Skip to main content

Advertisement

Log in

RETRACTED ARTICLE: Apelin-13 Prevents the Delayed Neuropathy Induced by Tri-ortho-cresyl Phosphate Through Regulation the Autophagy Flux in Hens

  • Original Paper
  • Published:
Neurochemical Research Aims and scope Submit manuscript

This article was retracted on 01 September 2016

Abstract

Organophosphate-induced delayed neuropathy (OPIDN) is pathologically characterized by the swollen axon containing aggregations of microtubules, neurofilaments, smooth endoplasmic reticulum and multivesicular vesicles. At present, the exact mechanism of OPIDN is unclear and the effective therapeutic methods is not available to counter this syndrome. Recent studies had shown that the autophagy was involved in OPIDN. The adipocytokine Apelin is a peptide, Apelin and its receptor are abundantly expressed in the nervous system. Recent researches illuminated that Apelin was neuroprotective factor and Apelin could regulate the autophagy in vivo and vitro model. So we investigated the effect of Apelin-13 on the OPIDN induced by Tri-ortho-cresyl phosphate (TOCP) in hens and explored the role of autophagy in Apelin-13 preventing OPIDN. Adult Roman hens were given a single dose of 750 mg/kg TOCP by gavage for 21 days to induce OPIDN, and neural dysfunction were detected, and the formation of autophagosomes in spinal cord neurons was observed by transmission electron microscopy, and the molecular markers of autophagy microtubule-associated protein light chain-3 (LC3) and the autophagy substrates p62/SQSTM1 were determined by Western blot analysis. The results demonstrated that the obvious neurological dysfunction such as hindlimb paralysis and paralysis of gait was present, the number of autophagosomes in the neurons of spinal cords was significantly increased, the level of LC3-II and p62 expressions and the ratio of LC3-II/LC3-I in spinal cords and sciatic nerve were significantly increased in the OPIDN model group compared with the control group. Compared with the OPIDN model group, the neurological dysfunction of tens was obviously reduced, the clinical signs scores was significantly decreased, the number of autophagosomes in the neurons of hen spinal cords was significantly decreased, the level of LC3-II and p62 expressions and the ratio of LC3-II/LC3-I in spinal cords and sciatic nerve were significantly decreased in Apelin-13 treatment group. Our results suggested that Apelin-13 prevented against the OPIDN induced by TOCP in hens, which the mechanism might be associated with regulation autophagy flux by Apelin-13.

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

Similar content being viewed by others

References

  1. Gao X, Lin H, Ray R, Ray P (2013) Toxicogenomic studies of human neural cells following exposure to organophosphorus chemical warfare nerve agent VX. Neurochem Res 38(5):916–934

    Article  CAS  PubMed  Google Scholar 

  2. Jiang Y, Liu X, Li S, Zhang Y, Piao F, Sun X (2013) Identification of differentially expressed proteins related to organophosphorus-induced delayed neuropathy in the brains of hens. J Appl Toxicol 34(12):1352–1360

    Article  PubMed  Google Scholar 

  3. Song F, Xie K (2012) Calcium-dependent neutral cysteine protease and organophosphate-induced delayed neuropathy. Chem Biol Interact 200(2–3):114–118

    Article  CAS  PubMed  Google Scholar 

  4. Jokanović M, Kosanović M, Brkić D, Vukomanović P (2011) Organophosphate induced delayed polyneuropathy in man: an overview. Clin Neurol Neurosurg 113(1):7–10

    Article  PubMed  Google Scholar 

  5. Lotti M, Moretto A (2005) Organophosphate-induced delayed polyneuropathy. Toxicol Rev 24(1):37–49

    Article  CAS  PubMed  Google Scholar 

  6. Masoud A, Sandhir R (2012) Increased oxidative stress is associated with the development of organophosphate-induced delayed neuropathy. Hum Exp Toxicol 31(12):1214–1227

    Article  CAS  PubMed  Google Scholar 

  7. Masoud A, Kiran R, Sandhir R (2009) Impaired mitochondrial functions in organophosphate induced delayed neuropathy in rats. Cell Mol Neurobiol 29(8):1245–1255

    Article  CAS  PubMed  Google Scholar 

  8. Song F, Yan Y, Zhao X, Dou D, Zhang C, Xie K (2009) Phenylmethylsulfonyl fluoride protects against the degradation of neurofilaments in tri-ortho-cresyl phosphate (TOCP) induced delayed neuropathy. Toxicology 262(3):258–264

    Article  CAS  PubMed  Google Scholar 

  9. Gambalunga A, Pasqualato F, Lotti M (2010) Soluble phenyl valerate esterases of hen sciatic nerve and the potentiation of organophosphate induced delayed polyneuropathy. Chem Biol Interact 187(1–3):340–343

