Skip to main content
Log in

Effects of DHA on Hippocampal Autophagy and Lysosome Function After Traumatic Brain Injury

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Traumatic brain injury (TBI) triggers endoplasmic reticulum (ER) stress and impairs autophagic clearance of damaged organelles and toxic macromolecules. In this study, we investigated the effects of the post-TBI administration of docosahexaenoic acid (DHA) on improving hippocampal autophagy flux and cognitive functions of rats. TBI was induced by cortical contusion injury in Sprague–Dawley rats, which received DHA (16 mg/kg in DMSO, intraperitoneal administration) or vehicle DMSO (1 ml/kg) with an initial dose within 15 min after the injury, followed by a daily dose for 3 or 7 days. First, RT-qPCR reveals that TBI induced a significant elevation in expression of autophagy-related genes in the hippocampus, including SQSTM1/p62 (sequestosome 1), lysosomal-associated membrane proteins 1 and 2 (Lamp1 and Lamp2), and cathepsin D (Ctsd). Upregulation of the corresponding autophagy-related proteins was detected by immunoblotting and immunostaining. In contrast, the DHA-treated rats did not exhibit the TBI-induced autophagy biogenesis and showed restored CTSD protein expression and activity. T2-weighted images and diffusion tensor imaging (DTI) of ex vivo brains showed that DHA reduced both gray matter and white matter damages in cortical and hippocampal tissues. DHA-treated animals performed better than the vehicle control group on the Morris water maze test. Taken together, these findings suggest that TBI triggers sustained stimulation of autophagy biogenesis, autophagy flux, and lysosomal functions in the hippocampus. Swift post-injury DHA administration restores hippocampal lysosomal biogenesis and function, demonstrating its therapeutic potential.

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.

Institutional subscriptions

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

Similar content being viewed by others

References

  1. Begum G, Yan HQ, Li L, Singh A, Dixon CE, Sun D (2014) Docosahexaenoic acid reduces ER stress and abnormal protein accumulation and improves neuronal function following traumatic brain injury. J Neurosci 34:3743–3755

    Article  CAS  PubMed  Google Scholar 

  2. Park Y, Liu C, Luo T, Dietrich WD, Bramlett H, Hu B (2015) Chaperone-mediated autophagy after traumatic brain injury. J Neurotrauma 32:1449–1457

    Article  PubMed  PubMed Central  Google Scholar 

  3. Yao X, Liu J, Lee E, Ling G, McCabe J (2006) Cullin 5 gene expression in the rat cerebral cortex and hippocampus following traumatic brain injury (TBI). Neurosci Lett 409:65–69

    Article  CAS  PubMed  Google Scholar 

  4. Lipinski MM, Wu J, Faden AI, Sarkar C (2015) Function and mechanisms of autophagy in brain and spinal cord trauma. Antioxid Redox Signal 23:565–577

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Liu CL, Chen S, Dietrich D, Hu BR (2008) Changes in autophagy after traumatic brain injury. J Cerb Blood Flow Metab 28:674–683

    Article  CAS  Google Scholar 

  6. Sarkar C, Zhao Z, Aungst S, Sabirzhanov B, Faden AI, Lipinski MM (2014) Impaired autophagy flux is associated with neuronal cell death after traumatic brain injury. Autophagy 10:2208–2222

    Article  CAS  PubMed  Google Scholar 

  7. Mayurasakorn K, Williams JJ, Ten VS, Deckelbaum RJ (2011) Docosahexaenoic acid: brain accretion and roles in neuroprotection after brain hypoxia and ischemia. Curr Opin Clin Nutr Metab Care 14:158–167

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Belayev L, Khoutorova L, Atkins KD, Bazan NG (2009) Robust docosahexaenoic acid-mediated neuroprotection in a rat model of transient focal cerebral ischemia. Stroke 40:3121–3126

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ariza M, Serra-Grabulosa JM, Junque C, Ramirez B, Mataro M, Poca A, Bargallo N, Sahuquillo J (2006) Hippocampal head atrophy after traumatic brain injury. Neuropsychologia 44:1956–1961

    Article  PubMed  Google Scholar 

  10. Dixon CE, Clifton GL, Lighthall JW, Yaghmai AA, Hayes RL (1991) A controlled cortical impact model of traumatic brain injury in the rat. J Neurosci Methods 39:253–262

