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:

HIF-driven SF3B1 induces KHK-C to enforce fructolysis and heart disease

Subjects

Abstract

Fructose is a major component of dietary sugar and its overconsumption exacerbates key pathological features of metabolic syndrome. The central fructose-metabolising enzyme is ketohexokinase (KHK), which exists in two isoforms: KHK-A and KHK-C, generated through mutually exclusive alternative splicing of KHK pre-mRNAs. KHK-C displays superior affinity for fructose compared with KHK-A and is produced primarily in the liver, thus restricting fructose metabolism almost exclusively to this organ. Here we show that myocardial hypoxia actuates fructose metabolism in human and mouse models of pathological cardiac hypertrophy through hypoxia-inducible factor 1α (HIF1α) activation of SF3B1 and SF3B1-mediated splice switching of KHK-A to KHK-C. Heart-specific depletion of SF3B1 or genetic ablation of Khk, but not Khk-A alone, in mice, suppresses pathological stress-induced fructose metabolism, growth and contractile dysfunction, thus defining signalling components and molecular underpinnings of a fructose metabolism regulatory system crucial for pathological growth.

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: Identification of Hif1α-dependent splicing factors and splicing events.
Figure 2: SF3B1 is a HIF1α-target gene that mediates splicing of KHK.
Figure 3: Anabolic metabolism and growth as a function of KHK-C.
Figure 4: SF3B1 is required for pathologic cardiac growth and function.
Figure 5: Khk-A/C−/− mice are protected from pathologic cardiac growth and contractile dysfunction.

Similar content being viewed by others

References

  1. Yip, G. W., Fung, J. W. H., Tan, Y.-T. & Sanderson, J. E. Hypertension and heart failure: a dysfunction of systole, diastole or both? J. Hum. Hypertens. 23, 295–306 (2009)

    CAS  PubMed  Google Scholar 

  2. Shiojima, I. et al. Disruption of coordinated cardiac hypertrophy and angiogenesis contributes to the transition to heart failure. J. Clin. Invest. 115, 2108–2118 (2005)

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Kaelin, W. G. & Ratcliffe, P. J. Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway. Mol. Cell 30, 393–402 (2008)

    CAS  PubMed  Google Scholar 

  4. Semenza, G. L. Oxygen sensing, hypoxia-inducible factors, and disease pathophysiology. Annu. Rev. Pathol. 9, 47–71 (2014)

    CAS  PubMed  Google Scholar 

  5. Hill, J. A. & Olson, E. N. Cardiac plasticity. N. Engl. J. Med. 358, 1370–1380 (2008)

    CAS  PubMed  Google Scholar 

  6. Grosso, A. R. et al. Tissue-specific splicing factor gene expression signatures. Nucleic Acids Res. 36, 4823–4832 (2008)

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Barbosa-Morais, N. L., Carmo-Fonseca, M. & Aparício, S. Systematic genome-wide annotation of spliceosomal proteins reveals differential gene family expansion. Genome Res. 16, 66–77 (2006)

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Rappsilber, J., Ryder, U., Lamond, A. I. & Mann, M. Large-scale proteomic analysis of the human spliceosome. Genome Res. 12, 1231–1245 (2002)

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Diggle, C. P. et al. Ketohexokinase: expression and localization of the principal fructose-metabolizing enzyme. J. Histochem. Cytochem. 57, 763–774 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Krishnan, J. et al. Activation of a HIF1α-PPARγ axis underlies the integration of glycolytic and lipid anabolic pathways in pathologic cardiac hypertrophy. Cell Metab. 9, 512–524 (2009)

    CAS  PubMed  Google Scholar 

  11. Wang, C. et al. Phosphorylation of spliceosomal protein SAP 155 coupled with splicing catalysis. Genes Dev. 12, 1409–1414 (1998)

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Bonnal, S., Vigevani, L. & Valcárcel, J. The spliceosome as a target of novel antitumour drugs. Nature Rev. Drug Discov. 11, 847–859 (2012)