    Article  CAS  PubMed  Google Scholar 

  10. Hou WY, Long DX, Wang HP, Wang Q, Wu YJ (2008) The homeostasis of phosphatidylcholine and lysophosphatidylcholine was not disrupted during tri-o-cresyl phosphate-induced delayed neurotoxicity in hens. Toxicology 252(1–3):56–63

    Article  CAS  PubMed  Google Scholar 

  11. Moretto A, Nicolli A, Lotti M (2005) Peripheral nerve esterases and the promotion of organophosphate-induced neuropathy in hens. Chem Biol Interact 157–158:285–291

    Article  PubMed  Google Scholar 

  12. Jortner BS (2000) Mechanisms of toxic injury in the peripheral nervous system: neuropathologic considerations. Toxicol Pathol 28(1):54–69

    Article  CAS  PubMed  Google Scholar 

  13. Jortner BS (2011) Preparation and analysis of the peripheral nervous system. Toxicol Pathol 39(1):66–72

    Article  PubMed  Google Scholar 

  14. Emerick GL, Peccinini RG, de Oliveira GH (2009) Organophosphorus-induced delayed neuropathy: a simple and efficient therapeutic strategy. Toxicol Lett 192(2):238–244

    Article  PubMed  Google Scholar 

  15. Su J, Zhang T, Wang K, Zhu T, Li X (2014) Autophagy activation contributes to the neuroprotection of remote ischemic perconditioning against focal cerebral ischemia in rats. Neurochem Res 39(11):2068–2077

    Article  CAS  PubMed  Google Scholar 

  16. He Y, Zhao X, Subahan NR, Fan L, Gao J, Chen H (2014) The prognostic value of autophagy-related markers beclin-1 and microtubule-associated protein light chain 3B in cancers: a systematic review and meta-analysis. Tumour Biol 35(8):7317–7326

    Article  CAS  PubMed  Google Scholar 

  17. Mizushima N (2007) Autophagy: process and function. Genes Dev 21(22):2861–2873

    Article  CAS  PubMed  Google Scholar 

  18. Song JX, Lu JH, Liu LF, Chen LL, Durairajan SS, Yue Z, Zhang HQ, Li M (2014) HMGB1 is involved in autophagy inhibition caused by SNCA/α-synuclein overexpression: a process modulated by the natural autophagy inducer corynoxine B. Autophagy 10(1):144–1454

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Dancourt J, Melia TJ (2014) Lipidation of the autophagy proteins LC3 and GABARAP is a membrane-curvature dependent process. Autophagy 10(8):1470–1471

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Song F, Kou R, Zou C, Gao Y, Zeng T, Xie K (2014) Involvement of autophagy in tri-ortho-cresyl phosphate-induced delayed neuropathy in hens. Neurochem Int 64:1–8

    Article  CAS  PubMed  Google Scholar 

  21. Song F, Han X, Zeng T, Zhang C, Zou C, Xie K (2012) Changes in beclin-1 and micro-calpain expression in tri-ortho-cresyl phosphate-induced delayed neuropathy. Toxicol Lett 210(3):276–284

    Article  CAS  PubMed  Google Scholar 

  22. Tatemoto K, Hosoya M, Habata Y, Fujii R, Kakegawa T, Zou MX, Kawamata Y, Fukusumi S, Hinuma S, Kitada C, Kurokawa T, Onda H, Fujino M (1998) Isolation and characterization of a novel endogenous peptide ligand for the human APJ receptor. Biochem Biophys Res Commun 251(2):471–476

    Article  CAS  PubMed  Google Scholar 

  23. O’Dowd BF, Heiber M, Chan A, Heng HH, Tsui LC, Kennedy JL, Shi X, Petronis A, George SR, Nguyen T (1993) A human gene that shows identity with the gene encoding the angiotensin receptor is located on chromosome 11. Gene 136(1–2):355–360

    Article  PubMed  Google Scholar 

  24. Rayalam S, Della-Fera MA, Kasser T, Warren W, Baile CA (2011) Emerging role of apelin as a therapeutic target in cancer: a patent review. Recent Pat Anticancer Drug Discov 6(3):367–372

    Article  CAS  PubMed  Google Scholar 

  25. Castan-Laurell I, Dray C, Attané C, Duparc T, Knauf C, Valet P (2011) Apelin, diabetes, and obesity. Endocrine 40(1):1–9

    Article  CAS  PubMed  Google Scholar 

  26. Bodineau L, Taveau C, Lê Quan Sang HH, Osterstock G, Queguiner I, Moos F, Frugière A, Llorens-Cortes C (2011) Data supporting a new physiological role for brain apelin in the regulation of hypothalamic oxytocin neurons in lactating rats. Endocrinology 152(9):3492–3503

