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.

  • Letter
  • Published:

C. elegans neurons jettison protein aggregates and mitochondria under neurotoxic stress

Abstract

The toxicity of misfolded proteins and mitochondrial dysfunction are pivotal factors that promote age-associated functional neuronal decline and neurodegenerative disease1,2. Accordingly, neurons invest considerable cellular resources in chaperones, protein degradation, autophagy and mitophagy to maintain proteostasis and mitochondrial quality3,4. Complicating the challenges of neuroprotection, misfolded human disease proteins and mitochondria can move into neighbouring cells via unknown mechanisms, which may promote pathological spread5,6. Here we show that adult neurons from Caenorhabditis elegans extrude large (approximately 4 μm) membrane-surrounded vesicles called exophers that can contain protein aggregates and organelles. Inhibition of chaperone expression, autophagy or the proteasome, in addition to compromising mitochondrial quality, enhances the production of exophers. Proteotoxically stressed neurons that generate exophers subsequently function better than similarly stressed neurons that did not produce exophers. The extruded exopher transits through surrounding tissue in which some contents appear degraded, but some non-degradable materials can subsequently be found in more remote cells, suggesting secondary release. Our observations suggest that exopher-genesis is a potential response to rid cells of neurotoxic components when proteostasis and organelle function are challenged. We propose that exophers are components of a conserved mechanism that constitutes a fundamental, but formerly unrecognized, branch of neuronal proteostasis and mitochondrial quality control, which, when dysfunctional or diminished with age, might actively contribute to pathogenesis in human neurodegenerative disease and brain ageing.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: C. elegans touch neurons can extrude cytoplasmic contents.
Figure 2: Touch neurons under proteotoxic stress jettison aggregation-prone proteins into exophers.
Figure 3: Disruption of multiple branches of proteostasis increases exopher formation.
Figure 4: Mitochondria can be extruded in exophers, and mitochondria with higher mitochondrial matrix oxidation might be preferentially extruded
Figure 5: Fluorescent mCherry escapes touch neurons and surrounding hypodermis to later concentrate in distant coelomocytes.

Similar content being viewed by others

References

  1. Yankner, B. A., Lu, T. & Loerch, P. The aging brain. Annu. Rev. Pathol. 3, 41–66 (2008)

    Article  CAS  Google Scholar 

  2. Federico, A. et al. Mitochondria, oxidative stress and neurodegeneration. J. Neurol. Sci. 322, 254–262 (2012)

    Article  CAS  Google Scholar 

  3. Morimoto, R. I. Proteotoxic stress and inducible chaperone networks in neurodegenerative disease and aging. Genes Dev. 22, 1427–1438 (2008)

    Article  CAS  Google Scholar 

  4. Ben-Gedalya, T. & Cohen, E. Quality control compartments coming of age. Traffic 13, 635–642 (2012)

    Article  CAS  Google Scholar 

  5. Lee, S. J., Desplats, P., Sigurdson, C., Tsigelny, I. & Masliah, E. Cell-to-cell transmission of non-prion protein aggregates. Nat. Rev. Neurol. 6, 702–706 (2010)

    Article  Google Scholar 

  6. Davis, C. H. et al. Transcellular degradation of axonal mitochondria. Proc. Natl Acad. Sci. USA 111, 9633–9638 (2014)

    Article  ADS  CAS  Google Scholar 

  7. Toth, M. L. et al. Neurite sprouting and synapse deterioration in the aging Caenorhabditis elegans nervous system. J. Neurosci. 32, 8778–8790 (2012)

    Article  CAS  Google Scholar 

  8. Craig, A. L., Moser, S. C., Bailly, A. P. & Gartner, A. Methods for studying the DNA damage response in the Caenorhabdatis elegans germ line. Methods Cell Biol. 107, 321–352 (2012)

    Article  CAS  Google Scholar 

  9. Perkins, L. A., Hedgecock, E. M., Thomson, J. N. & Culotti, J. G. Mutant sensory cilia in the nematode Caenorhabditis elegans. Dev. Biol. 117, 456–487 (1986)

    Article  CAS  Google Scholar 

  10. Parker, J. A. et al. Expanded polyglutamines in Caenorhabditis elegans cause axonal abnormalities and severe dysfunction of PLM mechanosensory neurons without cell death. Proc. Natl Acad. Sci. USA 98, 13318–13323 (2001)

