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:

CLN3 is required for the clearance of glycerophosphodiesters from lysosomes

Abstract

Lysosomes have many roles, including degrading macromolecules and signalling to the nucleus1. Lysosomal dysfunction occurs in various human conditions, such as common neurodegenerative diseases and monogenic lysosomal storage disorders (LSDs)2,3,4. For most LSDs, the causal genes have been identified but, in some, the function of the implicated gene is unknown, in part because lysosomes occupy a small fraction of the cellular volume so that changes in lysosomal contents are difficult to detect. Here we develop the LysoTag mouse for the tissue-specific isolation of intact lysosomes that are compatible with the multimodal profiling of their contents. We used the LysoTag mouse to study CLN3, a lysosomal transmembrane protein with an unknown function. In children, the loss of CLN3 causes juvenile neuronal ceroid lipofuscinosis (Batten disease), a lethal neurodegenerative LSD. Untargeted metabolite profiling of lysosomes from the brains of mice lacking CLN3 revealed a massive accumulation of glycerophosphodiesters (GPDs)—the end products of glycerophospholipid catabolism. GPDs also accumulate in the lysosomes of CLN3-deficient cultured cells and we show that CLN3 is required for their lysosomal egress. Loss of CLN3 also disrupts glycerophospholipid catabolism in the lysosome. Finally, we found elevated levels of glycerophosphoinositol in the cerebrospinal fluid of patients with Batten disease, suggesting the potential use of glycerophosphoinositol as a disease biomarker. Our results show that CLN3 is required for the lysosomal clearance of GPDs and reveal Batten disease as a neurodegenerative LSD with a defect in glycerophospholipid metabolism.

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

Fig. 1: LysoTag mouse for proteomic and metabolite profiling of tissue lysosomes.
Fig. 2: Loss of CLN3 leads to significant alterations in metabolite levels in brain lysosomes.
Fig. 3: GPDs accumulate in lysosomes after CLN3 loss.
Fig. 4: CLN3 is required for the efflux of GPDs out of lysosomes.

Similar content being viewed by others

Data availability

The MS proteomics data were deposited to the ProteomeXchange Consortium through the PRIDE partner repository with the dataset identifier PXD018624. Known lysosomal proteins in our proteomics analysis were defined based on Gene Ontology Cellular Component or UniProt subcellular localization annotation (https://www.uniprot.org/). The conditional LysoTag mouse was deposited at the Jackson Laboratory (strain 035401). Other unique biological materials in the form of plasmids or cell lines are available from the corresponding author on request. Detailed lipidomics and metabolomics data are provided in Supplementary Tables 3 and 4, respectively. Other data generated are available from the corresponding author on request. Source data are provided with this paper.

References

  1. Ballabio, A. & Bonifacino, J. S. Lysosomes as dynamic regulators of cell and organismal homeostasis. Nat. Rev. Mol. Cell Biol. 21, 101–118 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Medoh, U. N., Chen, J. Y. & Abu-Remaileh, M. Lessons from metabolic perturbations in lysosomal storage disorders for neurodegeneration. Curr. Opin. Syst. Biol. 29, 100408 (2022).

    Article  CAS  Google Scholar 

  3. Platt, F. M., d'Azzo, A., Davidson, B. L., Neufeld, E. F. & Tifft, C. J. Lysosomal storage diseases. Nat. Rev. Dis. Primers 4, 27 (2018).

    Article  PubMed  Google Scholar 

  4. Ferguson, S. M. Neuronal lysosomes. Neurosci. Lett. 697, 1–9 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Perera, R. M. & Zoncu, R. The lysosome as a regulatory hub. Annu. Rev. Cell Dev. Biol. 32, 223–253 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Savini, M., Zhao, Q. & Wang, M. C. Lysosomes: signaling hubs for metabolic sensing and longevity. Trends Cell Biol. 29, 876–887 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ballabio, A. & Gieselmann, V. Lysosomal disorders: from storage to cellular damage. Biochim. Biophys. Acta 1793, 684–696 (2009).

    Article  CAS  PubMed  Google Scholar 

  8. Boustany, R. M. Lysosomal storage diseases—the horizon expands. Nat. Rev. Neurol. 9, 583–598 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Marques, A. R. A. & Saftig, P. Lysosomal storage disorders—challenges, concepts and avenues for therapy: beyond rare diseases. J. Cell Sci. 132, jcs221739 (2019).

