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:

C13orf31 (FAMIN) is a central regulator of immunometabolic function

Abstract

Single-nucleotide variations in C13orf31 (LACC1) that encode p.C284R and p.I254V in a protein of unknown function (called 'FAMIN' here) are associated with increased risk for systemic juvenile idiopathic arthritis, leprosy and Crohn's disease. Here we set out to identify the biological mechanism affected by these coding variations. FAMIN formed a complex with fatty acid synthase (FASN) on peroxisomes and promoted flux through de novo lipogenesis to concomitantly drive high levels of fatty-acid oxidation (FAO) and glycolysis and, consequently, ATP regeneration. FAMIN-dependent FAO controlled inflammasome activation, mitochondrial and NADPH-oxidase-dependent production of reactive oxygen species (ROS), and the bactericidal activity of macrophages. As p.I254V and p.C284R resulted in diminished function and loss of function, respectively, FAMIN determined resilience to endotoxin shock. Thus, we have identified a central regulator of the metabolic function and bioenergetic state of macrophages that is under evolutionary selection and determines the risk of inflammatory and infectious disease.

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

Figure 1: FAMIN interacts with fatty acid synthase and localizes to peroxisomes.
Figure 2: FAMIN promotes glycolysis and flux of glucose into de novo lipogenesis.
Figure 3: FAMIN augments FAO.
Figure 4: FAMIN controls FAO of endogenously synthesized lipids.
Figure 5: FAMIN-deficient macrophages have impaired mROS production and exhibit features of mitochondrial injury and remodeling.
Figure 6: FAMIN(p.I254V) is hypomorphic and C284R) leads to complete loss of function.
Figure 7: FAMIN deficiency causes defective bacterial clearance and inflammasome activation in vitro.
Figure 8: FAMIN deficiency causes dysregulated response to endotoxin in vivo.

Similar content being viewed by others

References

  1. Wakil, S.M. et al. Association of a mutation in LACC1 with a monogenic form of systemic juvenile idiopathic arthritis. Arthritis Rheumatol. 67, 288–295 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Patel, N. et al. Study of Mendelian forms of Crohn's disease in Saudi Arabia reveals novel risk loci and alleles. Gut 63, 1831–1832 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Liu, H. et al. Discovery of six new susceptibility loci and analysis of pleiotropic effects in leprosy. Nat. Genet. 47, 267–271 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Jostins, L. et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491, 119–124 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hruz, T. et al. Genevestigator v3: a reference expression database for the meta-analysis of transcriptomes. Adv. Bioinformatics 2008, 420747 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Heng, T.S. & Painter, M.W. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Jensen-Urstad, A.P. & Semenkovich, C.F. Fatty acid synthase and liver triglyceride metabolism: housekeeper or messenger? Biochim. Biophys. Acta 1821, 747–753 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Jensen-Urstad, A.P. et al. Nutrient-dependent phosphorylation channels lipid synthesis to regulate PPARα. J. Lipid Res. 54, 1848–1859 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lodhi, I.J. & Semenkovich, C.F. Peroxisomes: a nexus for lipid metabolism and cellular signaling. Cell Metab. 19, 380–392 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Murray, P.J. et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 14–20 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Tannahill, G.M. et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 496, 238–242 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Huang, S.C. et al. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nat. Immunol. 15, 846–855 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Skarnes, W.C. et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337–342 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hillebrand, M. et al. Identification of a new fatty acid synthesis-transport machinery at the peroxisomal membrane. J. Biol. Chem. 287, 210–221 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Semenkovich, C.F. Regulation of fatty acid synthase (FAS). Prog. Lipid Res. 36, 43–53 (1997).

    Article  CAS  PubMed  Google Scholar 

  16. Jha, A.K. et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity 42, 419–430 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Grevengoed, T.J., Klett, E.L. & Coleman, R.A. Acyl-CoA metabolism and partitioning. Annu. Rev. Nutr. 34, 1–30 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Carracedo, A., Cantley, L.C. & Pandolfi, P.P. Cancer metabolism: fatty acid oxidation in the limelight. Nat. Rev. Cancer 13, 227–232 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wu, M. et al. Multiparameter metabolic analysis reveals a close link between attenuated mitochondrial bioenergetic function and enhanced glycolysis dependency in human tumor cells. Am. J. Physiol. Cell Physiol. 292, C125–C136 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Hao, W., Chang, C.P., Tsao, C.C. & Xu, J. Oligomycin-induced bioenergetic adaptation in cancer cells with heterogeneous bioenergetic organization. J. Biol. Chem. 285, 12647–12654 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Newsholme, E.A., Sugden, P.H. & Williams, T. Effect of citrate on the activities of 6-phosphofructokinase from nervous and muscle tissues from different animals and its relationships to the regulation of glycolysis. Biochem. J. 166, 123–129 (1977).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Fantin, V.R., St-Pierre, J. & Leder, P. Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell 9, 425–434 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Vats, D. et al. Oxidative metabolism and PGC-1β attenuate macrophage-mediated inflammation. Cell Metab. 4, 13–24 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Landree, L.E. et al. C75, a fatty acid synthase inhibitor, modulates AMP-activated protein kinase to alter neuronal energy metabolism. J. Biol. Chem. 279, 3817–3827 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Liesa, M. & Shirihai, O.S. Mitochondrial dynamics in the regulation of nutrient utilization and energy expenditure. Cell Metab. 17, 491–506 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 417, 1–13 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. Yu, L., Quinn, M.T., Cross, A.R. & Dinauer, M.C. Gp91phox is the heme binding subunit of the superoxide-generating NADPH oxidase. Proc. Natl. Acad. Sci. USA 95, 7993–7998 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ran, F.A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. West, A.P. et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature 472, 476–480 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kampmann, B. et al. Evaluation of human antimycobacterial immunity using recombinant reporter mycobacteria. J. Infect. Dis. 182, 895–901 (2000).