    Article  ADS  CAS  Google Scholar 

  11. Vayndorf, E. M. et al. Morphological remodeling of C. elegans neurons during aging is modified by compromised protein homeostasis. NPJ Aging Mech. Dis. 2, 16001 (2016)

    Article  Google Scholar 

  12. Labbadia, J. & Morimoto, R. I. Proteostasis and longevity: when does aging really begin? F1000Prime Rep. 6, 7 (2014)

    Article  Google Scholar 

  13. Labbadia, J. & Morimoto, R. I. Repression of the heat shock response is a programmed event at the onset of reproduction. Mol. Cell 59, 639–650 (2015)

    Article  CAS  Google Scholar 

  14. Liu, G., Rogers, J., Murphy, C. T. & Rongo, C. EGF signalling activates the ubiquitin proteasome system to modulate C. elegans lifespan. EMBO J. 30, 2990–3003 (2011)

    Article  CAS  Google Scholar 

  15. Sämann, J. et al. Caenorhabditits elegans LRK-1 and PINK-1 act antagonistically in stress response and neurite outgrowth. J. Biol. Chem. 284, 16482–16491 (2009)

    Article  Google Scholar 

  16. Springer, W., Hoppe, T., Schmidt, E. & Baumeister, R. A Caenorhabditis elegans Parkin mutant with altered solubility couples alpha-synuclein aggregation to proteotoxic stress. Hum. Mol. Genet. 14, 3407–3423 (2005)

    Article  CAS  Google Scholar 

  17. Benedetti, C., Haynes, C. M., Yang, Y., Harding, H. P. & Ron, D. Ubiquitin-like protein 5 positively regulates chaperone gene expression in the mitochondrial unfolded protein response. Genetics 174, 229–239 (2006)

    Article  CAS  Google Scholar 

  18. Cannon, M. B. & Remington, S. J. Redox-sensitive green fluorescent protein: probes for dynamic intracellular redox responses. A review. Methods Mol. Biol. 476, 50–64 (2008)

    Article  Google Scholar 

  19. Ghose, P., Park, E. C., Tabakin, A., Salazar-Vasquez, N. & Rongo, C. Anoxia-reoxygenation regulates mitochondrial dynamics through the hypoxia response pathway, SKN-1/Nrf, and stomatin-like protein STL-1/SLP-2. PLoS Genet. 9, e1004063 (2013)

    Article  Google Scholar 

  20. Laker, R. C. et al. A novel MitoTimer reporter gene for mitochondrial content, structure, stress, and damage in vivo. J. Biol. Chem. 289, 12005–12015 (2014)

    Article  CAS  Google Scholar 

  21. Ji, Y. B., Qu, Z. Y. & Zou, X. Juglone-induced apoptosis in human gastric cancer SGC-7901 cells via the mitochondrial pathway. Exp. Toxicol. Pathol. 63, 69–78 (2011)

    Article  CAS  Google Scholar 

  22. Patton, A. et al. Endocytosis function of a ligand-gated ion channel homolog in Caenorhabditis elegans. Curr. Biol. 15, 1045–1050 (2005)

    Article  CAS  Google Scholar 

  23. Nath, S. et al. Spreading of neurodegenerative pathology via neuron-to-neuron transmission of β-amyloid. J. Neurosci. 32, 8767–8777 (2012)

    Article  CAS  Google Scholar 

  24. Nussbaum-Krammer, C. I., Park, K. W., Li, L., Melki, R. & Morimoto, R. I. Spreading of a prion domain from cell-to-cell by vesicular transport in Caenorhabditis elegans. PLoS Genet. 9, e1003351 (2013)

    Article  CAS  Google Scholar 

  25. Costanzo, M. et al. Transfer of polyglutamine aggregates in neuronal cells occurs in tunneling nanotubes. J. Cell Sci. 126, 3678–3685 (2013)

    Article  CAS  Google Scholar 

  26. Abounit, S. & Zurzolo, C. Wiring through tunneling nanotubes—from electrical signals to organelle transfer. J. Cell Sci. 125, 1089–1098 (2012)

