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:

Dual roles of hexokinase 2 in shaping microglial function by gating glycolytic flux and mitochondrial activity

Abstract

Microglia continuously survey the brain parenchyma and actively shift status following stimulation. These processes demand a unique bioenergetic programme; however, little is known about the metabolic determinants in microglia. By mining large datasets and generating transgenic tools, here we show that hexokinase 2 (HK2), the most active isozyme associated with mitochondrial membrane, is selectively expressed in microglia in the brain. Genetic ablation of HK2 reduced microglial glycolytic flux and energy production, suppressed microglial repopulation, and attenuated microglial surveillance and damage-triggered migration in male mice. HK2 elevation is prominent in immune-challenged or disease-associated microglia. In ischaemic stroke models, however, HK2 deletion promoted neuroinflammation and potentiated cerebral damages. The enhanced inflammatory responses after HK2 ablation in microglia are associated with aberrant mitochondrial function and reactive oxygen species accumulation. Our study demonstrates that HK2 gates both glycolytic flux and mitochondrial activity to shape microglial functions, changes of which contribute to metabolic abnormalities and maladaptive inflammation in brain diseases.

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: Restrictive expression of hexokinase 2 in microglia in the brain.
Fig. 2: Hexokinase 2 regulates microglial metabolic flux.
Fig. 3: HK2 is required for microglial motility and dynamics.
Fig. 4: Hexokinase 2 deficiency delays microglial repopulation.
Fig. 5: Prominent upregulation of hexokinase 2 in activated microglia.
Fig. 6: Hexokinase 2 ablation potentiates brain damages in a stroke model.
Fig. 7: HK2 ablation impairs mitochondrial function and boosts pro-inflammatory responses in immune-challenged microglia.
Fig. 8: The dual roles of hexokinase 2 in shaping microglial functions.

Similar content being viewed by others

Data availability

RNA-seq data of repopulating microglia are available from the GEO under accession GSE188642. The data that support the finding of this study are available from the corresponding authors upon request. Source data are provided with this paper.

References

  1. O’Neill, L. A., Kishton, R. J. & Rathmell, J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 16, 553–565 (2016).

    Article  Google Scholar 

  2. Hu, Y. et al. mTOR-mediated metabolic reprogramming shapes distinct microglia functions in response to lipopolysaccharide and ATP. Glia 68, 1031–1045 (2020).

    Article  Google Scholar 

  3. Orihuela, R., McPherson, C. A. & Harry, G. J. Microglial M1/M2 polarization and metabolic states. Br. J. Pharmacol. 173, 649–665 (2016).

    Article  CAS  Google Scholar 

  4. Baik, S. H. et al. A breakdown in metabolic reprogramming causes microglia dysfunction in Alzheimer’s disease. Cell Metab. 30, 493–507 (2019).

    Article  CAS  Google Scholar 

  5. Ulland, T. K. et al. TREM2 maintains microglial metabolic fitness in Alzheimer’s disease. Cell 170, 649–663 (2017).

    Article  CAS  Google Scholar 

  6. Nugent, A. A. et al. TREM2 regulates microglial cholesterol metabolism upon chronic phagocytic challenge. Neuron 105, 837–854 (2020).

    Article  CAS  Google Scholar 

  7. Pan, R. Y. et al. Positive feedback regulation of microglial glucose metabolism by histone H4 lysine 12 lactylation in Alzheimer’s disease. Cell Metab. 34, 634–648 e6 (2022).

    Article  CAS  Google Scholar 

  8. Tay, T. L. et al. A new fate mapping system reveals context-dependent random or clonal expansion of microglia. Nat. Neurosci. 20, 793–803 (2017).

    Article  CAS  Google Scholar 

  9. Lawson, L. J., Perry, V. H. & Gordon, S. Turnover of resident microglia in the normal adult mouse brain. Neuroscience 48, 405–415 (1992).

    Article  CAS  Google Scholar 

  10. Davalos, D. et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 8, 752–758 (2005).

    Article  CAS  Google Scholar 

  11. Ransohoff, R. M. & Perry, V. H. Microglial physiology: unique stimuli, specialized responses. Annu Rev. Immunol. 27, 119–145 (2009).

    Article  CAS  Google Scholar 

  12. Shemer, A. et al. Microglia plasticity during health and disease: an immunological perspective. Trends Immunol. 36, 614–624 (2015).

