Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3349 2022-11-29 02:07:21 |
2 format corrected. Meta information modification 3349 2022-11-29 09:05:47 | |
3 format corrected. + 3 word(s) 3352 2022-11-29 09:07:57 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Wu, Y.;  Zou, H. Mitochondrial Dysfunction in Diabetic Retinopathy. Encyclopedia. Available online: https://encyclopedia.pub/entry/36924 (accessed on 27 July 2024).
Wu Y,  Zou H. Mitochondrial Dysfunction in Diabetic Retinopathy. Encyclopedia. Available at: https://encyclopedia.pub/entry/36924. Accessed July 27, 2024.
Wu, Yiwei, Haidong Zou. "Mitochondrial Dysfunction in Diabetic Retinopathy" Encyclopedia, https://encyclopedia.pub/entry/36924 (accessed July 27, 2024).
Wu, Y., & Zou, H. (2022, November 29). Mitochondrial Dysfunction in Diabetic Retinopathy. In Encyclopedia. https://encyclopedia.pub/entry/36924
Wu, Yiwei and Haidong Zou. "Mitochondrial Dysfunction in Diabetic Retinopathy." Encyclopedia. Web. 29 November, 2022.
Mitochondrial Dysfunction in Diabetic Retinopathy
Edit

Diabetic Retinopathy (DR) is one of the most important microvascular complications of diabetes mellitus, which can lead to blindness in severe cases. Mitochondria are energy-producing organelles in eukaryotic cells, which participate in metabolism and signal transduction, and regulate cell growth, differentiation, aging, and death. Metabolic changes of retinal cells and epigenetic changes of mitochondria-related genes under high glucose can lead to mitochondrial dysfunction and induce mitochondrial pathway apoptosis. In addition, mitophagy and mitochondrial dynamics also change adaptively. These mechanisms may be related to the occurrence and progression of DR, and also provide valuable clues for the prevention and treatment of DR. 

