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:

Alleviating symptoms of neurodegenerative disorders by astrocyte-specific overexpression of TMEM164 in mice

Abstract

Neuroinflammatory microglia secrete cytokines to induce neurotoxic reactive astrocytes, which are one of the major causes of neuronal death. However, the intrinsic key regulators underlying neurotoxic reactive astrocytes induction are unknown. Here we show that the transmembrane protein 164 (TMEM164) is an early-response intrinsic factor that regulates neurotoxic astrocyte reactivity. TMEM164 overexpression inhibits the induction of neurotoxic reactive astrocytes, maintains normal astrocytic functions and suppresses neurotoxic reactive astrocyte-mediated neuronal death by decreasing the secretion of neurotoxic saturated lipids. Adeno-associated virus-mediated, astrocyte-specific TMEM164 overexpression in male and female mice prevents the induction of neurotoxic reactive astrocytes, dopaminergic neuronal loss and motor deficits in a Parkinson’s disease model. Notably, brain-wide astrocyte-specific TMEM164 overexpression prevents the induction of neurotoxic reactive astrocytes, amyloid β deposition, neurodegeneration and memory decline in the 5XFAD Alzheimer’s disease mouse model, suggesting that TMEM164 could serve as a potential therapeutic target for neurodegenerative disorders.

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: The early-response gene Tmem164 identified in the bulk RNA-seq of mouse astrocytes is downregulated in the astrocytes of patients with several neurodegenerative diseases.
Fig. 2: TMEM164 overexpression virtually maintains normal astrocytic functions and blocks the induction of neurotoxic reactive astrocytes.
Fig. 3: TMEM164 overexpression reduces the secretion of neurotoxic saturated lipids via the CAPN15–ELOVL1 pathway.
Fig. 4: TMEM164 overexpression inhibits the induction of human neurotoxic reactive astrocytes.
Fig. 5: AAV-mediated, astrocyte-specific TMEM164 overexpression reduces dopaminergic neuronal loss in an LPS-induced model of PD.
Fig. 6: Brain-wide TMEM164 overexpression in astrocytes alleviates AD pathologies in 5XFAD mice.

Similar content being viewed by others

Data availability

All next-generation sequencing data have been deposited in the National Genomics Data Center (https://ngdc.cncb.ac.cn/) database (project ID: PRJCA011982). We selected snRNA-seq datasets of neurodegenerative diseases based on three criteria: (1) the sampling area should be the recognized core area of disease onset, for example, PD should be sampled in the substantia nigra48 and MS should be sampled in the white matter49; (2) there should be at least ten samples in the disease and healthy groups; (3) the datasets should be published in top-rated journals. The links to the publicly available datasets are as follows: AD snRNA-seq (from both the RADC Research Resource Sharing Hub (project IDs: syn18485175 and syn21125841) and the Gene Expression Omnibus (GEO) repository (accession nos. GSE174367, GSE138852, GSE129308, GSE163577, GSE185553, GSE185277 and GSE198323); MS snRNA-seq (accession no. GSE118257); ALS snRNA-seq (accession no. GSE174332); FTLD snRNA-seq (accession no. GSE174332); PD snRNA-seq (accession no. GSE184950); and HD snRNA-seq (accession nos. GSE152058 and GSE173731). Source data are provided with this paper.

Code availability

All original code used in this study is available from the corresponding authors upon reasonable request.

References

  1. Liddelow, S. A. & Barres, B. A. Reactive astrocytes: production, function, and therapeutic potential. Immunity 46, 957–967 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Clarke, L. E. & Barres, B. A. Emerging roles of astrocytes in neural circuit development. Nat. Rev. Neurosci. 14, 311–321 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sofroniew, M. V. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 32, 638–647 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zamanian, J. L. et al. Genomic analysis of reactive astrogliosis. J. Neurosci. 32, 6391–6410 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Guttenplan, K. A. et al. Neurotoxic reactive astrocytes induce cell death via saturated lipids. Nature 599, 102–107 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. Yun, S. P. et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson’s disease. Nat. Med. 24, 931–938 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Guttenplan, K. A. et al. Knockout of reactive astrocyte activating factors slows disease progression in an ALS mouse model. Nat. Commun. 11, 3753 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Guttenplan, K. A. et al. Neurotoxic reactive astrocytes drive neuronal death after retinal Injury. Cell Rep. 31, 107776 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhou, Y. et al. Molecular landscapes of human hippocampal immature neurons across lifespan. Nature 607, 527–533 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yang, A. C. et al. A human brain vascular atlas reveals diverse mediators of Alzheimer’s risk. Nature 603, 885–892 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Otero-Garcia, M. et al. Molecular signatures underlying neurofibrillary tangle susceptibility in Alzheimer’s disease. Neuron 110, 2929–2948 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Morabito, S. et al. Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer’s disease. Nat. Genet. 53, 1143–1155 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhou, Y. et al. Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimer’s disease. Nat. Med. 26, 131–142 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mathys, H. et al. Single-cell transcriptomic analysis of Alzheimer’s disease. Nature 570, 332–337 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Grubman, A. et al. A single-cell atlas of entorhinal cortex from individuals with Alzheimer’s disease reveals cell-type-specific gene expression regulation. Nat. Neurosci. 22, 2087–2097 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Wang, Q. et al. Single-cell transcriptomic atlas of the human substantia nigra in Parkinson’s disease. Preprint at bioRxiv https://doi.org/10.1101/2022.03.25.485846 (2022).

