Skip to main content
Log in

Modulation of virus-induced neuroinflammation by the autophagy receptor SHISA9 in mice

  • Article
  • Published:

From Nature Microbiology

View current issue Submit your manuscript

Abstract

Microglia and astrocytes are subgroups of brain glia cells that support and protect neurons within the central nervous system (CNS). At early stages of viral infection in the CNS, they are predominant responding cells and lead to recruitment of peripheral immune cells for viral clearance. Inhibitor of nuclear factor κB kinase subunit epsilon (IKKi) is critical for type I interferon signalling and inflammation, which modulate heterogenic immune responses during CNS infection. Balanced autophagy is vital to maintain brain integrity, yet regulation of autophagy and immune activity within brain glia cells is poorly understood. Here we identify SHISA9 as an autophagy cargo receptor that mediates the autophagy-dependent degradation of IKKi during herpes simplex virus type 1 infection. IKKi is recognized by SHISA9 through unanchored K48-linked poly-ubiquitin chains and bridged to autophagosome membrane components GABARAPL1. Single-cell RNA sequencing analysis shows that SHISA9 has temporal characteristics while modulating both antiviral and inflammatory responses in microglia and astrocytes at different stages during viral infection. We found that Shisa9−/− mice are highly susceptible to herpes simplex virus encephalitis, have pathogenic astrocytes and display more severe neuroinflammation compared with wild-type mice. Taken together, our study unravels a critical role of selective autophagy by orchestrating immune heterogeneity of different CNS resident cells through the SHISA9–IKKi axis.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Shisa9 deficiency resulted in enhanced innate immune response against viral infection.
Fig. 2: SHISA9 exerts distinct immune suppressive functions in microglia and astrocytes.
Fig. 3: SHISA9 targets IKKi for autophagic degradation during CNS infection.
Fig. 4: SHISA9 directly interacts with GABARAPL1 through LIR domain.
Fig. 5: Unanchored poly-ubiquitin chain is a recognition signal of SHISA9.
Fig. 6: SHISA9 protects mice from inflammatory damage of virus encephalitis through autophagy.

Similar content being viewed by others

Data availability

The MS proteomics data have been deposited to the ProteomeXchange Consortium (http://proteomecentral.proteomexchange.org) via the iProX partner repository30,31 with the dataset identifier PXD039377. RNA-seq data have been submitted to the SRA database under accession number PRJNA723487. Single cell RNA-seq data have been submitted to the GEO database under accession number GSE216271.The data that support the findings of this study are available from the corresponding authors upon reasonable request. Source data are provided with this paper.

References

  1. Klein, R. S. et al. Neuroinflammation during RNA viral infections. Annu. Rev. Immunol. 37, 73–95 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Rehwinkel, J. & Gack, M. U. RIG-I-like receptors: their regulation and roles in RNA sensing. Nat. Rev. Immunol. 20, 537–551 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zhang, X., Bai, X. C. & Chen, Z. J. Structures and mechanisms in the cGAS-STING innate immunity pathway. Immunity 53, 43–53 (2020).

