Skip to main content
Log in

Thioredoxin-Interacting Protein (TXNIP) Regulates Parkin/PINK1-mediated Mitophagy in Dopaminergic Neurons Under High-glucose Conditions: Implications for Molecular Links Between Parkinson’s Disease and Diabetes

  • Original Article
  • Published:
Neuroscience Bulletin Aims and scope Submit manuscript

Abstract

Patients with diabetes mellitus have a higher risk of developing Parkinson’s disease (PD). However, the molecular links between PD and diabetes remain unclear. In this study, we investigated the roles of thioredoxin-interacting protein (TXNIP) in Parkin/PINK1-mediated mitophagy in dopaminergic (DA) cells under high-glucose (HG) conditions. In streptozotocin-induced diabetic mice, TXNIP was upregulated and autophagy was inhibited in the midbrain, while the loss of DA neurons was accelerated by hyperglycemia. In cultured PC12 cells under HG, TXNIP expression was upregulated and the intracellular reactive oxygen species (ROS) levels increased, leading to cell death. Autophagic flux was further blocked and PINK1 expression was decreased under HG conditions. Parkin expression in the mitochondrial fraction and carbonyl cyanide 3-chlorophenylhydrazone (CCCP)-induced co-localization of COX IV (marker for mitochondria) and LAMP1 (marker for lysosomes) were also significantly decreased by HG. Overexpression of TXNIP was sufficient to decrease the expression of both PINK1 and Parkin in PC12 cells, while knockdown of the expression of TXNIP by siRNA decreased intracellular ROS and attenuated cellular injury under HG. Moreover, inhibition of TXNIP improved the CCCP-induced co-localization of COX IV and LAMP1 in PC12 cells under HG. Together, these results suggest that TXNIP regulates Parkin/PINK1-mediated mitophagy under HG conditions, and targeting TXNIP may be a promising therapeutic strategy for reducing the risk of PD under hyperglycemic conditions.

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

Similar content being viewed by others

References

  1. Luitse MJ, Biessels GJ, Rutten GE, Kappelle LJ. Diabetes, hyperglycaemia, and acute ischaemic stroke. Lancet Neurol 2012, 11: 261–271.

    PubMed  Google Scholar 

  2. Liu RY, Wang JJ, Qiu X, Wu JM. Acute hyperglycemia together with hematoma of high-glucose blood exacerbates neurological injury in a rat model of intracerebral hemorrhage. Neurosci Bull 2014, 30: 90–98.

    PubMed  Google Scholar 

  3. Biosa A, Outeiro TF, Bubacco L, Bisaglia M. Diabetes mellitus as a risk factor for Parkinson’s disease: a molecular point of view. Mol Neurobiol 2018, 55: 8754–8763.

    CAS  PubMed  Google Scholar 

  4. Kaplan M, Aviram M, Hayek T. Oxidative stress and macrophage foam cell formation during diabetes mellitus-induced atherogenesis: role of insulin therapy. Pharmacol Ther 2012, 136: 175–185.

    CAS  PubMed  Google Scholar 

  5. Pickering RJ, Rosado CJ, Sharma A, Buksh S, Tate M, de Haan JB. Recent novel approaches to limit oxidative stress and inflammation in diabetic complications. Clin Transl Immunol 2018, 7: e1016.

    PubMed  PubMed Central  Google Scholar 

  6. Nasoohi S, Ismael S, Ishrat T. Thioredoxin-interacting protein (TXNIP) in cerebrovascular and neurodegenerative diseases: regulation and implication. Mol Neurobiol 2018, 55: 7900–7920.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Li X, Kover KL, Heruth DP, Watkins DJ, Guo Y, Moore WV, et al. Thioredoxin-interacting protein promotes high-glucose-induced macrovascular endothelial dysfunction. Biochem Biophys Res Commun 2017, 493: 291–297.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Wei J, Wu H, Zhang H, Li F, Chen S, Hou B, et al. Anthocyanins inhibit high glucose-induced renal tubular cell apoptosis caused by oxidative stress in db/db mice. Int J Mol Med 2018, 41: 1608–1618.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Su CJ, Feng Y, Liu TT, Liu X, Bao JJ, Shi AM, et al. Thioredoxin-interacting protein induced alpha-synuclein accumulation via inhibition of autophagic flux: Implications for Parkinson’s disease. CNS Neurosci Ther 2017, 23: 717–723.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Macdonald R, Barnes K, Hastings C, Mortiboys H. Mitochondrial abnormalities in Parkinson’s disease and Alzheimer’s disease: can mitochondria be targeted therapeutically? Biochem Soc Trans 2018, 46: 891–909.

