Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Primary-cilium-dependent autophagy controls epithelial cell volume in response to fluid flow

Abstract

Autophagy is an adaptation mechanism that is vital for cellular homeostasis in response to various stress conditions. Previous reports indicate that there is a functional interaction between the primary cilium (PC) and autophagy. The PC, a microtubule-based structure present at the surface of numerous cell types, is a mechanical sensor. Here we show that autophagy induced by fluid flow regulates kidney epithelial cell volume in vitro and in vivo. PC ablation blocked autophagy induction and cell-volume regulation. In addition, inhibition of autophagy in ciliated cells impaired the flow-dependent regulation of cell volume. PC-dependent autophagy can be triggered either by mTOR inhibition or a mechanism dependent on the polycystin 2 channel. Only the LKB1–AMPK–mTOR signalling pathway was required for the flow-dependent regulation of cell volume by autophagy. These findings suggest that therapies regulating autophagy should be considered in developing treatments for PC-related diseases.

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

Access options

Buy this article

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

Figure 1: Fluid flow induces autophagy in MDCK cells.
Figure 2: Inhibition of ciliogenesis impairs flow-induced autophagy.
Figure 3: Inhibition of autophagy impairs regulation of cell volume.
Figure 4: Blockage of autophagic flux increases the size of tubular cells.
Figure 5: Inhibition of urinary fluid flow decreases the autophagic response and increases the size of tubular cells.
Figure 6: Impairment of autophagic activity and deregulation of cell size in PC-deficient kidney tubules.
Figure 7: Fluid-flow-induced autophagy and cell-volume regulation is dependent on the LKB1–AMPK–mTOR pathway.

Similar content being viewed by others

References

  1. Satir, P. & Christensen, S. T. Overview of structure and function of mammalian cilia. Annu. Rev. Physiol. 69, 377–400 (2007).

    CAS  PubMed  Google Scholar 

  2. Eggenschwiler, J. T. & Anderson, K. V. Cilia and developmental signaling. Annu. Rev. Cell Dev. Biol. 23, 345–373 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Goetz, S. C. & Anderson, K. V. The primary cilium: a signalling centre during vertebrate development. Nat. Rev. Genet. 11, 331–344 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Zimmerman, K. & Yoder, B. K. SnapShot: sensing and signaling by cilia. Cell 161, 692 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Fliegauf, M., Benzing, T. & Omran, H. When cilia go bad: cilia defects and ciliopathies. Nat. Rev. Mol. Cell Biol. 8, 880–893 (2007).

    CAS  PubMed  Google Scholar 

  6. Hildebrandt, F., Benzing, T. & Katsanis, N. Ciliopathies. N. Engl. J. Med. 364, 1533–1543 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Abdul-Majeed, S., Moloney, B. C. & Nauli, S. M. Mechanisms regulating cilia growth and cilia function in endothelial cells. Cell Mol. Life Sci. 69, 165–173 (2012).

    CAS  PubMed  Google Scholar 

  8. Kallakuri, S. et al. Endothelial cilia are essential for developmental vascular integrity in zebrafish. J. Am. Soc. Nephrol. 26, 864–875 (2015).

    CAS  PubMed  Google Scholar 

  9. Masyuk, A. I. et al. Cholangiocyte cilia detect changes in luminal fluid flow and transmit them into intracellular Ca2+ and cAMP signaling. Gastroenterology 131, 911–920 (2006).

    CAS  PubMed  Google Scholar 

  10. McGrath, J., Somlo, S., Makova, S., Tian, X. & Brueckner, M. Two populations of node monocilia initiate left-right asymmetry in the mouse. Cell 114, 61–73 (2003).

    CAS  PubMed  Google Scholar 

  11. Nauli, S. M. et al. Polycystins 1 and 2 mediate mechanosensation in the primary cilium of kidney cells. Nat. Genet. 33, 129–137 (2003).

    CAS  PubMed  Google Scholar 

  12. Weinbaum, S., Duan, Y., Satlin, L. M., Wang, T. & Weinstein, A. M. Mechanotransduction in the renal tubule. Am. J. Physiol. Renal Physiol. 299, F1220–F1236 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Raghavan, V. & Weisz, O. A. Discerning the role of mechanosensors in regulating proximal tubule function. Am. J. Physiol. Renal Physiol. 310, F1–F5 (2016).