    Article  CAS  PubMed  Google Scholar 

  31. Zhou, R., Yazdi, A.S., Menu, P. & Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 469, 221–225 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. Gattorno, M. et al. The pattern of response to anti-interleukin-1 treatment distinguishes two subsets of patients with systemic-onset juvenile idiopathic arthritis. Arthritis Rheum. 58, 1505–1515 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Mellins, E.D., Macaubas, C. & Grom, A.A. Pathogenesis of systemic juvenile idiopathic arthritis: some answers, more questions. Nat. Rev. Rheumatol. 7, 416–426 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Quartier, P. et al. A multicentre, randomised, double-blind, placebo-controlled trial with the interleukin-1 receptor antagonist anakinra in patients with systemic-onset juvenile idiopathic arthritis (ANAJIS trial). Ann. Rheum. Dis. 70, 747–754 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Pascual, V., Allantaz, F., Arce, E., Punaro, M. & Banchereau, J. Role of interleukin-1 (IL-1) in the pathogenesis of systemic onset juvenile idiopathic arthritis and clinical response to IL-1 blockade. J. Exp. Med. 201, 1479–1486 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Leist, M., Single, B., Castoldi, A.F., Kühnle, S. & Nicotera, P. Intracellular adenosine triphosphate (ATP) concentration: a switch in the decision between apoptosis and necrosis. J. Exp. Med. 185, 1481–1486 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hue, L. & Taegtmeyer, H. The Randle cycle revisited: a new head for an old hat. Am. J. Physiol. Endocrinol. Metab. 297, E578–E591 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Guan, H.P. et al. A futile metabolic cycle activated in adipocytes by antidiabetic agents. Nat. Med. 8, 1122–1128 (2002).

    Article  CAS  PubMed  Google Scholar 

  39. O'Sullivan, D. et al. Memory CD8+ T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity 41, 75–88 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. van der Windt, G.J. et al. CD8 memory T cells have a bioenergetic advantage that underlies their rapid recall ability. Proc. Natl. Acad. Sci. USA 110, 14336–14341 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Beloqui, A. et al. Novel polyphenol oxidase mined from a metagenome expression library of bovine rumen: biochemical properties, structural analysis, and phylogenetic relationships. J. Biol. Chem. 281, 22933–22942 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Kim, Y. et al. Crystal structure of hypothetical protein YfiH from Shigella flexneri at 2 A resolution. Proteins 63, 1097–1101 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Samudio, I. et al. Pharmacologic inhibition of fatty acid oxidation sensitizes human leukemia cells to apoptosis induction. J. Clin. Invest. 120, 142–156 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Berod, L. et al. De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat. Med. 20, 1327–1333 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Hall, C.J. et al. Immunoresponsive gene 1 augments bactericidal activity of macrophage-lineage cells by regulating β-oxidation-dependent mitochondrial ROS production. Cell Metab. 18, 265–278 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. O'Neill, L.A. & Pearce, E.J. Immunometabolism governs dendritic cell and macrophage function. J. Exp. Med. 213, 15–23 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Mulders-Manders, C.M. & Simon, A. Hyper-IgD syndrome/mevalonate kinase deficiency: what is new? Semin. Immunopathol. 37, 371–376 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Behringer, R., Gertsenstein, M., Vintersten Nagy, K. & Nagy, A. Manipulating the Mouse Embryo: A Laboratory Manual 4th edn. 92–93, 139–142 and 211–215 (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 2014).