    Article  PubMed  Google Scholar 

  10. Wallings, R. L., Humble, S. W., Ward, M. E. & Wade-Martins, R. Lysosomal dysfunction at the centre of Parkinson's disease and frontotemporal dementia/amyotrophic lateral sclerosis. Trends Neurosci. 42, 899–912 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang, C., Telpoukhovskaia, M. A., Bahr, B. A., Chen, X. & Gan, L. Endo-lysosomal dysfunction: a converging mechanism in neurodegenerative diseases. Curr. Opin. Neurobiol. 48, 52–58 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Abu-Remaileh, M. et al. Lysosomal metabolomics reveals V-ATPase- and mTOR-dependent regulation of amino acid efflux from lysosomes. Science 358, 807–813 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Thai, T. H. et al. Regulation of the germinal center response by microRNA-155. Science 316, 604–608 (2007).

    Article  ADS  CAS  PubMed  Google Scholar 

  14. Pisoni, R. L., Acker, T. L., Lisowski, K. M., Lemons, R. M. & Thoene, J. G. A cysteine-specific lysosomal transport system provides a major route for the delivery of thiol to human fibroblast lysosomes: possible role in supporting lysosomal proteolysis. J. Cell Biol. 110, 327–335 (1990).

    Article  CAS  PubMed  Google Scholar 

  15. Eiberg, H., Gardiner, R. M. & Mohr, J. Batten disease (Spielmeyer-Sjogren disease) and haptoglobins (HP): indication of linkage and assignment to chr. 16. Clin. Genet. 36, 217–218 (1989).

    Article  CAS  PubMed  Google Scholar 

  16. Lerner, T.J. et al. Isolation of a novel gene underlying Batten disease, CLN3. Cell 82, 949–957 (1995).

    Article  Google Scholar 

  17. Mirza, M. et al. The CLN3 gene and protein: what we know. Mol. Genet. Genomic Med. 7, e859 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Butz, E. S., Chandrachud, U., Mole, S. E. & Cotman, S. L. Moving towards a new era of genomics in the neuronal ceroid lipofuscinoses. Biochim. Biophys. Acta Mol. Basis Dis. 1866, 165571 (2019).

  19. Jarvela, I. et al. Biosynthesis and intracellular targeting of the CLN3 protein defective in Batten disease. Hum. Mol. Genet. 7, 85–90 (1998).

    Article  CAS  PubMed  Google Scholar 

  20. Storch, S., Pohl, S. & Braulke, T. A dileucine motif and a cluster of acidic amino acids in the second cytoplasmic domain of the batten disease-related CLN3 protein are required for efficient lysosomal targeting. J. Biol. Chem. 279, 53625–53634 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Mao, Q., Foster, B. J., Xia, H. & Davidson, B. L. Membrane topology of CLN3, the protein underlying Batten disease. FEBS Lett. 541, 40–46 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Ezaki, J. et al. Characterization of Cln3p, the gene product responsible for juvenile neuronal ceroid lipofuscinosis, as a lysosomal integral membrane glycoprotein. J. Neurochem. 87, 1296–1308 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Oetjen, S., Kuhl, D. & Hermey, G. Revisiting the neuronal localization and trafficking of CLN3 in juvenile neuronal ceroid lipofuscinosis. J. Neurochem. 139, 456–470 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Perland, E., Bagchi, S., Klaesson, A. & Fredriksson, R. Characteristics of 29 novel atypical solute carriers of major facilitator superfamily type: evolutionary conservation, predicted structure and neuronal co-expression. Open Biol. 7, 170142 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Mitchison, H. M. et al. Targeted disruption of the Cln3 gene provides a mouse model for Batten disease. The Batten Mouse Model Consortium [corrected]. Neurobiol. Dis. 6, 321–334 (1999).

