figure 7D), indicating that autophagy is required for normal biliary function. Autophagy can remove oxidatively damaged mitochondria; therefore, it may assist in cholangiocyte survival in damaged livers, where oxidative stresseinduced up-regulation of p21 inhibits hepatocyte proliferation.41 Selective growth advantage for cholangiocyte may serve as a permissive mechanism for an exaggerated DR. In support of this speculation, our data

12

indicated that autophagy was significantly associated with the expansion of the DR in liver cirrhosis. Thus, this study provides a new mechanism for explaining the preferential emergence of DR in many liver diseases. In conclusion, we provided the evidence of increased autophagy markers in clinical and AAF/CCL4-induced experimental cirrhotic livers. We also demonstrated that autophagy is significantly associated with the expansion of DR. Furthermore, to prove cause-effect, we used this AAF/ CCL4 rat model to validate that blocking autophagy by CQ inhibits the progression of DR and liver fibrosis. These findings suggest that pharmacological modulation of autophagy may be a potential therapeutic strategy for the management of liver cirrhosis.

ajp.amjpathol.org

-

The American Journal of Pathology

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

Q23

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 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 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547 1548 1549 1550

Autophagy Link for DR and Cirrhosis

Acknowledgment We thank the imaging core staff at the First Core Labs, National Taiwan University College of Medicine, for their technical assistance.

Supplemental Data Supplemental material for this article can be found at http://dx.doi.org/10.1016/j.ajpath.2015.05.010.

References 1. Yang Z, Klionsky DJ: Eaten alive: a history of macroautophagy. Nat Cell Biol 2010, 12:814e822 2. Choi AM, Ryter SW, Levine B: Autophagy in human health and disease. N Engl J Med 2013, 368:1845e1846 3. Deter RL, Baudhuin P, De Duve C: Participation of lysosomes in cellular autophagy induced in rat liver by glucagon. J Cell Biol 1967, 35:C11eC16 4. Czaja MJ, Ding WX, Donohue TM Jr, Friedman SL, Kim JS, Komatsu M, Lemasters JJ, Lemoine A, Lin JD, Ou JH, Perlmutter DH, Randall G, Ray RB, Tsung A, Yin XM: Functions of autophagy in normal and diseased liver. Autophagy 2013, 9:1131e1158 5. Rautou PE, Mansouri A, Lebrec D, Durand F, Valla D, Moreau R: Autophagy in liver diseases. J Hepatol 2010, 53:1123e1134 6. Ni HM, Williams JA, Yang H, Shi YH, Fan J, Ding WX: Targeting autophagy for the treatment of liver diseases. Pharmacol Res 2012, 66: 463e474 7. Lee UE, Friedman SL: Mechanisms of hepatic fibrogenesis. Best Pract Res Clin Gastroenterol 2011, 25:195e206 8. Hernández-Gea V, Ghiassi-Nejad Z, Rozenfeld R, Gordon R, Fiel MI, Yue Z, Czaja MJ, Friedman SL: Autophagy releases lipid that promotes fibrogenesis by activated hepatic stellate cells in mice and in human tissues. Gastroenterology 2012, 142:938e946 9. Thoen LF, Guimarães EL, Dollé L, Mannaerts I, Najimi M, Sokal E, van Grunsven LA: A role for autophagy during hepatic stellate cell activation. J Hepatol 2011, 55:1353e1360 10. Glaser SS, Gaudio E, Miller T, Alvaro D, Alpini G: Cholangiocyte proliferation and liver fibrosis. Expert Rev Mol Med 2009, 11:e7 11. Gouw ASH, Clouston AD, Theise ND: Ductular reactions in human liver: diversity at the interface. Hepatology 2011, 54:1853e1863 12. Roskams TA, Theise ND, Balabaud C, Bhagat G, Bhathal PS, BioulacSage P, Brunt EM, Crawford JM, Crosby HA, Desmet V, Finegold MJ, Geller SA, Gouw AS, Hytiroglou P, Knisely AS, Kojiro M, Lefkowitch JH, Nakanuma Y, Olynyk JK, Park YN, Portmann B, Saxena R, Scheuer PJ, Strain AJ, Thung SN, Wanless IR, West AB: Nomenclature of the finer branches of the biliary tree: canals, ductules, and ductular reactions in human livers. Hepatology 2004, 39: 1739e1745 13. Clouston AD, Powell EE, Walsh MJ, Richardson MM, Demetris AJ, Jonsson JR: Fibrosis correlates with a ductular reaction in hepatitis C: roles of impaired replication, progenitor cells and steatosis. Hepatology 2005, 41:809e818 14. Svegliati-Baroni G, Faraci G, Fabris L, Saccomanno S, Cadamuro M, Pierantonelli I, Trozzi L, Bugianesi E, Guido M, Strazzabosco M, Benedetti A, Marchesini G: Insulin resistance and necroinflammation drives ductular reaction and epithelial-mesenchymal transition in chronic hepatitis C. Gut 2011, 60:108e115 15. Roskams T, Yang SQ, Koteish A, Durnez A, DeVos R, Huang X, Achten R, Verslype C, Diehl AM: Oxidative stress and oval cell accumulation in mice and humans with alcoholic and nonalcoholic fatty liver disease. Am J Pathol 2003, 163:1301e1311