    Article  CAS  PubMed  Google Scholar 

  4. Goubau, D., Deddouche, S. & Reis e Sousa, C. Cytosolic sensing of viruses. Immunity 38, 855–869 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Williams, J. L., Holman, D. W. & Klein, R. S. Chemokines in the balance: maintenance of homeostasis and protection at CNS barriers. Front. Cell Neurosci. 8, 154 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Klein, R. S., Garber, C. & Howard, N. Infectious immunity in the central nervous system and brain function. Nat. Immunol. 18, 132–141 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Reinert, L. S. et al. Sensing of HSV-1 by the cGAS-STING pathway in microglia orchestrates antiviral defence in the CNS. Nat. Commun. 7, 13348 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Johansen, T. & Lamark, T. Selective autophagy: ATG8 family proteins, LIR motifs and cargo receptors. J. Mol. Biol. 432, 80–103 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Gatica, D., Lahiri, V. & Klionsky, D. J. Cargo recognition and degradation by selective autophagy. Nat. Cell Biol. 20, 233–242 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Stolz, A., Ernst, A. & Dikic, I. Cargo recognition and trafficking in selective autophagy. Nat. Cell Biol. 16, 495–501 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Kim, N. et al. Interferon-inducible protein SCOTIN interferes with HCV replication through the autolysosomal degradation of NS5A. Nat. Commun. 7, 10631 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Conway, O., Akpinar, H. A., Rogov, V. V. & Kirkin, V. Selective autophagy receptors in neuronal health and disease. J. Mol. Biol. 432, 2483–2509 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Yamamoto, A., Nagano, T., Takehara, S., Hibi, M. & Aizawa, S. Shisa promotes head formation through the inhibition of receptor protein maturation for the caudalizing factors, Wnt and FGF. Cell 120, 223–235 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Tenoever, B. R. et al. Multiple functions of the IKK-related kinase IKKepsilon in interferon-mediated antiviral immunity. Science 315, 1274–1278 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Rajsbaum, R. et al. Unanchored K48-linked polyubiquitin synthesized by the E3-ubiquitin ligase TRIM6 stimulates the interferon-IKKepsilon kinase-mediated antiviral response. Immunity 40, 880–895 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Reyes-Turcu, F. E. et al. The ubiquitin binding domain ZnF UBP recognizes the C-terminal diglycine motif of unanchored ubiquitin. Cell 124, 1197–1208 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Cadwell, K. Crosstalk between autophagy and inflammatory signalling pathways: balancing defence and homeostasis. Nat. Rev. Immunol. 16, 661–675 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sparrer, K. M. J. et al. TRIM23 mediates virus-induced autophagy via activation of TBK1. Nat. Microbiol. 2, 1543–1557 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Oakes, J. A., Davies, M. C. & Collins, M. O. TBK1: a new player in ALS linking autophagy and neuroinflammation. Mol. Brain 10, 5 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Zeng, W. et al. Reconstitution of the RIG-I pathway reveals a signaling role of unanchored polyubiquitin chains in innate immunity. Cell 141, 315–330 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Xia, Z. P. et al. Direct activation of protein kinases by unanchored polyubiquitin chains. Nature 461, 114–119 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kang, Z. et al. Astrocyte-restricted ablation of interleukin-17-induced Act1-mediated signaling ameliorates autoimmune encephalomyelitis. Immunity 32, 414–425 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nassour, J. et al. Autophagic cell death restricts chromosomal instability during replicative crisis. Nature 565, 659–663 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Galluzzi, L. et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 25, 486–541 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Agalave, N. M., Lane, B. T., Mody, P. H., Szabo-Pardi, T. A. & Burton, M. D. Isolation, culture, and downstream characterization of primary microglia and astrocytes from adult rodent brain and spinal cord. J. Neurosci. Methods 340, 108742 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ablasser, A. & Hur, S. Regulation of cGAS- and RLR-mediated immunity to nucleic acids. Nat. Immunol. 21, 17–29 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Chen, M. et al. TRIM14 inhibits cGAS degradation mediated by selective autophagy receptor p62 to promote innate immune responses. Mol. Cell 64, 105–119 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Lun, A. T. L. et al. EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 20, 63 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Chen, T. et al. iProX in 2021: connecting proteomics data sharing with big data. Nucleic Acids Res. 50, D1522–D1527 (2022).

    Article  CAS  PubMed  Google Scholar 

  31. Ma, J. et al. iProX: an integrated proteome resource. Nucleic Acids Res. 47, D1211–D1217 (2019).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Key R&D Program of China (2020YFA0908700 to J.C.), National Natural Science Foundation of China (92042303 and 31870862 to J.C.) and Guangdong Basic and Applied Basic Research Foundation (2020B1515120090 to J.C.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. HSV-1 (KOS strain), eGFP-HSV-1 (KOS strain) and VSV (Indiana Strain) were respectively provided by G. Zhou (Guangzhou Medical University, China), M. Zeng (Sun Yat-sen University Cancer Center, China) and X. Qin (Suzhou Institute of Systems Medicine, Suzhou, China), and amplified in Vero cells.

Author information

Authors and Affiliations

Authors

Contributions

Y.Z. and L.W. performed the experiments and analysed the results. Q.L., H.X., C.Z., S.C., S.Y. and S.J. provided reagents and technical assistance. J.C. initiated and designed the project and directed the research. Y.Z., Q.L., L.W. and J.C. wrote the manuscript.

Corresponding author

Correspondence to Jun Cui.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks Juan Carlos de la Torre, Stanley Perlman and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 The expression of Shisa9 is up-regulated during HSV-1 infection.

