Kinetics and protective role of autophagy in a mouse cecal ligation and puncture-induced sepsis

Waka Takahashi, Eizo Watanabe, Lisa Fujimura, Haruko Watanabe-Takano, Hiroyuki Yoshidome, Paul E Swanson, Takeshi Tokuhisa, Shigeto Oda, Masahiko Hatano, Waka Takahashi, Eizo Watanabe, Lisa Fujimura, Haruko Watanabe-Takano, Hiroyuki Yoshidome, Paul E Swanson, Takeshi Tokuhisa, Shigeto Oda, Masahiko Hatano

Abstract

Introduction: It is not well understood whether the process of autophagy is accelerated or blocked in sepsis, and whether it is beneficial or harmful to the immune defense mechanism over a time course during sepsis. Our aim was to determine both the kinetics and the role of autophagy in sepsis.

Methods: We examined autophagosome and autolysosome formation in a cecal ligation and puncture (CLP) mouse model of sepsis (in C57BL/6N mice and GFP-LC3 transgenic mice), using western blotting, immunofluorescence, and electron microscopy. We also investigated the effect of chloroquine inhibition of autophagy on these processes.

Results: Autophagy, as demonstrated by increased LC3-II/LC3-I ratios, is induced in the liver, heart, and spleen over 24 h after CLP. In the liver, autophagosome formation peaks at 6 h and declines by 24 h. Immunofluorescent localization of GFP-LC3 dots (alone and with lysosome-associated membrane protein type 1 (LAMP1)), as well as electron microscopic examination, demonstrate that both autophagosomes and autolysosomes are increased after CLP, suggesting that intact autophagy mechanisms operate in the liver in this model. Furthermore, inhibition of autophagy process by chloroquine administration immediately after CLP resulted in elevated serum transaminase levels and a significant increase in mortality.

Conclusions: All autophagy-related processes are properly activated in the liver in a mouse model of sepsis; autophagy appears to play a protective role in septic animals.