    CAS  Google Scholar 

  13. Corrionero, A., Minana, B. & Valcarcel, J. Reduced fidelity of branch point recognition and alternative splicing induced by the anti-tumor drug spliceostatin A. Genes Dev. 25, 445–459 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Dolatshad, H. et al. Disruption of SF3B1 results in deregulated expression and splicing of key genes and pathways in myelodysplastic syndrome hematopoietic stem and progenitor cells. Leukemia 29, 1092–1103 (2015)

    CAS  PubMed  Google Scholar 

  15. Ramasamy, R. & Goldberg, I. J. Aldose reductase and cardiovascular diseases, creating human-like diabetic complications in an experimental model. Circ. Res. 106, 1449–1458 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Lewis, C. A. et al. Tracing compartmentalized NADPH metabolism in the cytosol and mitochondria of mammalian cells. Mol. Cell 55, 253–263 (2014)

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Cirillo, P. et al. Ketohexokinase-dependent metabolism of fructose induces proinflammatory mediators in proximal tubular cells. J. Am. Soc. Nephrol. 20, 545–553 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Fang, M. et al. The ER UDPase ENTPD5 promotes protein N-glycosylation, the Warburg effect, and proliferation in the PTEN pathway. Cell 143, 711–724 (2010)

    CAS  PubMed  Google Scholar 

  19. Vander Heiden, M. G. et al. Evidence for an alternative glycolytic pathway in rapidly proliferating cells. Science 329, 1492–1499 (2010)

    ADS  CAS  PubMed  Google Scholar 

  20. Sharma, S. et al. Intramyocardial lipid accumulation in the failing human heart resembles the lipotoxic rat heart. FASEB J. 18, 1692–1700 (2004)

    CAS  PubMed  Google Scholar 

  21. Diggle, C. P. et al. Both isoforms of ketohexokinase are dispensable for normal growth and development. Physiol. Genomics 42A, 235–243 (2010)

    CAS  PubMed  Google Scholar 

  22. Gao, Y., Trivedi, S., Ferris, R. L. & Koide, K. Regulation of HPV16 E6 and MCL1 by SF3B1 inhibitor in head and neck cancer cells. Sci. Rep. 4, 6098 (2014)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. David, C. J., Chen, M., Assanah, M., Canoll, P. & Manley, J. L. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature 463, 364–368 (2010)

    ADS  CAS  PubMed  Google Scholar 

  24. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Shiota, M., Galassetti, P., Monohan, M., Neal, D. W. & Cherrington, A. D. Small amounts of fructose markedly augment net hepatic glucose uptake in the conscious dog. Diabetes 47, 867–873 (1998)

    CAS  PubMed  Google Scholar 

  26. Luo, W. et al. Pyruvate Kinase M2 Is a PHD3-Stimulated Coactivator for Hypoxia-Inducible Factor 1. Cell 145, 732–744 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Falcão-Pires, I. et al. Diabetes mellitus worsens diastolic left ventricular dysfunction in aortic stenosis through altered myocardial structure and cardiomyocyte stiffness. Circulation 124, 1151–1159 (2011)

    PubMed  Google Scholar 

  28. Kawasaki, T., Akanuma, H. & Yamanouchi, T. Increased fructose concentrations in blood and urine in patients with diabetes. Diabetes Care 25, 353–357 (2002)

    CAS  PubMed  Google Scholar 

  29. van Heerebeek, L. et al. Myocardial structure and function differ in systolic and diastolic heart failure. Circulation 113, 1966–1973 (2006)

    PubMed  Google Scholar 

  30. Kassiri, Z. et al. Combination of tumor necrosis factor-α ablation and matrix metalloproteinase inhibition prevents heart failure after pressure overload in tissue inhibitor of metalloproteinase-3 knock-out mice. Circ. Res. 97, 380–390 (2005)

    CAS  PubMed  Google Scholar 

  31. Nemir, M. et al. The Notch pathway controls fibrotic and regenerative repair in the adult heart. Eur. Heart J. 21, 2174–2185 (2014)

