The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress (original) (raw)

Nature Medicine volume 13, pages 619–624 (2007)Cite this article

Abstract

Autophagy, an evolutionarily conserved process for the bulk degradation of cytoplasmic components, serves as a cell survival mechanism in starving cells1,2. Although altered autophagy has been observed in various heart diseases, including cardiac hypertrophy3,4 and heart failure5,6, it remains unclear whether autophagy plays a beneficial or detrimental role in the heart. Here, we report that the cardiac-specific loss of autophagy causes cardiomyopathy in mice. In adult mice, temporally controlled cardiac-specific deficiency of Atg5 (autophagy-related 5), a protein required for autophagy, led to cardiac hypertrophy, left ventricular dilatation and contractile dysfunction, accompanied by increased levels of ubiquitination. Furthermore, Atg5-deficient hearts showed disorganized sarcomere structure and mitochondrial misalignment and aggregation. On the other hand, cardiac-specific deficiency of Atg5 early in cardiogenesis showed no such cardiac phenotypes under baseline conditions, but developed cardiac dysfunction and left ventricular dilatation one week after treatment with pressure overload. These results indicate that constitutive autophagy in the heart under baseline conditions is a homeostatic mechanism for maintaining cardiomyocyte size and global cardiac structure and function, and that upregulation of autophagy in failing hearts is an adaptive response for protecting cells from hemodynamic stress.

This is a preview of subscription content, access via your institution

Access options

Subscribe to this journal

Receive 12 print issues and online access

$209.00 per year

only $17.42 per issue

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Additional access options:

