DOI QR코드

DOI QR Code

Allithiamine Exerts Therapeutic Effects on Sepsis by Modulating Metabolic Flux during Dendritic Cell Activation

  • Choi, Eun Jung (Department of Biomedical Science, School of Medicine, Kyungpook National University) ;
  • Jeon, Chang Hyun (Department of Biomedical Science, School of Medicine, Kyungpook National University) ;
  • Park, Dong Ho (Department of Ophthalmology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University) ;
  • Kwon, Tae-Hwan (Department of Biomedical Science, School of Medicine, Kyungpook National University)
  • Received : 2020.10.11
  • Accepted : 2020.11.02
  • Published : 2020.11.30

Abstract

Recent studies have highlighted that early enhancement of the glycolytic pathway is a mode of maintaining the proinflammatory status of immune cells. Thiamine, a wellknown co-activator of pyruvate dehydrogenase complex, a gatekeeping enzyme, shifts energy utilization of glucose from glycolysis to oxidative phosphorylation. Thus, we hypothesized that thiamine may modulate inflammation by alleviating metabolic shifts during immune cell activation. First, using allithiamine, which showed the most potent anti-inflammatory capacity among thiamine derivatives, we confirmed the inhibitory effects of allithiamine on the lipopolysaccharide (LPS)-induced pro-inflammatory cytokine production and maturation process in dendritic cells. We applied the LPS-induced sepsis model to examine whether allithiamine has a protective role in hyper-inflammatory status. We observed that allithiamine attenuated tissue damage and organ dysfunction during endotoxemia, even when the treatment was given after the early cytokine release. We assessed the changes in glucose metabolites during LPS-induced dendritic cell activation and found that allithiamine significantly inhibited glucose-driven citrate accumulation. We then examined the clinical implication of regulating metabolites during sepsis by performing a tail bleeding assay upon allithiamine treatment, which expands its capacity to hamper the coagulation process. Finally, we confirmed that the role of allithiamine in metabolic regulation is critical in exerting anti-inflammatory action by demonstrating its inhibitory effect upon mitochondrial citrate transporter activity. In conclusion, thiamine could be used as an alternative approach for controlling the immune response in patients with sepsis.