    Google Scholar 

  32. Haefliger, J. A. et al. Connexin43-dependent mechanism modulates renin secretion and hypertension. J. Clin. Invest. 116, 405–413 (2006)

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Kuroda, H., Kutner, R. H., Bazan, N. G. & Reiser, J. Simplified lentivirus vector production in protein-free media using polyethylenimine-mediated transfection. J. Virol. Methods 157, 113–121 (2009)

    CAS  PubMed  Google Scholar 

  34. Ventura, A. et al. Cre-lox-regulated conditional RNA interference from transgenes. Proc. Natl Acad. Sci. USA 101, 10380–10385 (2004)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  35. Huang, L. E., Gu, J., Schau, M. & Bunn, H. F. Regulation of hypoxia-inducible factor 1α is mediated by an O2-dependent degradation domain via the ubiquitin-proteasome pathway. Proc. Natl Acad. Sci. USA 95, 7987–7992 (1998)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Troilo, A. et al. HIF1α deubiquitination by USP8 is essential for ciliogenesis in normoxia. EMBO Rep. 15, 77–85 (2014)

    CAS  PubMed  Google Scholar 

  37. Casonato, A. et al. A new L1446P mutation is responsible for impaired von Willebrand factor synthesis, structure, and function. J. Lab. Clin. Med. 144, 254–259 (2004)

    CAS  PubMed  Google Scholar 

  38. Krishnan, J. et al. Dietary obesity-associated Hif1 activation in adipocytes restricts fatty acid oxidation and energy expenditure via suppression of the Sirt2-NAD+ system. Genes Dev. 26, 259–270 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  39. König, J. et al. iCLIP reveals the function of hnRNP particles in splicing at individual nucleotide resolution. Nature Struct. Mol. Biol. 17, 909–915 (2010)

    Google Scholar 

  40. Hirschy, A., Schatzmann, F., Ehlers, E. & Perriard, J. C. Establishment of cardiac cytoarchitecture in the developing mouse heart. Dev. Biol. 289, 430–441 (2006)

    CAS  PubMed  Google Scholar 

  41. Ishimoto, T. et al. Opposing effects of fructokinase C and A isoforms on fructose-induced metabolic syndrome in mice. Proc. Natl Acad. Sci. USA 109, 4320–4325 (2012)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Buescher, J. M., Moco, S., Sauer, U. & Zamboni, N. Ultrahigh performance liquid chromatography-tandem mass spectrometry method for fast and robust quantification of anionic and aromatic metabolites. Anal. Chem. 82, 4403–4412 (2010)

    CAS  PubMed  Google Scholar 

  43. Fuhrer, T., Heer, D., Begemann, B. & Zamboni, N. High-throughput, accurate mass metabolome profiling of cellular extracts by flow injection-time-of-flight mass spectrometry. Anal. Chem. 83, 7074–7080 (2011)

    CAS  PubMed  Google Scholar 

  44. Fukuzawa, J. et al. Cardiotrophin-1 increases angiotensinogen mRNA in rat cardiac myocytes through STAT3: an autocrine loop for hypertrophy. Hypertension 35, 1191–1196 (2000)

    CAS  PubMed  Google Scholar 

  45. Son, N.-H. et al. Cardiomyocyte aldose reductase causes heart failure and impairs recovery from ischemia. PLoS ONE 7, e46549 (2012)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Radin, N. S. Extraction of tissue lipids with a solvent of low toxicity. Methods Enzymol. 72, 5–7 (1981)

    CAS  PubMed  Google Scholar 

  47. Koopman, R., Schaart, G. & Hesselink, M. K. C. Optimisation of oil red O staining permits combination with immunofluorescence and automated quantification of lipids. Histochem. Cell Biol. 116, 63–68 (2001)

    CAS  PubMed  Google Scholar 

  48. Kim, K.-Y. et al. Parkin is a lipid-responsive regulator of fat uptake in mice and mutant human cells. J. Clin. Invest. 121, 3701–3712 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Georgiev, T. Simka, S. Xu, C. Bischoff, J. M. Dominguez, W. Kovacs and M. Piontek and other members of the Krek laboratory for discussions, help and technical assistance. We are grateful to M. Stoffel for performing tail vein injections. K. Chien, A. Asipu and R. J. Johnson provided mouse lines. This work was supported by grants from Sinergia (Swiss National Science Foundation) to W.K., T.P. and J. U. and the Swiss Heart Foundation to W.K.