    Article  CAS  Google Scholar 

  27. Gousset, K. et al. Prions hijack tunnelling nanotubes for intercellular spread. Nat. Cell Biol. 11, 328–336 (2009)

    Article  CAS  Google Scholar 

  28. Pearce, M. M., Spartz, E. J., Hong, W., Luo, L. & Kopito, R. R. Prion-like transmission of neuronal huntingtin aggregates to phagocytic glia in the Drosophila brain. Nat. Commun. 6, 6768 (2015)

    Article  ADS  CAS  Google Scholar 

  29. Pasquier, J. et al. Preferential transfer of mitochondria from endothelial to cancer cells through tunneling nanotubes modulates chemoresistance. J. Transl. Med. 11, 94 (2013)

    Article  CAS  Google Scholar 

  30. Hayakawa, K. et al. Transfer of mitochondria from astrocytes to neurons after stroke. Nature 535, 551–555 (2016)

    Article  ADS  CAS  Google Scholar 

  31. Brenner, S. The genetics of Caenorhabditis elegans. Genetics 77, 71–94 (1974)

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Duan, Z. & Sesti, F. A Caenorhabditis elegans model system for amylopathy study. J. Vis. Exp. 75, 50435 (2013)

    Google Scholar 

  33. Mapes, J. et al. CED-1, CED-7, and TTR-52 regulate surface phosphatidylserine expression on apoptotic and phagocytic cells. Curr. Biol. 22, 1267–1275 (2012)

    Article  CAS  Google Scholar 

  34. Neumann, B. et al. EFF-1-mediated regenerative axonal fusion requires components of the apoptotic pathway. Nature 517, 219–222 (2015)

    Article  ADS  CAS  Google Scholar 

  35. Kachur, T. M., Audhya, A. & Pilgrim, D. B. UNC-45 is required for NMY-2 contractile function in early embryonic polarity establishment and germline cellularization in C. elegans. Dev. Biol. 314, 287–299 (2008)

    Article  CAS  Google Scholar 

  36. Kamath, R. S. & Ahringer, J. Genome-wide RNAi screening in Caenorhabditis elegans. Methods 30, 313–321 (2003)

    Article  CAS  Google Scholar 

  37. Calixto, A., Chelur, D., Topalidou, I., Chen, X. & Chalfie, M. Enhanced neuronal RNAi in C. elegans using SID-1. Nat. Methods 7, 554–559 (2010)

    Article  CAS  Google Scholar 

  38. Liu, J. et al. Beclin1 controls the levels of p53 by regulating the deubiquitination activity of USP10 and USP13. Cell 147, 223–234 (2011)

    Article  CAS  Google Scholar 

  39. Scerbak, C. et al. Insulin signaling in the aging of healthy and proteotoxically stressed mechanosensory neurons. Front. Genet. 5, 212 (2014)

    Article  Google Scholar 

  40. Topalidou, I. et al. Genetically separable functions of the MEC-17 tubulin acetyltransferase affect microtubule organization. Curr. Biol. 22, 1057–1065 (2012)

    Article  CAS  Google Scholar 

  41. Gabel, C. V., Antoine, F., Chuang, C. F., Samuel, A. D. & Chang, C. Distinct cellular and molecular mechanisms mediate initial axon development and adult-stage axon regeneration in C. elegans. Development 135, 1129–1136 (2008)

    Article  CAS  Google Scholar 

  42. Kopito, R. R. Aggresomes, inclusion bodies and protein aggregation. Trends Cell Biol. 10, 524–530 (2000)

    Article  CAS  Google Scholar 

  43. Hermann, G. J. et al. Genetic analysis of lysosomal trafficking in Caenorhabditis elegans. Mol. Biol. Cell 16, 3273–3288 (2005)

    Article  CAS  Google Scholar 

  44. Zhou, Z., Hartwieg, E. & Horvitz, H. R. CED-1 is a transmembrane receptor that mediates cell corpse engulfment in C. elegans. Cell 104, 43–56 (2001)

    Article  CAS  Google Scholar 

  45. Hong, Y., Roy, R. & Ambros, V. Developmental regulation of a cyclin-dependent kinase inhibitor controls postembryonic cell cycle progression in Caenorhabditis elegans. Development 125, 3585–3597 (1998)