    CAS  PubMed  Google Scholar 

  14. Chapin, H. C. & Caplan, M. J. The cell biology of polycystic kidney disease. J. Cell Biol. 191, 701–710 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Brown, N. E. & Murcia, N. S. Delayed cystogenesis and increased ciliogenesis associated with the re-expression of polaris in Tg737 mutant mice. Kidney Int. 63, 1220–1229 (2003).

    PubMed  Google Scholar 

  16. Grantham, J. J., Geiser, J. L. & Evan, A. P. Cyst formation and growth in autosomal dominant polycystic kidney disease. Kidney Int. 31, 1145–1152 (1987).

    CAS  PubMed  Google Scholar 

  17. Boehlke, C. et al. Primary cilia regulate mTORC1 activity and cell size through Lkb1. Nat. Cell Biol. 12, 1115–1122 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Takiar, V. & Caplan, M. J. Polycystic kidney disease: pathogenesis and potential therapies. Biochim. Biophys. Acta 1812, 1337–1343 (2011).

    CAS  PubMed  Google Scholar 

  19. Boya, P., Reggiori, F. & Codogno, P. Emerging regulation and functions of autophagy. Nat. Cell Biol. 15, 713–720 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Mizushima, N. & Komatsu, M. Autophagy: renovation of cells and tissues. Cell 147, 728–741 (2011).

    CAS  PubMed  Google Scholar 

  21. Yang, Z. & Klionsky, D. J. Eaten alive: a history of macroautophagy. Nat. Cell Biol. 12, 814–822 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Kroemer, G., Mariño, G. & Levine, B. Autophagy and the integrated stress response. Mol. Cell 40, 280–293 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Meijer, A. J., Lorin, S., Blommaart, E. F. & Codogno, P. Regulation of autophagy by amino acids and MTOR-dependent signal transduction. Amino Acids 47, 2037–2063 (2015).

    CAS  PubMed  Google Scholar 

  24. DuFort, C. C., Paszek, M. J. & Weaver, V. M. Balancing forces: architectural control of mechanotransduction. Nat. Rev. Mol. Cell Biol. 12, 308–319 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. French, A. S. Mechanotransduction. Annu. Rev. Physiol. 54, 135–152 (1992).

    CAS  PubMed  Google Scholar 

  26. He, C. et al. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature 481, 511–515 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Hussain, S. N. et al. Mechanical ventilation-induced diaphragm disuse in humans triggers autophagy. Am. J. Respir. Crit. Care Med. 182, 1377–1386 (2010).

    CAS  PubMed  Google Scholar 

  28. Zaglia, T. et al. Atrogin-1 deficiency promotes cardiomyopathy and premature death via impaired autophagy. J. Clin. Invest. 124, 2410–2424 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. King, J. S. Mechanical stress meets autophagy: potential implications for physiology and pathology. Trends Mol. Med. 18, 583–588 (2012).

    CAS  PubMed  Google Scholar 

  30. Pampliega, O. et al. Functional interaction between autophagy and ciliogenesis. Nature 502, 194–200 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Tang, Z. et al. Autophagy promotes primary ciliogenesis by removing OFD1 from centriolar satellites. Nature 502, 254–257 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Kotsis, F., Boehlke, C. & Kuehn, E. W. The ciliary flow sensor and polycystic kidney disease. Nephrol. Dial. Transplant. 28, 518–526 (2013).

    PubMed  PubMed Central  Google Scholar 

  33. Kabeya, Y. et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 19, 5720–5728 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Klionsky, D. J. et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12, 1–222 (2016).

    PubMed  PubMed Central  Google Scholar 

  35. Kimura, S., Noda, T. & Yoshimori, T. Dissection of the autophagosome maturation process by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy 3, 452–460 (2007).

    CAS  PubMed  Google Scholar 

  36. Heo, J., Sachs, F., Wang, J. & Hua, S. Z. Shear-induced volume decrease in MDCK cells. Cell Physiol. Biochem. 30, 395–406 (2012).

    CAS  PubMed  Google Scholar 

  37. Tanida, I., Minematsu-Ikeguchi, N., Ueno, T. & Kominami, E. Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy 1, 84–91 (2005).