The American Journal of Pathology

-

16. Richardson MM, Jonsson JR, Powell EE, Brunt EM, NeuschwanderTetri BA, Bhathal PS, Dixon JB, Weltman MD, Tilg H, Moschen AR, Purdie DM, Demetris AJ, Clouston AD: Progressive fibrosis in nonalcoholic steatohepatitis: association with altered regeneration and a ductular reaction. Gastroenterology 2007, 133:80e90 17. Wood MJ, Gadd VL, Powell LW, Ramm GA, Clouston AD: Ductular reaction in hereditary hemochromatosis: the link between hepatocyte senescence and fibrosis progression. Hepatology 2014, 59:848e857 18. Chobert MN, Couchie D, Fourcot A, Chobert MN, Couchie D, Fourcot A, Zafrani ES, Laperche Y, Mavier P, Brouillet A: Liver precursor cells increase hepatic fibrosis induced by chronic carbon tetrachloride intoxication in rats. Lab Invest 2012, 92:135e150 19. Haspel J, Shaik RS, Ifedigbo E, Nakahira K, Dolinay T, Englert JA, Choi AM: Characterization of macroautophagic flux in vivo using a leupeptin-based assay. Autophagy 2011, 7:629e642 20. Hung TM, Hu RH, Ho CM, Chiu YL, Lee JL, Jeng YM, Shih DT, Lee PH: Downregulation of alpha-fetoprotein expression by LHX4: a critical role in hepatocarcinogenesis. Carcinogenesis 2011, 32: 1815e1823 21. Kao YH, Lin YC, Tsai MS, Sun CK, Yuan SS, Chang CY, Jawan B, Lee PH: Involvement of the nuclear high mobility group B1 peptides released from injured hepatocytes in murine hepatic fibrogenesis. Biochim Biophys Acta 2014, 1842:1720e1732 22. Kirkin V, McEwan DG, Novak I, Dikic I: A role for ubiquitin in selective autophagy. Mol Cell 2009, 34:259e269 23. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, AcevedoArozena A, Adeli K, et al: Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 2012, 8:445e544 24. Fujita K, Maeda D, Xiao Q, Srinivasula SM: Nrf2-mediated induction of p62 controls Toll-like receptor-4-driven aggresome-like induced structure formation and autophagic degradation. Proc Natl Acad Sci U S A 2011, 108:1427e1432 25. Zhou H, Rogler LE, Teperman L, Morgan G, Rogler CE: Identification of hepatocytic and bile ductular cell lineages and candidate stem cells in bipolar ductular reactions in cirrhotic human liver. Hepatology 2007, 45:716e724 26. Wolf R, Wolf D, Ruocco V: Antimalarials: unapproved uses or indications. Clin Dermatol 2000, 18:17e35 27. Taguwa S, Kambara H, Fujita N, Noda T, Yoshimori T, Koike K, Moriishi K, Matsuura Y: Dysfunction of autophagy participates in vacuole formation and cell death in cells replicating hepatitis C virus. J Virol 2011, 85:13185e13194 28. Rautou PE, Cazals-Hatem D, Feldmann G, Mansouri A, Grodet A, Barge S, Martinot-Peignoux M, Duces A, Bièche I, Lebrec D, Bedossa P, Paradis V, Marcellin P, Valla D, Asselah T, Moreau R: Changes in autophagic response in patients with chronic hepatitis C virus infection. Am J Pathol 2011, 178:2708e2715 29. Sir D, Chen WL, Choi J, Wakita T, Yen TS, Ou JH: Induction of incomplete autophagic response by hepatitis C virus via the unfolded protein response. Hepatology 2008, 48:1054e1061 30. Vescovo T, Romagnoli A, Perdomo AB, Corazzari M, Ciccosanti F, Alonzi T, Nardacci R, Ippolito G, Tripodi M, Garcia-Monzon C, Lo Iacono O, Piacentini M, Fimia GM: Autophagy protects cells from HCV-induced defects in lipid metabolism. Gastroenterology 2012, 142:644e653 31. Yu L, Strandberg L, Lenardo MJ: The selectivity of autophagy and its role in cell death and survival. Autophagy 2008, 4:567e573 32. Sasaki M, Miyakoshi M, Sato Y, Nakanuma Y: Autophagy may precede cellular senescence of bile ductular cells in ductular reaction in primary biliary cirrhosis. Dig Dis Sci 2012, 57:660e666 33. Svegliati-Baroni G, De Minicis S, Marzioni M: Hepatic fibrogenesis in response to chronic liver injury: novel insights on the role of cell-to-cell interaction and transition. Liver Int 2008, 28: 1052e1064 34. Lorenzini S, Bird TG, Boulter L, Bellamy C, Samuel K, Aucott R, Clayton E, Andreone P, Bernardi M, Golding M, Alison MR, Iredale JP, Forbes SJ: Characterisation of a stereotypical cellular and