    Article  CAS  PubMed  Google Scholar 

  26. Kovacs, A. D. & Pearce, D. A. Finding the most appropriate mouse model of juvenile CLN3 (Batten) disease for therapeutic studies: the importance of genetic background and gender. Dis. Model. Mech. 8, 351–361 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lojewski, X. et al. Human iPSC models of neuronal ceroid lipofuscinosis capture distinct effects of TPP1 and CLN3 mutations on the endocytic pathway. Hum. Mol. Genet. 23, 2005–2022 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Platt, F. M. Sphingolipid lysosomal storage disorders. Nature 510, 68–75 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  29. Fuller, M. & Futerman, A. H. The brain lipidome in neurodegenerative lysosomal storage disorders. Biochem. Biophys. Res. Commun. 504, 623–628 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Hobert, J. A. & Dawson, G. A novel role of the Batten disease gene CLN3: association with BMP synthesis. Biochem. Biophys. Res. Commun. 358, 111–116 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Padilla-Lopez, S., Langager, D., Chan, C. H. & Pearce, D. A. BTN1, the Saccharomyces cerevisiae homolog to the human Batten disease gene, is involved in phospholipid distribution. Dis. Model. Mech. 5, 191–199 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Kopp, F. et al. The glycerophospho metabolome and its influence on amino acid homeostasis revealed by brain metabolomics of GDE1(−/−) mice. Chem. Biol. 17, 831–840 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Allen, F., Pon, A., Wilson, M., Greiner, R. & Wishart, D. CFM-ID: a web server for annotation, spectrum prediction and metabolite identification from tandem mass spectra. Nucleic Acids Res. 42, W94–W99 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sumner, L. W. et al. Proposed minimum reporting standards for chemical analysis Chemical Analysis Working Group (CAWG) Metabolomics Standards Initiative (MSI). Metabolomics 3, 211–221 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dang Do, A. N. et al. Neurofilament light chain levels correlate with clinical measures in CLN3 disease. Genet. Med. 23, 751–757 (2021).

    Article  CAS  PubMed  Google Scholar 

  36. Fowler, S. & De Duve, C. Digestive activity of lysosomes. 3. The digestion of lipids by extracts of rat liver lysosomes. J. Biol. Chem. 244, 471–481 (1969).

    Article  CAS  PubMed  Google Scholar 

  37. Schmidtke, C. et al. Lysosomal proteome analysis reveals that CLN3-defective cells have multiple enzyme deficiencies associated with changes in intracellular trafficking. J. Biol. Chem. 294, 9592–9604 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Corda, D. et al. The emerging physiological roles of the glycerophosphodiesterase family. FEBS J. 281, 998–1016 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Patton-Vogt, J. Transport and metabolism of glycerophosphodiesters produced through phospholipid deacylation. Biochim. Biophys. Acta 1771, 337–342 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Rigoni, M. et al. Equivalent effects of snake PLA2 neurotoxins and lysophospholipid-fatty acid mixtures. Science 310, 1678–1680 (2005).

    Article  ADS  CAS  PubMed  Google Scholar 

  41. Fallbrook, A., Turenne, S. D., Mamalias, N., Kish, S. J. & Ross, B. M. Phosphatidylcholine and phosphatidylethanolamine metabolites may regulate brain phospholipid catabolism via inhibition of lysophospholipase activity. Brain Res. 834, 207–210 (1999).

    Article  CAS  PubMed  Google Scholar 

  42. Storey, J. D. A direct approach to false discovery rates. J. R. Stat. Soc. B 64, 479–498 (2002).

    Article  MathSciNet  MATH  Google Scholar 

  43. Wyant, G. A. et al. NUFIP1 is a ribosome receptor for starvation-induced ribophagy. Science 360, 751–758 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    Article  CAS  PubMed  Google Scholar 