(a) Wild-type (WT) mice (n = 3 mice per group) were intravenously infected (HSV-1) or uninfected (UI) with HSV-1 (1 × 105 PFU) for 5 days, then real-time quantitative polymerase chain reaction (qPCR) analysis was performed of indicated Shisa genes in the brain. Results were normalized to gapdh and presented as log2 (values). (b) qPCR analysis of SHISA9 in indicated human brain cell lines with or without HSV-1 (MOI = 1) infection for 24 hrs. qPCR analysis of Shisa9 in mice microglia cell line BV2 with HSV-1(MOI = 1) infection for indicated time periods. (c) U87 cells were transfected with control siRNA or SHISA2-9 siRNA for 24 hrs, and then infected with HSV-1 (MOI = 1) for 24 hrs. Cell lysates were collected to determine SHISA2-9 knockdown efficiency by qPCR. Cell supernatants were collected to detect IL-6 production by ELISA. (d) Schematic presentation of generation of Shisa9−/− mice through CRISPR/Cas9 technology. (e) Sequencing analysis of Shisa9−/− mice. In (b-c), all error bars, mean values ± SEM, P-values were determined by unpaired two-tailed Student’s t test of 3 independent biological experiments.

Source data

Extended Data Fig. 2 Identification of SHISA9-regulated pathways in brain during HSV-1 infection.

(a) Global gene expression in WT and Shisa9−/− mice brains are presented as heat map. Microarray analysis for total RNA was performed for WT and Shisa9−/− mice brain before (day 0, HSV-1 uninfected, UI) and after intracranial infection with HSV-1(1 × 102 PFU) (day 3; day 7), results are presented as log2 (TPM + 1). (b) Relative expression of genes related to ‘Chemokine’, ‘Pattern Recognition Receptor Signaling’, ‘NF-κB’ and ‘Lysosome’ pathways in WT mice and Shisa9−/− mice, results are presented as mean of TPM from RNA seq analysis at day 0 (UI), 3, 7 after intracranial infection with HSV-1 (1 × 102 PFU). (c and d) KEGG (Kyoto Encyclopedia of Genes and Genomes) enrichment analysis of up-regulated genes in Shisa9−/− mice compared with WT mice at day 3 (c), and day 7 (d) after intracranial infection with HSV-1(1 × 102 PFU). TPM, transcripts per million.

Source data

Extended Data Fig. 3 Loss of Shisa9 promotes antiviral immune responses in microglia and astrocytes.

(a) Real-time quantitative polymerase chain reaction (qPCR) analysis of the expression of Shisa9 in wild-type (WT) microglia and astrocytes infected with HSV-1 (MOI = 1) for the indicated time periods. (b) Expression scatterplots of cell type markers in population from (Fig. 2a). (c) The expression of Shisa9 in different cell types is analyzed from the scRNA data of day 3 and day 7. (d) The proportion analysis of astrocytes in different samples. (e) The expression of inflammatory genes in 8 subclusters of astrocytes. (f-g) Pseudotime analysis of astrocytes from WT mice compared to Shisa9−/− mice. (f) The expression of Shisa9 in WT astrocytes along with pseudotime axis. (g) Pseudo time-line differential genes. (h) The expression of inflammatory genes was presented as a heatmap with pseudotime axis. For every time point, each group is a mix of brain tissues from 5 mice in Extended Data Fig. 3b–h. (i) qPCR analysis of indicated interferon-stimulated genes (ISGs, Mx1, Oas1, Ifit1 and Ifit2) or chemokines (Ccl2, Ccl5, Ccl7, Cxcl9 and Cxcl10) in microglia or astrocytes transfected with control siRNA or Shisa9 siRNA, followed by HSV-1 (MOI = 1, 48hrs) infection. (j and k) Immunofluorescence (j) and quantitation (k) for IFNβ or IL-6 (red) on Iba1+ microglia or GFAP+ astrocytes (green) to identify the source of indicated cytokines in the brains of WT and Shisa9-/- mice at day 0 (HSV-1 uninfected, UI). Scale bars (j), 100 μm. Data In (a and i), data are presented as mean ± SEM, P-values were determined by unpaired two-tailed Student’s t test of 3 independent biological experiments, ns: not significant. In (k), mean ± SD of 8 mice. P-values were determined by unpaired two-tailed Student’s t test.