Similar content being viewed by others

References

  1. Mizushima, N., Ohsumi, Y. & Yoshimori, T. Autophagosome formation in mammalian cells. Cell Struct. Funct. 27, 421–429 (2002).
    Article Google Scholar
  2. Levine, B. & Klionsky, D.J. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev. Cell 6, 463–477 (2004).
    Article CAS Google Scholar
  3. Dammrich, J. & Pfeifer, U. Cardiac hypertrophy in rats after supravalvular aortic constriction. II. Inhibition of cellular autophagy in hypertrophying cardiomyocytes. Virchows Arch. B Cell Pathol. Incl. Mol. Pathol. 43, 287–307 (1983).
    Article CAS Google Scholar
  4. Pfeifer, U., Fohr, J., Wilhelm, W. & Dammrich, J. Short-term inhibition of cardiac cellular autophagy by isoproterenol. J. Mol. Cell. Cardiol. 19, 1179–1184 (1987).
    Article CAS Google Scholar
  5. Shimomura, H. et al. Autophagic degeneration as a possible mechanism of myocardial cell death in dilated cardiomyopathy. Jpn. Circ. J. 65, 965–968 (2001).
    Article CAS Google Scholar
  6. Miyata, S. et al. Autophagic cardiomyocyte death in cardiomyopathic hamsters and its prevention by granulocyte colony-stimulating factor. Am. J. Pathol. 168, 386–397 (2006).
    Article CAS Google Scholar
  7. Kuma, A. et al. The role of autophagy during the early neonatal starvation period. Nature 432, 1032–1036 (2004).
    Article CAS Google Scholar
  8. Komatsu, M. et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J. Cell Biol. 169, 425–434 (2005).
    Article CAS Google Scholar
  9. Hara, T. et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441, 885–889 (2006).
    Article CAS Google Scholar
  10. Komatsu, M. et al. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature 441, 880–884 (2006).
    Article CAS Google Scholar
  11. Baehrecke, E.H. Autophagy: dual roles in life and death? Nat. Rev. Mol. Cell Biol. 6, 505–510 (2005).
    Article CAS Google Scholar
  12. Decker, R.S. & Wildenthal, K. Lysosomal alterations in hypoxic and reoxygenated hearts. I. Ultrastructural and cytochemical changes. Am. J. Pathol. 98, 425–444 (1980).
    CAS PubMed PubMed Central Google Scholar
  13. Yan, L. et al. Autophagy in chronically ischemic myocardium. Proc. Natl. Acad. Sci. USA 102, 13807–13812 (2005).
    Article CAS Google Scholar
  14. Nishino, I. et al. Primary LAMP-2 deficiency causes X-linked vacuolar cardiomyopathy and myopathy (Danon disease). Nature 406, 906–910 (2000).
    Article CAS Google Scholar
  15. Tanaka, Y. et al. Accumulation of autophagic vacuoles and cardiomyopathy in LAMP-2-deficient mice. Nature 406, 902–906 (2000).
    Article CAS Google Scholar
  16. Sohal, D.S. et al. Temporally regulated and tissue-specific gene manipulations in the adult and embryonic heart using a tamoxifen-inducible Cre protein. Circ. Res. 89, 20–25 (2001).
    Article CAS Google Scholar
  17. Kabeya, Y. et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 19, 5720–5728 (2000).
    Article CAS Google Scholar
  18. 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 15, 1101–1111 (2004).
    Article CAS Google Scholar
  19. Bjorkoy, G. et al. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J. Cell Biol. 171, 603–614 (2005).
    Article Google Scholar
  20. Hosokawa, N., Hara, Y. & Mizushima, N. Generation of cell lines with tetracycline-regulated autophagy and a role for autophagy in controlling cell size. FEBS Lett. 580, 2623–2629 (2006).
    Article CAS Google Scholar
  21. Nakagawa, T. et al. Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta. Nature 403, 98–103 (2000).
    Article CAS Google Scholar
  22. Boya, P. et al. Inhibition of macroautophagy triggers apoptosis. Mol. Cell. Biol. 25, 1025–1040 (2005).
    Article CAS Google Scholar
  23. Chen, J. et al. Selective requirement of myosin light chain 2v in embryonic heart function. J. Biol. Chem. 273, 1252–1256 (1998).
    Article CAS Google Scholar
  24. Yamaguchi, O. et al. Cardiac-specific disruption of the c-raf-1 gene induces cardiac dysfunction and apoptosis. J. Clin. Invest. 114, 937–943 (2004).
    Article CAS Google Scholar
  25. Lyons, G.E. et al. Developmental regulation of myosin gene expression in mouse cardiac muscle. J. Cell Biol. 111, 2427–2436 (1990).
    Article CAS Google Scholar
  26. Yamaguchi, O. et al. Targeted deletion of apoptosis signal-regulating kinase 1 attenuates left ventricular remodeling. Proc. Natl. Acad. Sci. USA 100, 15883–15888 (2003).
    Article CAS Google Scholar
  27. Wencker, D. et al. A mechanistic role for cardiac myocyte apoptosis in heart failure. J. Clin. Invest. 111, 1497–1504 (2003).
    Article CAS Google Scholar
  28. Hirotani, S. et al. Involvement of nuclear factor-κB and apoptosis signal-regulating kinase 1 in G-protein-coupled receptor agonist-induced cardiomyocyte hypertrophy. Circulation 105, 509–515 (2002).
    Article CAS Google Scholar
  29. Zhou, Y.Y. et al. Culture and adenoviral infection of adult mouse cardiac myocytes: methods for cellular genetic physiology. Am. J. Physiol. Heart Circ. Physiol. 279, H429–H436 (2000).
    Article CAS Google Scholar
  30. Mizushima, N. et al. Dissection of autophagosome formation using Apg5-deficient mouse embryonic stem cells. J. Cell Biol. 152, 657–668 (2001).
    Article CAS Google Scholar

Download references

Acknowledgements

We are grateful to K. Chien (Harvard University) for the gift of MLC-2v Cre mice, J. Molkentin (Cincinnati Children's Hospital Medical Center) for MerCreMer mice, T. Yoshimori (Osaka University) for antibody to LC3 and E. Lakatta for teaching us to isolate adult mouse cardiomyocytes. This work was supported by a Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology to K.O. (16590683). O.Y. held a postdoctoral fellowship from the Japan Society for the Promotion of Science. S.H. was the recipient of a postdoctoral fellowship from the Center of Excellence Research of the Ministry of Education, Culture, Sports, Science and Technology. T.T. received a postdoctoral fellowship from the Japan Health Science Foundation.

