Skip to main content

Advertisement

Log in

Functional Regulation of an Oxidative Stress Mediator, Rac1, in Diabetic Retinopathy

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Purpose

Early activation of cytosolic NADPH oxidase-2 (Nox2) in diabetes increases retinal ROS production, damaging their mitochondria. The assembly of Nox2 holoenzyme requires activation of a small molecular weight G protein Rac1. Rac1 activation is regulated by guanine exchange factors and guanine nucleotide-dissociation inhibitors, and post-translational modifications assist in its association with exchange factors and dissociation inhibitors. The goal of this study is to investigate the mechanisms of Rac1 activation in the development of diabetic retinopathy.

Methods

The levels of the dissociation inhibitor, prenylating enzyme (farnesyltransferase, FNTA), and exchange factor Vav2 were quantified in human retinal endothelial cells, incubated in normal or high glucose for 96 h. The roles of prenylation and Vav2 in Rac1-Nox2-ROS mitochondrial damage were confirmed in FNTA-siRNA–transfected cells and using the Vav2 inhibitor EHop, respectively. Retinal histopathology and functional changes associated with diabetic retinopathy were analyzed in diabetic mice receiving EHop for 6 months. Key parameters of Rac1 activation were confirmed in the retinal microvasculature from human donors with diabetic retinopathy.

Results

In HRECs, glucose increased FNTA and Vav2 and decreased the dissociation inhibitor. FNTA-siRNA and EHop inhibited glucose-induced activation of Rac1–Nox2–ROS signaling. In diabetic mice, EHop ameliorated the development of retinopathy and functional/structural abnormalities and attenuated Rac1–Nox2–mitochondrial damage. Similar alterations in Rac1 regulators were observed in retinal microvasculature from human donors with diabetic retinopathy. In diabetes, Rac1 prenylation and its interactions with Vav2 contribute to Nox2–ROS–mitochondrial damage, and the pharmacological inhibitors to attenuate Rac1 interactions with its regulators could have the potential to halt/inhibit the development of diabetic retinopathy.

Activation of prenylating enzyme farnesyltransferase (FNTA) in diabetes, prenylates Rac1. The binding of Rac1 with guanine nucleotide-dissociation inhibitor (GDI) is decreased, but its association with the guanine exchange factor, Vav2, is increased, resulting in Rac1 activation. Active Rac1 helps in the assembly of Nox2 holoenzyme, and Nox2 activation increases cytosolic ROS production, damaging the mitochondria. Damaged mitochondria accelerate capillary cell apoptosis, and ultimately, results in the development of diabetic retinopathy.

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

Similar content being viewed by others

References

  1. Frank RN (2004) Diabetic retinopathy. New Eng J Med 350:48–58

    Article  CAS  PubMed  Google Scholar 

  2. Kowluru RA, Kowluru A, Mishra M, Kumar B (2015) Oxidative stress and epigenetic modifications in the pathogenesis of diabetic retinopathy. Prog Retin Eye Res 48:40–61

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Guzman DC, Olguin HJ, Garcia EH, Peraza AV, de la Cruz DZ, Soto MP (2017) Mechanisms involved in the development of diabetic retinopathy induced by oxidative stress. Redox Rep 22:10–16

    Article  CAS  PubMed  Google Scholar 

  4. Kowluru RA, Kowluru A, Veluthakal R, Mohammad G, Syed I, Santos JM, Mishra M (2014) TIAM1-RAC1 signalling axis-mediated activation of NADPH oxidase-2 initiates mitochondrial damage in the development of diabetic retinopathy. Diabetologia 57:1047–1056

    Article  CAS  PubMed  Google Scholar 

  5. Kowluru A, Kowluru RA (2014) Phagocyte-like NADPH oxidase [Nox2] in cellular dysfunction in models of glucolipotoxicity and diabetes. Biochem Pharmacol 88:275–283

    Article  CAS  PubMed  Google Scholar 

  6. El-Benna J, Dang PM, Gougerot-Pocidalo MA, Marie JC, Braut-Boucher F (2009) p47phox, the phagocyte NADPH oxidase/NOX2 organizer: structure, phosphorylation and implication in diseases. Exp Mol Med 41:217–225

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pick E (2014) Role of the Rho GTPase Rac in the activation of the phagocyte NADPH oxidase: outsourcing a key task. Small GTPases 5:e27952

    Article  PubMed  PubMed Central  Google Scholar 

  8. Duraisamy AJ, Mishra M, Kowluru A, Kowluru RA (2018) Epigenetics and regulation of oxidative stress in diabetic retinopathy. Invest Ophthalmol Vis Sci 59:4831–4840

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kowluru RA, Mishra M, Kumar B (2016) Diabetic retinopathy and transcriptional regulation of a small molecular weight G-protein, Rac1. Exp Eye Res 147:72–77

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Haga RB, Ridley AJ (2016) Rho GTPases: regulation and roles in cancer cell biology. Small GTPases 7:207–221