    CAS  PubMed  Google Scholar 

  46. Kastelowitz, N. & Yin, H. Exosomes and microvesicles: identification and targeting by particle size and lipid chemical probes. ChemBioChem. 15, 923–928 (2014)

    Article  CAS  Google Scholar 

  47. Cocucci, E. & Meldolesi, J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 25, 364–372 (2015)

    Article  CAS  Google Scholar 

  48. Thery, C., Zitvogel, L. & Amigorena, S. Exosomes: composition, biogenesis and function. Nat. Rev. Immunol. 2, 569–579 (2002)

    Article  CAS  Google Scholar 

  49. Ma, L. et al. Discovery of the migrasome, an organelle mediating release of cytoplasmic contents during cell migration. Cell Res. 25, 24–38 (2015)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank B. Grant for expert advice, C. Reina for time-lapse microscopy help; N. Kane and J. Kramer for confocal microscopy assistance; and H. Ushakov for construction of some genetic lines. We thank G. Perumal and F. Macaluso for help with HPF fixations, and C. Crocker for the cartoon in Extended Data Fig. 2b. F. Sesti and M. Hilliard supplied C. elegans strains; A. Mendenhall, B. Sands and R. Brent constructed the MOSCI mitoTimer strain. Research was supported by the National Institutes of Health under award numbers 1R01NS086064 and 1R01AG046358. I.M. and R.J.G. were supported by the National Institute of General Medical Sciences under award number T32 GM008339. K.C.N. and D.H.H. were supported by NIH OD10943 (to D.H.H.); Core EM facilities (D.H.H.) NICHD P30 HD71593 for the RFK-IDDRC at Albert Einstein College of Medicine. Some strains were provided by the CGC, which is funded by NIH Office of Research Infrastructure Programs (P40 OD010440). Content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

I.M., M.L.T., M.L.A., R.J.G. and G.H. conducted and designed experiments along with M.D. I.M. and M.D. wrote the manuscript with input from R.J.G., M.L.A. and M.L.T. C.V.G. and D.T. carried out calcium connection experiments. K.C.N. and D.H.H. carried out electron microscopy. J.A.P. and C.N. supplied the Q128 reagent and manuscript critiques.

Corresponding author

Correspondence to Monica Driscoll.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks C. Bargmann, D. Divizio and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Morphological features of exophers derived from touch neurons.

a, An exopher is generated with evident filling and growth. The exopher size increased for more than 1 h, specifically in the exopher compartment, possibly via continual delivery of materials to the exopher after initial formation (see Supplementary Video 2). Strain is Is[Pmec-4mCh1], adult day 2. b, An ALMR soma with multiple exophers. Strain is Is[Pmec-4mCh2], adult day 2. c, A rare instance of an ALM neuron with exophers that appear extruded from the dendrite (arrows). Strain is Is[Pmec-4mCh1], adult day 2. d, Size measurements for somas (squares) and exophers (circles). Data are combined for exophers scored in different backgrounds, n = 35. Values analysed in Extended Data Table 2. e, Example of a exopher derived from a dye-filled amphid neuron. We identified exophers from dye-filled amphids, some of which appeared attached. Neuronal exophers are thus not induced solely in response to expression of foreign proteins, but can be produced from neurons that express only native proteins. See Extended Data Fig. 6c for a second example of a dye-filled exopher. f, Early adult longitudinal time-course on DiI-soaked wild-type N2. Dye-filled chemosensory amphid neurons also produce exophers with a peak early in life in wild-type animals. The production of exophers in this study reflects the extrusion of native neuronal contents, as no fluorescent transgene is introduced. Total n > 150, 3 trials. g, h, Dopaminergic PDE and CEP neurons can produce exophers. g, GFP-expressing PDE neuron with an anterior exopher indicated (8 out of 450 had exophers, typical of the low rate observed with GFP reporters in touch neurons). h, CEP neuron with an associated exopher. Strain is egIs1[Pdat-1GFP]; adult day 2. i, ASER neurons can form exophers. Strain is sesIs25[Pflp-6Aβ; Pgcy-5GFP]32; adult day 2. j, The onset of touch neuron exophers in an hsf-1 mutant background occurs 1 day earlier than wild-type touch neurons, beginning on adult day 1, but follows the general trend of high incidence early in adult life. Longitudinal study with Is[Pmec-4GFP]; hsf-1(sy441) (2 green trials, starting n = 25), and Is[Pmec-4mCh1]; hsf-1(sy441) (red, starting n = 25). We observed a similar temporal pattern for Is[Pmec-4mCh2]; [Pmec-3Q128CFP] (data not shown). A late onset peak might not be apparent owing to sickness of hsf-1 mutants later in life. Data in d and f are mean ± s.e.m. Data in j are from single longitudinal trials and thus error bars are not included. Scale bars, 2 μm.