diabetic retinopathy mitochondrial metabolism epigenetics

1. Metabolic Changes

1.1. Glucose Metabolism

Under the stress of hyperglycemia, the nerve cells, pigment epithelial cells, and capillary endothelial cells in the retina can undergo glycometabolic reprogramming. Dysfunction of the electron transport chain (ETC), as the core change, results in energy generation disturbance [1], oxidative stress, abnormal glucose metabolite production, retinal microcirculation disorders, and other pathophysiological changes, inducing the occurrence of retinopathy [2].
In the case of continuous hyperglycemia, the overload of NADH and FADH2 produced by TCA can induce high mitochondrial membrane potential, which makes the ETC stagnate in complex III, and the electrons and protons carried by coenzyme Q are difficult to transfer to the downstream of the respiratory chain. At this time, oxygen molecules as electron acceptors can generate superoxide, mediating changes in cell metabolism [3].
  • The activity of GAPDH is reduced, activating the polyol pathway
Excess accumulation of intracellular superoxide activates PARP with the depletion of nicotinamide adenine dinucleotide (NAD+) [4]. As a result, the activity of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is significantly reduced [5], and the glycolysis process is inhibited. At this time, the polyol pathway of glucose metabolism is over-activated; in other words, glucose is converted to sorbitol under the action of aldose reductase, and then sorbitol can be oxidized to fructose [6]. Due to the decrease of the NAD+/NADH ratio, sorbitol, the intermediate product of polyol pathway, accumulates, and its high hydrophilicity can lead to cell hypertonicity, damage retinal capillary endothelial cells, and cause micro-circulation disorders [7]. The activation of the polyol pathway is also accompanied by further depletion of NAD+, which has a positive feedback effect on oxidative stress [8]. The C106T polymorphism in the aldose reductase gene is associated with the severity of retinopathy in type 2 diabetes [9]. Inhibition of aldose reductase reduces neuronal apoptosis, glial response, and complement deposition and retinal ne-ovascularization in DR [10].
  • The inhibition of glycolysis also leads to overactivation of the hexosamine pathway
The inhibition of glycolysis also promotes the metabolism of fructose-6-phosphate via the hexosamine pathway to UDP-N-acetyl glucosamine (UDP-GlcNAc), which promotes the synthesis of proteoglycans and O-linked glycoproteins. O-GlcNAc can covalently modify the transcription factor Sp1, which activates the expression of glucose-responsive gene plasminogen activator inhibitor-1 (Pal-1) in vascular smooth muscle cells [11], thus promoting DR [12]. It has been found that the level of hexosamine in the retinal tissue of diabetic patients is increased [7], which promotes the downstream O-GlcNAc to play a role in signal transduction, transcription regulation, regulation of cytoskeletal dynamics, and cell division [13]. Glucosamine, a product of the hexosamine metabolic pathway, causes retinal pericyte loss and the formation of acellular capillaries in non-diabetic animals by inhibiting VEGFR2 and Ang2 in the normal retina [14]. High hexosamine levels can also enhance cellular oxidative stress by positive feedback, further damaging the mitochondrial respiratory chain by promoting ROS generation [15]. However, oral administration of glucosamine protected retinal neurons in a mouse model of DR. The mechanism of this bidirectional regulation remains to be clarified [14].
  • Triose phosphate accumulates, activating several PKC-related pathways
Attenuation of GAPDH activity results in increased concentration of intracellular triose phosphate, which can be decomposed and acylated to diacyl glycerol(DAG) [16], and also participates in the formation of various advanced glycation end products (AGEs) [17], all of which play an important role in the pathogenesis of DR. On the one hand, DAG can directly activate protein kinase C (PKC) in the retina, mainly β- and δ-type isozymes, while the activation of PKC-α and -ɛ is also found in the retina of diabetic rats [16]. On the other hand, AGE-mediated signaling pathways as well as metabolic products of the polyol pathway are also associated with PKC activation [18][19]. Activated PKC increases the activity of cytosolic phospholipase A2 and promotes the production of arachidonic acid and prostaglandin E2 (PGE2); the latter inhibits the activity of Na+/K+ ATPase [20], leading to cell edema. PKC-α is related to the increased permeability of vascular endothelial cells under the condition of high glucose [21], and the activation of PKC-β can mediate retinal vasoconstriction and blood flow reduction by inhibiting the production of nitric oxide and increasing the activity of endothelin-1 [22]. These mechanisms may result in the injury and necrosis of retinal cells and affect the function of the retina. In addition, PKC can induce the expression of vascular endothelial growth factor (VEGF) in retinal tissue, thus increasing vascular permeability and promoting angiogenesis, which is closely related to the non-proliferative and proliferative changes in DR, respectively [23][24].
  • Massive synthesis of AGEs exerts pathological effects inside and outside cells
AGEs refer to a series of proteins, of which amino-groups are modified by intracellular dicarbonyl products (including glyoxal, methylglyoxal, and 3-deoxyglucosone), can be produced massively and secreted to extracellular medium when glycolysis is blocked. AGEs can change the function of intracellular and extracellular proteins, and are associated with intracellular signal transduction, metabolic regulation and cell adhesion [3]. Stitt et al. found that the content of AGEs in retinal blood vessels of diabetic mice was increased [25], and AGEs could interact with the receptor for advanced glycation end products (RAGE) on the plasma membrane of adjacent cells, exerting pathological effects, including promoting the activation of nuclear factor Kappa B (NF-κB) in retinal pericytes to induce their apoptosis, up-regulating the expression of VEGF in retinal capillary endothelial cells to increase vascular permeability, and activating the RhoA/ROCK signaling pathway as well as inducing moesin phosphorylation to promote retinal neovascularization [26][27][28]. AGEs can also lead to retinal vascular hyperpermeability by disrupting intercellular adhesion and tight junctions [29]. AGEs can up-regulate the expression of intercellular adhesion molecule-1 (ICAM-1) on the surface of endothelial cells, resulting in leukocyte stasis in the microcirculation and causing microcirculation disorders [30]. Ying et al. proposed that the severity of DR can be predicted by measuring AGEs [31]. As a result, researchers have begun to explore the possibility of delaying the progression of DR by inhibiting AGEs. Hammes et al. found that AGE-inhibitor amino-guanidine can inhibit the proliferation of abnormal retinal endothelial cells and significantly reduce pericyte shedding, thereby inhibiting the progression of DR [32]. Endogenous Glyoxalase I can inhibit the production of AGEs, and Maisonpierr et al. have found that its overexpression could inhibit the increase of Angiopoietin-2 (Ang-2) expression in Müller cells induced by hyperglycemia, thereby reducing damage to pericytes and capillary endothelial cells [33].