    Article  CAS  Google Scholar 

  13. Elmore, M. R. et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 82, 380–397 (2014).

    Article  CAS  Google Scholar 

  14. Huang, Y. et al. Repopulated microglia are solely derived from the proliferation of residual microglia after acute depletion. Nat. Neurosci. 21, 530–540 (2018).

    Article  CAS  Google Scholar 

  15. Bruttger, J. et al. Genetic cell ablation reveals clusters of local self-renewing microglia in the mammalian central nervous system. Immunity 43, 92–106 (2015).

    Article  CAS  Google Scholar 

  16. Rao, J., Oz, G. & Seaquist, E. R. Regulation of cerebral glucose metabolism. Minerva Endocrinol. 31, 149–158 (2006).

    CAS  Google Scholar 

  17. Raichle, M. E., Posner, J. B. & Plum, F. Cerebral blood flow during and after hyperventilation. Arch. Neurol. 23, 394–403 (1970).

    Article  CAS  Google Scholar 

  18. Wilson, J. E. Isozymes of mammalian hexokinase: structure, subcellular localization and metabolic function. J. Exp. Biol. 206, 2049–2057 (2003).

    Article  CAS  Google Scholar 

  19. Wilson, J. E. Hexokinases. Rev. Physiol. Biochem. Pharmacol. 126, 65–198 (1995).

    Article  CAS  Google Scholar 

  20. Pastorino, J. G. & Hoek, J. B. Regulation of hexokinase binding to VDAC. J. Bioenerg. Biomembr. 40, 171–182 (2008).

    Article  CAS  Google Scholar 

  21. Moon, J. S. et al. mTORC1-Induced HK1-dependent glycolysis regulates NLRP3 inflammasome activation. Cell Rep. 12, 102–115 (2015).

    Article  CAS  Google Scholar 

  22. Everts, B. et al. TLR-driven early glycolytic reprogramming via the kinases TBK1-IKKvarepsilon supports the anabolic demands of dendritic cell activation. Nat. Immunol. 15, 323–332 (2014).

    Article  CAS  Google Scholar 

  23. Li, Y. et al. Hexokinase 2-dependent hyperglycolysis driving microglial activation contributes to ischemic brain injury. J. Neurochem. 144, 186–200 (2018).

    Article  CAS  Google Scholar 

  24. Pastorino, J. G., Shulga, N. & Hoek, J. B. Mitochondrial binding of hexokinase II inhibits Bax-induced cytochrome c release and apoptosis. J. Biol. Chem. 277, 7610–7618 (2002).

    Article  CAS  Google Scholar 

  25. Nederlof, R. et al. Targeting hexokinase II to mitochondria to modulate energy metabolism and reduce ischaemia-reperfusion injury in heart. Br. J. Pharmacol. 171, 2067–2079 (2014).

    Article  CAS  Google Scholar 

  26. Wolf, A. J. et al. Hexokinase is an innate immune receptor for the detection of bacterial peptidoglycan. Cell 166, 624–636 (2016).

    Article  CAS  Google Scholar 

  27. Hinrichsen, F. et al. Microbial regulation of hexokinase 2 links mitochondrial metabolism and cell death in colitis. Cell Metab. 33, 2355–2366 e8 (2021).

    Article  CAS  Google Scholar 

  28. Moehlman, A. T. & Youle, R. J. Mitochondrial quality control and restraining innate immunity. Annu. Rev. Cell Dev. Biol. 36, 265–289 (2020).

    Article  CAS  Google Scholar 

  29. Jin, H. S. et al. Mitochondrial control of innate immunity and inflammation. Immune Netw. 17, 77–88 (2017).

    Article  Google Scholar 

  30. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    Article  CAS  Google Scholar 

  31. Gosselin, D. et al., An environment-dependent transcriptional network specifies human microglia identity. Science 356, eaal3222 (2017).

  32. Stott, F. J. et al. The alternative product from the human CDKN2A locus, p14(ARF), participates in a regulatory feedback loop with p53 and MDM2. EMBO J. 17, 5001–5014 (1998).

    Article  CAS  Google Scholar 

  33. Gu, X. et al. Human Schlafen 5 inhibits proliferation and promotes apoptosis in lung adenocarcinoma via the PTEN/PI3K/AKT/mTOR pathway. Biomed. Res. Int. 6628682 (2021).