    Article  CAS  PubMed  Google Scholar 

  27. Zhang X, Peng X, Fang M, Zhou C, Zhao F, Zhang Y, Xu Y, Zhu Q, Luo J, Chen G, Wang X (2011) Up-regulation of apelin in brain tissue of patients with epilepsy and an epileptic rat model. Peptides 32(9):1793–1799

    Article  CAS  PubMed  Google Scholar 

  28. Xin Q, Cheng B, Pan Y, Liu H, Yang C, Chen J, Bai B (2014) Neuroprotective effects of apelin-13 on experimental ischemic stroke through suppression of inflammation. Peptides 63C:55–62

    Google Scholar 

  29. Zeng XJ, Yu SP, Zhang L, Wei L (2010) Neuroprotective effect of the endogenous neural peptide apelin in cultured mouse cortical neurons. Exp Cell Res 316(11):1773–1783

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhang H, Gong Y, Wang Z, Jiang L, Chen R, Fan X, Zhu H, Han L, Li X, Xiao J, Kong X (2014) Apelin inhibits the proliferation and migration of rat PASMCs via the activation of PI3K/Akt/mTOR signal and the inhibition of autophagy under hypoxia. J Cell Mol Med 18(3):542–553

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Bao HJ, Zhang L, Han WC, Dai DK (2015) Apelin-13 attenuates traumatic brain injury-induced damage by suppressing autophagy. Neurochem Res 40(1):89–97

    Article  CAS  PubMed  Google Scholar 

  32. Zou C, Kou R, Gao Y, Xie K, Song F (2013) Activation of mitochondria-mediated apoptotic pathway in tri-ortho-cresyl phosphate-induced delayed neuropathy. Neurochem Int 62(7):965–972

    Article  CAS  PubMed  Google Scholar 

  33. Honorato de Oliveira G, Moreira V, Ribeiro Goes SP (2002) Organophosphate induced delayed neuropathy in genetically dissimilar chickens: studies with tri-ortho-cresyl phosphate (TOCP) and trichlorfon. Toxicol Lett 136(2):143–150

    Article  CAS  PubMed  Google Scholar 

  34. Masoud A, Sandhir R (2012) Increased oxidative stress is associated with the development of organophosphate-induced delayed neuropathy. Hum Exp Toxicol 31(12):1214–1227

    Article  CAS  PubMed  Google Scholar 

  35. Makhaeva GF, Malygin VV, Strakhova NN, Sigolaeva LV, Sokolovskaya LG, Eremenko AV, Kurochkin IN, Richardson RJ (2007) Biosensor assay of neuropathy target esterase in whole blood as a new approach to OPIDN risk assessment: review of progress. Hum Exp Toxicol 26(4):273–282

    Article  CAS  PubMed  Google Scholar 

  36. Song F, Yan Y, Zhao X, Zhang C, Xie K (2009) Neurofilaments degradation as an early molecular event in tri-ortho-cresyl phosphate (TOCP) induced delayed neuropathy. Toxicology 258(2–3):94–100

    Article  CAS  PubMed  Google Scholar 

  37. Zhao XL, Zhang TL, Zhang CL, Han XY, Yu SF, Li SX, Cui N, Xie KQ (2006) Expression changes of neurofilament subunits in the central nervous system of hens treated with tri-ortho-cresyl phosphate (TOCP). Toxicology 223(1–2):127–135

    Article  CAS  PubMed  Google Scholar 

  38. Honorato de Oliveira G, Moreira V, Ribeiro Goes SP (2002) Organophosphate induced delayed neuropathy in genetically dissimilar chickens: studies with tri-ortho-cresyl phosphate (TOCP) and trichlorfon. Toxicol Lett 136(2):143–150

    Article  CAS  PubMed  Google Scholar 

  39. Simpkin JC, Yellon DM, Davidson SM, Lim SY, Wynne AM, Smith CC (2007) Apelin-13 and apelin-36 exhibit direct cardioprotective activity against ischemia-reperfusion injury. Basic Res Cardiol 102(6):518–528

    Article  CAS  PubMed  Google Scholar 

  40. O’Donnell LA, Agrawal A, Sabnekar P, Dichter MA, Lynch DR, Kolson DL (2007) Apelin, an endogenous neuronal peptide, protects hippocampal neurons against excitotoxic injury. J Neurochem 102(6):1905–1917

    Article  PubMed  Google Scholar 

  41. Masoud A, Sandhir R (2012) Increased oxidative stress is associated with the development of organophosphate-induced delayed neuropathy. Hum Exp Toxicol 31(12):1214–1227

    Article  CAS  PubMed  Google Scholar 

  42. Masoud A, Kiran R, Sandhir R (2011) Modulation of dopaminergic system and neurobehavioral functions in delayed neuropathy induced by organophosphates. Toxicol Mech Methods 21(1):1–5