1.2. Lipid Metabolism

Abnormal lipid metabolism occurs in more than 75% of individuals with type 2 diabetes [34]. Excessive lipid accumulation and abnormal lipid metabolites have been proved to be important mechanisms for the progression of DR. Mitochondria, as the site of lipid metabolism, incur damage related to abnormal lipid metabolism [35]. High concentration of palmitate culture and high glucose induction can play a synergistic role in retinal endothelial cells, aggravating mtDNA damage [36]. Excessive production of ceramide, acrolein and incomplete β-oxidation products of fatty acids in retinal cells under high glucose environment are also associated with mitochondrial damage.
Under physiological conditions, activated fatty acids generate acyl-CoA, which enters mitochondria for β-oxidation, followed by terminal metabolism through the Krebs cycle [37]. In mice with DR, the β-oxidation of fatty acids in the mitochondria of retinal cells is incomplete, and excessive intermediate products accumulate to produce toxic lipid peroxidation products, which in turn cause mitochondrial damage [38][39].
  • Ceramide accumulation causes ETC disorder and mitochondrial pathway apoptosis
There are many kinds of sphingolipids in mitochondria, including sphingomyelin and ceramide [40][41], as well as metabolism-related enzymes, such as ceramide synthetase, acidic and neutral sphingomyelinase, and neutral ceramidase [35]. In normal cells, ceramide is minimally expressed in the cytoplasmic membrane, while a high glucose level induces ceramide accumulation in a concentration- and time-dependent manner [42][43][44]. Levitsky et al. found that ceramide induced by acid sphingomyelinase increased in mitochondria of retinal pigment epithelial cells in streptozotocin-induced diabetic rats, resulting in respiratory chain dysfunction. Inhibition of acid sphingomyelinase restores the function of the respiratory chain [45]. In terms of mechanism, excessive ceramide can inhibit the electron transfer function of complex III [46], promote the production of Sphingosine-1-Phosphate (S1P) and hexadecenal and the activation of BAX/BAK, increasing the permeability of the mitochondrial outer membrane and the release of cytochrome C. Finally, apoptosis is induced [47][48].
  • Acrolein overproduction promotes oxidative stress, causing mitochondrial damage
Acrolein is overproduced in the retinal cells of diabetic individuals, and the production of its protein-bound products is associated with the progression of DR [49]. As a biomarker, FDP-lysine can reflect the content of acrolein, which is increased in hemoglobin and vitreous humor in patients with proliferative diabetic retinopathy [50][51], and its level in retinal Müller cells of diabetic animal models also parallels the progress of the disease [52]. Polyamine oxidation and lipid peroxidation are the main intrinsic pathways of acrolein generation during DR pathology [49][53]. By depleting antioxidants such as glutathione [54] and promoting oxidative stress by forming protein carbonyls [55], excessive acrolein reduces the membrane potential of mitochondria and the activity of complexes I, II, and IV, resulting in mitochondrial damage in retinal pigment epithelial cells [56][57]. The corresponding metabolic and biochemical changes also occur in pathological processes such as retinal inflammatory response, ganglion cell degeneration, blood-retinal barrier damage, and Müller cell dysfunction [49].
  • Extracellular accumulation of modified lipoprotein induces apoptosis
Extracellular accumulation of abnormal lipid metabolites can also promote DR. Highly oxidized and glycosylated low-density lipoprotein accumulation in the lumen and extravasation of retinal capillaries is an early feature of DR [58]. Abnormally modified lipoproteins can induce apoptosis by increasing mitochondrial outer membrane permeability through the Bax pathway [59].