ajp.amjpathol.org

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

13

1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593 1594 1595 1596 1597 1598 1599 1600 1601 1602 1603 1604 1605 1606 1607 1608 1609 1610 1611 1612

1613 1614 1615 1616 1617 1618 1619 1620 1621 1622 1623 1624 1625 1626 1627 1628 1629 1630 1631 1632 1633 1634 1635 1636 1637 1638 1639 1640 1641 1642 1643 1644 1645 1646 1647 1648 1649 1650 1651 1652 1653 1654 1655 1656 1657 1658 1659 1660 1661 1662 1663 1664 1665 1666 1667 1668 1669 1670 1671 1672 1673 1674

Hung et al

35.

36.

37.

38.

14

extracellular adult liver progenitor cell niche in rodents and diseased human liver. Gut 2010, 59:645e654 Van Hul NK, Abarca-Quinones J, Sempoux C, Horsmans Y, Leclercq IA: Relation between liver progenitor cell expansion and extracellular matrix deposition in a CDE-induced murine model of chronic liver injury. Hepatology 2009, 49:1625e1635 Robertson H, Kirby JA, Yip WW, Jones DE, Burt AD: Biliary epithelial-mesenchymal transition in posttransplantation recurrence of primary biliary cirrhosis. Hepatology 2007, 45:977e981 Rygiel KA, Robertson H, Marshall HL, Pekalski M, Zhao L, Booth TA, Jones DE, Burt AD, Kirby JA: Epithelial-mesenchymal transition contributes to portal tract fibrogenesis during human chronic liver disease. Lab Invest 2008, 88:112e123 Syn WK, Jung Y, Omenetti A, Abdelmalek M, Guy CD, Yang L, Wang J, Witek RP, Fearing CM, Pereira TA, Teaberry V, Choi SS,

