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:

Mitochondrial aspartate regulates TNF biogenesis and autoimmune tissue inflammation

Abstract

Misdirected immunity gives rise to the autoimmune tissue inflammation of rheumatoid arthritis, in which excess production of the cytokine tumor necrosis factor (TNF) is a central pathogenic event. Mechanisms underlying the breakdown of self-tolerance are unclear, but T cells in the arthritic joint have a distinctive metabolic signature of ATPlo acetyl-CoAhi proinflammatory effector cells. Here we show that a deficiency in the production of mitochondrial aspartate is an important abnormality in these autoimmune T cells. Shortage of mitochondrial aspartate disrupted the regeneration of the metabolic cofactor nicotinamide adenine dinucleotide, causing ADP deribosylation of the endoplasmic reticulum (ER) sensor GRP78/BiP. As a result, ribosome-rich ER membranes expanded, promoting co-translational translocation and enhanced biogenesis of transmembrane TNF. ERrich T cells were the predominant TNF producers in the arthritic joint. Transfer of intact mitochondria into T cells, as well as supplementation of exogenous aspartate, rescued the mitochondria-instructed expansion of ER membranes and suppressed TNF release and rheumatoid tissue inflammation.

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: Mitochondrial insufficiency and ER expansion in RA.
Fig. 2: Mitochondrial insufficiency promotes ER expansion.
Fig. 3: Mitochondria-derived aspartate controls ER size.
Fig. 4: Aspartate is anti-inflammatory.
Fig. 5: Aspartate is required for NAD+ regeneration and ADP ribosylation of BiP.
Fig. 6: Expansion of the rough ER increases co-translational translocation in RA T cells.
Fig. 7: ERrich RA T cells are TNF superproducers.
Fig. 8: TNF-producing CD4+ T cells function as arthritogenic effector cells.

Similar content being viewed by others

Data availability

All data in the present study are available within the article and its Supplementary files and from the corresponding author upon reasonable request. Source data are provided with this paper.

References

  1. Deane, K. D., Norris, J. M. & Holers, V. M. Preclinical rheumatoid arthritis: identification, evaluation, and future directions for investigation. Rheum. Dis. Clin. North Am. 36, 213–241 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Feldmann, M., Brennan, F. M. & Maini, R. N. Role of cytokines in rheumatoid arthritis. Annu. Rev. Immunol. 14, 397–440 (1996).

    Article  CAS  PubMed  Google Scholar 

  3. Maini, R. N. & Taylor, P. C. Anti-cytokine therapy for rheumatoid arthritis. Annu. Rev. Med 51, 207–229 (2000).

    Article  CAS  PubMed  Google Scholar 

  4. Zhang, F. et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat. Immunol. 20, 928–942 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Rubbert-Roth, A. et al. Failure of anti-TNF treatment in patients with rheumatoid arthritis: the pros and cons of the early use of alternative biological agents. Autoimmun. Rev. 18, 102398 (2019).

    Article  PubMed  CAS  Google Scholar 

  6. Grivennikov, S. I. et al. Distinct and nonredundant in vivo functions of TNF produced by T cells and macrophages/neutrophils: protective and deleterious effects. Immunity 22, 93–104 (2005).

    CAS  PubMed  Google Scholar 

  7. Bovensiepen, C. S. et al. TNF-producing Th1 cells are selectively expanded in liver infiltrates of patients with autoimmune hepatitis. J. Immunol. 203, 3148–3156 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Jain, A. et al. T cells instruct myeloid cells to produce inflammasome-independent IL-1β and cause autoimmunity. Nat. Immunol. 21, 65–74 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Schreurs, R. et al. Human fetal TNF-alpha-cytokine-producing CD4+ effector memory T cells promote intestinal development and mediate inflammation early in life. Immunity 50, 462–476.e468 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Weyand, C. M. & Goronzy, J. J. T-cell-targeted therapies in rheumatoid arthritis. Nat. Clin. Pr. Rheumatol. 2, 201–210 (2006).

    Article  CAS  Google Scholar 

  11. Weyand, C. M. & Goronzy, J. J. Immunometabolism in early and late stages of rheumatoid arthritis. Nat. Rev. Rheumatol. 13, 291–301 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Slowikowski, K., Wei, K., Brenner, M. B. & Raychaudhuri, S. Functional genomics of stromal cells in chronic inflammatory diseases. Curr. Opin. Rheumatol. 30, 65–71 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Isaacs, J. D. Therapeutic T-cell manipulation in rheumatoid arthritis: past, present and future. Rheumatology 47, 1461–1468 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Probert, L. et al. Wasting, ischemia, and lymphoid abnormalities in mice expressing T cell-targeted human tumor necrosis factor transgenes. J. Immunol. 151, 1894–1906 (1993).