2. Epigenetic Changes

High glucose can not only reprogramme the metabolism of cells, but also regulate epigenetics by changing the activity of corresponding modifying enzymes, resulting in changes in gene expression, including a number of nuclear-encoded mitochondrial function-related genes and mitochondrial genome genes. The reprogramming of the expression of these genes is closely related to mitochondrial damage.
The most prominent epigenetic change is increased methylation levels [60]. Among patients with type II diabetes, DNA methylation of key genes in islets increases, which inhibits their expression [61][62][63][64], and DNA methylation levels are altered in adipose tissue, liver, and skeletal muscle [65][66][67][68][69][70]. Methylation of these genes may be directly induced by high glucose and HbA1C [63][64][71]. Kowluru et al. found that in the rat model of type 2 diabetes induced by high-fat diet, DNA Methyltransferase 1 (DNMT1) in retinal capillaries was highly expressed in the early stage of diabetes, which could affect the methylation of a series of genes related to retinal damage [72].
Ras-related C3 botulinum toxin substrate 1 (Rac1) is a component of the NADPH oxidase 2 (Nox2) holoenzyme [73], and the latter is able to induce ROS production in mitochondria [74], mediating oxidative stress. In the rat model of type 2 diabetes mellitus induced by a high-fat diet, the Rac1 promoter was methylated in retinal microvascular endothelial cells, and a large number of Nox2 produced promoted the accumulation of intracellular ROS, resulting in the damage to mtDNA and the respiratory chain [72]. In retinal cells of DR patients, methylation of H3K4 in the Keap1 promoter was significantly increased, which activated Keap1 transcription, blocking the nuclear transport of nuclear factor erythroid-2 related factor 2 (Nrf2), thereby inhibiting its antioxidant effects and exacerbating mitochondrial damage [75][76][77]. Histone hypermethylation occurred in Sod2 promoter under high glucose conditions (H3K4 monomethylation and dimethylation decreased, while trimethylation increased), accompanied by an increase in acetylation level (H3K9ac). As a result, the down-regulation of Sod2 expression and the reduction of intracellular antioxidant MnSOD content also promote oxidative stress damage [78][79][80]. Mitofusin 2 (Mfn2), which mediates mitochondrial outer membrane fusion, is down-regulated in retinal endothelial cells cultured in high glucose due to hypermethylation of the promoter, interfering with mitochondrial homeostasis [81].
Mitochondrial DNA (mtDNA), which does not bind to histones, encodes a total of 13 proteins, all of which are important components of the respiratory chain. DNA polymerase γ, encoded by the nuclear gene POLG, is responsible for mtDNA replication. In DR, the CpG island of POLG regulatory region is highly methylated, which inhibits the transcription of POLG and affects the replication of mtDNA [82]. MutL homolog 1 (MLH1) is involved in mismatch repair during mtDNA replication. In human retinal endothelial cells cultured with high glucose, the methylation level of MLH1 promoter is increased, down-regulating its transcription and affecting the replication accuracy and function of mtDNA [83], which may reduce the activity of respiratory chain complex I and the antioxidant capacity of mitochondria [84].
The mtDNA itself is also susceptible to methylation under high glucose conditions. This mostly occurs in the Displacement loop (D-Loop) of mtDNA, which, as a non-coding region, contains transcription elements and also controls DNA polymerase γ-dependent mtDNA replication. D-Loop has a loose structure and is susceptible to various modifying enzymes [85]. Mitochondrial DNA methyltransferase (DNMT) is also highly expressed in retinal cells under high glucose condition, which can highly methylate the D-Loop region of mtDNA and cause mitochondrial damage in retinal cells [86][87]. Inhibition of DNMT can reduce mtDNA damage, improve transcriptional repression of mtDNA induced by high glucose, and restore ETC function [88]. At the same time, inhibition of promoter methylation may restore the activity of Sod2 (see above), which can inhibit the methylation of D-Loop in mouse retinal microvascular endothelial cells and ensure the operation of base mismatch repair mechanism [89].
Interestingly, Zhong et al. found that the promoter methylation level (H3K9me2) of Matrix Metalloproteinase-9 (MMP-9) in retinal microvascular endothelial cells of diabetes mice decreased, while the acetylation level (H3K9Ac) increased [77][90][91][92], resulting in high expression of MMP-9, damaging mitochondria and inducing apoptosis of retinal microvascular cells [90][93]. In view of this, the influence of a high glucose environment on epigenetic modification of the genome may be diverse and complex. More studies are needed to clarify the expression patterns of mitochondria-related nuclear coding genes and mitochondrial genome in retinal cells under high glucose environment, and the exploration of omics can also provide valuable reference.