Author information

Authors and Affiliations

Authors

Contributions

P.M, J.K. and W.K. designed and P.M. executed most experiments. F.G. performed ketohexokinase assays, ex vivo glucose/fructose uptake and lipid loading studies. A.S. performed mouse surgeries, echocardiography and necropsy analysis under the supervision of T.P. M.H. and N.Z. performed metabolomic analysis. J.U. and M.K. performed splice junction microarrays and initial data analysis. C.C. performed ATP measurements and Sf3b1-rescue experiments, O.F. quantified lipids and A.V. performed biodistribution experiments. N.F. and Y.C. generated the splice factor list and analysed splice junction microarray data. S.S. and S.N.S. provided human left ventricular biopsies. P.M. and W.K, with help from J. K., wrote the paper.

Corresponding author

Correspondence to Wilhelm Krek.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 HIF1α induces SF3B1 gene expression that mediates KHK-C isoform expression.

a, Schematic experimental workflow. b, Comparison of C1QBP, HNRNPH3, JMJD6 and SF3B1 promoter sequences of human and mouse illustrate putative HREs up to 1,000 bp upstream of the transcription start site (TSS). The core consensus HRE motif is shown in colour. c, d, qPCR (c) and immunoblotting (d) for Sf3b1 expression in NMCs transduced with scrambled shNs viruses or viruses bearing two distinct shRNAs against Sf3b1. All values obtained by qPCR are presented in relation to Sf3b1 expression in shNs (set as 1.0) (n = 3 biological replicates per group). e, RT–PCR analysis of Khk mRNA as in Fig. 1g in NMCs transduced as indicated. f, RT–PCR analyses of Khk mRNA from NMCs infected as indicated and stimulated with mock or isoproterenol (ISO). PCR products were digested with HincII as in Fig. 1c . Splicing is quantified as percent of the Khk-C isoform (%Khk-C). g, RT–PCR based assessment of Khk-A and Khk-C isoform expression as done in Fig. 1g. h, i, Ventricular samples of sham- or 1K1C-operated mice were analysed for Khk isoform expression (h) as done in Fig. 1g or Sf3b1, Khk-A and Khk-C mRNA expression (i) by qPCR; n = 6 for each group. j, k, Ventricular samples of sham- or TAC-treated mice (j) and NaCl- or isoproterenol-treated mice (k) were analysed for expression of Sf3b1, Khk-A and Khk-C mRNA expression by qPCR (n = 6 for each individual group). l, NMCs transduced as specified were validated for protein expression of indicated proteins by using immunoblots. In addition lysates of NMCs overexpressing KHK-A or KHK-C were processed for immunoblotting Khk antibodies. m, n, Ventricular samples of sham- or TAC-treated (m) and sham- or isoproterenol-treated mice (n) were analysed for indicated proteins by immunoblotting. o, Probes from left ventricles of 1K1C-, TAC- or isoproterenol-treated mice and their corresponding controls were analysed for mRNA expression of Hif1α targets. All values were presented in relation to sham-operated controls set as 1.0 (n = 6 for each individual group). p, q, Left-ventricular biopsies of HCM patients and healthy controls were analysed for mRNA expression of SF3B1, KHK-A and KHK-C (p) and HIF1α targets (q). All values are presented in relation to healthy controls set as 1.0 (n = 6 for controls and n = 16 for patient samples). Error bars are s.d. (c) or s.e.m. (ik, oq). *P < 0.05; **P < 0.01; ***P < 0.001; two-tailed unpaired t-test (c, ik, oq).