Figures

Figure 1
Figure 1
Cecal ligation and puncture (CLP) induces autophagy in the liver of CLP model mice. For B-D and F, data are expressed as mean ± SD; data were analyzed for statistical significance using the Mann-Whitney test. (A) Western blotting analysis of microtubule-associated protein light chain 3 (LC3) in the liver. Sepsis was induced by CLP. Liver samples were prepared from sham-operated and CLP mice at each indicated time point post surgery. Results are representative of seven independent experiments. β-tubulin served as standard. (B) The ratio between the levels of LC3-II and LC3-I at each time point. *P <0.05, as compared to sham-operated group (n = 7/group). (C) The ratio of LC3-II/LC3-I expression in various organs at 6 and 24 h after CLP. Values in CLP mice are expressed as fold-change relative to each sham-operated group. MLN, mesenteric lymph node. (D)LC3 mRNA expression in the liver at each time point assessed by real-time PCR. *P <0.05, as compared to sham-operated group (n = 7/group). (E) Confocal images of liver samples obtained from GFP-LC3 transgenic mice. Green-fluorescing GFP-LC3 dots were present in the cytosol indicated by the arrow head. (F) The number of GFP-LC3 dots per cellular confocal image was quantified. Differences in the number of GFP-LC3 dots in CLP mice at 6 h compared with sham-operated mice at 6 h and CLP mice at 24 h were statistically significant (*P <0.05; n = 50 cells /animal; n = 4 animals).
Figure 2
Figure 2
Co-localization of green fluorescent protein (GFP)-Microtubule-associated protein light chain 3 (LC3) dots with lysosome-associated membrane protein type 1 (LAMP1) in the liver after cecal ligation and puncture (CLP). (A) Confocal images of liver samples obtained from GFP-LC3 transgenic mice. Green-fluorescing GFP-LC3 dots were present in the cytosol. LAMP1 was stained using Cy3-conjugated IgG secondary antibodies. Merged images demonstrate co-localization of GFP-LC3 dots and LAMP1. (B) The number of GFP-LC3 or GFP-LC3 + LAMP1 dots per cellular confocal image was quantified at 6 and 24 h after CLP. All data are expressed as mean ± SD. Data were analyzed for statistical significance using the Mann-Whitney test. Differences in the number of GFP-LC3 or GFP-LC3 dots in CLP mice at 6 h compared with CLP mice at 24 h were statistically significant (*P <0.05; n = 50 cells /animal; n = 4 animals). (C) Relative expression of p62 protein in the liver at 6 and 24 h after sham or CLP operation. The amount of p62 protein was normalized to that of β-tubulin by evaluation of band intensity from western blotting. All data were expressed as the mean ± SD. Data were analyzed for statistical significance using the Mann-Whitney test (*P <0.05; n = 5 in each group).
Figure 3
Figure 3
Electron microscopic analysis of the liver. (A) The number of autophagosomes and autolysosomes are compared in CLP and sham animals. All data are expressed as the mean ± SD. Data were analyzed for statistical significance using the Mann-Whitney test. Increase in autolysosomes in the CLP group was statistically significant (*P <0.05; n = 3); mean increases in autophagosomes in CLP compared to sham did not reach statistical significance. NS, not significant. (B) Images of electron microscopy of the liver; a: Liver sample obtained from sham-operated mice. Organelles in the hepatocyte are generally intact and lysosomes do not contain discrete membrane structures, although the inhomogeneous electron-dense material often seen in (hetero)lyosomes most certainly represent end-stage degradation of phospholipid and other cytoplasmic materials (a material at the light microscopic level referred to as lipofuscin); b-e: CLP-operated mice. Double arrow heads identify complex structures bounded by two membranes (autophagosomes); arrow heads identify single membrane-bound lysosomal complexes with degraded organellar content (autolysosomes); e: the double arrow head identifies an autophagosome that clearly contains an injured mitochondrion.
Figure 4
Figure 4
Blockade of the autophagic process by chloroquine. Green fluorescent protein- microtubule-associated protein light chain 3 (GFP-LC3) transgenic mice were administered saline or chloroquine (CQ) (60 mg/kg intraperitoneally) at 1 h after CLP or sham surgery. Samples were obtained from 6 or 24 h after surgery. (A) Confocal images of liver samples obtained from GFP-LC3 transgenic mice with or without CQ treatment. Blockade of autophagic process is indicated by the arrow head. (B) The number of GFP-LC3 or GFP-LC3 + lysosome-associated membrane protein type 1 (LAMP1) dots per cellular confocal image obtained from GFP-LC3 transgenic mice with or without CQ treatment was quantified at 6 and 24 h after cecal ligation and puncture (CLP). All data are expressed as the mean ± SD. Data were analyzed for statistical significance using the Student-Newman-Keuls test. Differences in the number of GFP-LC3 or GFP-LC3 + LAMP1 dots in CLP mice at 24 h compared with CLP mice at 24 h were statistically significant (*P <0.05; n = 50 cells /animal; n = 4 animals).
Figure 5
Figure 5
Inhibition of autophagy enhances cecal ligation and puncture (CLP)-induced liver injury. (A) Histological findings of mouse liver by hematoxylin and eosin staining (original magnification 100×) with a larger view (insets, 400×). Liver tissue was obtained from either sham-operated or CLP mice with or without chloroquine (CQ) treatment 6 h after surgery. (B) Hepatocellular damage as defined by serum aspartate aminotransferase (AST) and arginin aminotransferase (ALT) levels. Samples were obtained from either sham-operated or CLP mice with or without CQ treatment 6 and 24 h after surgery (n = 7/group). Data are shown as mean ± SD. *P <0.05 versus saline treatment (one-way analysis of variance); individual group means were compared using the Student-Newman-Keuls test. (C) Survival after CQ or sterile saline administration (n = 18/group) in sham-operated or CLP mice; 3/18 mice in the CLP + saline group and 12/18 mice in the CLP + CQ group were sacrificed because they were moribund. Comparisons of overall survival were performed using the log-rank test (P = 0.003; CLP + saline versus CLP + CQ).