  34. Sen, S. et al. Role of hexokinase-1 in the survival of HIV-1-infected macrophages. Cell Cycle 14, 980–989 (2015).

    Article  CAS  Google Scholar 

  35. Roiniotis, J. et al. Hypoxia prolongs monocyte/macrophage survival and enhanced glycolysis is associated with their maturation under aerobic conditions. J. Immunol. 182, 7974–7981 (2009).

    Article  CAS  Google Scholar 

  36. Nowak, N., Kulma, A. & Gutowicz, J. Up-regulation of key glycolysis proteins in cancer development. Open Life Sci. 13, 569–581 (2018).

    Article  CAS  Google Scholar 

  37. Qin, C. et al. Dual functions of microglia in ischemic stroke. Neurosci. Bull. 35, 921–933 (2019).

    Article  Google Scholar 

  38. Schaar, K. L., Brenneman, M. M. & Savitz, S. I. Functional assessments in the rodent stroke model. Exp. Transl. Stroke Med. 2, 13 (2010).

    Article  Google Scholar 

  39. Soto-Heredero, G. et al. Glycolysis—a key player in the inflammatory response. FEBS J. 287, 3350–3369 (2020).

    Article  CAS  Google Scholar 

  40. Shoshan-Barmatz, V. et al. VDAC, a multi-functional mitochondrial protein regulating cell life and death. Mol. Asp. Med 31, 227–285 (2010).

    Article  CAS  Google Scholar 

  41. Ghosh, S. et al. Bioenergetic regulation of microglia. Glia 66, 1200–1212 (2018).

    Article  Google Scholar 

  42. Xiang, X. et al. Microglial activation states drive glucose uptake and FDG-PET alterations in neurodegenerative diseases. Sci. Transl. Med. 13, eabe5640 (2021).

    Article  CAS  Google Scholar 

  43. Bernier, L. P. et al. Microglial metabolic flexibility supports immune surveillance of the brain parenchyma. Nat. Commun. 11, 1559 (2020).

    Article  CAS  Google Scholar 

  44. Gimenez-Cassina, A. et al. Mitochondrial hexokinase II promotes neuronal survival and acts downstream of glycogen synthase kinase-3. J. Biol. Chem. 284, 3001–3011 (2009).

    Article  CAS  Google Scholar 

  45. Chow, H. M. et al. Age-related hyperinsulinemia leads to insulin resistance in neurons and cell-cycle-induced senescence. Nat. Neurosci. 22, 1806–1819 (2019).

    Article  CAS  Google Scholar 

  46. Chao, C. C. et al. Metabolic control of astrocyte pathogenic activity via cPLA2-MAVS. Cell 179, 1483–1498 (2019).

    Article  CAS  Google Scholar 

  47. Eyo, U. & Dailey, M. E. Effects of oxygen-glucose deprivation on microglial mobility and viability in developing mouse hippocampal tissues. Glia 60, 1747–1760 (2012).

    Article  Google Scholar 

  48. Palsson-McDermott, E. M. & O’Neill, L. A. The Warburg effect then and now: from cancer to inflammatory diseases. Bioessays 35, 965–973 (2013).

    Article  CAS  Google Scholar 

  49. Apostolova, I. et al. Hypermetabolism in the hippocampal formation of cognitively impaired patients indicates detrimental maladaptation. Neurobiol. Aging 65, 41–50 (2018).

    Article  CAS  Google Scholar 

  50. Mao, C. Y. et al. Brain glucose metabolism changes in Parkinson’s disease patients with CHCHD2 mutation based on [18F]-FDG PET imaging. J. Neurol. Sci. 369, 303–305 (2016).

    Article  CAS  Google Scholar 

  51. O’Connell, M. T. et al. Glucose metabolism in traumatic brain injury: a combined microdialysis and [18F]-2-fluoro-2-deoxy-d-glucose-positron emission tomography (FDG-PET) study. Acta Neurochir. Suppl. 95, 165–168 (2005).

    Article  Google Scholar 

  52. Sokoloff, L. Energetics of functional activation in neural tissues. Neurochem. Res. 24, 321–329 (1999).

    Article  CAS  Google Scholar 

  53. Leng, L. et al. Microglial hexokinase 2 deficiency increases ATP generation through lipid metabolism leading to beta-amyloid clearance. Nat. Metab. 4, 1287–1305 (2022).