Extended Data Figure 2 SF3B1 is a HIF1α-target gene, whose product regulates growth and metabolic shift in vitro.

a, NMCs infected as denoted, were stimulated with either mock, phenylephrine and isoproterenol, respectively. Sf3b1 mRNA expression was examined by qPCR. (n = 3 biological replicates). b, NMCs were transduced as indicated and processed for qPCR analysis of Sf3b1 mRNA. (n = 3 biological replicates). c, Comparative analysis of a conserved HRE (shown in red) in the promoter of SF3B1 of diverse mammalian species located 122 bp downstream of the transcription start site (TSS). dg, Immunoblot detection of indicated proteins of NMCs transduced with shNs, and shSf3b lentiviruses treated with phenylephrine or isoproterenol (d) co-infected with empty vector or HIF1αΔODD (e) cultured under normoxic (21% O2) or hypoxic (3% O2) conditions (f) and co-transduced with shVhl lentiviruses (g). ho, Evaluation of [3H]leucine incorporation and the myosin heavy chain α/β mRNA expression ratio in NMCs treated with either phenylephrine or isoproterenol (h, l), HIF1αΔODD overexpression (i, m), hypoxia (j, n) or pVhl deletion (k, o), with or without shSf3b1 mediated Sf3b1 depletion. Obtained values are shown relative to incorporated radioactivity in control (shNS) NMCs (set as 1.0) (n = 4 per individual group). p, NMCs treated as in i were microscopically analysed for cell size using CellProfiler. Data represents 1 of 3 independent experiments with approximately 200 analysed cells per experiment. q, Lysates of NMCs treated as indicated, were assessed for protein expression of specified proteins by immunoblots. r, Evaluation of [3H]leucine incorporation of NMCs infected as indicated. Obtained values are shown relative to incorporated radioactivity in control (shNs/empty) NMCs (set as 1.0) (n = 3 per individual group). sz, Assessment of extracellular acidification rate (ECAR), and oleic acid- and carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone (FCCP)-induced oxygen consumption rate (OCR) in NMCs transduced and treated as indicated. Highlighted ECAR measurements were done at baseline. All data are compared to shNs-transduced NMCs. (n = 8 biological replicates in the group shNs + isoproterenol; n = 10 biological replicates in all other groups). Error bars are s.d. (a, b, ho, r) or s.e.m. (p, sz). *P < 0.05; **P < 0.01; ***P < 0.001. Two-tailed unpaired t-test (b, t, u, wz). One-way ANOVA followed by Dunnett’s multiple comparison post-test (a, hp, r).

Extended Data Figure 3 Influence of U2snRNP-complex members on KHK isoform expression and validation of KHK-C-mediated fructose uptake and metabolism.