  50. Bougaki, M. et al. Nos3 protects against systemic inflammation and myocardial dysfunction in murine polymicrobial sepsis. Shock 34, 281–290 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank G. Brown for help with laccase assays; J. Murkin and M. Deery for proteomics; L. Porter for metabolic-flux assays; J. Skepper for electron microscopy; I. Purvis for help with in vivo procedures; R. Rodrigues, M. Md-Ibrahim and J. Jones for cellular assays; S. Dhillon for the generation of constructs; T. Lawley, M. Pardo, J. Choudhary, K. Smith, J. Lee, D. Thomas, G. Schneditz, L. Haag, M. Parkes and R. Blumberg for discussions; all National Institute for Health Research Cambridge BioResource volunteers for the participation; the Cambridge BioResource staff for help with volunteer recruitment; members of the Cambridge BioResource SAB and Management Committee for support of this study; and the National Institute for Health Research Cambridge BRC Cell Phenotyping Hub for expertise and help. Access to Cambridge BioResource volunteers and their data and samples is governed by the Cambridge BioResource SAB (documents on access arrangements and contact details, http://www.cambridgebioresource.org.uk/). Supported by the European Research Council under the European Community's Seventh Framework Programme (FP7/2007-2013)/ERC Grant agreement 260961 (A.K.), the Wellcome Trust (investigator award 106260/Z/14/Z to A.K.; a PhD fellowship for clinicians to M.Z.C.; and a Career Re-Entry Fellowship to N.C.K.), the Wellcome Trust Sanger Institute (G.D., A.B., S.M., S.C. and K.B.), the US National Institutes of Health (5U420D011174 and 5U54HG006348 to A.B. and K.B.), the Biotechnology and Biological Sciences Research Council (M.J.O.W.), the National Institute for Health Research Cambridge Biomedical Research Centre, the European Crohn's and Colitis Organisation (M.T.) and the Swedish Medical Research Council and the Olle Engkvist foundation (M.D'A.).

Author information

Authors and Affiliations

Authors

Contributions

M.Z.C., together with S.L.K., G.W.S., S.S., J.W.A., M.T., T.R. and N.C.K., designed and performed most of the experiments; K.B., B.D. and A.B. designed, generated and confirmed the genotype of CRISPR-Cas9–generated mouse lines; Q.Z. and M.J.O.W. provided lipidomics experiments and analysis; G.A. and M.D'A. identified the cellular localization of FAMIN; S.C., S.M. and G.D. contributed Salmonella and part of the in vivo experimentation; K.P.B. and R.A.F. contributed to mycobacterial experiments; E.R.C. helped with metabolic-flux assays and ROS experimentation; J.L.G. contributed metabolomics experimentation and analysis; and A.K. devised and coordinated the project and, together with M.Z.C. and G.D., and with contributions from all authors, designed experiments, interpreted data and wrote the manuscript.

Corresponding author

Correspondence to Arthur Kaser.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Expression of Strep-tagged FAMIN in HEK293T cells.

Immunoblots (IB) of HEK293T lysates expressing N- and C-terminally Strep-tagged FAMIN(p.254I) and FAMIN(p.254V) variants for FAMIN and Strep-tag; β-actin loading control. Data are representative of three independent experiments.

Supplementary Figure 2 FAMIN localizes to peroxisomes.

(a) Co-localization by immunofluorescence (IF) of FAMIN (red) with PMP70 (green) in primary human macrophages (MΦ). DAPI nuclear staining, blue; scale bar = 10 μm. White box corresponds to enlarged images shown in Fig. 1d. (be) Co-localization by IF of FAMIN (red) with PMP70 (green), catalase (green), cytochrome oxidase IV (green) and calreticulin (green) in THP-1 macrophages. DAPI, blue; scale bar = 10 μm for original images and 5 μm for enlarged images. Upper panels: original image, middle panels: enlargement of the area shown in the white box, lower panels: further enlargement as indicated. No significant co-localization of FAMIN was detected with CENP-A, centromere; caveolin-2, cholesterol/sphingolipid enriched plasma membrane; EEA1 and RAB5, early endosome; LAMP1, lysosomes; NUP98, nuclear envelope; fibrillin, nucleolus and syntaxin 6, trans-Golgi network (data not shown). (f) Proximity-ligation assay (PLA) of FAMIN in combination with PMP70, Catalase (red) or centromere protein A (red) as negative control in THP-1 macrophages. DAPI, blue; scale bar = 10 μm. White box corresponds to enlarged images shown in Fig. 1e. Data are representative of three independent experiments.

Supplementary Figure 3 Generation of mFamin–/–, mFaminp254I and mFaminp284R mice.