Keywords

References

  1. Armstrong, J.A., Cash, N.J., Ouyang, Y., Morton, J.C., Chvanov, M., Latawiec, D., Awais, M., Tepikin, A.V., Sutton, R., and Criddle, D.N. (2018). Oxidative stress alters mitochondrial bioenergetics and modifies pancreatic cell death independently of cyclophilin D, resulting in an apoptosis-tonecrosis shift. J. Biol. Chem. 293, 8032-8047. https://doi.org/10.1074/jbc.RA118.003200
  2. Bozic, I., Savic, D., Laketa, D., Bjelobaba, I., Milenkovic, I., Pekovic, S., Nedeljkovic, N., and Lavrnja, I. (2015). Benfotiamine attenuates inflammatory response in LPS stimulated BV-2 microglia. PLoS One 10, e0118372. https://doi.org/10.1371/journal.pone.0118372
  3. Cubillos-Ruiz, J.R., Silberman, P.C., Rutkowski, M.R., Chopra, S., Perales-Puchalt, A., Song, M., Zhang, S., Bettigole, S.E., Gupta, D., Holcomb, K., et al. (2015). ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell 161, 1527-1538. https://doi.org/10.1016/j.cell.2015.05.025
  4. Dellinger, R.P., Levy, M.M., Rhodes, A., Annane, D., Gerlach, H., Opal, S.M., Sevransky, J.E., Sprung, C.L., Douglas, I.S., Jaeschke, R., et al. (2013). Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit. Care Med. 41, 580-637. https://doi.org/10.1097/CCM.0b013e31827e83af
  5. den Brok, M.H., Raaijmakers, T.K., Collado-Camps, E., and Adema, G.J. (2018). Lipid droplets as immune modulators in myeloid cells. Trends Immunol. 39, 380-392. https://doi.org/10.1016/j.it.2018.01.012
  6. Donnino, M.W., Andersen, L.W., Chase, M., Berg, K.M., Tidswell, M., Giberson, T., Wolfe, R., Moskowitz, A., Smithline, H., Ngo, L., et al. (2016). Randomized, double-blind, placebo-controlled trial of thiamine as a metabolic resuscitator in septic shock: a pilot study. Crit. Care Med. 44, 360-367.
  7. Fleischmann, C., Scherag, A., Adhikari, N.K., Hartog, C.S., Tsaganos, T., Schlattmann, P., Angus, D.C., Reinhart, K., and International Forum of Acute Care Trialists (2016). Assessment of global incidence and mortality of hospital-treated sepsis. Current estimates and limitations. Am. J. Respir. Crit. Care Med. 193, 259-272. https://doi.org/10.1164/rccm.201504-0781OC
  8. Friedman, E.A., Ogletree, M.L., Haddad, E.V., and Boutaud, O. (2015). Understanding the role of prostaglandin E2 in regulating human platelet activity in health and disease. Thromb. Res. 136, 493-503. https://doi.org/10.1016/j.thromres.2015.05.027
  9. Hanberry, B.S., Berger, R., and Zastre, J.A. (2014). High-dose vitamin B1 reduces proliferation in cancer cell lines analogous to dichloroacetate. Cancer Chemother. Pharmacol. 73, 585-594. https://doi.org/10.1007/s00280-014-2386-z
  10. Infantino, V., Iacobazzi, V., Menga, A., Avantaggiati, M.L., and Palmieri, F. (2014). A key role of the mitochondrial citrate carrier (SLC25A1) in TNFalpha- and IFNgamma-triggered inflammation. Biochim. Biophys. Acta 1839, 1217-1225. https://doi.org/10.1016/j.bbagrm.2014.07.013
  11. Infantino, V., Pierri, C.L., and Iacobazzi, V. (2019). Metabolic routes in inflammation: the citrate pathway and its potential as therapeutic target. Curr. Med. Chem. 26, 7104-7116. https://doi.org/10.2174/0929867325666180510124558
  12. Liu, Y., Jennings, N.L., Dart, A.M., and Du, X.J. (2012). Standardizing a simpler, more sensitive and accurate tail bleeding assay in mice. World J. Exp. Med. 2, 30-36. https://doi.org/10.5493/wjem.v2.i2.30
  13. MacPherson, S., Horkoff, M., Gravel, C., Hoffmann, T., Zuber, J., and Lum, J.J. (2017). STAT3 regulation of citrate synthase is essential during the initiation of lymphocyte cell growth. Cell Rep. 19, 910-918. https://doi.org/10.1016/j.celrep.2017.04.012
  14. McCall, C.E., Zabalawi, M., Liu, T., Martin, A., Long, D.L., Buechler, N.L., Arts, R.J.W., Netea, M., Yoza, B.K., Stacpoole, P.W., et al. (2018). Pyruvate dehydrogenase complex stimulation promotes immunometabolic homeostasis and sepsis survival. JCI Insight 3, e99292. https://doi.org/10.1172/jci.insight.99292
  15. Merz, T.M., Pereira, A.J., Schurch, R., Schefold, J.C., Jakob, S.M., Takala, J., and Djafarzadeh, S. (2017). Mitochondrial function of immune cells in septic shock: a prospective observational cohort study. PLoS One 12, e0178946. https://doi.org/10.1371/journal.pone.0178946
  16. Nayak, M.K., Dhanesha, N., Doddapattar, P., Rodriguez, O., Sonkar, V.K., Dayal, S., and Chauhan, A.K. (2018). Dichloroacetate, an inhibitor of pyruvate dehydrogenase kinases, inhibits platelet aggregation and arterial thrombosis. Blood Adv. 2, 2029-2038. https://doi.org/10.1182/bloodadvances.2018022392
  17. Nemzek, J.A., Xiao, H.Y., Minard, A.E., Bolgos, G.L., and Remick, D.G. (2004). Humane endpoints in shock research. Shock 21, 17-25.
  18. Nuzzo, E., Berg, K.M., Andersen, L.W., Balkema, J., Montissol, S., Cocchi, M.N., Liu, X., and Donnino, M.W. (2015). Pyruvate dehydrogenase activity is decreased in the peripheral blood mononuclear cells of patients with sepsis. A prospective observational trial. Ann. Am. Thorac. Soc. 12, 1662-1666.
  19. O'Neill, L.A. (2014). Glycolytic reprogramming by TLRs in dendritic cells. Nat. Immunol. 15, 314-315. https://doi.org/10.1038/ni.2852
  20. Singer, M. (2014). The role of mitochondrial dysfunction in sepsis-induced multi-organ failure. Virulence 5, 66-72. https://doi.org/10.4161/viru.26907
  21. Van den Bossche, J., O'Neill, L.A., and Menon, D. (2017). Macrophage immunometabolism: where are we (going)? Trends Immunol. 38, 395-406. https://doi.org/10.1016/j.it.2017.03.001
  22. Wang, H. and Ma, S. (2008). The cytokine storm and factors determining the sequence and severity of organ dysfunction in multiple organ dysfunction syndrome. Am. J. Emerg. Med. 26, 711-715. https://doi.org/10.1016/j.ajem.2007.10.031
  23. Woolum, J.A., Abner, E.L., Kelly, A., Thompson Bastin, M.L., Morris, P.E., and Flannery, A.H. (2018). Effect of thiamine administration on lactate clearance and mortality in patients with septic shock. Crit. Care Med. 46, 1747-1752. https://doi.org/10.1097/CCM.0000000000003311
  24. Yadav, U.C., Kalariya, N.M., Srivastava, S.K., and Ramana, K.V. (2010). Protective role of benfotiamine, a fat-soluble vitamin B1 analogue, in lipopolysaccharide-induced cytotoxic signals in murine macrophages. Free Radic. Biol. Med. 48, 1423-1434. https://doi.org/10.1016/j.freeradbiomed.2010.02.031

Cited by

  1. Lipid Droplets, the Central Hub Integrating Cell Metabolism and the Immune System vol.12, 2020, https://doi.org/10.3389/fphys.2021.746749