Author information

Author notes

  1. Atsuko Nakai and Osamu Yamaguchi: These authors contributed equally to this work.

Authors and Affiliations

  1. Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, 565-0871, Osaka, Japan
    Atsuko Nakai, Osamu Yamaguchi, Toshihiro Takeda, Yoshiharu Higuchi, Shungo Hikoso, Masayuki Taniike, Shigemiki Omiya, Isamu Mizote, Kazuhiko Nishida, Masatsugu Hori & Kinya Otsu
  2. Department of Medical Information Science, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, 565-0871, Osaka, Japan
    Yasushi Matsumura
  3. Department of Biochemistry, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, 565-0871, Osaka, Japan
    Michio Asahi
  4. Department of Bioregulation and Metabolism, Tokyo Metropolitan Institute of Medical Science, Bunkyoku, Tokyo, 113-8613, Japan
    Noboru Mizushima
  5. Department of Physiology and Cell Biology, Tokyo Medical and Dental University, Bunkyoku, Tokyo, 113-8519, Japan
    Noboru Mizushima
  6. Solution-Oriented Research for Science and Technology, Japan Science and Technology Agency, 4-1-8 Honmachi, Kawaguchi, 332-0012, Saitama, Japan
    Noboru Mizushima

Authors

  1. Atsuko Nakai
    You can also search for this author inPubMed Google Scholar
  2. Osamu Yamaguchi
    You can also search for this author inPubMed Google Scholar
  3. Toshihiro Takeda
    You can also search for this author inPubMed Google Scholar
  4. Yoshiharu Higuchi
    You can also search for this author inPubMed Google Scholar
  5. Shungo Hikoso
    You can also search for this author inPubMed Google Scholar
  6. Masayuki Taniike
    You can also search for this author inPubMed Google Scholar
  7. Shigemiki Omiya
    You can also search for this author inPubMed Google Scholar
  8. Isamu Mizote
    You can also search for this author inPubMed Google Scholar
  9. Yasushi Matsumura
    You can also search for this author inPubMed Google Scholar
  10. Michio Asahi
    You can also search for this author inPubMed Google Scholar
  11. Kazuhiko Nishida
    You can also search for this author inPubMed Google Scholar
  12. Masatsugu Hori
    You can also search for this author inPubMed Google Scholar
  13. Noboru Mizushima
    You can also search for this author inPubMed Google Scholar
  14. Kinya Otsu
    You can also search for this author inPubMed Google Scholar

Contributions

A.N. worked on the in vitro analysis of the mice; O.Y. conducted the in vivo analysis of the mice and wrote the manuscript; T.T. performed adult cardiomyocyte isolation and Ca2+ transient experiments; Y.H. performed ischemia-reperfusion surgery; S.H. assisted with RT-PCR experiments; M.T., S.O. and I.M. contributed to the in vitro experiments; Y.M. performed statistical analysis of the data; M.A. contributed to Ca2+ transient measurements; K.N. contributed to the in vivo experiments; M.H. supervised this project; N.M. provided advice on designing and conducting experiments; K.O. conceived, designed and directed the study.

Corresponding author

Correspondence toKinya Otsu.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Fig. 1

Inhibition of autophagy using adenovirus expressing shRNA against Atg7. (PDF 617 kb)

Supplementary Fig. 2

Characterization of _Atg5_flox/flox; MLC2v-Cre+ mice. (PDF 235 kb)

Supplementary Fig. 3

Pressure overload induces cardiac dysfunction in _Atg5_flox/flox; α-MyHC-Cre+ mice. (PDF 372 kb)

Supplementary Fig. 4

Autophagy in pressure overload-induced cardiac remodeling. (PDF 236 kb)

Supplementary Fig. 5

Biochemical analysis of _Atg5_flox/flox; MLC2v-Cre+ hearts after TAC. (PDF 287 kb)

Supplementary Fig. 6

β-adrenergic stress induces cardiac dysfunction in _Atg5_flox/flox; MLC2v-Cre+ mice. (PDF 627 kb)

Rights and permissions

About this article

Cite this article

Nakai, A., Yamaguchi, O., Takeda, T. et al. The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress.Nat Med 13, 619–624 (2007). https://doi.org/10.1038/nm1574

Download citation

Associated content

Eat your heart out

Nature Medicine News & Views 01 May 2007