Extended Data Figure 2 Electron microscopy images of an extruded exopher.

a, A membrane GFP reporter in a PVM neuron reveals that exophers contain membrane. Strain is Pmec-4PH(plcDelta)::GFP (ref. 35). b, Relationship of exopher to ALMR soma. Schematic view from a lateral aspect (anterior to the left). ALMR soma remains connected to its primary dendrite. Several smaller membrane-bound tubes extend away from the soma, containing small expelled items, such as the large vesicle shown. The ALMR nucleus (N) is intact, but pushed to an eccentric position by cytoplasmic inclusions. The soma contains intact rough endoplasmic reticulum (RER), mitochondria (M), small vesicles (not shown), and larger protein aggregates (A). The exopher comprises heterogeneous contents. Exopher exterior is completely bounded by hypodermal plasma membrane, so exopher contents are not in immediate contact with hypodermal cytoplasm. A double membrane is often observed where the exopher is likely to supply the inner bilayer, and the hypodermis contributes the outer bilayer. Additional, separate membrane-bound items lie peripherally (not shown; but see h), which may be breakdown products from an earlier, larger stage of the exopher. Most internal exopher contents have their own membrane boundaries, but diffuse material (not shown; but see c, e) fills spaces around those membrane-bound objects. Membrane-bound contents include portions of neuronal cytoplasm holding intact RER, large protein aggregates, and complex whorls of membrane (W) that seem to enclose empty space. Two lysosomes (L) are shown, one in the process of fusing to exopher outer membrane. A tube is shown extending (far right) towards the pseudocoelom, which might offer a route for elimination of contents that cannot be degraded during hypodermal transit. Cartoon designed by and published with permission of C. Crocker. c, d, f, h, TEM views of an exopher, emitted from cell body in e. c, The exopher is fully embedded within hypodermis (H), seen from anterior aspect (thus left/right reversed). Strain is Is[Pmec-4mCh1]; Is[Pmec-4GFP]. C, animal cuticle. Exopher is 1.5 μm, similar in size to the excretory canal (EC), and lies closer to the pseudocoelom (P), whereas the ALMR neuron soma lies closer to cuticle (see e). Jagged white spaces running vertically through hypodermis are an artefact where tissue cracked during processing. d, The exopher is characterized by many small round protrusions and involuted portions, with multiple membrane layers. The main exopher complex has a complete plasma membrane surrounding it. e, The originating ALMR neuron still has intact cell and nuclear membranes. Aggregate within soma is not membrane bound, resembling a mammalian aggresome42. Electron density of the neuronal cytoplasm is darker than that of surrounding hypodermis, and mitochondria of hypodermis are far larger than those of the neuron. f, The exopher is surrounded by a continuous membrane and contains electron-lucent materials and electron-dense membrane whorls. g, Fluorescent microscopy of exopher in a day 2 animal, expressing soluble YFP, CFP-tagged Q128 fusion, and aggregation-prone mCherry. mCherry has a bright spot that excludes the other two signals (arrowhead). Q128–CFP and YFP are localized in the middle and bottom of exopher, respectively. YFP signal also forms a dim ring around mCherry spot. Lateral aspect shown, as in a. h, Thin sections (1–7, from 50 serial sections) through an exopher reveal a complex and heterogeneous structure. Additional membrane-bound objects at fringe of exopher main body (see panels 6, 7) may represent portions that have decayed from the original larger object, and are perhaps more easily phagocytosed by hypodermis, or shuttled along a tube for release into pseudocoelom. A small electron dense lysosome (L) lies beside exopher in panel 5. Scale bars, 2 μm (a, c, e, g), 1 μm (d, f, h).