    CAS  PubMed  Google Scholar 

  38. Jin, G. et al. Skp2-mediated RagA ubiquitination elicits a negative feedback to prevent amino-acid-dependent mTORC1 hyperactivation by recruiting GATOR1. Mol. Cell 58, 989–1000 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Orhon, I., Dupont, N., Pampliega, O., Cuervo, A. M. & Codogno, P. Autophagy and regulation of cilia function and assembly. Cell Death Differ. 22, 389–397 (2015).

    CAS  PubMed  Google Scholar 

  40. Wang, S., Livingston, M. J., Su, Y. & Dong, Z. Reciprocal regulation of cilia and autophagy via the MTOR and proteasome pathways. Autophagy 11, 607–616 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Pazour, G. J. et al. Chlamydomonas IFT88 and its mouse homologue, polycystic kidney disease gene tg737, are required for assembly of cilia and flagella. J. Cell Biol. 151, 709–718 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Hirokawa, N., Noda, Y., Tanaka, Y. & Niwa, S. Kinesin superfamily motor proteins and intracellular transport. Nat. Rev. Mol. Cell Biol. 10, 682–696 (2009).

    CAS  PubMed  Google Scholar 

  43. Seglen, P. O. & Gordon, P. B. 3-Methyladenine: specific inhibitor of autophagic/lysosomal protein degradation in isolated rat hepatocytes. Proc. Natl Acad. Sci. USA 79, 1889–1892 (1982).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Briscoe, J. & Therond, P. P. The mechanisms of Hedgehog signalling and its roles in development and disease. Nat. Rev. Mol. Cell Biol. 14, 416–429 (2013).

    PubMed  Google Scholar 

  45. Hofherr, A. & Kottgen, M. TRPP channels and polycystins. Adv. Exp. Med. Biol. 704, 287–313 (2011).

    CAS  PubMed  Google Scholar 

  46. Jin, X. et al. Cilioplasm is a cellular compartment for calcium signaling in response to mechanical and chemical stimuli. Cell Mol. Life Sci. 71, 2165–2178 (2014).

    CAS  PubMed  Google Scholar 

  47. Cao, M. & Zhong, Q. Cilia in autophagy and cancer. Cilia 5, 4 (2016).

    PubMed  PubMed Central  Google Scholar 

  48. Pampliega, O. & Cuervo, A. M. Autophagy and primary cilia: dual interplay. Curr. Opin. Cell Biol. 39, 1–7 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Kim, E. S. et al. Inhibition of autophagy suppresses sertraline-mediated primary ciliogenesis in retinal pigment epithelium cells. PLoS ONE 10, e0118190 (2015).

    PubMed  PubMed Central  Google Scholar 

  50. Lam, H. C. et al. Histone deacetylase 6-mediated selective autophagy regulates COPD-associated cilia dysfunction. J. Clin. Invest. 123, 5212–5230 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Meijer, A. J. et al. Cell swelling and the sensitivity of autophagic proteolysis to inhibition by amino acids in isolated rat hepatocytes. Eur. J. Biochem. 215, 449–454 (1993).

    CAS  PubMed  Google Scholar 

  52. vom Dahl, S. et al. Involvement of integrins in osmosensing and signaling toward autophagic proteolysis in rat liver. J. Biol. Chem. 278, 27088–27095 (2003).

    CAS  PubMed  Google Scholar 

  53. Kimura, T. et al. Autophagy protects the proximal tubule from degeneration and acute ischemic injury. J. Am. Soc. Nephrol. 22, 902–913 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Komatsu, M. et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J. Cell Biol. 169, 425–434 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Chang, T. K. et al. Uba1 functions in Atg7- and Atg3-independent autophagy. Nat. Cell Biol. 15, 1067–1078 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Miettinen, T. P. & Bjorklund, M. Mevalonate pathway regulates cell size homeostasis and proteostasis through autophagy. Cell Rep. 13, 2610–2620 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Fader, C. M., Sanchez, D., Furlan, M. & Colombo, M. I. Induction of autophagy promotes fusion of multivesicular bodies with autophagic vacuoles in k562 cells. Traffic 9, 230–250 (2008).