References

    1. Russell JA. Management of sepsis. N Engl J Med. 2006;17:1699–1713. doi: 10.1056/NEJMra043632.
    1. Kumar G, Kumar N, Taneja A, Kaleekal T, Tarima S, McGinley E, Jimenez E, Mohan A, Khan RA, Whittle J, Jacobs E, Nanchal R. Milwaukee Initiative in Critical Care Outcomes Research Group of I: Nationwide trends of severe sepsis in the 21st century (2000–2007) Chest. 2011;17:1223–1231. doi: 10.1378/chest.11-0352.
    1. Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis. 2013;17:260–268. doi: 10.1016/S1473-3099(13)70001-X.
    1. Hotchkiss RS, Strasser A, McDunn JE, Swanson PE. Cell death. N Engl J Med. 2009;17:1570–1583. doi: 10.1056/NEJMra0901217.
    1. Levine B, Klionsky DJ. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell. 2004;17:463–477. doi: 10.1016/S1534-5807(04)00099-1.
    1. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;17:27–42. doi: 10.1016/j.cell.2007.12.018.
    1. Mizushima N, Ohsumi Y, Yoshimori T. Autophagosome formation in mammalian cells. Cell Struct Funct. 2002;17:421–429. doi: 10.1247/csf.27.421.
    1. Levine B, Mizushima N, Virgin HW. Autophagy in immunity and inflammation. Nature. 2011;17:323–335. doi: 10.1038/nature09782.
    1. Mizushima N, Levine B. Autophagy in mammalian development and differentiation. Nat Cell Biol. 2010;17:823–830. doi: 10.1038/ncb0910-823.
    1. Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H, Yoshimori T, Ohsumi Y, Tokuhisa T, Mizushima N. The role of autophagy during the early neonatal starvation period. Nature. 2004;17:1032–1036. doi: 10.1038/nature03029.
    1. Miller BC, Zhao Z, Stephenson LM, Cadwell K, Pua HH, Lee HK, Mizushima NN, Iwasaki A, He YW, Swat W, Virgin HW. The autophagy gene ATG5 plays an essential role in B lymphocyte development. Autophagy. 2008;17:309–314.
    1. Mortensen M, Ferguson DJ, Edelmann M, Kessler B, Morten KJ, Komatsu M, Simon AK. Loss of autophagy in erythroid cells leads to defective removal of mitochondria and severe anemia in vivo. Proc Natl Acad Sci USA. 2010;17:832–837. doi: 10.1073/pnas.0913170107.
    1. Shibata M, Yoshimura K, Tamura H, Ueno T, Nishimura T, Inoue T, Sasaki M, Koike M, Arai H, Kominami E, Uchiyama. LC3, a microtubule-associated protein1A/B light chain3, is involved in cytoplasmic lipid droplet formation. Biochem Biophys Res Commun. 2010;17:274–279. doi: 10.1016/j.bbrc.2010.01.121.
    1. Goldberg AL. Protein degradation and protection against misfolded or damaged proteins. Nature. 2003;17:895–899. doi: 10.1038/nature02263.
    1. Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, Yokoyama M, Mishima K, Saito I, Okano H, Mizushima N. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature. 2006;17:885–889. doi: 10.1038/nature04724.
    1. Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, Ueno T, Koike M, Uchiyama Y, Kominami E, Tanaka K. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature. 2006;17:880–884. doi: 10.1038/nature04723.
    1. Crotzer VL, Blum JS. Autophagy and adaptive immunity. Immunology. 2010;17:9–17.
    1. Nakagawa I, Amano A, Mizushima N, Yamamoto A, Yamaguchi H, Kamimoto T, Nara A, Funao J, Nakata M, Tsuda K, Hamada S, Yoshimori T. Autophagy defends cells against invading group A Streptococcus. Science. 2004;17:1037–1040. doi: 10.1126/science.1103966.