Source data

Extended Data Fig. 4 The function of Shisa9 in microglia and astrocytes during HSV-1 infection.

(ad) Immunofluorescence (a and c) and quantitation (b and d) for IFNβ, IL-6 or HSV-1 (red) on Iba1+ microglia or GFAP+ astrocytes (green) to identify the source of indicated cytokines in the brains of WT and Shisa9−/− mice at day 3 or day 18 after intracranial infection with HSV-1(1 × 102 PFU). Scale bars (a and c), 100 μm. In (b and d), all error bars, mean values ± SD of 8 mice. P-values were determined by unpaired two-tailed Student’s t test.

Source data

Extended Data Fig. 5 SHISA9 regulates antiviral and pro-inflammatory genes expression through IKKi.

(a) U87 was infected without or with HSV-1 (MOI = 1) for 24 hrs, the cell lysates were then immunoprecipitated (IP) by SHISA9, and the proteins binding with SHISA9 were eluted and send for mass spectrometry analysis (MS). The IKKi peptide was detected by IP-MS. Green color represents the b- fragment ions (come from the N-terminus) while orange color represents y - fragment ions (come from the C-terminus). (b) Co-immunoprecipitation (IP) and immunoblot analysis of extracts of BV2 cells with or without HSV-1 (MOI = 1) infection for 24 hrs. WCL, whole-cell lysates. (c) List of the top 100 upregulated immune genes in the brain of Shisa9/ mice after intracranial infection with HSV-1 (1 × 102 PFU) for 7 days. IKKi-dependent genes are labelled. (d) Real-time quantitative polymerase chain reaction (qPCR) analysis of indicated genes in wild-type (WT) and Shisa9/ astrocytes transfected with control siRNA or Ikbke siRNA, followed by HSV-1 (MOI = 1) infection for 24 hrs. The efficiency of Ikbke siRNA was confirmed by immunoblot. (e) Immunoblotting analysis of extracts of HEK293T cells transfected with HA-IKKi and Myc-SHISA9, followed by HSV-1 (MOI = 1) infection for 24 hrs, then treated with bafilomycin A1 (Baf A1, 0.2 μM), MG132 (10 μM), 3-methyladenine (3-MA, 10 mM) or lactacystin (5 μM) for 6 hrs. NT, not treated. Similar results were obtained by three independent biological experiments. In (d) all error bars, mean values ± SEM, P-values were determined by unpaired two-tailed Student’s t test of 3 independent biological experiments.

Source data

Extended Data Fig. 6 SHISA9 bridges IKKi to autophagy membrane protein GABARAPL1.

(a) Immunoblotting analysis of extracts of wild-type (WT), SQSTM1 knockout (KO), OPTN KO or CALCOCO2 KO HEK293T cells transfected with empty vector or increasing amounts of Myc-SHISA9, with EBSS treatment. (b) Immunoblotting analysis of extracts of U87 cells treated with DMSO or 3-MA (10 mM). (c) Co-immunoprecipitation (IP) and immunoblotting analysis of extracts of HEK293T cells transfected with HA-SHISA9 and indicated Flag tagged ATG8 family members. (d) Co-immunoprecipitation and immunoblotting analysis of extracts of Flag-SHISA9-inducible cells treated with or without doxycycline (Dox, 100 ng/ml) for 12 hrs, followed by immunoprecipitation with anti-Flag. (e) Purified Flag-GABARAP1/ GABARAPL1 and His-tagged SHISA9 were incubated together, followed by immunoprecipitation with anti-His and immunoblotting analysis. (f) Domain organization of SHISA9 protein. (g) Co-immunoprecipitation and immunoblotting analysis of extracts of U87 cells transfected with Flag-GABARAPL1, and Myc-SHISA9 or its indicated mutations, followed by HSV-1 (MOI = 1) infection for 24 hrs. (h) Immunoblotting analysis of extracts of U87 cells transfected with WT Flag-SHISA9 or its F170A mutant, with or without HSV-1 (MOI = 1) infection for 24 hrs. (h) Co-immunoprecipitation and immunoblotting analysis of extracts of U87 cells transfected with HA-GABARAPL1, Flag-IKKi and Myc-SHISA9 with indicated combinations, followed by HSV-1(MOI = 1) infection for 24 hrs. (j) Co-immunoprecipitation and immunoblotting analysis of extracts of HEK293T cells transfected with Flag-SHISA9 (WT, F170A) and HA-IKKi. (k) Immunoblotting analysis of extracts of U87 cells transfected with Flag-SHISA9 and its indicated mutations. (l) Quantification of the indicated proteins levels of Fig. 5k by using ImageJ software. P-values were determined by unpaired two-tailed Student’s t test. For (a-c, j and k), EBSS treatment were for 6 hrs. For (c, g, h and j), chloroquine (CQ, 50 μM) was added in all groups for 6 hrs. For (a-e, g-k), similar results were obtained by three independent biological experiments.