    Article  CAS  PubMed  Google Scholar 

  11. Dixon CE, Kraus MF, Kline AE, Ma X, Yan HQ, Griffith RG et al (1999) Amantadine improves water maze performance without affecting motor behavior following traumatic brain injury in rats. Restor Neurol Neurosci 14:285–294

    CAS  PubMed  Google Scholar 

  12. Tanida I, Ueno T, Kominami E (2004) LC3 conjugation system in mammalian autophagy. Int J Biochem Cell Biol 36:2503–2518

    Article  CAS  PubMed  Google Scholar 

  13. Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Øvervatn A, Bjørkøy G et al (2007) p62SQSTM1 binds directly to Atg8LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282:24131–24145

    Article  CAS  PubMed  Google Scholar 

  14. Sardiello M, Palmieri M, di Ronza A, Medina DL, Valenza M, Gennarino VA, Di Malta C, Donaudy F et al (2009) A gene network regulating lysosomal biogenesis and function. Science 325(5939):473–477

    CAS  PubMed  Google Scholar 

  15. Peña KA, Kiselyov K (2015) Transition metals activate TFEB in overexpressing cells. Biochem J 470(1):65–76

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zhang X, Cheng X, Yu L, Yang J, Calvo R, Patnaik S, Hu X, Gao Q et al (2016) MCOLN1 is a ROS sensor in lysosomes that regulates autophagy. Nat Commun 7:12109

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gieselmann V, Pohlmann R, Hasilik A, Von Figura K (1983) Biosynthesis and transport of cathepsin D in cultured human fibroblasts. J Cell Biol 97:1–5

    Article  CAS  PubMed  Google Scholar 

  18. Rudenko G, Bonten E, d’Azzo A, Hol WG (1995) Three-dimensional structure of the human ‘protective protein’: structure of the precursor form suggests a complex activation mechanism. Structure 3(11):1249–1259

    Article  CAS  PubMed  Google Scholar 

  19. Rosenfeld MG, Kreibich G, Popov D, Kato K, Sabatini DD (1982) Biosynthesis of lysosomal hydrolases: their synthesis in bound polysomes and the role of co- and post-translational processing in determining their subcellular distribution. J Cell Biol 93(1):135–143

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Colletti GA, Miedel MT, Quinn J, Andharia N, Weisz OA, Kiselyov K (2012) Loss of lysosomal ion channel transient receptor potential channel mucolipin-1 (TRPML1) leads to cathepsin B-dependent apoptosis. J Biol Chem 287(11):8082–8091

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Nakanishi H, Zhang J, Koike M, Nishioku T, Okamoto Y, Kominami E, von Figura K, Peters C et al (2001) Involvement of nitric oxide released from microglia-macrophages in pathological changes of cathepsin D-deficient mice. J Neurosci 21:7526–7533

    CAS  PubMed  Google Scholar 

  22. Carlsson SR, Roth J, Piller F, Fukuda M (1988) Isolation and characterization of human lysosomal membrane glycoproteins, h-lamp- 1 and h-lamp-2. J Biol Chem 263:18911–18919

    CAS  PubMed  Google Scholar 

  23. Zhao J et al (2015) Docosahexaenoic acid attenuated experimental chronic colitis in interleukin 10-deficient mice by enhancing autophagy through inhibition of the mTOR pathway. JPEN J Parenter Enteral Nutr 20:1–6

    CAS  Google Scholar 

  24. Pettersen K, Monsen VT, Hakvåg Pettersen CH et al (2016) DHA-induced stress response in human colon cancer cells—focus on oxidative stress and autophagy. Free Radic Biol Med 90:158–172

    Article  CAS  PubMed  Google Scholar 

  25. Johansson I, Monsen VT, Pettersen K et al (2015) The marine n-3 PUFA DHA evokes cytoprotection against oxidative stress and protein misfolding by inducing autophagy and NFE2L2 in human retinal pigment epithelial cells. Autophagy 11:1636–1651

    Article  PubMed  PubMed Central  Google Scholar 

  26. Harvey LD, Yin Y, Attarwala IY et al (2015) Administration of DHA reduces endoplasmic reticulum stress-associated inflammation and alters microglial or macrophage activation in traumatic brain injury. ASN Neuro 7:1–15

    Article  Google Scholar 

  27. Calandria JM, Asatryan A, Balaszczuk V et al (2015) NPD1-mediated stereoselective regulation of BIRC3 expression through cREL is decisive for neural cell survival. Cell Death Diff 22:1363–1377