  18. Garcia, F. J. et al. Single-cell dissection of the human brain vasculature. Nature 603, 893–899 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lee, H. et al. Cell type-specific transcriptomics reveals that mutant huntingtin leads to mitochondrial RNA release and neuronal innate immune activation. Neuron 107, 891–908 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Jäkel, S. et al. Altered human oligodendrocyte heterogeneity in multiple sclerosis. Nature 566, 543–547 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Pineda, S. S. et al. Single-cell profiling of the human primary motor cortex in ALS and FTLD. Preprint at bioRxiv https://doi.org/10.1101/2021.07.07.451374 (2021).

  22. Barbar, L. et al. CD49f is a novel marker of functional and reactive human iPSC-derived astrocytes. Neuron 107, 436–453 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Varadi, M. et al. AlphaFold Protein Structure Database: massively expanding the structural coverage of protein-sequence space with high-accuracy models. Nucleic Acids Res. 50, D439–D444 (2022).

    Article  CAS  PubMed  Google Scholar 

  24. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. He, D. et al. Activation of HCA2 regulates microglial responses to alleviate neurodegeneration in LPS-induced in vivo and in vitro models. J. Neuroinflammation 20, 86 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Han, X. et al. Small molecule-driven NLRP3 inflammation inhibition via interplay between ubiquitination and autophagy: implications for Parkinson disease. Autophagy 15, 1860–1881 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mendes-Oliveira, J., Lopes Campos, F., Videira, R. A. & Baltazar, G. GPER activation is effective in protecting against inflammation-induced nigral dopaminergic loss and motor function impairment. Brain Behav. Immun. 64, 296–307 (2017).

    Article  CAS  PubMed  Google Scholar 

  28. Hoban, D. B. et al. Further characterisation of the LPS model of Parkinson’s disease: a comparison of intra-nigral and intra-striatal lipopolysaccharide administration on motor function, microgliosis and nigrostriatal neurodegeneration in the rat. Brain Behav. Immun. 27, 91–100 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Herrera, A. J., Castaño, A., Venero, J. L., Cano, J. & Machado, A. The single intranigral injection of LPS as a new model for studying the selective effects of inflammatory reactions on dopaminergic system. Neurobiol. Dis. 7, 429–447 (2000).

    Article  CAS  PubMed  Google Scholar 

  30. Lee, Y., Messing, A., Su, M. & Brenner, M. GFAP promoter elements required for region-specific and astrocyte-specific expression. Glia 56, 481–493 (2008).

    Article  PubMed  Google Scholar 

  31. Escartin, C. et al. Reactive astrocyte nomenclature, definitions, and future directions. Nat. Neurosci. 24, 312–325 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hasel, P., Aisenberg, W. H., Bennett, F. C. & Liddelow, S. A. Molecular and metabolic heterogeneity of astrocytes and microglia. Cell Metab. 35, 555–570 (2023).

    Article  CAS  PubMed  Google Scholar 

  33. Sadick, J. S. et al. Astrocytes and oligodendrocytes undergo subtype-specific transcriptional changes in Alzheimer’s disease. Neuron 110, 1788–1805 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hasel, P., Rose, I. V. L., Sadick, J. S., Kim, R. D. & Liddelow, S. A. Neuroinflammatory astrocyte subtypes in the mouse brain. Nat. Neurosci. 24, 1475–1487 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Oakley, H. et al. Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J. Neurosci. 26, 10129–10140 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chan, K. Y. et al. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20, 1172–1179 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Caccamo, A. et al. Necroptosis activation in Alzheimer’s disease. Nat. Neurosci. 20, 1236–1246 (2017).