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bai Y, Xiang X, Liang C, Shi L (2015) Regulating Rac in the nervous system: molecular function and disease implication of Rac GEFs and GAPs. Biomed Res Int 2015:632450

    PubMed  PubMed Central  Google Scholar 

  12. DerMardirossian C, Bokoch GM (2005) GDIs: central regulatory molecules in Rho GTPase activation. Trends Cell Biol 15:356–363

    Article  CAS  PubMed  Google Scholar 

  13. Syed I, Kyathanahalli CN, Kowluru A (2011) Phagocyte-like NADPH oxidase generates ROS in INS 832/13 cells and rat islets: role of protein prenylation. Am J Physiol Regul Integr Comp Physiol 300:R756–R762

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. van Rijssel J, van Buul JD (2012) The many faces of the guanine-nucleotide exchange factor trio. Cell Adhes Migr 6:482–487

    Article  Google Scholar 

  15. Abdrabou A, Wang Z (2018) Post-translational modification and subcellular distribution of Rac1: an update. Cells 7

  16. Mishra M, Duraisamy AJ, Bhattacharjee S, Kowluru RA (2019) Adaptor protein p66Shc: a link between cytosolic and mitochondrial dysfunction in the development of diabetic retinopathy. Antioxid Redox Signal 30:1621–1634

    Article  CAS  PubMed  Google Scholar 

  17. Schlegel N, Waschke J (2014) cAMP with other signaling cues converges on Rac1 to stabilize the endothelial barrier—a signaling pathway compromised in inflammation. Cell Tissue Res 355:587–596

    Article  CAS  PubMed  Google Scholar 

  18. Kowluru A (2017) Tiam1/Vav2-Rac1 axis: a tug-of-war between islet function and dysfunction. Biochem Pharmacol 132:9–17

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Veluthakal R, Tunduguru R, Arora DK, Sidarala V, Syeda K, Vlaar CP, Thurmond DC, Kowluru A (2015) VAV2, a guanine nucleotide exchange factor for Rac1, regulates glucose-stimulated insulin secretion in pancreatic beta cells. Diabetologia 58:2573–2581

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mishra M, Kowluru RA (2017) Role of PARP-1 as a novel transcriptional regulator of MMP-9 in diabetic retinopathy. Biochim Biophys Acta 1863:1761–1769

    Article  CAS  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  22. Mishra M, Duraisamy AJ, Kowluru RA (2018) Sirt1—a guardian of the development of diabetic retinopathy. Diabetes 67:745–754

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Castillo-Pichardo L, Humphries-Bickley T, De La Parra C, Forestier-Roman I, Martinez-Ferrer M, Hernandez E, Vlaar C, Ferrer-Acosta Y et al (2014) The Rac inhibitor EHop-016 inhibits mammary tumor growth and metastasis in a nude mouse model. Transl Oncol 7:546–555

    Article  PubMed  PubMed Central  Google Scholar 

  24. Mishra M, Flaga J, Kowluru RA (2016) Molecular mechanism of transcriptional regulation of matrix metalloproteinase-9 in diabetic retinopathy. J Cell Physiol 231:1709–1718

    Article  CAS  PubMed  Google Scholar 

  25. Kumar B, Kowluru A, Kowluru RA (2015) Lipotoxicity augments glucotoxicity-induced mitochondrial damage in the development of diabetic retinopathy. Invest Ophtahlmol Vis Sci 56:2985–2992

    Article  CAS  Google Scholar 

  26. Kowluru RA, Mohammad G, dos Santos JM, Zhong Q (2011) Abrogation of MMP-9 gene protects against the development of retinopathy in diabetic mice by preventing mitochondrial damage. Diabetes 60:3023–3033

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Liu CH, Sun Y, Li J, Gong Y, Tian KT, Evans LP, Morss PC, Fredrick TW et al (2015) Endothelial microRNA-150 is an intrinsic suppressor of pathologic ocular neovascularization. Proc Natl Acad Sci 112:12163–12168

    Article  CAS  PubMed  Google Scholar 

  28. Bokoch GM, Bohl BP, Chuang TH (1994) Guanine nucleotide exchange regulates membrane translocation of Rac/Rho GTP-binding proteins. J Biol Chem 269:31674–31679

    CAS  PubMed  Google Scholar 

  29. Engerman RL, Kern TS (1987) Progression of incipient diabetic retinopathy during good glycemic control. Diabetes 36:808–812

    Article  CAS  PubMed  Google Scholar 

  30. Mizutani M, Kern TS, Lorenzi M (1996) Accelerated death of retinal microvascular cells in human and experimental diabetic retinopathy. J Clin Invest 97:2883–2890

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kern TS, Miller CM, Tang J, Du Y, Ball SL, Berti-Matera L (2010) Comparison of three strains of diabetic rats with respect to the rate at which retinopathy and tactile allodynia develop. Mol Vis 16:1629–1639

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Kern TS, Barber AJ (2008) Retinal ganglion cells in diabetes. J Physiol 586:4401–4408