Schematic representation of the targeting strategy for creating a mFamin–/– allele in murine ES cells. The targeting vector, L1L2_Bact_P (International Mouse Knockout Consortium), contains a reading frame-independent LacZ gene trap cassette consisting of an En2 splice acceptor (SA), an internal ribosomal entry site (IRES), LacZ gene and a polyadenylation site (PA). The vector also contains a selectable marker consisting of a human β-actin promoter (hBactP), neomycin resistance gene (neo) and a PA site. The vector is flanked by flippase recognition target (FRT) sites to allow removal of the targeting cassette and conditional mFamin deletion upon Cre-mediated recombination of the loxP sites. mFaminp254I and mFaminp284R mice were generated by CRISPR/Cas9 genome editing to introduce nucleotide changes to encode p.254I or p.284R amino acids, respectively, at indicated positions. Nucleotides in mFamin exon 5 were targeted using guide RNAs, ‘line’ 7 and ‘line’ 9, respectively as outlined in the methods. Two different targeting oligodeoxynucleotides were used for each: one containing only the nucleotide changes leading to the amino acid substitutions, the other containing 2 additional synonymous nucleotide changes (‘wobble’) in the underlined region.

Supplementary Figure 4 Famin expression is highest in M1 macrophages, and FAMIN deficiency does not affect FASN expression.

(a) mRNA expression of Famin in M0, M1 and M2 macrophages (MΦ). (b) mRNA expression of Fasn in M0, M1 and M2 macrophages. (c) Immunoblots (IB) of FASN in M1 and M2 macrophage cell lysates; β-actin loading control. Data are from one experiment with three mice representative of two (b,c; mean ± S.E.M.) or three independent experiments (c).

Supplementary Figure 5 FAMIN does not directly affect the Krebs cycle.

(a) Schematic representation of 1,2-13C-glucose incorporation, via citrate, into fatty acyl species. (b) Basal extracellular acidification rate (ECAR) of mFamin–/– and mFamin+/+ M1 and M2 macrophages (MΦ) in the presence of exogenous pyruvate (n = 5/14). (c) Relative levels of malate, fumarate, succinate and α-ketoglutarate in M1 and M2 macrophages. (d) Oxygen consumption rate (OCR) of mFamin–/– and mFamin+/+ M2 macrophages treated as indicated with 40 μM etomoxir (ETO) 1 h prior to OCR measurement and followed by sequential treatment (dotted vertical lines) with oligomycin (Oligo), FCCP, and rotenone
plus antimycin (Rot + ant). *P < 0.05, **P < 0.01 (Unpaired, two-tailed Student’s t-test). Data are pooled from three independent experiments (b; mean ± S.E.M.), from one experiment with seven mice per group (c; mean ± S.E.M.) or from one experiment with three mice representative of two independent experiments (d; mean ± S.E.M.).

Supplementary Figure 6 FAMIN-deficient M2 macrophages exhibit impaired mitochondrial ROS- and FAO-dependent production of extracellular ROS.

(a,b) Intracellular ROS measurement in unstimulated M1 and M2 mFamin–/– and mFamin+/+ macrophages (MΦ) stained with the cytosolic ROS indicator, 5-(and-6)-chloromethyl-2-7-dichlorodihydrofluorescein diacetate, acetyl ester (CM-H2DCFDA) and measured in relative fluorescence units, RFU. (ci) Zymosan induced eROS production in M1 and M2 macrophages treated as indicated for 16 h with 20 μM C75; or 1 h 40 μM etomoxir (ETO) or 500 μM mitoTEMPO prior to stimulation; or silenced for Fasn (Fasn siRNA), Cpt1a (Cpt siRNA) or Cybb (Cybb siRNA) or scrambled siRNA (Ctrl siRNA); Left, eROS kinetic plots measured in relative light units, RLU and right, area under curve, AUC. (j) NADPH quantification in M1 and M2 macrophages cell lysates. (k) PMA-induced eROS production in mFamin–/– and mFamin+/+ neutrophils. *P < 0.05, **P < 0.01 (Unpaired, two-tailed Student’s t-test). Data are from one experiment with three mice representative of two independent experiments (ak; mean ± S.E.M.).

Supplementary Figure 7 FAMIN p.I254V and p.C284R M2 macrophage have impaired eROS production.

(a) Zymosan-stimulated eROS production in mFamin–/–, mFaminp254V, mFaminp254I and mFaminp284R murine M2 macrophages (MΦ). (b) Zymosan induced eROS production in mFaminp254I and mFaminp254V M2 macrophages treated as indicated for 16 h with 20 μM C75. (c) FAMIN mRNA expression in M2 macrophages and neutrophils from healthy donors homozygous for the Crohn’s disease and leprosy risk (‘rs3764147G/G’) and non-risk (‘rs3764147A/A’) haplotypes. Data are from one experiment with three mice representative of three independent experiments (a,b; mean ± S.E.M.) or pooled from ten independent experiments (c; mean ± S.E.M.).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 and Supplementary Tables 1–5 (PDF 1676 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cader, M., Boroviak, K., Zhang, Q. et al. C13orf31 (FAMIN) is a central regulator of immunometabolic function. Nat Immunol 17, 1046–1056 (2016). https://doi.org/10.1038/ni.3532

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3532

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