3. Mitophagy Changes

Autophagy is a highly conserved biological process, which can be induced by growth factor deficiency, hypoxia, cell starvation, oxidative stress, and other conditions, and maintains cell survival and homeostasis by degrading and reusing some intracellular proteins and organelles.
Mitophagy is a process in which cells selectively degrade damaged mitochondria by acting on themselves to control the number and quality of mitochondria, mediate cell differentiation and metabolic reprogramming [94], and the most typical pathway is PINK1-Parkin-mediated ubiquitin-dependent mitophagy [95]. Under stress conditions, when the mitochondrial outer membrane is damaged, the transmembrane transport of PINK1 is blocked and retained on the outer membrane, which recruits and activates the ubiquitin ligase Parkin1; the latter constructs a polyubiquitinated modification chain on the mitochondrial outer membrane protein, recruits the adaptor protein, and then the adaptor protein binds to LC3 to form a double-membrane-wrapped autophagosome. Autophagosomes subsequently bind to lysosomes and damaged mitochondria are engulfed [96].
Retinal cells can also protect themselves through mitophagy in the environment of high glucose.
Many studies have shown that mitophagy is upregulated during DR [97]. Devi et al. found that Parkin accumulated in mitochondria of retinal Müller cell line rMC1 under high glucose induction, and then induced autophagy [98]. Mitophagy plays a protective role in DR, and notoginsenoside R1 (NGR1) can alleviate the damage of retinal Müller cells by further enhancing PINK1-Parkin-dependent mitophagy [99]. Mitophagy is also critical for the function and survival of cone cells under hyperglycemic conditions [100]. In retinal vascular endothelial cells, activating bile acid G-protein-coupled membrane receptor (TGR5) enhances mitophagy, thereby reducing endothelial dysfunction and slowing the progression of DR [101]. In retinal pigment epithelial cells, Sirt3, a deacetylase in mitochondria, can activate mitophagy through Foxo3a/PINK1-Parkin pathway and protect the retina in a high glucose environment [102]. Normal mitophagy is essential for the maintenance of mitochondrial oxidative phosphorylation and ATP synthesis, which may be one of the mechanisms for mitophagy to exert cytoprotective effects in DR [103].
Thioredoxin-interacting protein (TXNIP) plays an important role in the maintenance of cellular homeostasis in the pathological process of DR. Under high glucose stress, the expression of TXNIP increases, which can antagonize the oxidative stress [104]. On the other hand, TXNIP can also play a cytoprotective role in promoting mitochondrial division and autophagy [105]. Up-regulated TXNIP in high glucose induces nitroso-modification of dynamin related protein 1 (Drp1) to promote mitochondrial fission [106], while modified Drp1 promotes TXNIP translocation to mitochondria, mediating mitophagy through multiple pathways [107]. For example, TXNIP can promote LC3BII-mediated mitophagy in retinal Müller cells of diabetic rats [98]. TXNIP causes nitrosylation, nuclear export, and cytoplasm localization of high mobility group box 1 protein (HMGB1), which competes with the cytoplasm-localized anti-apoptotic protein B-cell lymphoma 2 (BCL-2) for binding to the autophagy-related protein Beclin 1 [108], thereby inducing autophagy.

4. Mitochondrial Pathway Apoptosis

As mentioned previously, oxidative stress is prone to occur in cells under high glucose conditions. Antioxidant substances in retinal cells of diabetic individuals are reduced (e.g., MnSOD is reduced [109][110]), while the oxidative system is hyperactive (e.g., Nox2 is increased [111]), and a large amount of ROS is generated to cause mitochondrial damage. Mitochondrial damage can induce apoptosis of retinal capillary endothelial cells, pericytes, and neurons through content extravasation and activation of apoptosis-related signaling pathways [112][113][114][115]. Among them, pericytes are more sensitive to high glucose and more prone to apoptosis than endothelial cells [116].
  • Extravasation of proapoptotic substances
The opening of the PT pore in the outer mitochondrial membrane and the extravasation of proapoptotic substances are considered to be the characteristic events of mitochondrial pathway apoptosis. In retinal endothelial cells, high glucose can induce the release of mitochondrial cytochrome C, which may be related to the decrease of Cx43 channel activity in the outer mitochondrial membrane [82]. The release of cytochrome C triggers a caspase-mediated cascade that ultimately induces apoptosis. Santiago et al. found that high glucose-induced apoptosis in retinal neurons was related to the release of apoptosis-inducing factor (AIF) by mitochondria, but not dependent on the activation of caspases [117]. It is worth mentioning that RNA in mitochondria can also be released into the cytosol during mitochondrial damage induced by high glucose and exist as double-stranded RNA [118], which can interact with RNA-dependent protein kinase (PKR), mediating apoptosis of retinal neurons [119]. High glucose activates the NF-κB signaling pathway, enhances oxidative stress in Müller cells, and is also involved in inducing apoptosis in the mitochondrial pathway [120]. As a transcription factor, NF-kB can promote the expression of Matrix Metalloproteinase-2 (MMP-2) and MMP-9 [121], both of which are involved in inducing apoptosis of retinal microvascular endothelial cells [90][93][122]. In the diabetic state, MMP-2 is activated, which promotes the production of superoxide, accelerating mitochondrial membrane damage and cytochrome C leakage [122]. MMP-9 is also upregulated in high glucose and accumulates in mitochondria, increasing mitochondrial membrane permeability, which in turn promotes the entry of the pro-apoptotic protein BCL2-associated X protein (Bax) into mitochondria [90][93], mediating the occurrence of apoptosis.
  • Disequilibrium of calcium homeostasis
High glucose can also induce apoptosis through the mitochondrial pathway by triggering disequilibrium of calcium homeostasis. Liu et al. found that sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2) is inactivated by irreversible oxidative modification of Cys674 residue, which disrupts intracellular calcium homeostasis and promotes apoptosis [123][124].
  • Mitochondrial dynamic imbalance