    Article  CAS  PubMed  Google Scholar 

  38. Chen, R. et al. Monoacylglycerol lipase is a therapeutic target for Alzheimer’s disease. Cell Rep. 2, 1329–1339 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhang, L. et al. BAD-mediated neuronal apoptosis and neuroinflammation contribute to Alzheimer’s disease pathology. iScience 24, 102942 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Liu, J. et al. TMEM164 is a new determinant of autophagy-dependent ferroptosis. Autophagy 19, 945–956 (2023).

    Article  CAS  PubMed  Google Scholar 

  41. Reed, A., Ware, T., Li, H., Fernando Bazan, J. & Cravatt, B. F. TMEM164 is an acyltransferase that forms ferroptotic C20:4 ether phospholipids. Nat. Chem. Biol. 19, 378–388 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Dunkley, P. R., Jarvie, P. E. & Robinson, P. J. A rapid Percoll gradient procedure for preparation of synaptosomes. Nat. Protoc. 3, 1718–1728 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Beaudoin, G. M. J. 3rd et al. Culturing pyramidal neurons from the early postnatal mouse hippocampus and cortex. Nat. Protoc. 7, 1741–1754 (2012).

    Article  CAS  PubMed  Google Scholar 

  44. Zhang, Y. et al. Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78, 785–798 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Leger, M. et al. Object recognition test in mice. Nat. Protoc. 8, 2531–2537 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Martorell, A. J. et al. Multi-sensory gamma stimulation ameliorates Alzheimer’s-associated pathology and improves cognition. Cell 177, 256–271 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhou, H. et al. Glia-to-neuron conversion by CRISPR-CasRx alleviates symptoms of neurological disease in mice. Cell 181, 590–603 (2020).

    Article  CAS  PubMed  Google Scholar 

  48. Poewe, W. et al. Parkinson disease. Nat. Rev. Dis. Primers 3, 17013 (2017).

    Article  PubMed  Google Scholar 

  49. Compston, A. & Coles, A. Multiple sclerosis. Lancet 359, 1221–1231 (2002).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Dr. M. Poo for helpful comments. We thank the Optical Imaging (Y. Wang, Y. Zhang and Q. Hu) and FACS facilities (H. Wu and L. Quan) for their technical assistance. We thank the National Health and Disease Human Brain Tissue Resource Center for providing the samples. This study was supported by the STI2030-Major Projects of China (no. 2021ZD0200900 to H.Z.), the National Key Research and Development Program of China (no. 2022YFC3400100 to H.Z.), the Shanghai Municipal Science and Technology Major Project (no. 2018SHZDZX05 to H.Z.), the Shanghai 2023 Special Biopharmaceutical Science and Technology Support Projects (no. 23S41900300 to H.Z.) and the State Key Laboratory of Neuroscience of China.

Author information

Authors and Affiliations

Authors

Contributions

L.Z., Z.X. and H.Z. conceived the study. L.Z., Z.X., Z.J. and Q.W. carried out the study. L.Z. and Z.J. performed the experiments, analysed the data and wrote the manuscript with valuable revision from all authors. Z.X. performed the bioinformatics data analysis. Q.W. and X.L. conducted the AAV injection and the LPS-induced model of PD. T.B. analysed LPS-induced neurotoxicity and AAV expression. X.H. contributed to the in vivo delivery. B.W. contributed to AAV testing. T.L. contributed to the enzyme-linked immunosorbent assay, lentiviral packaging and plasmid construction. Y.C. and X.D. provided the human ESCs. J.F. and Z.L. prepared the human ESC-derived neurons. H.Z. supervised the project and wrote the manuscript.

Corresponding authors

Correspondence to Zhengzheng Xu or Haibo Zhou.

Ethics declarations

Competing interests

H.Z., L.Z., Z.J. and Z.X. are mentioned as inventors on a patent application (no. CN202310486745.2) related to this work. H.Z. is a founder and scientific adviser of Genemagic Biosciences.

Peer review

Peer review information

Nature Metabolism thanks Liliane Tenenbaum, Marc Freeman and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Ashley Castellanos-Jankiewicz, in collaboration with the Nature Metabolism team.

