Skip to content
2000
Volume 22, Issue 1
  • ISSN: 1567-2026
  • E-ISSN: 1875-5739

Abstract

Background

Sepsis-associated encephalopathy (SAE) is the most severe complication of sepsis. Ubiquitin-specific protease 8 (USP8) could improve cognitive and motor disorders in SAE.

Objective

This study explored the mechanism of USP8 in SAE mice to provide new therapeutic targets for SAE.

Methods

C57BL/6 mice were selected to establish SAE models by caecal ligation and puncture (CLP) and injected with lentivirus overexpressing USP8 one week before SAE modeling. Mouse weight changes were monitored, cognitive and learning abilities were tested by the Morris water maze test, behaviors were evaluated by open-field tests, and pathological changes in brain tissue were analyzed by H&E staining. Levels of USP8, TNF-α, IL-1β, IL-6, and IL-10, and SOD, GSH-Px activities, and MDA levels were detected by Western blot, ELISA, and kits. Co-immunoprecipitation assay verified the interaction between USP8 and SIRT1 and SIRT1 ubiquitination level.

Results

In CLP mice, the body weight, cognitive function, and learning ability were decreased, along with motor disorder, abnormal morphological structure of neurons, and obvious inflammatory infiltration. USP8 protein in brain tissue was decreased, the levels of TNF-α, IL-1β, and IL-6 were increased, IL-10 was decreased, SOD and GSH-Px activities were decreased, and MDA level was increased. USP8 treatment improved cognitive dysfunction and inhibited inflammation and oxidative stress in CLP mice. USP8 promoted SIRT1 expression by direct deubiquitination. SIRT1 knockdown partially reversed the regulation of USP8 on SAE mice.

Conclusion

USP8 can directly deubiquitinate SIRT1 and inhibit inflammatory reactions and oxidative stress, thus improving cognitive dysfunction in SAE mice.

Loading

Article metrics loading...

/content/journals/cnr/10.2174/1567202620666230712112521
2023-10-06
2025-09-09
Loading full text...

Full text loading...