Extended Data Figure 3 Fluorescence recovery after photo-bleaching (FRAP) and post-axotomy calcium imaging indicate that exophers that appear connected to the soma can fill with cytoplasmic materials.

a, Both connected and unconnected exophers can be identified at high frequency in the same strain with a 40× objective. Strain is Is[Pmec-4mCh1], adult day 2; n = 77 total, 3 trials. Data are mean ± s.e.m. b, Example of a connected ALMR exopher recovering after laser bleaching. Strain is Is[Pmec-4mCh1]; Is[Pmec-4GFP], adult day 2. Before laser treatment is ‘0 s’, other times are post-treatment. c, Example of a detached ALMR exopher photo-bleaching and failing to refill. Strain is Is[Pmec-4mCh1]; Is[Pmec-4GFP], adult day 2. d, Fluorescence recovery measurements reveal that some connected exophers are able to transport fluorescent material from the cytoplasm to the exopher, whereas disconnected exophers do not appear to transport fluorescent material. Shown are data for examples in b (red, connected) and c (blue, unconnected) above. e, Time-lapse measurements of fluorescence intensity of the soma (blue trace) and the exopher (red trace) during the laser axotomy experiment in f show that the injury-induced calcium wave in the soma is followed by a pulse of calcium increase in the exopher (red arrow, laser axotomy at t = 0 s). We analysed two individual neurons with exophers connected to the soma and three individual neurons with what appeared to be non-connected exophers. Only the clearly connected exophers gave a calcium response comparable (~100% signal) to that measured at the cell soma as in this panel. Strain is ZB4059 bzIs163[Pmec-4::GCaMP3.0::SL2::mCherry]. f, The soma calcium wave induced by laser axotomy is followed by a calcium wave to connected exophers. We laser-cut an ALMR neuron that had a connected exopher in a day 2 adult that expressed both mCherry and the calcium-sensitive fluorophore, GCaMP3. We made the laser cut 20 μm along the axon (yellow arrow) at time t = 0, while taking simultaneous time-lapse images (1 frame per 5 s). Selected frames are shown at t = −20 s (before laser axotomy), right after laser axotomy at t = 5 s (note increased fluorescence in the soma; white arrow), and at t = 15 s and 25 s post-axotomy (note the later exopher increase in fluorescence; white arrowheads). Signal quantification is in e. Supplementary Video 3 shows the calcium wave that travels from soma to exopher.

Extended Data Figure 4 Lysosomes can be found in exophers.

GFP-tagged lysosomes in touch neurons (bzIs168[Pmec-7LMP-1::GFP]) can be extruded in exophers in two types of lysosomal arrangements: (1) those that have small lysosome-like concentrated fluorescence, with mCherry dispersed (d), and (2) those that are nearly fully loaded with lysosome-like staining in which mCherry is also present throughout. a, Neuron soma featuring typical two large LMP-1::GFP-tagged pericentric lysosome domains, with no smaller ones evident. Strain is ZB4509 Is[Pmec-4mCh1]; bzIs168[Pmec-7LMP-1::GFP], green channel shown. As observed previously43, the LMP-1::GFP signal clearly marks the plasma membrane (M), but less intensely than the lysosomes (L). b, LMP-1::GFP reveals lysosome inclusions are frequent and sometimes prominent in exophers. Strain is ZB4070 bzIs168[Pmec-7LMP-1::GFP]. We found that 18 out of 25 (72%) of exophers scored contained lysosomes in day 2 adults. Note that LMP-1::GFP faintly labels membrane43 and rings the exopher in bd, supporting that the exopher is membrane-bound. c, d, Co-labelling of aggregating mCherry and lysosome compartments suggests two types of lysosomal organization in exophers. Strain is ZB4509 Is[Pmec-4mCh1]; bzIs168[Pmec-7LMP-1::GFP]. c, Some exophers appear to be filled with LMP-1::GFP and coincident mCherry. d, LMP-1::GFP-tagged lysosomes included in exophers can be small and differentially localized from mCherry. In the absence of stress, neurons typically feature two large intensely fluorescent pericentric lysosome domains with LMP-1::GFP, with few smaller ones evident (see a). Neurons that had an exopher tended to also have additional small mobile lysosomes that we did not observe in cells without an exopher (see bd). Additionally, neurons that featured ‘large lysosome’ exophers generally appeared to have fewer of the large perinuclear lysosomes in the soma (example in d). Scale bars, 2 μm. The inclusion of lysosomes in exophers suggests that some elimination of expelled material might be accomplished via internal degradation. Alternatively, dysfunctional lysosomes might be expelled via exophers.