    CAS  PubMed  Google Scholar 

  58. Knodler, A. et al. Coordination of Rab8 and Rab11 in primary ciliogenesis. Proc. Natl Acad. Sci. USA 107, 6346–6351 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Zeng, H. et al. mTORC1 couples immune signals and metabolic programming to establish T(reg)-cell function. Nature 499, 485–490 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Huber, T. B. et al. Emerging role of autophagy in kidney function, diseases and aging. Autophagy 8, 1009–1031 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Ravichandran, K. & Edelstein, C. L. Polycystic kidney disease: a case of suppressed autophagy? Semin. Nephrol. 34, 27–33 (2014).

    CAS  PubMed  Google Scholar 

  62. Wang, Z. & Choi, M. E. Autophagy in kidney health and disease. Antioxid. Redox Signal. 20, 519–537 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Patel, A. & Honore, E. Polycystins and renovascular mechanosensory transduction. Nat. Rev. Nephrol. 6, 530–538 (2010).

    CAS  PubMed  Google Scholar 

  64. Cebotaru, V. et al. Polycystin-1 negatively regulates Polycystin-2 expression via the aggresome/autophagosome pathway. J. Biol. Chem. 289, 6404–6414 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Delling, M., DeCaen, P. G., Doerner, J. F., Febvay, S. & Clapham, D. E. Primary cilia are specialized calcium signalling organelles. Nature 504, 311–314 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Thomas, G. & Hall, M. N. TOR signalling and control of cell growth. Curr. Opin. Cell Biol. 9, 782–787 (1997).

    CAS  PubMed  Google Scholar 

  67. Raghavan, V., Rbaibi, Y., Pastor-Soler, N. M., Carattino, M. D. & Weisz, O. A. Shear stress-dependent regulation of apical endocytosis in renal proximal tubule cells mediated by primary cilia. Proc. Natl Acad. Sci. USA 111, 8506–8511 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Tooze, S. A., Abada, A. & Elazar, Z. Endocytosis and autophagy: exploitation or cooperation? Cold Spring Harb. Perspect. Biol. 6, a018358 (2014).

    PubMed  PubMed Central  Google Scholar 

  69. Puri, C., Renna, M., Bento, C. F., Moreau, K. & Rubinsztein, D. C. Diverse autophagosome membrane sources coalesce in recycling endosomes. Cell 154, 1285–1299 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Dupont, N., Orhon, I., Bauvy, C. & Codogno, P. Autophagy and autophagic flux in tumor cells. Methods Enzymol. 543, 73–88 (2014).

    CAS  PubMed  Google Scholar 

  71. El Boustany, C. et al. Capacitative calcium entry and transient receptor potential canonical 6 expression control human hepatoma cell proliferation. Hepatology 47, 2068–2077 (2008).

    CAS  PubMed  Google Scholar 

  72. Yoo, K. H. et al. Osteopontin regulates renal apoptosis and interstitial fibrosis in neonatal chronic unilateral ureteral obstruction. Kidney Int. 70, 1735–1741 (2006).

    CAS  PubMed  Google Scholar 

  73. Viau, A. et al. Lipocalin 2 is essential for chronic kidney disease progression in mice and humans. J. Clin. Invest. 120, 4065–4076 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Canaud, G. et al. AKT2 is essential to maintain podocyte viability and function during chronic kidney disease. Nat. Med. 19, 1288–1296 (2013).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank F. Bienaimé, T. Blanc, C. Nguyen and C. Bauvy for assistance. We also thank T. Yoshimori (Osaka University, Osaksa, Japan) for providing GFP–LC3 and RFP–GFP–LC3 constructs. We are grateful to A. M. Cuervo (Albert Einstein College, Bronx, New York, USA) for sharing KECs and IFT88−/− KECs. We acknowledge the design of the transgenic mouse strains by L. Goldstein, San Diego, California, USA (Kif3atm1Gsn), R. Koesters, Heidelberg, Germany (Tg(Pax8–rtTA2SM2)1Koes), and D. Tenen, Boston, USA (Tg(TetOCre)1Dgt). We also acknowledge the Necker Institute Imaging Facility, the Fondation Imagine, the Cochin Imaging Electron Microscopy facility, and the Animal Histology and Morphology Core Facility (SFR Necker INSERM US24, CNRS UMS 3633). The work was supported by institutional funding from INSERM, CNRS, and University Paris Descartes and grants from ANR and INCa to P.C. N.D. is supported by a fellowship from Association pour la Recherche sur le Cancer (ARC), and A.V. was supported by an ERA-EDTA fellowship. E.W.K. is supported by the DFG (KU1504-5/1) and Else–Kröner–Fresenius Stiftung (2011_A87).