  45. Salek, R. M., Steinbeck, C., Viant, M. R., Goodacre, R. & Dunn, W. B. The role of reporting standards for metabolite annotation and identification in metabolomic studies. Gigascience 2, 13 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Bird, S. S., Marur, V. R., Sniatynski, M. J., Greenberg, H. K. & Kristal, B. S. Serum lipidomics profiling using LC-MS and high-energy collisional dissociation fragmentation: focus on triglyceride detection and characterization. Anal. Chem. 83, 6648–6657 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Taguchi, R. & Ishikawa, M. Precise and global identification of phospholipid molecular species by an Orbitrap mass spectrometer and automated search engine Lipid Search. J. Chromatogr. A 1217, 4229–4239 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Yamada, T. et al. Development of a lipid profiling system using reverse-phase liquid chromatography coupled to high-resolution mass spectrometry with rapid polarity switching and an automated lipid identification software. J. Chromatogr. A 1292, 211–218 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Hankin, J. A., Murphy, R. C., Barkley, R. M. & Gijon, M. A. Ion mobility and tandem mass spectrometry of phosphatidylglycerol and bis(monoacylglycerol)phosphate (BMP). Int. J. Mass Spectrom. 378, 255–263 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang, T., Wei, J. J., Sabatini, D. M. & Lander, E. S. Genetic screens in human cells using the CRISPR-Cas9 system. Science 343, 80–84 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  51. Krink-Koutsoubelis, N. et al. Engineered production of short-chain acyl-coenzyme A esters in Saccharomyces cerevisiae. ACS Synth. Biol. 7, 1105–1115 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Zheng, B., Berrie, C. P., Corda, D. & Farquhar, M. G. GDE1/MIR16 is a glycerophosphoinositol phosphodiesterase regulated by stimulation of G protein-coupled receptors. Proc. Natl Acad. Sci. USA 100, 1745–1750 (2003).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the members of the Abu-Remaileh and Sabatini laboratories for insights; J. M. Kirkpatrick and the FLI Core Proteomics; and the participants and families in this study for their invaluable contributions. This work was supported by grants from the NIH (DP2-CA271386), Stanford Alzheimer’s Disease Research Center (ADRC), Charles King fellowship, BeatBatten (Netherlands) and NCL-Stiftung (Germany) foundations to M.A.-R. and from the NIH (R01 CA103866 and R01 CA129105) to D.M.S. N.N.L. and W.D. were also supported by the BeatBatten (Netherlands) and NCL-Stiftung (Germany) foundations; U.N.M. by the Stanford ChEM-H Chemistry/Biology Interface Program, O’Leary-Thiry Graduate Fellowship and NIH T32 training grant (T32GM120007); A.L.C. by the F31 predoctoral NRSA (5F31DK113665); and C.G. and R.T. were partially supported by funding from the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation), Projektnummer 23928380—TRR 152 and the NCL-Stiftung (Germany); A.O. by DFG through the Research Training Group ProMoAge (GRK 2155), the Else Kröner Fresenius Stiftung (no. 2019_A79), the Fritz-Thyssen foundation (no. 10.20.1.022MN) and the Chan Zuckerberg Initiative Neurodegeneration Challenge Network (nos 2020-221617 and 2021-230967). The FLI is a member of the Leibniz Association and is financially supported by the Federal Government of Germany and the State of Thuringia. The CLN3 natural history study is funded by an NIH Clinical Center Bench to Bedside award and the NICHD intramural research programme (ZIA HD008989). M.A.-R. is a Terman Faculty Fellow at Stanford University.

Author information

Authors and Affiliations

Authors

Contributions

M.A.-R. and D.M.S. initiated the project and, with help from N.N.L., designed the research plan. N.N.L. and W.D. performed most of the experiments and analysed the data with help from A.L.C., V.D. and S.H.C.; S.H.C., T.K. and C.A.L. had a critical role in LC–MS analysis. W.D. also helped with analysing LC–MS data and U.N.M. generated GPD chemical standards. I.H. and A.O. designed the proteomic experiments and analysed the lysosomal proteomic data. R.T. and C.G. were consulted on CLN3 function and edited the manuscript. A.N.D.D. and F.D.P. supervised the CLN3 natural history study and provided the CSF samples. M.A.-R. and N.N.L. wrote the manuscript and D.M.S. edited it.

Corresponding author

Correspondence to Monther Abu-Remaileh.

Ethics declarations

Competing interests

M.A.-R. is a scientific advisory board member of Lycia Therapeutics. A.N.D.D. and F.D.P. have a collaborative research agreement with Amicus Therapeutics. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks David Pearce, Peter Vangheluwe and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 The effects of LysoTag expression on lysosomes in vivo.