    CAS  PubMed  Google Scholar 

  15. Weyand, C. M. & Goronzy, J. J. The immunology of rheumatoid arthritis. Nat. Immunol. 22, 10–18 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Yang, Z. et al. Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Sci. Transl. Med. 8, 331ra338 (2016).

    Google Scholar 

  17. Yang, Z., Fujii, H., Mohan, S. V., Goronzy, J. J. & Weyand, C. M. Phosphofructokinase deficiency impairs ATP generation, autophagy, and redox balance in rheumatoid arthritis T cells. J. Exp. Med. 210, 2119–2134 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wu, B. et al. Succinyl-CoA ligase deficiency in pro-inflammatory and tissue-invasive T cells. Cell Metab. 32, 967–980 e965 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Shen, Y. et al. Metabolic control of the scaffold protein TKS5 in tissue-invasive, proinflammatory T cells. Nat. Immunol. 18, 1025–1034 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Li, Y. et al. The DNA repair nuclease MRE11A functions as a aitochondrial protector and prevents T cell pyroptosis and tissue inflammation. Cell Metab. 30, 477–492.e476 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wen, Z. et al. N-Myristoyltransferase deficiency impairs activation of kinase AMPK and promotes synovial tissue inflammation. Nat. Immunol. 20, 313–325 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Bommiasamy, H. et al. ATF6alpha induces XBP1-independent expansion of the endoplasmic reticulum. J. Cell Sci. 122, 1626–1636 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sriburi, R., Jackowski, S., Mori, K. & Brewer, J. W. XBP1: a link between the unfolded protein response, lipid biosynthesis, and biogenesis of the endoplasmic reticulum. J. Cell Biol. 167, 35–41 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Martinez-Reyes, I. & Chandel, N. S. Mitochondrial TCA cycle metabolites control physiology and disease. Nat. Commun. 11, 102 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zaslona, Z. & O’Neill, L. A. J. Cytokine-like roles for metabolites in immunity. Mol. Cell 78, 814–823 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Chinopoulos, C. Acute sources of mitochondrial NAD+ during respiratory chain dysfunction. Exp. Neurol. 327, 113218 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Korennykh, A. & Walter, P. Structural basis of the unfolded protein response. Annu. Rev. Cell Dev. Biol. 28, 251–277 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Jager, R., Bertrand, M. J., Gorman, A. M., Vandenabeele, P. & Samali, A. The unfolded protein response at the crossroads of cellular life and death during endoplasmic reticulum stress. Biol. Cell 104, 259–270 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Voorhees, R. M. & Hegde, R. S. Toward a structural understanding of co-translational protein translocation. Curr. Opin. Cell Biol. 41, 91–99 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Elvekrog, M. M. & Walter, P. Dynamics of co-translational protein targeting. Curr. Opin. Chem. Biol. 29, 79–86 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Schwarz, D. S. & Blower, M. D. The endoplasmic reticulum: structure, function and response to cellular signaling. Cell. Mol. Life Sci. 73, 79–94 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Sullivan, L. B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rubbert-Roth, A. et al. TNF inhibitors in rheumatoid arthritis and spondyloarthritis: are they the same? Autoimmun. Rev. 17, 24–28 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Li, Y. et al. Deficient activity of the nuclease MRE11A induces T cell aging and promotes arthritogenic effector functions in patients with rheumatoid arthritis. Immunity 45, 903–916 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Shaffer, A. L. et al. XBP1, downstream of Blimp-1, expands the secretory apparatus and other organelles, and increases protein synthesis in plasma cell differentiation. Immunity 21, 81–93 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Phillips, M. J. & Voeltz, G. K. Structure and function of ER membrane contact sites with other organelles. Nat. Rev. Mol. Cell Biol. 17, 69–82 (2016).

    Article  CAS  PubMed  Google Scholar 

  37. Hayashi, T. & Su, T. P. Sigma-1 receptor chaperones at the ER–mitochondrion interface regulate Ca2+ signaling and cell survival. Cell 131, 596–610 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Sebastian, D. et al. Mitofusin 2 (Mfn2) links mitochondrial and endoplasmic reticulum function with insulin signaling and is essential for normal glucose homeostasis. Proc. Natl Acad. Sci. USA 109, 5523–5528 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Simmen, T. et al. PACS-2 controls endoplasmic reticulum-mitochondria communication and Bid-mediated apoptosis. EMBO J. 24, 717–729 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Knupp, J., Arvan, P. & Chang, A. Increased mitochondrial respiration promotes survival from endoplasmic reticulum stress. Cell Death Differ. 26, 487–501 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Bravo, R. et al. Increased ER–mitochondrial coupling promotes mitochondrial respiration and bioenergetics during early phases of ER stress. J. Cell Sci. 124, 2143–2152 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation Is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Davila, A. et al. Nicotinamide adenine dinucleotide is transported into mammalian mitochondria. eLife https://doi.org/10.7554/eLife.33246 (2018).