    Article  CAS  Google Scholar 

  54. Maes, M. E. et al. Targeting microglia with lentivirus and AAV: recent advances and remaining challenges. Neurosci. Lett. 707, 134310 (2019).

    Article  CAS  Google Scholar 

  55. Bellver-Landete, V. et al. Microglia are an essential component of the neuroprotective scar that forms after spinal cord injury. Nat. Commun. 10, 518 (2019).

    Article  CAS  Google Scholar 

  56. Lloyd-Burton, S. M. et al. SPARC regulates microgliosis and functional recovery following cortical ischemia. J. Neurosci. 33, 4468–4481 (2013).

    Article  CAS  Google Scholar 

  57. Minhas, P. S. et al. Restoring metabolism of myeloid cells reverses cognitive decline in ageing. Nature 590, 122–128 (2021).

    Article  CAS  Google Scholar 

  58. Pajak, B, et al. 2-Deoxy-d-glucose and its analogs: from diagnostic to therapeutic agents. Int. J. Mol. Sci. 21, 234 (2019).

  59. Shoshan-Barmatz, V. & Golan, M. Mitochondrial VDAC1: function in cell life and death and a target for cancer therapy. Curr. Med. Chem. 19, 714–735 (2012).

    Article  CAS  Google Scholar 

  60. Ciscato, F. et al. Hexokinase 2 displacement from mitochondria-associated membranes prompts Ca2+-dependent death of cancer cells. EMBO Rep. 21, e49117 (2020).

    Article  CAS  Google Scholar 

  61. Cai, M., He, P. & Fang, D. L. Hypoxiainduced mitochondrial translocation of DNM1L increases mitochondrial fission and triggers mPTP opening in HCC cells via activation of HK2. Oncol. Rep. 42, 1125–1132 (2019).

    CAS  Google Scholar 

  62. Pasdois, P., Parker, J. E. & Halestrap, A. P. Extent of mitochondrial hexokinase II dissociation during ischemia correlates with mitochondrial cytochrome c release, reactive oxygen species production, and infarct size on reperfusion. J. Am. Heart Assoc. 2, e005645 (2012).

    Article  Google Scholar 

  63. De Jesus, A. et al. Hexokinase 1 cellular localization regulates the metabolic fate of glucose. Mol. Cell 82, 1261–1277 (2022).

  64. Mucke, L. et al. High-level neuronal expression of Aβ(1–42) in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J. Neurosci. 20, 4050–4058 (2000).

    Article  CAS  Google Scholar 

  65. Liu, C. et al. Mosaic analysis with double markers reveals tumor cell of origin in glioma. Cell 146, 209–221 (2011).

    Article  CAS  Google Scholar 

  66. Zhang, X. et al. Aged microglia promote peripheral T cell infiltration by reprogramming the microenvironment of neurogenic niches. Immun. Ageing 19, 34 (2022).

    Article  Google Scholar 

  67. Sipe, G. O. et al. Microglial P2Y12 is necessary for synaptic plasticity in mouse visual cortex. Nat. Commun. 7, 10905 (2016).

    Article  CAS  Google Scholar 

  68. Paris, I. et al. ProMoIJ: a new tool for automatic three-dimensional analysis of microglial process motility. Glia 66, 828–845 (2018).

    Article  Google Scholar 

Download references

Acknowledgements

We greatly appreciate A. Gitler at Stanford University for comments and suggestions on the manuscript. We thank P. Shi at the Institute of Translational Medicine of Zhejiang University for help with microglia sorting, and M. -X. Guan at the Division of Medical Genetics and Genomics of Zhejiang University School of Medicine for help with Metabolic Extracellular Flux analysis, W. Wang at the Charles Institute of Dermatology of University College Dublin and L. Chen at the First Affiliated Hospital of Zhejiang University for generously providing hydrogel, C. Liu at the School of Brain Science and Brain Medicine of Zhejiang University for generously providing transgenic mice. We are thankful to S. Fang at the Core Facilities of the School of Brain Science and Brain Medicine of Zhejiang University, H. Wu at the Center of Cryo-Electron Microscopy of Zhejiang University, Y. Huang, Y. Li and J. Wang at the Core Facilities of Zhejiang University for technical assistance. We greatly appreciate X. Zhang and A. Ping at the Second Affiliated Hospital of Zhejiang University for help with human microglia sorting. This work was supported by grants from the National Key Research and Development Program of China (2017YFA0104200 to Z.G.), National Natural Science Foundation of China (32070974 to Z.G., 82090033 to Z.G. and 82101413 to Y.H.), major programme of the National Natural Science Foundation of China (82090033 to S.D.) and China Postdoctoral Science Foundation (2020M671693 to Y.H.).