Author information

Authors and Affiliations

Authors

Contributions

I.O., N.D., M.Z., M.B. and V.B. performed most of the experiments with the exception of electron microscopy experiments, which were performed by A.S. I.B. contributed to the initial part of the project. A.V. and E.W.K. provided the mouse tissues. I.O., N.D., M.Z., T.C., G.F., F.T. and P.C. conceived and planned the experiments and interpreted data. I.O., N.D. and P.C. wrote the manuscript.

Corresponding author

Correspondence to Patrice Codogno.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 2 Kinetics of LC3-II and actin expression determined by western blot analyses of WT and IFT88−/− KECs subjected to fluid flow.

(a) WT KEC and (b) IFT88−/− KECs were subjected to flow from 4 h to 4 days (d0, h4, d1, d2, d4) with or without chloroquine (CQ) then LC3-II and actin levels were determined by immunoblotting independently repeated three times. Unprocessed original scans of blots are shown in Supplementary Fig. 8.

Supplementary Figure 3 Recruitment of ATG16L1 to the basal body on fluid flow induction.

(ad) WT KEC were subjected to flow or not (d0) for 1 day (d1) then fixed and labelled with γ-tubulin and ATG16L1 antibodies. (a) Cells were analysed by confocal fluorescence microscopy. Arrowheads indicate co-localization of ATG16L1 (green) and γ-tubulin (red). (b) 3D reconstruction of WT KECs subjected to flow or not (dark blue, small cells; white, large cells). (c) Two-fluorescence channel line tracings corresponding to dashed lines in the merge of panel A. (d) Recruitment of ATG16L1 to basal bodies was analysed by quantification of ATG16L1+ basal bodies using ImageJ; n = 125 cells from 5 fields of view per experiment pooled from 3 independent experiments. Data in (d) are means ± s.e.m. Statistical significance was determined using an unpaired Student’s t-test; P values are shown.

Supplementary Figure 4 Ultrastucture analyses of WT and IFT88−/− KECs subjected to fluid flow.

(ac) WT KEC and IFT88−/− KECs were subjected to flow or not (d0) for 1 day (d1) then fixed and processed for ultrastructure electron microscopy. (a) Representative images. (b) Magnification of double membrane structures (autophagosome) shown in the white inset in panel A. (c) Quantification of autophagic vacuoles (AV) in WT KEC and IFT88−/− KECs with and without (ctrl) flow; n = 15 cells from a single experiment that was independently repeated 3 times. (d) Immunogold electron microscopy for LC3. Data in (c) are means ± s.e.m. Statistical significance was determined using an unpaired Student’s t-test; P values are shown.

Supplementary Figure 5 Effect of fluid flow on ciliogenesis.

(ad) WT KECs were not subjected to flow (d0) or were subjected to flow for 4 h (h4) or 4 days (d4) then fixed, labelled with acetylated-tubulin (red) and LC3 antibody (green), then analysed by fluorescence microscopy. (a) Representative images of ciliated cells. (b) Number of ciliated cells was quantified using Imaris software. Not significant indicated by ‘ns’. n = 45 cells pooled from 3 independent experiments. (c) Representative images. (d) Cilium length was quantified using Imaris software; n = 45 cells pooled from 3 independent experiments. Data in (b) and (d) are means ± s.e.m. Statistical significance was determined using an unpaired Student’s t-test; P values are shown.

Supplementary Figure 6 Effect of inhibition of ciliogenesis on flow-induced autophagy.