  44. Bettigole, S. E. & Glimcher, L. H. Endoplasmic reticulum stress in immunity. Annu. Rev. Immunol. 33, 107–138 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Gardner, B. M., Pincus, D., Gotthardt, K., Gallagher, C. M. & Walter, P. Endoplasmic reticulum stress sensing in the unfolded protein response. Cold Spring Harb. Perspect. Biol. 5, a013169 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Chambers, J. E., Petrova, K., Tomba, G., Vendruscolo, M. & Ron, D. ADP ribosylation adapts an ER chaperone response to short-term fluctuations in unfolded protein load. J. Cell Biol. 198, 371–385 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wu, B., Goronzy, J. J. & Weyand, C. M. Metabolic fitness of T cells in autoimmune disease. Immunometabolism https://doi.org/10.20900/immunometab20200017 (2020).

  48. Kalliolias, G. D. & Ivashkiv, L. B. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat. Rev. Rheumatol. 12, 49–62 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Yang, Z. et al. Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Sci. Transl. Med. 8, 331ra338 (2016).

    Google Scholar 

  50. Li, Y. et al. Deficient activity of the nuclease MRE11A induces T cell aging and promotes arthritogenic effector functions in patients with rheumatoid arthritis. Immunity 45, 903–916 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Institutes of Health (grant nos. R01AR042527, R01AI108906, R01HL142068 and P01HL129941 to C.M.W. and nos. R01AI108891, R01AG045779, U19AI057266 and R01AI129191 to J.J.G.).

Author information

Authors and Affiliations

Authors

Contributions

C.M.W., J.J.G. and B.W. conceived the project. B.W., T.V.Z., Z.H. and K.J. formally analyzed and investigated the data. K.J.W. and M.P.A. recruited the patients. C.M.W., J.J.G. and B.W. wrote the original manuscript. C.M.W. and J.J.G. supervised the study. C.M.W. and J.J.G. acquired the funds.

Corresponding author

Correspondence to Cornelia M. Weyand.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Navdeep Chandel and George Tsokos for their contribution to the peer review of this work. N. Bernard was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team. Peer review reports are available.

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

Extended data

Extended Data Fig. 1 Mitochondrial mass in healthy and RA T cells.

CD4+CD45RA+ T cells were stimulated for 72 h. Flow cytometric quantification of mitochondria mass (MitoTrack Green MFI); n = 6 healthy and 6 RA. Data are mean ± SEM. Two-tailed unpaired Mann-Whitney-Wilcoxon rank test.

Source data

Extended Data Fig. 2 Gating strategy to analyze ER and mitochondrial function on the single cell level.

CD4+CD45RA+ T cells were stimulated for 72 h. ER biomass was determined with ER tracker and mitochondrial function was assessed with the mitochondrial membrane potential. Gate #1: small cellular size. Gate #2: medium cellular size. Gate #3: large cellular size.

Extended Data Fig. 3 XBP1S overexpression induces ER expansion.

Naive CD4+ T cells were stimulated and transfected with control or XBP1S overexpression plasmid before the ER size was determined. (a, b) Flow cytometry for ER Tracker staining; n = 4. (c) Confocal microscopy imaging of the ER protein calnexin. Scale bar, 10 μm, n = 3 independent experiments. (d, e) Flow cytometry for calnexin expression; n = 3. (f) XBP1S expression in T cells from patients treated with or w/o Methotrexate (MTX) (MTX: n = 13; w/o MTX: n = 12). All data are mean ± SEM. Two-tailed paired t test (b, e). Unpaired Mann-Whitney-Wilcoxon rank test (f). *P < 0.05, ***P < 0.001.

Source data

Extended Data Fig. 4 Inhibitors of mitochondrial respiration promote ER expansion.

Healthy naive CD4+ T cells were stimulated for 72 h in the presence of the mitochondrial respiration inhibitors Piericidin A (10 pM), Antimycin A (10 nM) or Oligomycin (1 nM). ER size was determined by flow cytometry measuring ER tracker (n = 4). Data are mean ± SEM. One-way ANOVA and post-ANOVA pair-wise two-group comparisons conducted with Tukey’s method. **P < 0.01.

Source data

Extended Data Fig. 5 Experimental scheme for mitochondria transfer.