Author information

Authors and Affiliations

Authors

Contributions

Z.G., S.D., Y.H., K.C. and F.W. conceived the project and designed experiments; Y.H., K.C., F.W. and L.Q. carried out experiments; Y.H., K.C., F.W., W.W., W.M. and Y.L. analysed the data; B.S., L.S., J.Z. and J.Z. provide technical assistance. Z.G., Y.H., K.C., F.W., and W.W. wrote the paper; Z.G., S.D., Y.H., K.C., F.W., W.W., Y.L., W.G., Z.W., Y.X. and S.D. reviewed and edited the paper; Z.G., S.D. and Y.H. provided funding acquisition.

Corresponding authors

Correspondence to Shumin Duan or Zhihua Gao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Marco Colonna and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Alfredo Giménez-Cassina, 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 HK expression in human and mouse microglia.

(a) A correlation network graph of genes in metabolic pathways (1147 genes) expressed in different types of brain cells (data retrieved from GSE52564). Nodes represent transcripts, while edges represent the degree of correlation in expression. Heatmaps and line charts showing the expression profile of transcripts contained within 3 clusters corresponding to astrocytes, endothelial cells and microglia clusters are presented. (b) Principal component analysis (PCA) of the metabolic genes in glycolysis/gluconeogenesis in 7 cell types of the brain (data retrieved from GSE52564). (c) A comparison of metabolic gene expression in human and mouse microglia (data retrieved from GSE89960). (d-f) The expression of Hk1, Hk2 and Hk3 in mouse brain cells (data retrieved from GSE52564), n = 2 mice per group. MG, Microglia; ECs, Endothelial cells; OPC, Oligodendrocyte progenitor cells; Neu, Neurons; MO, Myelinating oligodendrocytes; NFO, Newly formed oligodendrocytes; Ast, Astrocytes. Data are presented as means ± SEM.

Source data

Extended Data Fig. 2 HK2 is selectively expressed in microglia.

(a) Representative micrographs (from 3 independent experiments) showing single-molecule fluorescence in situ hybridization (FISH) of Hk2 mRNA with different cellular markers, NeuN (neurons), Olig2 (oligodendrocytes) and GFAP (astrocytes) in the mouse brain. White arrowheads point to Hk2+ and Iba1+ microglia. Dotted boxes show regions of higher magnification.

Extended Data Fig. 3 Characterization of Hk2CreERT2-tdTomato mice.

(a) Flow cytometry analyses of tdT+ cells and CD11b+CD45low microglia in Hk2CreERT2-tdTomato mice. (b) Representative micrographs (from 3 independent experiments) showing the co-labeling of tdT+, Iba1+and Hk2+ in brain slices from Hk2CreERT2-tdTomato mice. Dotted boxes show regions of higher magnification. (c) FISH of Hk2 and tdT staining in brain slices from Hk2CreERT2-tdTomato mice and quantification. Dotted boxes show regions of higher magnification. n = 3 mice per group. (d) Representative micrographs (from 3 independent experiments) showing the three-dimension (3D) reconstruction of a tdT+ cell in brain slices from Hk2CreERT2-tdTomato mice using Imaris. (e) Representative micrographs (from 3 independent experiments) of tdT and different cellular markers, NeuN (neurons), Sox10 (oligodendrocytes) and GFAP (astrocytes) in the brains of Hk2CreERT2-tdTomato mice. (f) Representative micrographs (from 3 independent experiments) showing full co-localization of GFP, tdT and Iba1 in brain slices from Hk2CreERT2-tdTomato::Ai3fl/+ mice after tamoxifen induction. TAM, tamoxifen; D, day; tdT, tdTomato. Data are presented as means ± SEM.

Extended Data Fig. 4 HK2 is similarly expressed in microglia in different brain regions.

(a) Representative gating strategies for flow cytometry-based measurement of fluorescence intensity of microglial tdTomato in different brain regions. (b) Representative Western blot (from 2 independent experiments) analysis of HK2 protein in lysates from FACS-sorted microglia from adult cortex, midbrain and cerebellum and quantification (c). n = 2 from 16 mice per group. Data are presented as means ± SEM.