Source data

Extended Data Fig. 7 Enhanced cerebral pathogenesis of HSE in Shisa9-/- mice.

(a) Survival curve of wild-type (WT) and Shisa9/ mice (n = 14 per group) post HSV-1 infection; P-values were determined by the log-rank test. (b) HSV-1 yield in WT and Shisa9/ mice post HSV-1 infection. (c) WT and Shisa9/ Mice (n = 9 per group) were intracranial infected with HSV-1 (1 × 102 PFU), scored for neuroinflammation symptoms. Data are presented as mean ± SD of 9 mice. Similar results were obtained by three independent experiments. Stars refers to difference between WT and Shisa9/ for total 9 days. (d) ELISA of indicated cytokines in brain homogenates in WT and Ikbke/ mice at 12 days after intracranial infection with HSV-1 (1 × 102 PFU). All error bars, mean ± SD of 12 mice. (e) Representative image (left) and quantification (right) of histology of WT and Shisa9/ mice brain. Scale bars, 200 μm (main panels), and 50 μm (right panel, enlarged view). Data are presented as mean ± SD of 6 mice. (f-h) WT and Shisa9/ mice were intracranial infected with VSV (1 × 102 PFU) (f) Survival of wild-type (WT) and Shisa9/ mice (n = 32 per group) at various times (horizontal axis), P-values were determined by the log-rank test. (g) ELISA of indicated cytokines in brain homogenates were detected 12 days post infection. (h) VSV-G staining and H&E staining of hippocampi area. Scale bars, 200 μm. Data are presented as mean ± SD of 12 mice in (g) and mean ± SD of 9 mice in (h). (i) Real-time quantitative polymerase chain reaction (qPCR) analysis of brain homogenates of WT and Shisa9/ mice (12 days after inoculated with a combination of HSV-1 (2 × 102 PFU) and lysosome inhibitor Lys05 (16 mg/kg)). Data are presented as mean ± SD of 6 mice. For (b, d, e, g-i), P-values were determined by unpaired two-tailed Student’s t test, for (c), by Two-way ANOVA.

Source data

Supplementary information

Supplementary Information

Breeding statistic, gating strategy and supplementary methods.

Reporting Summary

Peer Review File

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Raw images for Fig. 2i.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Raw images for Fig. 3h.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Raw images for Fig. 4b,e,h.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 6

Raw images for Fig. 6d.

Source Data Extended Data Fig./Table 1

Statistical source data.

Source Data Extended Data Fig./Table 2

Statistical source data.

Source Data Extended Data Fig./Table 3

Statistical source data.

Source Data Extended Data Fig./Table 3

Raw images for Extended Data Fig. 3j.

Source Data Extended Data Fig./Table 4

Statistical source data.

Source Data Extended Data Fig./Table 4

Raw images for Extended Data Fig. 4a,c.

Source Data Extended Data Fig./Table 5

Statistical source data + mass data table.

Source Data Extended Data Fig./Table 5

Unprocessed western blots.

Source Data Extended Data Fig./Table 6

Statistical source data.

Source Data Extended Data Fig./Table 6

Unprocessed western blots.

Source Data Extended Data Fig./Table 7

Statistical source data.

Source Data Extended Data Fig./Table 7

Raw images for Extended Data Fig. 7e,h.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

Zheng, Y., Wang, L., Liu, Q. et al. Modulation of virus-induced neuroinflammation by the autophagy receptor SHISA9 in mice. Nat Microbiol 8, 958–972 (2023). https://doi.org/10.1038/s41564-023-01357-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-023-01357-3

  • Springer Nature Limited

Navigation