(a) Experimental scheme for mitochondria transfer into RA T cells. (b) Experimental scheme for mitochondria transfer in Jurkat T cells. Mitochondria were labeled with MitoTrackerRed and isolated, then transferred into recipient cells. (c) Flow cytometric analysis of MitoTracker Red intensity after mitochondria transfer. Ratio indicates donor cell number/recipient cell number. (d) Representative confocal imaging of exogenous mitochondria transferred into Jurkat T cells, n = 3 independent experiments, scale bar, 20 μm.

Extended Data Fig. 6 Expression of GOT1 and GOT2 in healthy and RA T cells.

Peripheral blood CD4+CD45RA+ T cells from patients with RA and age-matched healthy individuals were isolated and stimulated for 72 h. mRNA levels of GOT1 and GOT2 were determined by qPCR. n = 4 in each group. All data are mean ± SEM. Two-tailed unpaired Mann-Whitney-Wilcoxon rank test.

Source data

Extended Data Fig. 7 Mitochondrial complex I inhibitor Piercidin A inhibits ADP-ribosylation of BiP.

ADP-ribosylation of BiP in healthy CD4+ T cells treated with or w/o Piercidin A (10 pM) for 24 h. n = 2.

Source data

Extended Data Fig. 8 Isolation of Rough ER.

(a) Naïve CD4+ T cells were purified from peripheral blood mononuclear cells and stimulated with anti-CD3/CD28 for 72 h. The rough ER was isolated by calcium precipitation and the isolate was immunoblotted for the ER protein calnexin, the ribosomal protein L17 and the cytoplasmic protein a-actin. (b) Healthy CD4+ T cells were activated with PMA/Ionomycin for 2 h before isolation of the rough ER and immunoblotting of the ER protein calnexin, the ribosomal protein S7 and the cytosolic protein β-actin.

Source data

Extended Data Fig. 9 Effects of Tunicamycin, aspartate, asparagine, pyruvate, and α-ketobutyrate on ER size and TNF production.

Naïve CD4+ T cells were purified from PBMCs and stimulated with anti-CD3/CD28 beads for 72 h in the presence of the indicated molecules. ER size quantified by MFI of ER tracker staining and TNF production measured by intracellular staining of TNF after PMA/ION stimulation for 2 h in the presence of the secretion inhibitor BFA. (a) Fold change of ER size after Tunicamycin treatment compared to the control group, n = 6. (b) Fold change of TNF production after Tunicamycin treatment compared to the control group, n = 4. (c) ER size change after Aspartate (1 mM) or Asparagine (1 mM) treatment, n = 6. (d) TNF production after aspartate and asparagine treatment, n = 3. (e) ER size change after pyruvate (1 mM) or α-KB (1 mM) treatment, n = 6. (f) TNF production after pyruvate or α-KB treatment, n = 4. All data are mean ± SEM, one-way ANOVA and post-ANOVA pair-wise two-group comparisons conducted with Tukey’s method. *P < 0.05, **P < 0.01, ***P < 0.001.

Source data

Extended Data Fig. 10 Mitochondria transfer into CD4+ T cells protects synovial tissue from inflammation.

Mitochondria were isolated from healthy T cells and transferred into RA CD4+ T cells prior to their adoptive transfer into synovium-NSG chimeras. Explanted synovial grafts were analyzed by immunohistochemical staining and tissue transcriptomics (RT-PCR). 8 tissues in each group. (a) H&E staining of synovial tissue sections. Scale bar; 50 μm. (b) Immunofluorescence staining for CD3+ T cells in synovial infiltrates. Representative images. Scale bar; 10 μm. (c) Gene expression profiling (RT-PCR) of TRB, TBET, RORG and other key inflammatory markers (n = 8). All data are mean ± SEM. Two-tailed unpaired Mann-Whitney-Wilcoxon rank test. *P < 0.05, **P < 0.01, ***P < 0.001.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2, and Supplementary Figs. 1–3.

Reporting Summary

Peer Review Information

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. 4

Statistical source data.

Source Data Fig. 5

Statistical source data and unprocessed immunobots.

Source Data Fig. 6

Statistical source data and unprocessed immunobots.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

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. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data and unprocessed immunobots.

Source Data Extended Data Fig. 8

Unprocessed immunoblots.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, B., Zhao, T.V., Jin, K. et al. Mitochondrial aspartate regulates TNF biogenesis and autoimmune tissue inflammation. Nat Immunol 22, 1551–1562 (2021). https://doi.org/10.1038/s41590-021-01065-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01065-2

This article is cited by

Search

Quick links

Nature Briefing

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

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