    CAS  PubMed  Google Scholar 

  11. Weil R, Laplantine E, Curic S, Genin P. Role of optineurin in the mitochondrial dysfunction: potential implications in neurodegenerative diseases and cancer. Front Immunol 2018, 9: 1243.

    PubMed  PubMed Central  Google Scholar 

  12. Fivenson EM, Lautrup S, Sun N, Scheibye-Knudsen M, Stevnsner T, Nilsen H, et al. Mitophagy in neurodegeneration and aging. Neurochem Int 2017, 109: 202–209.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Yuan Y, Zhang X, Zheng Y, Chen Z. Regulation of mitophagy in ischemic brain injury. Neurosci Bull 2015, 31: 395–406.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Ordureau A, Paulo JA, Zhang W, Ahfeldt T, Zhang J, Cohn EF, et al. Dynamics of PARKIN-dependent mitochondrial ubiquitylation in induced neurons and model systems revealed by digital snapshot proteomics. Mol Cell 2018, 70: 211–227 e218.

    Google Scholar 

  15. Yamada T, Murata D, Adachi Y, Itoh K, Kameoka S, Igarashi A, et al. Mitochondrial stasis reveals p62-mediated ubiquitination in Parkin-independent mitophagy and mitigates nonalcoholic fatty liver disease. Cell Metab 2018, 28: 588–604.e5.

    Google Scholar 

  16. Burte F, Carelli V, Chinnery PF, Yu-Wai-Man P. Disturbed mitochondrial dynamics and neurodegenerative disorders. Nat Rev Neurol 2015, 11: 11–24.

    CAS  PubMed  Google Scholar 

  17. Piano I, Novelli E, Della Santina L, Strettoi E, Cervetto L, Gargini C. Involvement of autophagic pathway in the progression of retinal degeneration in a mouse model of diabetes. Front Cell Neurosci 2016, 10: 42.

    PubMed  PubMed Central  Google Scholar 

  18. Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998, 392: 605–608.

    CAS  PubMed  Google Scholar 

  19. Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, et al. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science 2004, 304: 1158–1160.

    CAS  PubMed  Google Scholar 

  20. Kumar A, Mittal R. Mapping Txnip: key connexions in progression of diabetic nephropathy. Pharmacol Rep 2018, 70: 614–622.

    CAS  PubMed  Google Scholar 

  21. Andrews ZB, Horvath B, Barnstable CJ, Elsworth J, Yang L, Beal MF, et al. Uncoupling protein-2 is critical for nigral dopamine cell survival in a mouse model of Parkinson’s disease. J Neurosci 2005, 25: 184–191.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Lu M, Su C, Qiao C, Bian Y, Ding J, Hu G. Metformin prevents dopaminergic neuron death in MPTP/P-induced mouse model of Parkinson’s disease via autophagy and mitochondrial ROS clearance. Int J Neuropsychopharmacol 2016, 19. https://doi.org/10.1093/ijnp/pyw047

  23. Youn CK, Kim HB, Wu TT, Park S, Cho SI, Lee JH. 53BP1 contributes to regulation of autophagic clearance of mitochondria. Sci Rep 2017, 7: 45290.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Suen DF, Narendra DP, Tanaka A, Manfredi G, Youle RJ. Parkin overexpression selects against a deleterious mtDNA mutation in heteroplasmic cybrid cells. Proc Natl Acad Sci U S A 2010, 107: 11835–11840.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Kwon KJ, Lee EJ, Kim MK, Kim SY, Kim JN, Kim JO, et al. Diabetes augments cognitive dysfunction in chronic cerebral hypoperfusion by increasing neuronal cell death: implication of cilostazol for diabetes mellitus-induced dementia. Neurobiol Dis 2015, 73: 12–23.