Source data

Extended Data Fig. 5 ECAR and OCR analyses of microglia after HK2 inhibition or deletion.

(a) ECAR measurement of primary mouse microglia (untreated or pretreated with 3-BrPA). (b) Quantification of basal ECAR and maximal ECAR from a. n = 7 or 4 independent cultures in Hk2fl/fl or Hk2-cKO groups, respectively. (c) OCR measurement of primary mouse microglia (untreated or pretreated with 3-BrPA). (d) Quantification of basal OCR and maximal OCR from c. n = 5 independent cultures per group. (e) A scheme of the standard Seahorse XF Mito-stress test. (f) OCR measurement of primary microglia from Hk2fl/fl (untreated or pretreated with 2-DG) and Hk2-cKO mouse brains in regular assay media containing pyruvate and glutamine. (g) Quantification of basal OCR and maximal OCR from f. n = 5 independent cultures per group. ECAR, extracellular acidification rate; OCR, oxygen consumption rate; OXPHOS, oxidative phosphorylation; TCA, tricarboxylic acid cycle; Glc, glucose; 3-BrPA, 3-Bromopyruvate; FCCP, Carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone; Olig, oligomycin; Antim, antimycin A; Rot, rotenone; 2-DG, 2-Deoxy-D-glucose; Data are presented as means ± SEM. Two-tailed Student’s t-tests for b, d. One-way ANOVA followed by Bonferroni’s post hoc tests for g, *p < 0.05, **p < 0.01 and ***p < 0.001.

Source data

Extended Data Fig. 6 Ablation of HK2 did not affect microglial density and morphology in the adult stage.

(a) A scheme of the experimental timeline for the analysis of microglial density and morphology from Hk2fl/fl and Hk2-cKO mice at 1 week after TAM administration. (b) Representative micrographs (from 3 independent experiments) showing the Iba1+ microglia and proliferative (BrdU+) microglia in adult Hk2fl/fl and Hk2-cKO mouse brains at 1 week after TAM administration. (c, d) Quantification of microglial number (c) and percentage of BrdU+ microglia (d) in the cortex from Hk2fl/fl and Hk2-cKO mice. n = 3 mice per group. (e) A scheme of the experimental timeline for analysis of microglial density and morphology from Hk2fl/fl and Hk2-cKO mice at 4 weeks after TAM administration. (f, g) Flow cytometry analysis and quantification of CD11b+ CD45low microglia in adult mice at 4 weeks after TAM administration. n = 3 mice per group. (h) Representative micrographs (from 3 independent experiments) showing 3D reconstruction of microglia from Hk2fl/fl and Hk2-cKO mice in the cortex using Imaris. (i) Scholl analysis of all reconstructed cells. n = 3 mice per group (total 36 cells). (j-l) Quantification of microglial morphology in the cortex from Hk2fl/fl and Hk2-cKO mice by Sholl analysis. n = 3 mice per group (total 36 cells). TAM, tamoxifen; D, day. Data are presented as means ± SEM. Two-tailed Student’s t-tests for c, d, f, g, j-l, *p < 0.05.

Source data

Extended Data Fig. 7 Ablation of HK2 did not affect Hk1 and Hk3 levels.

(a) The expression of Hk1, Hk2 and Hk3 in FACS-sorted microglia from Hk2fl/fl and Hk2-cKO mice at adult. n = 4 or 3 mice in Hk2fl/fl or Hk2-cKO groups, respectively. (b-d) The expression of Hk1, Hk2 and Hk3 in sorted microglia during repopulation in Hk2fl/fl and Hk2-cKO mice. n = 1481 or 264 cells for HomoMG, 162 or 264 cells for Precursor MG, 121 or 91 cells for G1/S MG, 200 or 108 cells for G2/M MG in Hk2fl/fl or Hk2-cKO groups, respectively. (e) Quantitative analyses of Hk1, Hk2, Hk3 and Cx3cr1 expression in the brain tissues from Hk2fl/fl and Hk2-cKO mice. n = 4 or 3 mice in Hk2fl/fl or Hk2-cKO groups, respectively. (f) CD11b+CD45low microglia and CD11b-CD45 non-microglial cells sorting from Hk2fl/fl and Hk2-cKO mice by flow cytometry. (g) Quantitative analyses of Hk2 and Cx3cr1 expression in CD11bCD45 non-microglial cells and CD11b+CD45low microglia from Hk2fl/fl and Hk2-cKO mice. n = 3 mice for microglia and non-microglial cells or 4 mice for microglia in Hk2fl/fl or Hk2-cKO groups, respectively. Data are presented as means ± SEM. Two-tailed student t-tests for a, e, g. Wilcoxon rank sum test and Bonferroni correction for b-d, *p < 0.05, **p < 0.01 and ***p < 0.001.