a, NMCs were transduced with either shNs or shSf3b1 and transfected with codon optimized Sf3b1 or empty construct and expression of denoted proteins was assessed. b, NMCs were transduced with either shNs or shSf3b1 and transfected with codon-optimized Sf3b1 or empty construct, respectively. Exogenous Sf3b1-expression was evaluated by qRT–PCR. Three different primer pairs were used. Primer pair A detects both endogenous and exogenous codon-optimized Sf3b1 (top). Primer pair B recognizes Sf3b1 close to the shRNA binding site and is specific for endogenous Sf3b1 (left). Primer pair C is located in the codon-optimized region (right). Values were normalized to shNs/empty-vector-infected NMCs (n = 3 biological replicates per group). c, [3H]leucine incorporation normalized to cell number in NMCs treated as in b (n = 4 biological replicates per group). d, NMCs transduced with empty vector or human SF3A3 or SF3B3 were assessed for mRNA expression of Khk-A and Khk-C by qRT–PCR. Values were normalized to empty-vector-transduced NMCs (n = 3 biological replicates per group). e, NMCs transduced as in d were immunoblotted for indicated proteins. f, NMCs transduced as indicated were probed for mRNA expression of Khk-A or Khk-C by qRT–PCR. Values were normalized to shNs/empty-vector-transduced NMCs (n = 3 biological replicates). g, NMCs treated with either scrambled shRNA or two different shRNAs targeting either Sf3a3, Sf3b2 or Sf3b3 were co-transduced with empty vector or HIF1αΔODD. Protein expression of Khk-A/C or Khk-C specifically was evaluated by immunoblotting. h, Evaluation of [3H]leucine incorporation of NMCs infected as in d normalized to empty-vector-transduced NMCs (n = 4 biological replicates per group). i, Lysates of NMCs treated as indicated were immunoblotted for specified proteins. j, KHK-activity in lysates of NMCs transduced with a combination of lentiviruses expressing either shKhk targeting mouse Khk and human KHK-A or shKhk and human KHK-C or a scrambled shRNA control together with an empty vector. k, Evaluation of the kinetics of [13C]fructose-1-phosphate accumulation in NMCs treated as in i at the respective time points (n = 3 biological replicates). l, Glut5 mRNA expression in NMCs transduced as indicated. (n = 3 biological replicates per group). m, NMCs transduced as indicated were microscopically analysed for cell size. Data represents 1 of 3 experiments with approximately 50 analysed cells per experiment n, Khk-A- and Khk-C-specific shRNAs were evaluated for their potential to inhibit Khk-A and Khk-C isoform expression, respectively. NMCs were infected with shNs, shKhk-A or shKhk-C lentiviruses. Data are shown in relation to shNs NMCs (set to 1.0) (n = 3 biological replicates). Error bars are s.d. (bd, f, h, k, l, n) or s.e.m. (m). *P < 0.05; **P < 0.01; ***P < 0.001. One-way ANOVA followed by Dunnett’s multiple comparison post-test (bd, f, k, l, m, n).

Extended Data Figure 4 Downstream metabolic effects of fructose metabolism in NMCs.

a, log2 fold change of metabolites in NMCs overexpressing KHK-C and cultured with glucose or glucose and fructose compared to the corresponding control transduced NMCs (n = 4 biological replicates per group). b, Incorporation of [3H]fructose into RNA (top), DNA (middle) and protein (bottom) of NMCs treated as indicated (n = 4 biological replicates per group). c, Uptake of [14C]deoxyglucose in NMCs infected as in b at depicted time points (n = 4 biological replicates per group). d, [3H]leucine incorporation in NMCs transduced with shNs or shKhk and co-transduced with either empty overexpression vector or KHK-A or KHK-C respectively. NMCs were cultured in media with increasing fructose concentrations, under physiologic glucose amounts or with increasing glucose concentrations under physiologic fructose amounts. Data are presented relative to shNs/empty-vector-transduced NMCs at 5 mM glucose/0 μM fructose (n = 4 biological replicates). e, ADP/ATP ratios in NMCs transduced as indicated. (n = 4 biological replicates, data show 1 of 3 representative experiments). f, Immunofluorescence images of NMCs transduced with empty vector (upper panel) or HIF1αΔODD (middle panel) lentiviruses or exposed to hypoxia (lower panel) were additionally transduced as indicated. Prior to staining for sarcomeric α-actinin, Oil Red O and DAPI, NMCs were incubated with oleic acid. g, Quantification of lipid droplets/cell in NMCs of immunofluorescent images shown in f. h, Ratio of lipid droplets/cell area of NMCs shown in g. il, NMCs were infected as indicated and processed for oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) measurements under basal conditions or after injection of OA or FCCP. Depicted are rates expressed as OCR to ECAR ratios (upper panels) or individual ECAR (for measurements at baseline) or OCR values (n = 8 biological replicates for HIF1αΔODD/shKhk-A; n = 10 biological replicates for all other groups). Error bars are s.d. (b–e) or s.e.m. (g–l). *P < 0.05; **P < 0.01; ***P < 0.001. One-way ANOVA followed by Dunnett’s multiple comparison post-test (b–e, g, h). Two-tailed unpaired t-test (i–l).