    CAS  PubMed  Google Scholar 

  26. Pagano G, Polychronis S, Wilson H, Giordano B, Ferrara N, Niccolini F, et al. Diabetes mellitus and Parkinson disease. Neurology 2018, 90: e1654–e1662.

    PubMed  Google Scholar 

  27. Yue X, Li H, Yan H, Zhang P, Chang L, Li T. Risk of Parkinson disease in diabetes mellitus: an updated meta-analysis of population-based cohort studies. Medicine (Baltimore) 2016, 95: e3549.

    Google Scholar 

  28. Burbulla LF, Song P, Mazzulli JR, Zampese E, Wong YC, Jeon S, et al. Dopamine oxidation mediates mitochondrial and lysosomal dysfunction in Parkinson’s disease. Science 2017, 357: 1255–1261.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Chitnis T, Weiner HL. CNS inflammation and neurodegeneration. J Clin Invest 2017, 127: 3577–3587.

    PubMed  PubMed Central  Google Scholar 

  30. Shimobayashi M, Albert V, Woelnerhanssen B, Frei IC, Weissenberger D, Meyer-Gerspach AC, et al. Insulin resistance causes inflammation in adipose tissue. J Clin Invest 2018, 128: 1538–1550.

    PubMed  PubMed Central  Google Scholar 

  31. Yuan H, Zheng JC, Liu P, Zhang SF, Xu JY, Bai LM. Pathogenesis of Parkinson’s disease: oxidative stress, environmental impact factors and inflammatory processes. Neurosci Bull 2007, 23: 125–130.

    CAS  PubMed  Google Scholar 

  32. Namazi Sarvestani N, Saberi Firouzi S, Falak R, Karimi MY, Davoodzadeh Gholami M, Rangbar A, et al. Phosphodiesterase 4 and 7 inhibitors produce protective effects against high glucose-induced neurotoxicity in PC12 cells via modulation of the oxidative stress, apoptosis and inflammation pathways. Metab Brain Dis 2018, 33: 1293–1306.

    CAS  PubMed  Google Scholar 

  33. Aminzadeh A. Protective effect of tropisetron on high glucose induced apoptosis and oxidative stress in PC12 cells: roles of JNK, P38 MAPKs, and mitochondria pathway. Metab Brain Dis 2017, 32: 819–826.

    CAS  PubMed  Google Scholar 

  34. Chen M, Zheng H, Wei T, Wang D, Xia H, Zhao L, et al. High glucose-induced PC12 cell death by increasing glutamate production and decreasing methyl group metabolism. Biomed Res Int 2016, 2016: 4125731.

    PubMed  PubMed Central  Google Scholar 

  35. Wang L, Zhai YQ, Xu LL, Qiao C, Sun XL, Ding JH, et al. Metabolic inflammation exacerbates dopaminergic neuronal degeneration in response to acute MPTP challenge in type 2 diabetes mice. Exp Neurol 2014, 251: 22–29.

    CAS  PubMed  Google Scholar 

  36. Nishikawa T, Araki E. Impact of mitochondrial ROS production in the pathogenesis of diabetes mellitus and its complications. Antioxid Redox Signal 2007, 9: 343–353.

    CAS  PubMed  Google Scholar 

  37. Wei W, Liu Q, Tan Y, Liu L, Li X, Cai L. Oxidative stress, diabetes, and diabetic complications. Hemoglobin 2009, 33: 370–377.

    CAS  PubMed  Google Scholar 

  38. Alhawiti NM, Al Mahri S, Aziz MA, Malik SS, Mohammad S. TXNIP in metabolic regulation: physiological role and therapeutic outlook. Curr Drug Targets 2017, 18: 1095–1103.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Lu L, Lu Q, Chen W, Li J, Li C, Zheng Z. Vitamin D3 protects against diabetic retinopathy by inhibiting high-glucose-induced activation of the ROS/TXNIP/NLRP3 inflammasome pathway. J Diabetes Res 2018, 2018: 8193523.

    PubMed  PubMed Central  Google Scholar 

  40. Mishra M, Kowluru RA. DNA methylation-a potential source of mitochondria DNA base mismatch in the development of diabetic retinopathy. Mol Neurobiol 2019, 56: 88–101.