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 Identification of potential regulators of neurotoxic reactive astrocytes via bulk RNA-seq analysis. (Related to Fig. 1).

a, Volcano plot and Venn diagram showing the RNA-seq analysis of 36 upregulated regulators in neurotoxic reactive astrocytes. b, Volcano plot and Venn diagram showing the RNA-seq analysis of 69 downregulated regulators in neurotoxic reactive astrocytes. The DESeq2 R package, along with the Wald test, was utilized to identify differentially expressed genes (DEGs). Genes were categorized as upregulated DEGs when they exhibited a p-value < 0.05 and a log2Foldchange > 0. Conversely, genes were classified as downregulated DEGs when they displayed a p-value < 0.05 and a log2Foldchange < 0. Genes with a p-value ≥ 0.05 were considered stable. Schematic illustrations were created with BioRender.com.

Extended Data Fig. 2 Unique 6 h DEGs in bulk RNA-seq. (Related to Fig. 1).

Heatmap displaying 69 downregulated and 36 upregulated factors in the unique 6 h DEGs dataset from primary astrocyte RNA-seq results. Schematic illustration was created with BioRender.com.

Source data

Extended Data Fig. 3 Expression of Tmem164 in different cell types of human brains. (Related to Fig. 1).

a-c, Analysis for the expression levels of Tmem164 in different brain cell types of post-mortem tissue from individuals without neurological disorders in AD snRNA-seq datasets (n = 3194 astrocytes, 1559 microglia, 10911 neurons, 16552 oligodendrocytes, and 1091 OPCs from 11 heath cases for the dataset of Zhou et al., 2020; n = 1562 astrocytes, 965 microglia, 21923 neurons, 9200 oligodendrocytes, and 1337 OPCs from 24 heath cases for the dataset of Mathys et al., 2019; n = 5610 astrocytes, 1603 microglia, 768 neurons, 4183 oligodendrocytes, and 56 OPCs from the cortex of 4 heath cases for the dataset of Yang et al., 2022), showing higher expression level of Tmem164 in astrocytes than other cell types. All data were presented as minima, maxima, median, 25th and 75th percentiles. p < 0.0001 for three datasets. ***p < 0.001, Kruskal−Wallis test.

Source data

Extended Data Fig. 4 Expression of Tmem164 is reduced in reactive astrocytes. (Related to Fig. 1).

a-c, Comparison of Tmem164 expression levels between non-reactive astrocytes and reactive astrocytes (see Methods for the definitions) in the snRNA-seq datasets of AD patients (n = 5568 non-reactive astrocytes and 901 reactive astrocytes from 21 AD patients for the dataset of Zhou et al., 2020, p = 0.01; n = 2007 non-reactive astrocytes and 965 reactive astrocytes from 11 AD patients for the dataset of Morabito et al., 2021, p < 0.0001; n = 9358 non-reactive astrocytes and 1466 reactive astrocytes from 4 AD patients for dataset of Yang et al., 2022, p < 0.0001), 7-month-old 5XFAD mice (n = 585 non-reactive astrocytes and 67 reactive astrocytes from 3 mice, p = 0.031), and PD patients (n = 3988 non-reactive astrocytes and 1022 reactive astrocytes from 20 PD patients, p = 0.00019), respectively. All data were presented as minima, maxima, median, 25th and 75th percentiles. *p < 0.05, ***p < 0.001, two-sided Wilcoxon test.

Source data

Extended Data Fig. 5 CAPN15 is the downstream transcriptional factor of TMEM164. (Related to Fig. 3).

a, Protein concentrations of CXCL1, CXCL2, and CXCL10 in the culture medium of EGFP-PBS, EGFP-ITC, and TMEM164-ITC astrocytes were measured by ELISA, n = 3 replicates per group. b, Relative mRNA expression levels of C3 among the groups of astrocytes with TMEM164 overexpression and individual transcriptional factor co-overexpression, n = 3 replicates per group. c, Relative mRNA expression levels of Tmem164, Capn15, and Elovl1 in primary mouse astrocytes treated with PBS or ITC, n = 3 replicates per group. d, Relative mRNA expression levels of Capn15 when knocking down Capn15 using CasRx/gRNA system, n = 5 replicates per group. e, Schematic illustration of the possible mechanism underlying TMEM164 overexpression-mediated suppression of neuronal death. Created with BioRender.com. All data were presented as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, p values were provided in Source data, one-way ANOVA and Tukey’s multiple comparison test in panels a, b, and d; unpaired two-tailed student’s t-test in panel c.