Extended Data Figure 5 Mitochondria GFP markers exhibit a normal mitochondrial appearance in exophers.

a, Mitochondria in exophers can form a network. Strain is Is[Pmec-4mitoLS::ROGFP]; Is[Pmec-4mCh1]). Shown is an exopher budding off from the ALMR soma. The exopher contains a disproportionate number of punctate mCherry aggregates, and also includes a GFP signal typical in size for neuronal mitochondria. The mitochondria in the exopher exhibit a filamentous structure similar to those in the soma, and the signal does not co-localize with the mCherry signal but may remain within a distinct sub-cellular domain. These two observations are consistent with the mitoGFP label localized to actual mitochondria as opposed to representing mislocalized GFP-labelled protein. b, Exophers can contain punctate mitochondria, networked mitochondria, or no mitochondria. Strain is Is[Pmec-4mitoLS::ROGFP]; Is[Pmec-4mCh1]). In the left exopher, the mitoROGFP signal is localized to two puncta. The middle exopher contains networked mitochondria. The right exopher contains no visible mitoROGFP signal. c, Zoomed out view of b, to show location of exophers relative to the touch neuron soma. Scale bars, 2 μm (ac). d, MitoTimer fluorescent reporter reveals a difference in the mitochondrial matrix oxidation environment in exophers versus somas. We used a single-copy Pmec-4MitoTimer reporter to measure the relative red/green signal in exopher–soma pairs. Exophers have proportionately more oxidized signal, suggesting ‘older’ mitochondria (with more oxidation of the matrix-localized reporter) are preferentially expelled. n = 7 exophers, single exopher mean ± s.e.m. *P < 0.05, paired t-test. e, Pharmacological disruption of mitochondria leads to higher rates of mitochondrial inclusion in exophers. Strain Is[Pmec-4mCh1]; zhsEx17[Pmec-4mitoLS::ROGFP] was treated with 230 μM juglone, which increases ROS production. Exophers from ALMR neurons exposed to juglone (blue, n = 30 total exophers) were significantly more likely to include at least one mitochondrion than untreated control exophers (white, n = 22 total exophers); 3 trials. Mitochondrial extrusion increases under conditions of juglone-induced oxidative stress. Data are mean ± s.e.m. **P < 0.01, unpaired t-test.

Extended Data Figure 6 RNAi knockdown of ced-1 and ced-6, but not other engulfment machinery, increases occurrence of mutiple exopher detection.