    CAS  PubMed  Google Scholar 

  41. Wang S, Zhao Z, Fan Y, Zhang M, Feng X, Lin J, et al. Mst1 inhibits Sirt3 expression and contributes to diabetic cardiomyopathy through inhibiting Parkin-dependent mitophagy. Biochim Biophys Acta 2019, 1865: 1905–1914.

    CAS  Google Scholar 

  42. Park JS, Davis RL, Sue CM. Mitochondrial dysfunction in Parkinson’s disease: new mechanistic insights and therapeutic perspectives. Curr Neurol Neurosci Rep 2018, 18: 21.

    PubMed  PubMed Central  Google Scholar 

  43. He L, Chen L, Li L. The TBK1-OPTN axis mediates crosstalk between mitophagy and the innate immune response: a potential therapeutic target for neurodegenerative diseases. Neurosci Bull 2017, 33: 354–356.

    PubMed  PubMed Central  Google Scholar 

  44. Wei H, Liu L, Chen Q. Selective removal of mitochondria via mitophagy: distinct pathways for different mitochondrial stresses. Biochim Biophys Acta 2015, 1853: 2784–2790.

    CAS  PubMed  Google Scholar 

  45. Greene AW, Grenier K, Aguileta MA, Muise S, Farazifard R, Haque ME, et al. Mitochondrial processing peptidase regulates PINK1 processing, import and Parkin recruitment. EMBO Rep 2012, 13: 378–385.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Lin W, Kang UJ. Characterization of PINK1 processing, stability, and subcellular localization. J Neurochem 2008, 106: 464–474.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Gladkova C, Maslen SL, Skehel JM, Komander D. Mechanism of parkin activation by PINK1. Nature 2018, 559: 410–414.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Oh CK, Sultan A, Platzer J, Dolatabadi N, Soldner F, McClatchy DB, et al. S-Nitrosylation of PINK1 attenuates PINK1/Parkin-dependent mitophagy in hiPSC-based Parkinson’s disease models. Cell Rep 2017, 21: 2171–2182.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, et al. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol 2010, 8: e1000298.

    PubMed  PubMed Central  Google Scholar 

  50. Singh LP. Thioredoxin interacting protein (TXNIP) and pathogenesis of diabetic retinopathy. J Clin Exp Ophthalmol 2013, 4. https://doi.org/10.4172/2155-9570.1000287

  51. Lee S, Kim SM, Lee RT. Thioredoxin and thioredoxin target proteins: from molecular mechanisms to functional significance. Antioxid Redox Signal 2013, 18: 1165–1207.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Huang C, Zhang Y, Kelly DJ, Tan CY, Gill A, Cheng D, et al. Thioredoxin interacting protein (TXNIP) regulates tubular autophagy and mitophagy in diabetic nephropathy through the mTOR signaling pathway. Sci Rep 2016, 6: 29196.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Xiao B, Goh JY, Xiao L, Xian H, Lim KL, Liou YC. Reactive oxygen species trigger Parkin/PINK1 pathway-dependent mitophagy by inducing mitochondrial recruitment of Parkin. J Biol Chem 2017, 292: 16697–16708.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Science Foundation of China (81601098 and 81603181), the Natural Science Foundation of Jiangsu Province (BK20150302, BK20170004), the Natural Science Foundation of the Jiangsu Higher Education Institutions of China (19KJB310016), Suzhou Science and Technology for People’s Livelihood (SYS201706), and the Natural Science Foundation of Suzhou (SYSD2018099).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Tong Liu or Yun-Li Yu.

Ethics declarations

Conflict of interest

The authors declare no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Su, CJ., Shen, Z., Cui, RX. et al. Thioredoxin-Interacting Protein (TXNIP) Regulates Parkin/PINK1-mediated Mitophagy in Dopaminergic Neurons Under High-glucose Conditions: Implications for Molecular Links Between Parkinson’s Disease and Diabetes. Neurosci. Bull. 36, 346–358 (2020). https://doi.org/10.1007/s12264-019-00459-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12264-019-00459-5

Keywords

Navigation