Conde-Vancells J, Karaca GF, Diehl AM: Hedgehog-mediated epithelial-to-mesenchymal transition and fibrogenic repair in nonalcoholic fatty liver disease. Gastroenterology 2009, 137:1478e1488 39. Acloque H, Adams MS, Fishwick K, Bronner-Fraser M, Nieto MA: Epithelial-mesenchymal transitions: the importance of changing cell state in development and disease. J Clin Invest 2009, 119: 1438e1449 40. Li J, Yang B, Zhou Q, Wu Y, Shang D, Guo Y, Song Z, Zheng Q, Xiong J: Autophagy promotes hepatocellular carcinoma cell invasion through activation of epithelial-mesenchymal transition. Carcinogenesis 2013, 34:1343e1351 41. Lunz JG 3rd, Tsuji H, Nozaki I, Murase N, Demetris AJ: An inhibitor of cyclin-dependent kinase, stress-induced p21Waf-1/Cip-1, mediates hepatocyte mito-inhibition during the evolution of cirrhosis. Hepatology 2005, 41:1262e1271

ajp.amjpathol.org

-

The American Journal of Pathology

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

1675 1676 1677 1678 1679 1680 1681 1682 1683 1684 1685 1686 1687 1688 1689 1690 1691 1692 1693 1694 1695 1696 1697 1698 1699 1700 1701 1702 1703 1704 1705 1706 1707 1708 1709 1710 1711 1712 1713 1714 1715 1716 1717 1718 1719 1720 1721 1722 1723 1724 1725 1726 1727 1728 1729 1730 1731 1732 1733 1734 1735 1736

1737 1738 1739 1740 1741 1742 1743 1744 1745 1746 1747 1748 1749 1750 1751 1752

Supplemental Figure S1 Changes in 2-acetylaminofluorene (AAF)/carbon tetrachloride (CCL4)einduced p62 expression by chloroquine (CQ). Normal and AAF/CCL4 rats were given phosphate-buffered saline (PBS) or CQ 4 hours before sacrifice. Liver tissues homogenized with PBS containing 1% Triton X-100 were separated into supernatant (Triton-soluble) and pellet (Triton-insoluble) fractions after centrifugation. Representative Western blots are shown. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Q16

Supplemental Figure S2 Impact of chloroquine (CQ) on lysosomal activities. A: Treatment with 50 mg/kg per day CQ was initiated when rats were pretreated with 2-acetylaminofluorene (AAF)/carbon tetrachloride (CCL4) for 2 weeks, followed by 4 weeks of cotreatment. Total liver lysates were subjected to Western blot analysis for examining level of cathepsin D (CathD), followed by densitometry analysis. B: Rats were treated as for A, and liver lysates were prepared and analyzed for the cathepsin D activities by Student’s unpaired t-test. Data are means  SEM. n Z 4 to 6 rats per group. *P < 0.05 versus normal group; yP < 0.05 versus AAF/CCL4 group. GAPDH, glyceraldehyde-3-phosphate dehydrogenase. LC3B-positive ductular structures were intimately surrounded by a-smooth muscle actin (a-SMA)epositive myofibroblasts. A: a-SMAepositive cells are observed in close proximity to LC3B-positive ductular structures in cirrhotic livers of different etiology. Note no yellow signals in merged images. Representative images of two experiments on liver sections from two patients in each disease etiology. B: LC3B does not co-express in aSMAepositive cells in 2-acetylaminofluorene (AAF)/carbon tetrachloride (CCL4)-treated livers. Representative images of two experiments on liver sections from two rats. ALD, alcoholic liver disease; CHB, chronic hepatitis B; CHC, chronic hepatitis C; PBC, primary biliary cirrhosis.

Supplemental Figure S3

FLA 5.2.0 DTD  AJPA2092_proof  25 July 2015  7:11 pm  EO: AJP14_0736

1753 1754 1755 1756 1757 1758 1759 1760 1761 1762 1763 1764 1765 1766 1767 1768