Source data

Extended Data Fig. 8 A comparison of repopulated microglia in Hk2fl/fl and Hk2-cKO mice.

(a) Pseudotime trajectory was constructed using Monocle 2 by re-ordering microglia population and single cells on the trajectory. Each microglia on the minimum spanning tree (MST) were colored by pseudotime. (b) Pseudotime trajectory was constructed and each microglia on the MST were colored by annotated microglial cell types during repopulation in Hk2fl/fl and Hk2-cKO mice. (c) The number of cells in different subsets of microglia from Hk2fl/fl and Hk2-cKO mice. (d) t-SNE plots of Hk2, Cx3cr1, Cdkn1a and Slfn5 gene expression between Hk2fl/fl and Hk2-cKO mice. n = 1481 or 264 cells for HomoMG, 162 or 264 cells for Precursor MG, 121 or 91 cells for G1/S MG, 200 or 108 cells for G2/M MG in Hk2fl/fl or Hk2-cKO groups, respectively. The color key indicates the expression levels.

Extended Data Fig. 9 Analyses of microglial number, phagocytosis, motor behavior and inflammation in Hk2fl/fl and Hk2-cKO mice before and after stroke.

(a) Representative micrographs (from 3 independent experiments) showing the density of microglia in the peri-infarct zone from Hk2fl/fl and Hk2-cKO mice at 3 and 7 Dpi. Dashed lines and dashed boxes indicate the border of ischemic core and regions of higher magnification, respectively. Rotarod (b, c) and open field (d, e) tests of adult Hk2fl/fl and Hk2-cKO mice. n = 10 mice per group. (f) Representative micrographs (from 3 independent experiments) showing CD68+ labeling in Iba1+ microglia in the peri-infarct zone from Hk2fl/fl and Hk2-cKO mice under sham and ischemic conditions (3 Dpi) and quantification of microglial volume and CD68+ volume in microglia (g, h). Dashed boxes show regions of higher magnification of microglia. n = 30 cells from 3 mice per group. RT-qPCR analysis of Tnfα, Il-6 and Il-1β levels (i) and Ifitm3, Stat1 levels (j) in the infarcted tissues at 1 Dpi. n = 5 mice per group for sham-operated animals or 6 mice per group for ischemic animals. RPM, rotations per minute; Dpi, days post ischemia. Data are presented as means ± SEM. Two-tailed student t-tests for b-e. Two-way ANOVA followed by Bonferroni’s post hoc tests for g, h, *p < 0.05, **p < 0.01 and ***p < 0.001.

Source data

Extended Data Fig. 10 Analyses of mitochondrial membrane potential and ROS levels in Hk2fl/fl and Hk2-deficient microglia upon LPS/ poly(I:C) stimulation.

(a-d) Analysis of microglial mitochondrial membrane potential and mitochondrial ROS after stimulation of LPS or poly(I:C) from Hk2fl/fl and Hk2-cKO mice in the presence or absence of Mito-TEPMO. n = 6 or 7 (a), 7 or 6 (b), 7 or 5 (c), 6 or 6(d) independent cultures for each treatment in Hk2fl/fl or Hk2-cKO groups, respectively. Data are presented as means ± SEM. Two-way ANOVA followed by Bonferroni’s post hoc tests for a-d, *p < 0.05, **p < 0.01 and ***p < 0.001.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 3–5 and authors statements

Reporting Summary

Supplementary Table 1

List of metabolic genes related to Fig. 1.

Supplementary Table 2

List of genes related to Fig. 5.

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. 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 Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots. gels

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

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

Hu, Y., Cao, K., Wang, F. et al. Dual roles of hexokinase 2 in shaping microglial function by gating glycolytic flux and mitochondrial activity. Nat Metab 4, 1756–1774 (2022). https://doi.org/10.1038/s42255-022-00707-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s42255-022-00707-5

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