Source data

Extended Data Fig. 6 Expression of TMEM164 is decreased during the induction of human neurotoxic reactive astrocytes. (Related to Fig. 4).

a, Predicted structure of human TMEM164 with seven transmembrane motifs by AlphaFold. b, Representative staining of TMEM164, the cell junction marker ZO1, and α-tubulin in human non-reactive astrocytes, showing that TMEM164 is mainly distributed in the cytoplasm and cell membrane, especially at cell junctions. Scale bar, 10 μm. Experiments were independently repeated twice with similar results. c, Representative staining of TMEM164, ZO1, and α-tubulin in human astrocytes treated with ITC for a time course, showing a decreased protein level of TMEM164 during the induction of human neurotoxic reactive astrocytes. Scale bar, 10 μm. Experiments were independently repeated twice with similar results.

Extended Data Fig. 7 AAV-mediated TMEM164 overexpression inhibits the induction of reactive astrocytes but not inflammatory microglia. (Related to Fig. 5).

a, Double staining of reactive astrocyte markers of GFAP and C3 in the SNc of untreated control and AAV-injected PD mice. The astrocytes infected with TMEM164-overexpressed AAV showed downregulated GFAP and C3 signals (the area below the white dashed line in the AAV-Tmem164 + LPS group). Scale bar, 100 μm. b, Representative images of Iba1 staining in the LPS-induced PD model. Scale bar, 50 μm. c, Quantification for the area covered by Iba1-positive microglia in the SNc, showing similar microglial activation between the AAV-mCherry + LPS and AAV-Tmem164 + LPS groups, n = 6 mice per group. Data were presented as mean ± SEM., p value was provided in Source data, unpaired two-tailed student’s t-test.

Source data

Extended Data Fig. 8 LPS-induced neurotoxicity in different brain regions. (Related to Fig. 5).

a, Representative images of NeuN and GFAP staining in the cortex of mice injected with AAV and LPS. Scale bar, 50 μm. b, Quantification for the neuron number in the cortex, n = 3 mice per group. c, Representative images of NeuN and GFAP staining in the striatum of mice injected with AAV and LPS. Scale bar, 50 μm. d, Quantification for the neuron number in the striatum, n = 3 mice per group. Data were presented as mean ± SEM. *p < 0.05, **p < 0.01, p values were provided in Source data, one-way ANOVA and Tukey’s multiple comparison test.

Source data

Extended Data Fig. 9 Evaluation of AAV expression in the brain and peripheral organs of 5XFAD mice. (Related to Fig. 6).

a, Representative images showing the expression of AAV-sGFAP-Tmem164-P2A-mCherry-WPRE in different brain regions of 9-month-old 5XFAD mice at 6 months after AAV injection. Scale bar, 20 μm. b, Representative images showing the expression of AAV-sGFAP-Tmem164-P2A-mCherry-WPRE in the peripheral organs of 9-month-old 5XFAD mice at 6 months after AAV injection. Scale bar, 20 μm. c, Percentage of mCherry-positive astrocytes in different brain regions or mCherry-positive cells in the peripheral organs of 5XFAD mice, n = 3 mice per group. Data were presented as mean ± SEM.

Source data

Extended Data Fig. 10 Effects of TMEM164 overexpression on neurodegeneration and Aβ deposition in 6-month-old 5XFAD mice at 3 months after AAV injection. (Related to Fig. 6).

a, Representative images showing Fluoro-Jade C-labeled degenerated neurons in the cortex and hippocampus of 6-month-old 5XFAD mice at 3 months after AAV injection. Scale bar, 50 μm. b, Quantification of the number of degenerated neurons per field in the cortex and hippocampus in panel a. c, Representative images showing Iba1 and Aβ staining in the cortex and hippocampus of 6-month-old 5XFAD mice at 3 months after AAV injection. Scale bar, 50 μm. d, Quantification of the area covered by Aβ in the cortex and hippocampus, n = 4 mice per group. e, Quantification of the area covered by Iba1-positive microglia in the cortex and hippocampus, n = 4 mice per group. Data were presented as mean ± SEM. *p < 0.05, **p < 0.01, p values were provided in Source data, unpaired two-tailed student’s t-test.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–3.

Reporting Summary

Supplementary Table 1

List of DEGs in the RNA-seq data of the primary astrocytes

Supplementary Table 2

List of DEGs and KEGG pathway in the RNA-seq data of 5XFAD model

Supplementary Table 3

Oligonucleotides used in this study

Supplementary Data 1

Raw gas chromatography–mass spectrometry data

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

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

Zhang, L., Jia, Z., Wu, Q. et al. Alleviating symptoms of neurodegenerative disorders by astrocyte-specific overexpression of TMEM164 in mice. Nat Metab 5, 1787–1802 (2023). https://doi.org/10.1038/s42255-023-00887-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-023-00887-8

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