    Article  CAS  PubMed  Google Scholar 

  43. Chen A, Xiong LJ, Tong Y, Mao M (2013) Neuroprotective effect of brain-derived neurotrophic factor mediated by autophagy through the PI3K/Akt/mTOR pathway. Mol Med Rep 8(4):1011–1016

    CAS  PubMed  Google Scholar 

  44. Carloni S, Girelli S, Scopa C, Buonocore G, Longini M, Balduini W (2010) Activation of autophagy and Akt/CREB signaling play an equivalent role in the neuroprotective effect of rapamycin in neonatal hypoxia-ischemia. Autophagy 6(3):366–377

    Article  CAS  PubMed  Google Scholar 

  45. Fu MM, Holzbaur EL (2014) MAPK8IP1/JIP1 regulates the trafficking of autophagosomes in neurons. Autophagy 10(11):2079–2081

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Yoon SY, Choi JE, Kweon HS, Choe H, Kim SW, Hwang O, Lee H, Lee JY, Kim DH (2008) Okadaic acid increases autophagosomes in rat neurons: implications for Alzheimer’s disease. J Neurosci Res 86(14):3230–3239

    Article  CAS  PubMed  Google Scholar 

  47. Nixon RA (2013) The role of autophagy in neurodegenerative disease. Nat Med 19(8):983–997

    Article  CAS  PubMed  Google Scholar 

  48. Son SM, Song H, Byun J, Park KS, Jang HC, Park YJ, Mook-Jung I (2012) Accumulation of autophagosomes contributes to enhanced amyloidogenic APP processing under insulin-resistant conditions. Autophagy 8(12):1842–1844

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Tanida I, Ueno T, Kominami E (2008) LC3 and autophagy. Methods Mol Biol 445:77–88

    Article  CAS  PubMed  Google Scholar 

  50. Giménez-Xavier P, Francisco R, Platini F, Pérez R, Ambrosio S (2008) LC3-I conversion to LC3-II does not necessarily result in complete autophagy. Int J Mol Med 22(6):781–785

    PubMed  Google Scholar 

  51. Huang AC, Lien JC, Lin MW, Yang JS, Wu PP, Chang SJ, Lai TY (2013) Tetrandrine induces cell death in SAS human oral cancer cells through caspase activation-dependent apoptosis and LC3-I and LC3-II activation-dependent autophagy. Int J Oncol 43(2):485–494

    CAS  PubMed  Google Scholar 

  52. Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Øvervatn A, Bjørkøy G, Johansen T (2007) p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282(33):24131–24145

    Article  CAS  PubMed  Google Scholar 

  53. Strappazzon F, Nazio F, Corrado M, Cianfanelli V, Romagnoli A, Fimia GM, Campello S, Nardacci R, Piacentini M, Campanella M, Cecconi F (2015) AMBRA1 is able to induce mitophagy via LC3 binding, regardless of PARKIN and p62/SQSTM1. Cell Death Differ 22(3):419–432

    Article  CAS  PubMed  Google Scholar 

  54. 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(12):2208–2222

    Article  CAS  PubMed  Google Scholar 

  55. Jiang P, Mizushima N (2015) LC3- and p62-based biochemical methods for the analysis of autophagy progression in mammalian cells. Methods 75:13–18

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The work was supported by the construct program of the key discipline in hunan province, China (Basic Medicine Sciences in University of South China), Zhengxiang Scholar Program of University of South China (2014-004) and the National Natural Science Foundation of China (Grant No. 81171281).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shou-hong Zhou.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical Standards

The manuscript does not contain clinical studies or patient data. Standards of animal care were in accordance with the ARRIVE guidelines and China law.

Additional information

An erratum to this article can be found online at http://dx.doi.org/10.1007/s11064-016-2049-z.

The article has been retracted by the Editor-in-Chief due to the fact that Figure 2 was copied from the earlier publication by Song et al., Involvement of autophagy in tri-ortho-cresyl phosphate-induced delayed neuropathy in hens, Neurochem. Int. (2014) 64:1-8 (DOI: 10.1016/j.neuint.2013.10.017 ). The figure does not reflect the experiment described. The corresponding author has acknowledged this and agrees to the retraction.

An erratum to this article can be found at http://dx.doi.org/10.1007/s11064-016-2049-z.

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, Sh., Ouyang, Xp., Tian, Sw. et al. RETRACTED ARTICLE: Apelin-13 Prevents the Delayed Neuropathy Induced by Tri-ortho-cresyl Phosphate Through Regulation the Autophagy Flux in Hens. Neurochem Res 40, 2374–2382 (2015). https://doi.org/10.1007/s11064-015-1725-8

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-015-1725-8

Keywords

Navigation