a, In the constitutive LysoTag mice, TMEM192–3×HA is expressed across all tissues examined as determined by immunoblotting using antibodies to the HA epitope. No difference in the expression of lysosomal markers LAMP1, Cathepsin B, C, and D is observed upon the expression of TMEM192–3×HA. Vinculin and AKT were used as loading controls. Numbers indicate molecular weights in kDa according to protein standards run on the same gel. Immunoblot is representative of two independent experiments. b, Normalized body weights of mice with the indicated genotypes at 5-9 weeks of age. Mice were weighed individually. The body mass of each mouse was normalized to the mean mass of sex-matched wild-type littermates before pooling by genotype for statistical analyses using a two-tailed unpaired t-test. (n = 25, 19, and 10 for Wild type, LysoTag+, and LysoTagLSL mice, respectively.). c,d, Measurement of lysosomal hydrolase activity in brain and liver tissues of the constitutive LysoTag mice shows no effect of TMEM192–3×HA expression on lysosomal function. For measuring Cathepsin B activity, its fluorogenic substrate was incubated with homogenates of the corresponding tissues (see methods). For determining β-hexosaminidase activity, fluorogenic 4-methylumbelliferyl (MUF)- N-acetyl-β-Glucosaminide was used as a substrate (see methods). Fluorescence was measured at 37 °C and data were shown following the subtraction of background fluorescence observed in the absence of homogenate (mean ± s.e.m.; n=4). e,f, Representative transmission electron micrographs showing lysosomes from brain and liver tissues of LysoTag- (control) and LysoTag+ mice. Lysosomes were identified by the presence of a single membrane and granular, electron-dense appearance. White and Red arrowheads indicate lysosomes in cells from LysoTag- and LysoTag+ mice, respectively. n= 2 mice per group and scale bar, 500 nm. The graph showing the mean diameter of lysosomes in multiple quantified cells. The diameter of lysosome in each group was generated by measuring lysosomes using ImageJ v1.52. (For brain, n = 42 and n = 55 measured lysosomes from LysoTag- and LysoTag+ mice, respectively. For liver, n = 51 and n = 56 measured lysosomes LysoTag- and LysoTag+ mice, respectively. Data presented as mean ± s.e.m. n.s: non-significant; Two-tailed unpaired t-test (e,f). g, Heatmap presentation of the enrichment of early- and late-endosomal markers as well as those for lysosome in the LysoIP performed from mouse liver tissue. Enrichment values are derived from Supplementary Table 1. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 2 Generation and validation of LysoTag mouse model for Batten disease studies.

a, Schematic depicting the breeding strategy to generate LysoTag models for Batten disease studies. Cln3−/− mice were crossed with heterozygous constitutive LysoTag mice and their progeny were then crossed back to Cln3−/− animals to generate the indicated experimental mouse genotypes. Mouse drawing was created with BioRender. b, Expression and localization of the TMEM192–3×HA fusion protein to lysosomes in brain neurons of Cln3+/+, Cln3+/− and Cln3−/− LysoTag mice. TMEM192–3×HA, lysosomes and neurons were detected in an immunofluorescence assay using antibodies to the HA epitope the lysosomal marker, LAMP1 and the neuronal marker Neu-N, respectively. Scale bar = 5 µm, and magnified insets are labelled with white boxes in the main image. Percentage of colocalization between TMEM192–3×HA and LAMP1 shown in the right panel was measured in n = 7 cells per genotype. Data are shown as mean ± s.e.m. Micrographs are representative of at least three independent experiments. , Volcano plot comparing the lipidomic data from whole brain homogenates of Cln3+/− and Cln3−/− mice shows that only few lipids change significantly and they belong to phospholipids (yellow) and lysophosphatidylglycerol (LPG) in red. Loss of CLN3 also led to a decrease in the brain of bis(monoacylglycero)phosphate (BMP), a class of lysosomal lipids (blue). BMP/PG (cyan) annotation was used when MS/MS fragmentation was not acquired (see Supplementary Table 3 and methods for details). Horizontal line indicates a p-value of 0.05, and vertical dotted lines a fold change of 2. Each dot represents a lipid species. Data were acquired in negative ion mode and normalized to internal lipid standards for the best-matched lipid class (n = 5 and 4 for Cln3+/− and Cln3−/− mice, respectively). Female mice with an average age of 7 months were used. Data are in Supplementary Table 3. Two-tailed unpaired t-test was used.

Source data

Extended Data Fig. 3 Validation of glycerophosphodiesters (GPDs) as the metabolites that accumulate in lysosomes upon CLN3 loss.