    1. Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature. 2008;17:1069–1075. doi: 10.1038/nature06639.
    1. Hsieh CH, Pai PY, Hsueh HW, Yuan SS, Hsieh YC. Complete induction of autophagy is essential for cardioprotection in sepsis. Ann Surg. 2011;17:1190–1200. doi: 10.1097/SLA.0b013e318214b67e.
    1. Carchman EH, Rao J, Loughran PA, Rosengart MR, Zuckerbraun BS. Heme oxygenase-1-mediated autophagy protects against hepatocyte cell death and hepatic injury from infection/sepsis in mice. Hepatology. 2011;17:2053–2062. doi: 10.1002/hep.24324.
    1. Chien WS, Chen YH, Chiang PC, Hsiao HW, Chuang SM, Lue SI, Hsu C. Suppression of autophagy in rat liver at late stage of polymicrobial sepsis. Shock. 2011;17:506–511. doi: 10.1097/SHK.0b013e31820b2f05.
    1. Mofarrahi M, Sigala I, Guo Y, Godin R, Davis EC, Petrof B, Sandri M, Burelle Y, Hussain SN. Autophagy and skeletal muscles in sepsis. PLoS One. 2012;17:e47265. doi: 10.1371/journal.pone.0047265.
    1. Lo S, Yuan SS, Hsu C, Cheng YJ, Chang YF, Hsueh HW, Lee PH, Hsieh YC. Lc3 transgene improves survival and attenuates lung injury through increasing autophagosomal clearance in septic mice. Ann Surg. 2013;17:352–363. doi: 10.1097/SLA.0b013e318269d0e2.
    1. Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, Fitzgerald KA, Ryter SW, Choi A. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011;17:222–230.
    1. Gunst J, Derese I, Aertgeerts A, Ververs EJ, Wauters A, Van den Berghe G, Vanhorebeek I. Insufficient autophagy contributes to mitochondrial dysfunction, organ failure, and adverse outcome in an animal model of critical illness. Crit Care Med. 2013;17:182–194. doi: 10.1097/CCM.0b013e31828e928d.
    1. Crouser ED, Julian MW, Huff JE, Struck J, Cook CH. Carbamoyl phosphate synthase-1: A marker of mitochondrial damage and depletion in the liver during sepsis. Crit Care Med. 2006;17:2439–2446.
    1. Watanabe E, Muenzer JT, Hawkins WG, Davis CG, Dixon DJ, McDunn JE, Brackett DJ, Lerner MR, Swanson PE, Hotchkiss RS. Sepsis induces extensive autophagic vacuolization in hepatocytes: a clinical and laboratory-based study. Lab Invest. 2009;17:549–561. doi: 10.1038/labinvest.2009.8.
    1. Hubbard WJ, Choudhry M, Schwacha MG, Kerby JD, Rue LW 3rd, Bland KI, Chaudry IH. Cecal ligation and puncture. Shock. 2005;17:52–57. doi: 10.1097/01.shk.0000191414.94461.7e.
    1. Rittirsch D, Huber-Lang MS, Flierl MA, Ward PA. Immunodesign of experimental sepsis by cecal ligation and puncture. Nat Protoc. 2009;17:31–36.
    1. Ding WX, Li M, Chen X, Ni HM, Lin CW, Gao W, Lu B, Stolz DB, Clemens DL, Yin XM. Autophagy reduces acute ethanol-induced hepatotoxicity and steatosis in mice. Gastroenterology. 2010;17:1740–1752. doi: 10.1053/j.gastro.2010.07.041.
    1. Morrison TB, Weis JJ, Wittwer CT. Quantification of low-copy transcripts by continuous SYBR Green I monitoring during amplification. Biotechniques. 1998;17:954–958. 960.
    1. Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell. 2004;17:1101–1111.