Extended Data Figure 5 Fructose and glucose uptake ex vivo and validation of AAV9-fl/fl-shSf3b1 viruses.

a, Ratio of left-ventricular weight to body weight of sham- or TAC-treated C57/Bl6J mice; (n = 7 per group). b, c, [3H]fructose (b) and [14C]deoxy-glucose (c) were co-applied by oral gavage and biodistribution was measured at 45 min post injection (n = 7 per group). d, Ratio of left-ventricular weight to tibia length (LV/TL) of sham- or 1K1C-treated C57/Bl6J wild-type mice (n = 6 for sham and n = 7 for 1K1C-treated mice). e, Fructose levels in probes from left ventricles of indicated mice as determined by a quantitative colorimetric assay, normalized to protein amount (n = 4 for sham and n = 5 for hearts of 1K1C-treated mice). f, g, Freshly isolated left-ventricular pieces of sham- or 1K1C-treated mice incubated in medium containing either [14C]fructose (f) or [14C]deoxyglucose (g) as indicated were processed for uptake measurements. Counts were normalized to tissue weight (n = 6 per time point and treatment). hj, qPCR analyses of Glut5 mRNA expression in left ventricles of 1K1C- (h), TAC- (i) or isoproterenol-treated (j) mice. All values are presented in relation to Glut5 expression in shams (set as 1.0). n = 5 for sham-treated mice as controls for TAC-treated mice (n = 6), all other groups n = 7. k, l, Relative mRNA expression of GLUT5 and aldose reductase (AKR1B1) in left-ventricular biopsies of patients diagnosed with aortic stenosis (k) or HCM (l) in relation to healthy controls (set as 1.0) was analysed by qPCR (n = 6 for controls; n = 16 for HCM and n = 17 for patients with aortic stenosis). m, Integration of AAV9-fl/fl-shSf3b1#1 viruses in the myocardium of Mlc2-cre or Mlc2v-cre+ mice was examined 12 weeks after intravenous injection by confocal microscopy (using 20× magnification) of left-ventricular cryoslices stained against GFP and sarcomeric α-actinin and counterstained for DAPI. n, Ventricular samples of Mlc2v-cre or Mlc2-cre+ mice transduced with AAV9-fl/fl-shSf3b1#1 or AAV9-fl/fl-shSf3b1#2 viruses were assessed for expression of indicated proteins by immunoblotting. o, p, Ratio of left-ventricular weight to tibia length (o) and ejection fraction (%EF) (p) of sham- or TAC-treated Mlc2v-cre and Mlc2v-cre+ mice injected with AAV9-fl/fl-shSf3b1#1 (number of mice per group is given in Extended Data Table 1). q, Lysates of hearts from sham- or TAC-operated Mlc2v-cre and Mlc2v-cre+ mice transduced with AAV9-fl/fl-shSf3b1#1 viruses were processed for immunoblotting with antibodies as denoted. Error bars are s.e.m. (al, o, p). *P < 0.05; **P < 0.01; ***P < 0.001, two-tailed unpaired t-test (al, o, p).

Extended Data Figure 6 Pathologic growth is dependent on HIF1α-driven and SF3B1-mediated KHK-C expression.