    Article  CAS  Google Scholar 

  28. Ogata M, Hino S, Saito A et al (2006) Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol 26:9220–9231

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Saftig P, Klumperman J (2009) Lysosome biogenesis and lysosomal membrane proteins: trafficking meets function. Nat Rev Mol Cell Biol 10:623–635

    Article  CAS  PubMed  Google Scholar 

  30. Schober ME, Requena DF, Abdullah OM, Casper TC, Beachy J, Malleske D, Pauly JR (2016) Dietary docosahexaenoic acid improves cognitive function, tissue sparing, and magnetic resonance imaging indices of edema and white matter injury in the immature rat after traumatic brain injury. J Neurotraum 33:390–402

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported in part by the National Institutes of Health [R01NS089051] (DS) and the US Department of Veterans Affairs VA [RR&D#B6761R] (CED). Y.Y. was supported by the Chinese Dalian Municipal Bureau Study Abroad Research Award.

Author information

Authors and Affiliations

Authors

Contributions

Q.H.Y. and C.E.D. conducted CCI. Y.Y. and I.Y.A. performed immunostaining and quantified immunofluorescent images. Y.Y. performed CTSD activity assays. E.L. conducted immunoblotting assays. L.A.A. and K.K. performed and quantified RT-PCR assays. T.K.H. and L.M.F. conducted MRI experiment. Y.Y. and D.S. designed the experiments. Y.Y., G.S., and D.S. completed the manuscript writing.

Corresponding author

Correspondence to Dandan Sun.

Ethics declarations

All studies were in compliance with the guidelines outlined in the Guide for the Care and Use of Laboratory Animals from the US Department of Health and Human Services and were approved by the University of Pittsburgh Medical Center Institutional Animal Care and Use Committee.

Conflict of Interest

The authors declare that they have no conflicts of interest.

Electronic Supplementary Material

Supplemental Figure 1.

No changes of CTSD protein expression in CA2 or CA3 regions after TBI. Representative confocal immnunofluorescence images show no changes in CTSD protein expression in CA2 or CA3 regions in the TBI + Vehicle and TBI + DHA rats at 3 days post-TBI. (GIF 302 kb)

High resolution image (TIFF 59225 kb)

Supplemental Figure 2.

No differences of LAMP1 and CTSD protein expression in naive and SHAM hippocampus. Representative confocal immnunofluorescence images show changes in LAMP1 (a) or CTSD (b) protein expression in CA1 regions in the naïve, SHAM, or TBI + Vehicle rats at 3 days post-TBI. Representative Western blots of LAMP1 (c) expression in the CL and IL hippocampi of naïve, Sham, or TBI + Vehicle-treated rats at 3 days after TBI. The same blot was probed with the antibody against β-actin as a loading control. (GIF 385 kb)

High resolution image (TIFF 66624 kb)

Supplemental Figure 3.

Changes of hippocampal protein ubiquitination and microtubule-associated light chain 3 protein after TBI. (a) Representative Western blots of LC3 expression in the CL and IL hippocampi of Sham, TBI + Vehicle, and TBI + DHA-treated rats at 3 days after TBI. The same blot was probed with the antibody against β-actin as a loading control. Summary data expressed as the band intensity ratio between LC3I and β-actin. The values are the mean ± SE (n = 6). * p < 0.05. No significant changes in LC3 were observed in the TBI+ Vehicle and TBI+DHA rats. (b) Representative Western blots of protein ubiquitination in the CL and IL hippocampi of Sham, TBI + Vehicle, and TBI + DHA-treated rats at 3 days TBI. The intensity of proteins in a range of molecular weights from 250 kDa to 50 kDa was calculated. The same blot was probed with the antibody against β-actin as a loading control. Summary data were normalized with β-actin. The values are the mean ± SE (n = 8). **p < 0.01 vs Sham, ***p < 0.001 vs Sham. No significant changes in protein ubiquitination were observed in the TBI+ Vehicle and TBI+DHA rats. (GIF 51 kb)

High resolution image (TIFF 37535 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yin, Y., Li, E., Sun, G. et al. Effects of DHA on Hippocampal Autophagy and Lysosome Function After Traumatic Brain Injury. Mol Neurobiol 55, 2454–2470 (2018). https://doi.org/10.1007/s12035-017-0504-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-017-0504-8

Keywords

Navigation