    1. Mizushima N, Yoshimoto T, Levine B. Methods in mammalian autophagy research. Cell. 2010;17:313–326. doi: 10.1016/j.cell.2010.01.028.
    1. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K, Agholme L, Agnello M, Agostinis P, Aguirre-Ghiso JA, Ahn HJ, Ait-Mohamed O, Ait-Si-Ali S, Akematsu T, Akira S, Al-Younes HM, Al-Zeer MA, Albert ML, Albin RL, Alegre-Abarrategui J, Aleo MF, Alirezaei M, Almasan A, Almonte-Becerril M, Amano A, Amaravadi R, Amarnath S, Amer AO, Andrieu-Abadie N, Anantharam V. et al.Guidlines for the use and interpretation of assays for monitoring autophagy. Autophagy. 2012;17:445–544. doi: 10.4161/auto.19496.
    1. Iwai-Kanai E, Yuan H, Huang C, Sayen MR, Perry-Garza CN, Kim L, Gottlieb RA. A method to measure cardiac autophagic flux in vivo. Autophagy. 2008;17:322–329.
    1. Ni H-M, Bockus A, Wozniak AL, Jones K, Weinman S, Yin X-M, Ding W-X. Dissecting the dynamic turnover of gfp-lc3 in the autolysosome. Autophagy. 2011;17:188–204. doi: 10.4161/auto.7.2.14181.
    1. Hsiao HW, Tsai KL, Wang LF, Chen YH, Chiang PC, Chuang SM, Hsu C. The decline of autophagy contributes to proximal tubular dysfunction during sepsis. Shock. 2012;17:289–296. doi: 10.1097/SHK.0b013e318240b52a.
    1. Ni HM, Bockus A, Boggess N, Jaeschke H, Ding WX. Activation of autophagy protects against acetaminophen-induced hepatotoxicity. Hepatology. 2012;17:222–232. doi: 10.1002/hep.24690.
    1. Wang Y, Singh R, Xiang Y, Czaja MJ. Macroautophagy and chaperone-mediated autophagy are required for hepatocyte resistance to oxidant stress. Hepatology. 2010;17:266–277. doi: 10.1002/hep.23645.
    1. Komatsu M, Waguri S, Koike M, Sou YS, Ueno T, Hara T, Mizushima N, Iwata J, Ezaki J, Murata S, Hamazaki J, Nishito Y, Iemura S, Natsume T, Yanagawa T, Uwayama J, Warabi E, Yoshida H, Ishii T, Kobayashi A, Yamamoto M, Yue Z, Uchiyama Y, Kominami E, Tanaka K. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell. 2007;17:1149–1163. doi: 10.1016/j.cell.2007.10.035.
    1. Takamura A, Komatsu M, Hara T, Sakamoto A, Kishi C, Waguri S, Eishi Y, Hino O, Tanaka K, Mizushima N. Autophagy-deficient mice develop multiple liver tumors. Genes Dev. 2011;17:795–800. doi: 10.1101/gad.2016211.
    1. Yin XM, Ding WX, Gao W. Autophagy in the liver. Hepatology. 2008;17:1773–1785. doi: 10.1002/hep.22146.
    1. Araki K, Ellebedy AH, Ahmed R. TOR in the immune system. Curr Opin Cell Biol. 2011;17:707–715. doi: 10.1016/j.ceb.2011.08.006.
    1. Waickman AT, Powell JD. mTOR, metabolism, and the regulation of T-cell differentiation and function. Immunol Rev. 2012;17:43–58. doi: 10.1111/j.1600-065X.2012.01152.x.
    1. Steel HC, Theron AJ, Cockeran R, Anderson R, Feldman C. Pathogen- and host-directed anti-inflammatory activities of macrolide antibiotics. Mediators Inflamm. 2012;17:584262.
    1. Casaer MP, Mesotten D, Hermans G, Wouters PJ, Schetz M, Meyfroidt G, Van Cromphaut S, Ingels C, Meersseman P, Muller J, Vlasselaers D, Debaveye Y, Desmet L, Dubois J, Van Assche A, Vanderheyden S, Wilmer A, Van den Berghe G. Early versus late parenteral nutrition in critically ill adults. N Engl J Med. 2011;17:506–517. doi: 10.1056/NEJMoa1102662.

Source: PubMed

3
Prenumerera