a, RNAi knockdown of ced-1 and ced-6 engulfment genes increases the number of ALMR neurons with ≥2 exophers near the touch neurons, supporting conclusions from mutants. Control is empty vector, strain is ZB4071 bzIs169[Pmec-18sid-1Psng-1YFP]; bzIs101[Pmec-4mCherry], at least 3 trials each, n > 30 ALMRs measured per trial; n > 15 cells with exopher per condition graphed. Data are mean ± s.e.m. *P < 0.05, **P < 0.01, one-way ANOVA with Dunnett’s test. ced-1 and ced-6 RNAi do not increase the percentage of ALMRs that produce exophers (data not shown). b, Phosphatidylserine indicator annexinV::GFP (ref. 44) labels apoptotic corpses, but not mCherry-labelled exophers in strain ZB4083 smIS76[Phsp-16ANV::GFP]; Is[Pmec-4mCh1]. Phosphatidylserine can be recognized on corpses of necrotic touch neurons, showing that touch neurons can produce surface phosphatidylserine and be recognized by annexinV-tagging, when inappropriately induced to die33,44. 0 out of 18 fluorescent mCherry-labelled exophers were co-labelled with annexinV::GFP. ced-1 RNAi in the annexinV::GFP line did not improve phosphatidylserine detection on exophers (n = 0 out of 25 additional observations; not shown). c, In a DiI-soaked N2 animal, an amphid exopher originating from the ASIR soma can be seen proximal to the terminal bulb of the pharynx (P). The anterior coelomocytes ccAR and ccPR (C) also contain DiI, which must have been taken in and then jettisoned by the chemosensory amphid neurons and subsequently engulfed, analogously to the mCherry detected in Is[Pmec-4mCh1] coelomocytes. Thus, coelomocytes can scavenge the contents of exophers that are generated under normal physiological conditions, without the added stress of a potentially aggregating product of a transgene. Scale bar, 5 μm. d, In ZB4082 cup-4(ok837); Is[Pmec-4mCh1] mutants in which coelomocyte uptake is disrupted, an increased incidence of dispersed (D) fluorescence occurs (bracket denotes ‘starry night phenotype’, present in 29 out 200 animals, adult day 4). Similar dispersions are rare in cup-4(+) lines. Scale bar, 2 μm. AVM soma is also visible. e, Young adult animals that produced an exopher often later exhibit the starry night phenotype, suggesting that mCherry material can move through the body. Is[Pmec-4mCh1] animals were separated into populations that had an ALMR exopher on adult days 2 or 3 (blue arrows), and a population that had no ALMR exopher on days 2 or 3 (white arrows). Animals were scored again on day 5 for presence of the starry night phenotype. In the exopher-producing and non-exopher-producing groups, 42% and 6%, respectively, of animals exhibited a starry night phenotype. n = 60 total per group, 3 trials. Data are mean ± s.e.m. *P < 0.05, unpaired t-test. Arrow thickness is weighted according to relative incidence. Note differences are likely to be underestimated here, as the ‘no exopher’ category should include animals that have produced exophers, but were not present at the time of sampling.

Extended Data Figure 7 Working model for a proposed exopher role in proteostasis.

As neurotoxic events such as protein aggregation or mitochondrial dysfunction occur in the cell, several homeostatic mechanisms clear them (left panel). At the young adult transition point to adult proteostasis (heat shock response down, unfolded protein response down, proteasome activity up12,13,14) or when basal levels of damage reach a threshold and overwhelm neuronal proteostasis, aggregates and organelles such as mitochondria and lysosomes are sequestered into a compartment that can be jettisoned from the cell. This compartment might include aggresomes described in mammalian cells42. For touch neurons, extruded exopher contents may be degraded by accompanying lysosomes, digested by the surrounding hypodermis, or may be re-extruded and reach the pseudocoelom to be taken up by coelomocytes. The process of exopher-genesis appears to be neuroprotective in young adults, but when dysregulated, might induce toxicity in neighbouring tissues. We speculate that exopher contents that cannot be degraded or passed on could remain in the neighbouring cell, where they could contribute to dysfunction. Exopher-genesis may be akin to the process by which protein aggregates and mitochondria become localized to neighbouring cells in humans, promoting the spread of disease.

Extended Data Table 1 Effect of RNAi knockdown of genes functioning in exosome biogenesis and cell cycle progression on exopher detection
Extended Data Table 2 Comparison of features of exophers to other characterized extracellular vesicles

Supplementary information

An exopher is generated with a striking concentration of fluorescence segregated to the extrusion.

Strain is Is[pmec-4mCh2]. ALM neuron with mCherry-visualized cytoplasm and aggregates. (MP4 355 kb)

An exopher is generated with evident filling and growth.

S indicates the soma of an ALM neuron on adult day 2 with mCherry visualized; E indicates the significant extrusion of a balloon-like exopher, which grows with time. We noted that the size of this exopher increased for more than an hour, with fluorescence intensity increasing specifically in the exopher compartment, possibly via continual delivery of materials to the exopher after the initial formation. Strain is Is[pmec-4mCh1]. (MP4 98 kb)

The soma calcium wave induced by laser axotomy is followed by a calcium wave to connected exophers.

We laser-cut an ALMR neuron that had a connected exopher in a day 2 adult that expressed both mCherry(bottom) and the calcium sensitive fluorophore, GCaMP3(top). Video shows the calcium wave that travels from soma to exopher. (MP4 236 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Melentijevic, I., Toth, M., Arnold, M. et al. C. elegans neurons jettison protein aggregates and mitochondria under neurotoxic stress. Nature 542, 367–371 (2017). https://doi.org/10.1038/nature21362

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature21362

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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