a, Identification and annotation of metabolites using untargeted metabolomics analyses. Overview of our untargeted metabolomics experimental workflow. MS1 data were collected for every sample using full scan mode on a high-resolution accurate-mass instrument. MS/MS data were collected from pooled samples only to aid with compound identification. Data were then analysed using Thermo Compound Discoverer v3.1 to generate a list of unique features which were deconvoluted into a list of unique compounds. Differential and statistical analyses were performed. Compounds which were statistically significant in the differential analyses were validated and identified using authentic standards, when available. If no authentic standard was available, compounds were annotated by comparing MS/MS data to a database, or in silico fragmentation prediction (competitive fragmentation modelling-ID, CFM-ID). Data were then reported according to the Metabolomics Standards Initiative (MSI) guidelines. b,c, Mirror plots for GPC and GPI in negative ion mode, respectively. Fragments common to the glycerol-phosphate group are indicated with an asterisk*. Fragments common to those found in the MS/MS spectra reported by Kopp et al.32 are indicated with †. d, EICs for GPE and GPG across a range of samples were analysed alongside in-house generated standards (Std) and showed a matching retention time (RT). e,f, Mirror plots for GPE and GPG, respectively. Fragments common to the glycerol-phosphate group are indicated with an asterisk*. Fragments common to those found in the MS/MS spectra reported by Kopp et al.32 are indicated with †. g, Mirror plots showing MS/MS spectra for glycerophosphoserine from experimental samples compared to MS/MS spectra generated in silico using CFM-ID. Fragments common to the glycerol-phosphate group are indicated with an asterisk*. Fragments common to those found in the MS/MS spectra reported in Kopp et al.32 are indicated with †. h, Amplicon sequencing was used to validate CLN3 knock out cells generated using CRISPR-Cas9. Three mutant alleles were identified and all were found to generate frameshift deletions in the CLN3 coding sequence as compared to the wild-type sequence. i, Localization of Flag-CLN3 protein to lysosomes. Flag-CLN3 and lysosomes were detected in an immunofluorescence assay using antibodies to the Flag epitope and the lysosomal marker LAMP2, respectively. Scale bar = 5 μm. Micrographs are representative images of three experiments. j, Changes in GPD levels in CLN3-deficient cells expressing TMEM192–3×HA tag or LAMP1–3×HA tag. Data presented as a comparison between the increase in the lysosomal abundance of GPDs upon CLN3 loss in LAMP1–3×HA tagged lysosomes relative to that in TMEM192–3×HA tagged lysosomes (mean ± s.e.m.; n= 4 biologically independent samples). n.s: non-significant; Two-tailed unpaired t-test.

Source data

Extended Data Fig. 4 Testing the effects of CLN3 loss on lysosomal pH and cellular lipid metabolism.

a to d, CLN3 loss does not increase lysosomal pH. a, A standard calibration curve of the ratiometric dye LysoSensor Yellow/Blue DND-160. See methods for experimental details. b, Lysosomal pH in wild-type and CLN3 KO HEK293T cells as calculated using the standard curve in a. Data are presented as mean ± s.d., n=12 biologically independent samples. c, Targeted analyses of the fold changes in whole-cell and lysosomal levels of GPDs upon treatment with 500 nM Bafilomycin A1 for 6 h in CLN3 expressing HEK293T cells. Data are presented as mean ± s.d., n=3 biologically independent samples, (Two-tailed unpaired t-test). d, The levels of several amino acids whose egress from lysosomes is sensitive to the proton gradient across the lysosomal membrane are not affected by CLN3 loss. The levels of proline, alanine, and glutamate in whole cells and lysosomes were compared between CLN3 KO cells with and without CLN3 cDNA addback and upon treatment with 500 nM Bafilomycin A1 (BafA1) for 6 h. Data are presented as mean ± s.d., n=3 biologically independent samples, (Two-tailed unpaired t-test). e, The recombinant CLN3 protein does not have glycerophosphodiesterase activity. Deuterated GPC (D9-GPC) was incubated with recombinant 3×Flag–CLN3 or the positive control glycerophosphodiesterase1 (3×Flag–GDE-1) for the indicated time points. The extent of D9-GPC hydrolysis was determined by measuring the decline in its level and the increase in the levels of the product D9-choline. Data presented as mean ± s.e.m. of n = 3 biologically independent samples. f, CLN3 loss does not decrease tracer uptake. Fold change in tracer levels (D9-16:0-16:0 PC) normalized to total protein for samples measured in Fig. 4f and g. Data presented as mean ± s.e.m. of n = 4 biologically independent samples. g, CLN3 loss does not affect the biosynthesis or turnover of PC and sphingomyelin (SM) in cells. Free D9-choline was used as a tracer. Data are presented as fold changes in the whole-cell molar percent enrichment (MPE) of D9-choline-containing lipids in cortical neuron cultures prepared from Cln3−/− mice relative to those from Cln3+/− animals (mean ± s.e.m., n = 3 biologically independent samples, (Two-tailed unpaired t-test)).

Source data

Supplementary information

Source data

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Laqtom, N.N., Dong, W., Medoh, U.N. et al. CLN3 is required for the clearance of glycerophosphodiesters from lysosomes. Nature 609, 1005–1011 (2022). https://doi.org/10.1038/s41586-022-05221-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05221-y

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