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. van Dijk HW, Verbraak FD, Kok PH, Stehouwer M, Garvin MK, Sonka M, DeVries JH, Schlingemann RO et al (2012) Early neurodegeneration in the retina of type 2 diabetic patients. Invest Ophtahlmol Vis Sci 53:2715–2719

    Article  Google Scholar 

  34. Kowluru RA (2005) Diabetic retinopathy, oxidative stress and antioxidants. Curr Top Nutraceut Res 3:209–218

    CAS  Google Scholar 

  35. Doly M, Droy-Lefaix MT, Braquet P (1992) Oxidative stress in diabetic retina. Basel, Birkhauser

    Book  Google Scholar 

  36. Madsen-Bouterse SA, Kowluru RA (2008) Oxidative stress and diabetic retinopathy: pathophysiological mechanisms and treatment perspectives. Rev Endocr Metab Disord 9:315–327

    Article  CAS  PubMed  Google Scholar 

  37. Calderon GD, Juarez OH, Hernandez GE, Punzo SM, De la Cruz ZD (2017) Oxidative stress and diabetic retinopathy: development and treatment. Eye 31:1122–1130

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kowluru RA, Mishra M, Kowluru A, Kumar B (2016) Hyperlipidemia and the development of diabetic retinopathy: comparison between type 1 and type 2 animal models. Metabolism 65:1570–1581

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Di-Poi N, Faure J, Grizot S, Molnar G, Pick E, Dagher MC (2001) Mechanism of NADPH oxidase activation by the Rac/Rho-GDI complex. Biochemistry 40:10014–10022

    Article  CAS  PubMed  Google Scholar 

  40. Chen GP, Zhang XQ, Wu T, Li L, Han J, Du CQ (2015) Alteration of mevalonate pathway in proliferated vascular smooth muscle from diabetic mice: possible role in high-glucose-induced atherogenic process. J Diabetes Res 2015:379287

    PubMed  PubMed Central  Google Scholar 

  41. Ugolev Y, Berdichevsky Y, Weinbaum C, Pick E (2008) Dissociation of Rac1(GDP).RhoGDI complexes by the cooperative action of anionic liposomes containing phosphatidylinositol 3,4,5-trisphosphate, Rac guanine nucleotide exchange factor, and GTP. J Biol Chem 283:22257–22271

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kowluru A, Kowluru RA (2018) RACking up ceramide-induced islet beta-cell dysfunction. Biochem Pharmacol 154:161–169

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu Y, Collins C, Kiosses WB, Murray AM, Joshi M, Shepherd TR, Fuentes EJ, Tzima E (2013) A novel pathway spatiotemporally activates Rac1 and redox signaling in response to fluid shear stress. J Cell Biol 201:863–873

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Phaneuf S, Leeuwenburgh C (2002) Cytochrome c release from mitochondria in the aging heart: a possible mechanism for apoptosis with age. Am J Physiol Regul Integr Comp Physiol 282:R423–R430

    Article  CAS  PubMed  Google Scholar 

  45. Simo R, Stitt AW, Gardner TW (2018) Neurodegeneration in diabetic retinopathy: does it really matter? Diabetologia 61:1902–1912

    Article  PubMed  PubMed Central  Google Scholar 

  46. Chen M, Knifley T, Subramanian T, Spielmann HP, O’Connor KL (2014) Use of synthetic isoprenoids to target protein prenylation and Rho GTPases in breast cancer invasion. PLoS One 9:e89892

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lacbay CM, Waller DD, Park J, Gomez Palou M, Vincent F, Huang XF, Ta V, Berghuis AM et al (2018) Unraveling the prenylation-cancer paradox in multiple myeloma with novel geranylgeranyl pyrophosphate synthase (GGPPS) inhibitors. J Med Chem 61:6904–6917

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This study was supported in parts by grants from the National Institutes of Health (RAK: EY014370 and EY017313; RAK, AK: EY022230), Thomas Foundation (RAK), Department of Veterans Affairs (1BX000469, AK), and an unrestricted grant to the Ophthalmology Department from Research to Prevent Blindness. AK is the recipient of a Senior Research Career Scientist Award from the Department of Veterans Affairs (13S-RCS-006).

Author information

Authors and Affiliations

Authors

Contributions

GM: researched data, literature search and manuscript editing; AJD: researched data, literature search and manuscript editing; AK: literature search, manuscript writing/editing; RAK: experimental plan, literature search, manuscript writing/editing. RAK is the guarantor of this work and, as such, had full access to all the data in this manuscript.

Corresponding author

Correspondence to Renu A. Kowluru.

Ethics declarations

The treatment of animals conformed to the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research and was approved by the Wayne State University’s Institutional Animal Care and Use Committee.

Conflict of Interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mohammad, G., Duraisamy, A.J., Kowluru, A. et al. Functional Regulation of an Oxidative Stress Mediator, Rac1, in Diabetic Retinopathy. Mol Neurobiol 56, 8643–8655 (2019). https://doi.org/10.1007/s12035-019-01696-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-019-01696-5

Keywords

Navigation