af, Ratio of left-ventricular weight to tibia length (LV/TL) (a), mRNA expression of the hypertrophic markers Nppa (b), myosin heavy chain β (βMhc) (c) and myosin heavy chain α (αMhc) (d), ejection fraction (%EF) (e) and left-ventricular ATP content (f) of sham- or TAC-treated Khk-A+/+ and Khk-A−/− mice. gl, Ratio of left-ventricular weight to tibia length (LV/TL) (g), mRNA expression of the hypertrophic markers Nppa (h), myosin heavy chain β (βMhc) (i) and myosin heavy chain α (αMhc) (j), ejection fraction (%EF) (k) and left-ventricular ATP content (l) of Khk-A/C+/+ and Khk-A/C−/− mice. m, n, Lysates from ventricles of sham- or TAC-operated Khk-A+/+ and Khk-A−/− mice (m) and Khk-A/C+/+ and Khk-A/C−/− mice (n) were processed for immunoblotting with antibodies as indicated. ot, Assessment of hypertrophic marker gene expression (Nppa, βMhc and αMhc) in ventricles of TAC- vs sham-treated Khk-A−/− mice and their corresponding Khk-A+/+ controls (oq) as well as Khk-A/C−/− mice and their corresponding Khk-A/C+/+ controls (rt). u, v, Mean blood pressure (BP) in mmHg of Khk-A+/+ and Khk-A−/− (u) as well as Khk-A/C+/+ and Khk-A/C−/− mice (v) 8 weeks after sham or 1K1C treatment. For al, ov, the number of mice per group is given in Extended Data Table 2. Error bars are s.e.m. *P < 0.05; **P < 0.01; ***P < 0.001; two-tailed unpaired t-test.

Extended Data Figure 7 Model explaining how HIF1α activation of SF3B1-dependent splicing of KHK regulates fructose and glucose metabolism to promote cardiac hypertrophy in response to pathologic stress.

In this model, pathologic stress leads to increased expression of HIF1α and HIF1α-dependent activation of genes encoding glycolytic enzymes and the splicing factor SF3B1. SF3B1, in turn, assembles at the branch-point sequence upstream of exon 3C of KHK pre-mRNA leading to an inclusion of this exon and KHK-C protein production. This shift in isoform expression from KHK-A to KHK-C in response to HIF1α–SF3B1 pathway activation drives KHK-C-dependent fructose uptake via stimulation of GLUT5 expression, the conversion of fructose to fructose-1-phosphate (F1P) and contributes simultaneously to the activation of glucose uptake and metabolism through a yet-to-be-determined mechanism. The model holds that the unrestrained conversion of fructose to F1P by KHK-C limits ATP levels, thereby alleviating potential allosteric inhibition of phosphofructokinase (PFK) by ATP to maintain a high glycolytic flux. F1P is further metabolized to dehydroxyacetone phosphate (DHAP) and glyceraldehyde (GA). While DHAP serves as a precursor for glycerol synthesis, GA can be further converted to glyceraldehyde-3-phosphate (G3P), a key glycolytic intermediate. G3P can be channelled into the non-oxidative pentose phosphate pathway (PPP) supporting nucleic and amino acid biosynthesis. This metabolic constellation, created by the activation of the HIF1α–SF3B1–KHK-C axis, increases macromolecular biosynthetic capacity essential for hypertrophic growth, steatosis and cardiac dysfunction. HKII and F-1,6-BP denotes hexokinase II and fructose-1,6-bisphosphate, respectively.

Extended Data Table 1 Echocardiographic analysis of sham-, 1K1C- or TAC-treated Mlc2v-cre/-cre+ mice injected with AAV9-fl/fl-shSf3b1 viruses.
Extended Data Table 2 Echocardiographic analysis of sham- or TAC-operated Khk-A+/+/Khk-A−/− and Khk-A/C+/+/Khk-A/C−/− mice.
Extended Data Table 3 Echocardiographic analysis of sham- or 1K1C-operated Khk-A+/+/Khk-A−/− and Khk-A/C+/+/Khk-A/C−/− mice.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-7, Supplementary Table 4, Supplementary Data 1 and Supplementary Western Blots and gels. (PDF 4756 kb)

Supplementary Information

This file contains Supplementary Table 1. (XLSX 69 kb)

Supplementary Information

This file contains Supplementary Table 2. (XLSX 103 kb)

Supplementary Information

This file contains Supplementary Table 3. (XLSX 721 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mirtschink, P., Krishnan, J., Grimm, F. et al. HIF-driven SF3B1 induces KHK-C to enforce fructolysis and heart disease. Nature 522, 444–449 (2015). https://doi.org/10.1038/nature14508

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14508

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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