(a) WT KEC and IFT88−/− KECs were subjected to flow or not from 4 h to 4 days (d0, h4, d1, d2, d4). Mean cell volumes were determined by microscopy; , P < 0.05, n = 125 cells from 5 fields of view per experiment pooled from 3 independent experiments. Not significant indicated by ‘ns’. (bd) KECs engineered to inducibly express an shRNA designed to inhibit expression of kif3a were treated or not with IPTG and subjected to flow for 4 days (d4) or not (d0). (b) Reductions in levels of Kif3a on induction of shRNA expression were confirmed by immunoblotting independently repeated three times. (c) KECs treated or not with IPTG were fixed, labelled with LC3 antibody, and analysed by fluorescence microscopy. Number of endogenous LC3 puncta per cell were quantified using ImageJ software; n = 125 cells from 5 fields of view per experiment pooled from 3 independent experiments. (d,e) KECs engineered to inducibly express shRNA targeting kif3a were treated or not with IPTG and subjected to flow for 4 days (d4) or not (d0). (d) 3D reconstructions of cells (dark blue indicates small cells and white indicates large cells). (e) Mean cell volumes; n = 125 cells from 5 fields of view per experiment pooled from 3 independent experiments. Data in (a), (c), and (e) are means ± s.e.m. Statistical significance was determined using an unpaired Student’s t-test; P values are shown. Unprocessed original scans of blots are shown in Supplementary Fig. 8.

Supplementary Figure 7 Western blot analysis of KECs expressing shRNAs targeting Atg5 or Atg16L1.

Expression of shRNA targeting (a) Atg5 or (b) Atg16L1 in KECs treated or not with IPTG and subjected to flow for 4 days reduced levels of ATG5 and ATG16L1, respectively, as shown by immunoblotting independently repeated three times. (c) KECs engineered to inducibly express shRNA targeting Atg5 were treated or not with IPTG and subjected to flow from 4 h to 4 days (d0, h4, d1, d2, d4), then LC3-II and actin levels were determined by immunoblotting. (d) KECs engineered to inducibly express shRNA targeting Atg16L1 were treated or not with IPTG and subjected to flow from 4 h to 4 days (d0, h4, d1, d2, d4), then LC3-II and actin levels were determined by immunoblotting independently repeated three times. Unprocessed original scans of blots are shown in Supplementary Fig. 8.

Supplementary Figure 8 Role of Hedgehog signalling and PC2 calcium channel in fluid flow-induced autophagy.

(a) Time course of expression of Gli1 and Ptch1in WT KECs subjected to flow from 4 h to 4 days (4 h, d1, d2, d4) or not (d0). As a positive control, cells were treated with purmophamine for 24 h. Gli1 and Ptch1 mRNA levels were quantified by real-time RT-PCR, normalized with respect to the β-actin gene, and presented as fold increases; from n = 3 independent experiments as shown in i. Not significant indicated by ‘ns’. (b) Cells were subjected to flow for 4 h (h4) or not (d0) or treated with purmophamine for 24 h, then fixed and stained for Smo (green) and acetylated-tubulin (red) to evaluate localization of Smo to the axoneme of cilia. Cells were then analysed by fluorescence microscopy. (c) KECs engineered to inducibly express an shRNA targeting Pkd2 were treated or not with IPTG, and Pkd2 mRNA levels were quantified by real-time RT-PCR, normalized with respect to the β-actin gene, and presented as fold increases; from n = 3 independent experiments as shown in i. (d,e) KECs engineered to inducibly express an shRNA targeting Pkd2 were treated with IPTG (right panel) or not (left panel), labelled with Fura-2, and analysed by fluorescence microscopy. Black arrowheads indicate when cells were subjected to flow or to ATP (to enable evaluation of cell viability). (f,g) KECs engineered to inducibly express an shRNA targeting Pkd2 were treated or not with IPTG and subjected to flow for 4 h or not (d0) with or without CQ. LC3-II/actin ratios visualized by immunoblotting were quantified by densitometry; , P < 0.05, from n = 3 independent experiments as shown in i. (h,i) KECs engineered to inducibly express shRNA targeting Pkd2 were treated or not with IPTG and subjected to flow for 4 days (d4) or not (d0). (h) 3D reconstructions of cells (dark blue indicates small cells and white indicates large cells). (i) Mean cell volumes; n = 125 cells from 5 fields of view per experiment pooled from 3 independent experiments. Data in (a), (c), (g), and (i) are means ± s.e.m. Statistical significance was determined using an unpaired Student’s t-test; P values are shown. Unprocessed original scans of blots are shown in Supplementary Fig. 8.

Supplementary Table 1 Lentivirus vectors and shRNAs.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1814 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Orhon, I., Dupont, N., Zaidan, M. et al. Primary-cilium-dependent autophagy controls epithelial cell volume in response to fluid flow. Nat Cell Biol 18, 657–667 (2016). https://doi.org/10.1038/ncb3360

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3360

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing