Skip to main content

Advertisement

Log in

Plasmalogens Inhibit Endocytosis of Toll-like Receptor 4 to Attenuate the Inflammatory Signal in Microglial Cells

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Microglial activation is a pathological feature of many neurodegenerative diseases and the role of cellular lipids in these diseases is mostly unknown. It was known that the special ether lipid plasmalogens (Pls) were reduced in the brain and blood samples of Alzheimer’s disease (AD) patients. It has recently been reported that the oral ingestion of scallop-derived Pls (sPls) improved cognition among mild AD patients, which led us to investigate the role of sPls in the microglial activation. We used the lipopolysaccharides (LPS)-induced microglial activation model and found that sPls inhibit the LPS-mediated TLR4 endocytosis and the downstream caspases activation. By using the specific inhibitors, we also confirmed that the TLR4 endocytosis and the caspases activation strictly controlled the pro-inflammatory cytokine expression. In addition, the reduction of cellular Pls by sh-RNA-mediated knockdown of GNPAT (glyceronephosphate O-acyltransferase), a Pls synthesizing enzyme, enhanced the endocytosis of TLR4 and activation of caspase-3 which resulted in the enhanced pro-inflammatory cytokine expression. We also report for the first time that the TLR4 endocytosis was significantly higher in the cortex of aged mice and AD model mice brains, proposing a significant link between the age-related reduction of Pls and microglial activation. Interestingly, the sPls drinking in AD model mice significantly reduced the TLR4 endocytosis. Our cumulative data indicates that the cellular Pls attenuate the microglial activation by maintaining the endocytosis of TLR4, suggesting a possible mechanism of the cognition improvement effect of sPls among mild AD patients.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11

Similar content being viewed by others

Abbreviations

AD:

Alzheimer’s disease

ANOVA:

one-way analysis of variance

BCA:

bicinchoninic acid

BSA:

bovine serum albumin

CDE:

clathrin-dependent endocytic

CHO:

Chinese hamster ovary

CIE:

clathrin-independent endocytic

C/LR:

caveolin/lipid raft-mediated endocytic

DAPI:

4′,6-diamidino-2-phenylindole

DEVD:

Z-DEVD-fmk

DMEM:

Dulbecco’s modified Eagle medium

FBS:

fetal bovine serum

GAPDH:

glyceraldehyde-3-phosphae dehydrogenase

GNPAT:

glycerone phosphate O-acyltransferase

HEK:

human embryonic kidney

IL-1β:

interleukin-1β

IL-1R:

interleukin-I receptor

IRF:

interferon regulatory factor-3

LPS:

lipopolysaccharide

MβCD:

methyl-β-cyclodextrin

NF-κB:

nuclear factor-κB

PARP1:

polyadenosine diphosphate ribosepolymerase1

PBS:

phosphate-buffered saline

PCR:

polymerase chain reaction

Pls:

plasmalogens

Pls-Etn:

ethanolamine plasmalogens

RCDP:

rhizomelic chondrodysplasia punctate

RIPA:

radio-immunoprecipitation assay

SDS-PAGE:

sodium dodecyl sulfate–polyacrylamide gel electrophoresis

sh-RNA:

short hairpin-RNA

TBS:

Tris-buffered saline

TDU:

transduction unites

TIRAP:

Toll/IL-1R domain-containing adaptor protein

TLR4:

toll-like receptor 4

TNF-α:

tumor necrosis factor-α

TRAM:

Toll/interleukin-1 receptor domain-containing adaptor inducing type I interferons-related adaptor molecule

TRIF:

Toll/interleukin-1 receptor domain-containing adaptor inducing type I interferons

VAD:

Z-VAD-fmk

References

  1. Tanzi RE, Bertram L (2005) Twenty years of the Alzheimer’s disease amyloid hypothesis: a genetic perspective. Cell 120(4):545–555. https://doi.org/10.1016/j.cell.2005.02.008

    Article  CAS  PubMed  Google Scholar 

  2. Haass C, Selkoe DJ (2007) Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat Rev Mol Cell Biol 8(2):101–112. https://doi.org/10.1038/nrm2101

    Article  CAS  PubMed  Google Scholar 

  3. Tahara K, Kim HD, Jin JJ, Maxwell JA, Li L, Fukuchi K (2006) Role of toll-like receptor signalling in Abeta uptake and clearance. Brain 129(Pt 11):3006–3019. https://doi.org/10.1093/brain/awl249

    Article  PubMed  PubMed Central  Google Scholar 

  4. Apelt J, Schliebs R (2001) Beta-amyloid-induced glial expression of both pro- and anti-inflammatory cytokines in cerebral cortex of aged transgenic Tg2576 mice with Alzheimer plaque pathology. Brain Res 894(1):21–30

    Article  CAS  Google Scholar 

  5. Salminen A, Ojala J, Suuronen T, Kaarniranta K, Kauppinen A (2008) Amyloid-beta oligomers set fire to inflammasomes and induce Alzheimer’s pathology. J Cell Mol Med 12(6A):2255–2262. https://doi.org/10.1111/j.1582-4934.2008.00496.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Fahy E, Subramaniam S, Brown HA, Glass CK, Merrill AH Jr, Murphy RC, Raetz CR, Russell DW et al (2005) A comprehensive classification system for lipids. J Lipid Res 46(5):839–861. https://doi.org/10.1194/jlr.E400004-JLR200

    Article  CAS  PubMed  Google Scholar 

  7. Maeba R, Ueta N (2003) Ethanolamine plasmalogens prevent the oxidation of cholesterol by reducing the oxidizability of cholesterol in phospholipid bilayers. J Lipid Res 44(1):164–171

    Article  CAS  Google Scholar 

  8. Guan Z, Wang Y, Cairns NJ, Lantos PL, Dallner G, Sindelar PJ (1999) Decrease and structural modifications of phosphatidylethanolamine plasmalogen in the brain with Alzheimer disease. J Neuropathol Exp Neurol 58(7):740–747

    Article  CAS  Google Scholar 

  9. Han X, Holtzman DM, McKeel DW Jr (2001) Plasmalogen deficiency in early Alzheimer’s disease subjects and in animal models: molecular characterization using electrospray ionization mass spectrometry. J Neurochem 77(4):1168–1180

    Article  CAS  Google Scholar 

  10. Hossain MS, Abe Y, Ali F, Youssef M, Honsho M, Fujiki Y, Katafuchi T (2017) Reduction of ether-type glycerophospholipids, plasmalogens, by NF-κB signal leading to microglial activation. J Neurosci 37(15):4074–4092. https://doi.org/10.1523/JNEUROSCI.3941-15.2017

    Article  CAS  PubMed  Google Scholar 

  11. Wood PL, Mankidy R, Ritchie S, Heath D, Wood JA, Flax J, Goodenowe DB (2010) Circulating plasmalogen levels and Alzheimer disease assessment scale-cognitive scores in Alzheimer patients. J Psychiatry Neurosci 35(1):59–62

    Article  Google Scholar 

  12. Hossain MS, Ifuku M, Take S, Kawamura J, Miake K, Katafuchi T (2013) Plasmalogens rescue neuronal cell death through an activation of AKT and ERK survival signaling. PLoS One 8(12):e83508. https://doi.org/10.1371/journal.pone.0083508

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ifuku M, Katafuchi T, Mawatari S, Noda M, Miake K, Sugiyama M, Fujino T (2012) Anti-inflammatory/anti-amyloidogenic effects of plasmalogens in lipopolysaccharide-induced neuroinflammation in adult mice. J Neuroinflammation 9:197. https://doi.org/10.1186/1742-2094-9-197

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Akira S, Takeda K (2004) Functions of toll-like receptors: lessons from KO mice. C R Biol 327(6):581–589

    Article  CAS  Google Scholar 

  15. Kagan JC, Su T, Horng T, Chow A, Akira S, Medzhitov R (2008) TRAM couples endocytosis of Toll-like receptor 4 to the induction of interferon-β. Nat Immunol 9(4):361–368. https://doi.org/10.1038/ni1569

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wong SW, Kwon MJ, Choi AM, Kim HP, Nakahira K, Hwang DH (2009) Fatty acids modulate toll-like receptor 4 activation through regulation of receptor dimerization and recruitment into lipid rafts in a reactive oxygen species-dependent manner. J Biol Chem 284(40):27384–27392. https://doi.org/10.1074/jbc.M109.044065

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Husebye H, Halaas O, Stenmark H, Tunheim G, Sandanger O, Bogen B, Brech A, Latz E et al (2006) Endocytic pathways regulate Toll-like receptor 4 signaling and link innate and adaptive immunity. EMBO J 25(4):683–692. https://doi.org/10.1038/sj.emboj.7600991

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Shuto T, Kato K, Mori Y, Viriyakosol S, Oba M, Furuta T, Okiyoneda T, Arima H et al (2005) Membrane-anchored CD14 is required for LPS-induced TLR4 endocytosis in TLR4/MD-2/CD14 overexpressing CHO cells. Biochem Biophys Res Commun 338(3):1402–1409. https://doi.org/10.1016/j.bbrc.2005.10.102

    Article  CAS  PubMed  Google Scholar 

  19. Pascual-Lucas M, Fernandez-Lizarbe S, Montesinos J, Guerri C (2014) LPS or ethanol triggers clathrin- and rafts/caveolae-dependent endocytosis of TLR4 in cortical astrocytes. J Neurochem 129(3):448–462. https://doi.org/10.1111/jnc.12639

    Article  CAS  PubMed  Google Scholar 

  20. Cai W, Du A, Feng K, Zhao X, Qian L, Ostrom RS, Xu C (2013) Adenylyl cyclase 6 activation negatively regulates TLR4 signaling through lipid raft-mediated endocytosis. J Immunol 191 (12):6093-6100. doi:https://doi.org/10.4049/jimmunol.1301912

    Article  CAS  Google Scholar 

  21. Budihardjo I, Oliver H, Lutter M, Luo X, Wang X (1999) Biochemical pathways of caspase activation during apoptosis. Annu Rev Cell Dev Biol 15:269–290. https://doi.org/10.1146/annurev.cellbio.15.1.269

    Article  CAS  PubMed  Google Scholar 

  22. Wu X, Guo R, Chen P, Wang Q, Cunningham PN (2009) TNF induces caspase-dependent inflammation in renal endothelial cells through a Rho- and myosin light chain kinase-dependent mechanism. Am J Physiol Renal Physiol 297(2):F316–F326. https://doi.org/10.1152/ajprenal.00089.2009

    Article  CAS  PubMed  Google Scholar 

  23. Su JH, Zhao M, Anderson AJ, Srinivasan A, Cotman CW (2001) Activated caspase-3 expression in Alzheimer’s and aged control brain: correlation with Alzheimer pathology. Brain Res 898(2):350–357

    Article  CAS  Google Scholar 

  24. D'Amelio M, Cavallucci V, Middei S, Marchetti C, Pacioni S, Ferri A, Diamantini A, De Zio D et al (2011) Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer’s disease. Nat Neurosci 14(1):69–76. https://doi.org/10.1038/nn.2709

    Article  CAS  PubMed  Google Scholar 

  25. Stone JR, Okonkwo DO, Singleton RH, Mutlu LK, Helm GA, Povlishock JT (2002) Caspase-3-mediated cleavage of amyloid precursor protein and formation of amyloid β peptide in traumatic axonal injury. J Neurotrauma 19(5):601–614. https://doi.org/10.1089/089771502753754073

    Article  PubMed  Google Scholar 

  26. Burguillos MA, Deierborg T, Kavanagh E, Persson A, Hajji N, Garcia-Quintanilla A, Cano J, Brundin P et al (2011) Caspase signalling controls microglia activation and neurotoxicity. Nature 472(7343):319–324. https://doi.org/10.1038/nature09788

    Article  CAS  PubMed  Google Scholar 

  27. Hossain MS, Abe Y, Ali F, Youssef M, Honsho M, Fujiki Y, Katafuchi T (2017) Reduction of ether-type glycerophospholipids, plasmalogens, by NF-kappaB signal leading to microglial activation. J Neurosci 37(15):4074–4092. https://doi.org/10.1523/JNEUROSCI.3941-15.2017

    Article  CAS  PubMed  Google Scholar 

  28. Fujino T, Yamada T, Asada T, Tsuboi Y, Wakana C, Mawatari S, Kono S (2017) Efficacy and blood plasmalogen changes by oral administration of plasmalogen in patients with mild Alzheimer’s disease and mild cognitive impairment: a multicenter, randomized, double-blind, placebo-controlled trial. EBioMedicine 17:199–205. https://doi.org/10.1016/j.ebiom.2017.02.012

    Article  PubMed  PubMed Central  Google Scholar 

  29. Mawatari S, Yunoki K, Sugiyama M, Fujino T (2009) Simultaneous preparation of purified plasmalogens and sphingomyelin in human erythrocytes with phospholipase A1 from Aspergillus orizae. Biosci Biotechnol Biochem 73(12):2621–2625. https://doi.org/10.1271/bbb.90455

    Article  CAS  PubMed  Google Scholar 

  30. Braverman NE, Moser AB (2012) Functions of plasmalogen lipids in health and disease. Biochim Biophys Acta 1822(9):1442–1452. https://doi.org/10.1016/j.bbadis.2012.05.008

    Article  CAS  PubMed  Google Scholar 

  31. Oettinghaus B, Schulz JM, Restelli LM, Licci M, Savoia C, Schmidt A, Schmitt K, Grimm A et al (2016) Synaptic dysfunction, memory deficits and hippocampal atrophy due to ablation of mitochondrial fission in adult forebrain neurons. Cell Death Differ 23(1):18–28. https://doi.org/10.1038/cdd.2015.39

    Article  CAS  PubMed  Google Scholar 

  32. Ifuku M, Hossain SM, Noda M, Katafuchi T (2014) Induction of interleukin-1β by activated microglia is a prerequisite for immunologically induced fatigue. Eur J Neurosci 40(8):3253–3263. https://doi.org/10.1111/ejn.12668

    Article  PubMed  Google Scholar 

  33. Cohen GM (1997) Caspases: the executioners of apoptosis. Biochem J 326(Pt 1):1–16

    Article  CAS  Google Scholar 

  34. Schmitz ML, Baeuerle PA (1991) The p65 subunit is responsible for the strong transcription activating potential of NF-κB. EMBO J 10(12):3805–3817

    Article  CAS  Google Scholar 

  35. Pike LJ, Han X, Chung KN, Gross RW (2002) Lipid rafts are enriched in arachidonic acid and plasmenylethanolamine and their composition is independent of caveolin-1 expression: a quantitative electrospray ionization/mass spectrometric analysis. Biochemistry 41(6):2075–2088

    Article  CAS  Google Scholar 

  36. Rodemer C, Thai TP, Brugger B, Kaercher T, Werner H, Nave KA, Wieland F, Gorgas K et al (2003) Inactivation of ether lipid biosynthesis causes male infertility, defects in eye development and optic nerve hypoplasia in mice. Hum Mol Genet 12(15):1881–1895

    Article  CAS  Google Scholar 

  37. Thai TP, Rodemer C, Jauch A, Hunziker A, Moser A, Gorgas K, Just WW (2001) Impaired membrane traffic in defective ether lipid biosynthesis. Hum Mol Genet 10(2):127–136

    Article  CAS  Google Scholar 

  38. Wanders RJ, Waterham HR (2005) Peroxisomal disorders I: biochemistry and genetics of peroxisome biogenesis disorders. Clin Genet 67(2):107–133. https://doi.org/10.1111/j.1399-0004.2004.00329.x

    Article  CAS  PubMed  Google Scholar 

  39. Kagan JC, Medzhitov R (2006) Phosphoinositide-mediated adaptor recruitment controls Toll-like receptor signaling. Cell 125(5):943–955. https://doi.org/10.1016/j.cell.2006.03.047

    Article  CAS  PubMed  Google Scholar 

  40. Tanimura N, Saitoh S, Matsumoto F, Akashi-Takamura S, Miyake K (2008) Roles for LPS-dependent interaction and relocation of TLR4 and TRAM in TRIF-signaling. Biochem Biophys Res Commun 368(1):94–99. https://doi.org/10.1016/j.bbrc.2008.01.061

    Article  CAS  PubMed  Google Scholar 

  41. Doherty GJ, McMahon HT (2009) Mechanisms of endocytosis. Annu Rev Biochem 78:857–902. https://doi.org/10.1146/annurev.biochem.78.081307.110540

    Article  CAS  PubMed  Google Scholar 

  42. Grant BD, Donaldson JG (2009) Pathways and mechanisms of endocytic recycling. Nat Rev Mol Cell Biol 10(9):597–608. https://doi.org/10.1038/nrm2755

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Nichols BJ (2002) A distinct class of endosome mediates clathrin-independent endocytosis to the Golgi complex. Nat Cell Biol 4(5):374–378. https://doi.org/10.1038/ncb787

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We appreciate the technical assistance of the Research Support Center, Graduate School of Medical Sciences, Kyushu University. We thank Dr. A. Ibrahim for insightful discussion and suggestions to continue this study. We appreciate the technical assistance from Ayako Tajima to perform experiments.

Funding

This work was supported by JSPS KAKENHI grant number 26460320 to Toshihiko Katafuchi, JSPS Wakate B (16K19007) to MSH and Egyptian ministry of higher education scholarship for young scientist fellowship to F. Ali.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Md. Shamim Hossain.

Electronic supplementary material

Figure S1.

Pls pretreatments attenuate the LPS-mediated cleavages of caspase-8 and caspase-3 proteins in microglial cells. Western blotting data showed that LPS (1 μg/ml) treatments for 6 hours induced cleavage of caspase-8 and caspase-3 (as shown in Fig. 1) in BV2 microglial cells, which were attenuated by pretreatments with 5 μg/ml of sPls for 12 hours. Here, we show the representative data of two independent experiments. (PPTX 153 kb)

Figure S2.

MβCD, an inhibitor of lipid raft function, abolishes the attenuating effect of Pls on LPS-induced IL-1β expression. BV2 cells were pretreated with Pls (5 μg/ml) for 12 hours then MβCD (5 μM) was applied 1 hour before LPS (1 μg/ml) treatment. Real time PCR analysis performed 6 hours after LPS treatment demonstrated that the LPS-induced increase in IL-1β mRNA expression was attenuated by the pretreatment with Pls. However, the attenuation was completely blocked by application of MβCD. MβCD treatment itself did not affect the LPS-induced IL-1β expression (**P<0.01 vs. control group, ##P<0.01 vs. LPS group, n=5) (PPTX 57 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ali, F., Hossain, M.S., Sejimo, S. et al. Plasmalogens Inhibit Endocytosis of Toll-like Receptor 4 to Attenuate the Inflammatory Signal in Microglial Cells. Mol Neurobiol 56, 3404–3419 (2019). https://doi.org/10.1007/s12035-018-1307-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-018-1307-2

Keywords

Navigation