References

  1. RelloJ. Valenzuela-SánchezF. Ruiz-RodriguezM. MoyanoS. Sepsis: A review of advances in management.Adv. Ther.201734112393241110.1007/s12325‑017‑0622‑8 29022217
    [Google Scholar]
  2. RocheteauP. ChatreL. BriandD. Sepsis induces long-term metabolic and mitochondrial muscle stem cell dysfunction amenable by mesenchymal stem cell therapy.Nat. Commun.2015611014510.1038/ncomms10145 26666572
    [Google Scholar]
  3. IwashynaT.J. CookeC.R. WunschH. KahnJ.M. Population burden of long-term survivorship after severe sepsis in older Americans.J. Am. Geriatr. Soc.20126061070107710.1111/j.1532‑5415.2012.03989.x 2264254
    [Google Scholar]
  4. GaieskiD.F. EdwardsJ.M. KallanM.J. CarrB.G. Benchmarking the incidence and mortality of severe sepsis in the United States.Crit. Care Med.20134151167117410.1097/CCM.0b013e31827c09f8 2344298
    [Google Scholar]
  5. TangY.F. DuanY.J. GeR.D. Environmental enrichment protects against cognition deficits caused by sepsis-associated encephalopathy.J. Integr. Neurosci.2022221510.31083/j.jin2201005 36722228
    [Google Scholar]
  6. GoftonT.E. YoungG.B. Sepsis-associated encephalopathy.Nat. Rev. Neurol.201281055756610.1038/nrneurol.2012.183 22986430
    [Google Scholar]
  7. HemingN. MazeraudA. VerdonkF. BozzaF.A. ChrétienF. SharsharT. Neuroanatomy of sepsis-associated encephalopathy.Crit. Care20172116510.1186/s13054‑017‑1643‑z 28320461
    [Google Scholar]
  8. ChungH.Y. WickelJ. BrunkhorstF.M. GeisC. Sepsis-associated encephalopathy: From delirium to dementia?J. Clin. Med.20209370310.3390/jcm9030703 32150970
    [Google Scholar]
  9. HuangM. CaiS. SuJ. The pathogenesis of sepsis and potential therapeutic targets.Int. J. Mol. Sci.20192021537610.3390/ijms20215376 31671729
    [Google Scholar]
  10. GallasteguiN. GrollM. The 26S proteasome: Assembly and function of a destructive machine.Trends Biochem. Sci.2010351163464210.1016/j.tibs.2010.05.005 20541423
    [Google Scholar]
  11. BiW. LanX. ZhangJ. USP8 ameliorates cognitive and motor impairments via microglial inhibition in a mouse model of sepsis-associated encephalopathy.Brain Res.20191719404810.1016/j.brainres.2019.05.009 31075263
    [Google Scholar]
  12. ZhaoL. AnR. YangY. Melatonin alleviates brain injury in mice subjected to cecal ligation and puncture via attenuating inflammation, apoptosis, and oxidative stress: The role of SIRT1 signaling.J. Pineal Res.201559223023910.1111/jpi.12254 26094939
    [Google Scholar]
  13. TianY. WangL. FanX. ZhangH. DongZ. TaoT. β-patchoulene alleviates cognitive dysfunction in a mouse model of sepsis associated encephalopathy by inhibition of microglia activation through Sirt1/Nrf2 signaling pathway.PLoS One2023181e027996410.1371/journal.pone.0279964 36608000
    [Google Scholar]
  14. ZhuY. WangK. MaZ. SIRT1 activation by butein attenuates sepsis-induced brain injury in mice subjected to cecal ligation and puncture via alleviating inflammatory and oxidative stress.Toxicol. Appl. Pharmacol.2019363344610.1016/j.taap.2018.10.013 30336174
    [Google Scholar]
  15. HaleyT.J. McCORMICK WG. Pharmacological effects produced by intracerebral injection of drugs in the conscious mouse.Br. J. Pharmacol. Chemother.1957121121510.1111/j.1476‑5381.1957.tb01354.x 13413144
    [Google Scholar]
  16. ChenH. PengY. WangL. WangX. Sevoflurane attenuates cognitive dysfunction and NLRP3-dependent caspase-1/11-GSDMD pathway-mediated pyroptosis in the hippocampus via upregulation of SIRT1 in a sepsis model.Arch. Physiol. Biochem.202212851413142010.1080/13813455.2020.1773860 32538180
    [Google Scholar]
  17. TianM. QingzhenL. ZhiyangY. Attractylone attenuates sepsis‐associated encephalopathy and cognitive dysfunction by inhibiting microglial activation and neuroinflammation.J. Cell. Biochem.201912057101710810.1002/jcb.27983 30672013
    [Google Scholar]
  18. PanC. SiY. MengQ. Suppression of the RAC1/MLK3/p38 signaling pathway by β-elemene alleviates sepsis-associated encephalopathy in mice.Front. Neurosci.20191335810.3389/fnins.2019.00358 31068775
    [Google Scholar]
  19. ShenY. ZhangY. DuJ. CXCR5 down-regulation alleviates cognitive dysfunction in a mouse model of sepsis-associated encephalopathy: Potential role of microglial autophagy and the p38MAPK/NF-κB/STAT3 signaling pathway.J. Neuroinflammation202118124610.1186/s12974‑021‑02300‑1 34711216
    [Google Scholar]
  20. CaiM. DuB. SiY. Knockdown of VDAC1 alleviates the cognitive dysfunction secondary to sepsis-associated encephalopathy.Am. J. Transl. Res.20211377538755534377234
    [Google Scholar]
  21. ChenQ. YuW. ShiJ. Insulin alleviates the inflammatory response and oxidative stress injury in cerebral tissues in septic rats.J. Inflamm.20141111810.1186/1476‑9255‑11‑18 25093012
    [Google Scholar]
  22. BonfanteS. JoaquimL. FiletiM.E. Stanniocalcin 1 inhibits the inflammatory response in microglia and protects against sepsis-associated encephalopathy.Neurotox. Res.202139211913210.1007/s12640‑020‑00293‑y 33025358
    [Google Scholar]
  23. TauberS.C. DjukicM. GossnerJ. EiffertH. BrückW. NauR. Sepsis-associated encephalopathy and septic encephalitis: An update.Expert Rev. Anti Infect. Ther.202119221523110.1080/14787210.2020.1812384 32808580
    [Google Scholar]
  24. Matutino SantosP. Pereira CamposG. NascimentoC. Endo-lysosomal and autophagy pathway and ubiquitin-proteasome system in mood disorders: A review article.Neuropsychiatr. Dis. Treat.20231913315110.2147/NDT.S376380 36684613
    [Google Scholar]
  25. ZhaoJ. BiW. ZhangJ. USP8 protects against lipopolysaccharide-induced cognitive and motor deficits by modulating microglia phenotypes through TLR4/MyD88/NF-κB signaling pathway in mice.Brain Behav. Immun.20208858259610.1016/j.bbi.2020.04.052 32335193
    [Google Scholar]
  26. GrangerJ.I. RattiP.L. DattaS.C. RaymondR.M. OppM.R. Sepsis-induced morbidity in mice: Effects on body temperature, body weight, cage activity, social behavior and cytokines in brain.Psychoneuroendocrinology20133871047105710.1016/j.psyneuen.2012.10.010 23146654
    [Google Scholar]
  27. CatarinaA.V. BranchiniG. BettoniL. De OliveiraJ.R. NunesF.B. Sepsis-associated encephalopathy: From pathophysiology to progress in experimental studies.Mol. Neurobiol.20215862770277910.1007/s12035‑021‑02303‑2 33495934
    [Google Scholar]
  28. YinH. HuM. LiangX. Deletion of SIRT1 from hepatocytes in mice disrupts lipin-1 signaling and aggravates alcoholic fatty liver.Gastroenterology2014146380181110.1053/j.gastro.2013.11.008 24262277
    [Google Scholar]
  29. WangR.H. LiC. DengC.X. Liver steatosis and increased ChREBP expression in mice carrying a liver specific SIRT1 null mutation under a normal feeding condition.Int. J. Biol. Sci.20106768269010.7150/ijbs.6.682 21103071
    [Google Scholar]
  30. ZhaoY. ZhangJ. ZhengY. NAD+ improves cognitive function and reduces neuroinflammation by ameliorating mitochondrial damage and decreasing ROS production in chronic cerebral hypoperfusion models through Sirt1/PGC-1α pathway.J. Neuroinflammation202118120710.1186/s12974‑021‑02250‑8 3453086
    [Google Scholar]
/content/journals/cnr/10.2174/1567202620666230712112521
Loading
/content/journals/cnr/10.2174/1567202620666230712112521
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error
Please enter a valid_number test