In addition, the dynamic imbalance of mitochondrial fusion and fission is also closely related to apoptosis. High glucose stimulation can change the activity of related enzymes, reducing the fusion process of mitochondria in cells, and increasing the division, eventually stimulating the release of cytochrome C, and causing apoptosis of retinal cells (such as vascular endothelial cells and Müller cells) [125]. At the molecular level, high glucose can down-regulate the expression of mitochondrial fusion protein Mfn2 and fusion-related optic atrophy protein 1 (OPA1) in human retinal capillary endothelial cells [126][127], resulting in the reduction of mitochondrial fusion [128]. High glucose can also promote the phosphorylation and activation of PKCδ as well as upregulate TXNIP, regulating the phosphorylation and nitroso modification of a motility related protein Drp1, respectively, thus activating Drp1, which is related to mitochondrial fission [106][107]. Overexpression of Mfn2 or knockdown of Drp1 can inhibit apoptosis of retinal endothelial cells induced by high glucose [129][130][131].

References

  1. Miller, D.J.; Cascio, M.A.; Rosca, M.G. Diabetic Retinopathy: The Role of Mitochondria in the Neural Retina and Microvascular Disease. Antioxidant 2020, 9, 905.
  2. Ferrington, D.A.; Fisher, C.R.; Kowluru, R.A. Mitochondrial Defects Drive Degenerative Retinal Diseases. Trends. Mol. Med. 2020, 26, 105–118.
  3. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001, 414, 813–820.
  4. Cohen, M.S. Interplay between compartmentalized NAD(+) synthesis and consumption: A focus on the PARP family. Genes Dev. 2020, 34, 254–262.
  5. Du, X.L.; Edelstein, D.; Rossetti, L.; Fantus, I.G.; Goldberg, H.; Ziyadeh, F.; Wu, J.; Brownlee, M. Hyperglycemia-induced mitochondrial superoxide overproduction activates the hexosamine pathway and induces plasminogen activator inhibitor-1 expression by increasing Sp1 glycosylation. Proc. Natl. Acad. Sci. USA 2000, 97, 12222–12226.
  6. Garg, S.S.; Gupta, J. Polyol pathway and redox balance in diabetes. Pharm. Res. 2022, 182, 106326.
  7. Kang, Q.; Yang, C. Oxidative stress and diabetic retinopathy: Molecular mechanisms, pathogenetic role and therapeutic implications. Redox Biol. 2020, 37, 101799.
  8. Fan, L.; Cacicedo, J.M.; Ido, Y. Impaired nicotinamide adenine dinucleotide (NAD(+)) metabolism in diabetes and diabetic tissues: Implications for nicotinamide-related compound treatment. J. Diabetes Investig. 2020, 11, 1403–1419.
  9. Abu-Hassan, D.W.; Al-Bdour, M.D.; Saleh, I.; Freihat, M.; El-Khateeb, M. The relationship between aldose reductase gene C106T polymorphism and the severity of retinopathy in Type 2 diabetic patients: A case-control study. J. Res. Med. Sci. 2021, 26, 2.
  10. Singh, M.; Kapoor, A.; Bhatnagar, A. Physiological and Pathological Roles of Aldose Reductase. Metabolites 2021, 11, 655.
  11. Chen, Y.Q.; Su, M.; Walia, R.R.; Hao, Q.; Covington, J.W.; Vaughan, D.E. Sp1 sites mediate activation of the plasminogen activator inhibitor-1 promoter by glucose in vascular smooth muscle cells. J. Biol. Chem. 1998, 273, 8225–8231.
  12. Altalhi, R.; Pechlivani, N.; Ajjan, R.A. PAI-1 in Diabetes: Pathophysiology and Role as a Therapeutic Target. Int. J. Mol. Sci. 2021, 22, 3170.
  13. Peterson, S.B.; Hart, G.W. New insights: A role for O-GlcNAcylation in diabetic complications. Crit. Rev. Biochem. Mol. Biol. 2016, 51, 150–161.
  14. Eshwaran, R.; Kolibabka, M.; Poschet, G.; Jainta, G.; Zhao, D.; Teuma, L.; Murillo, K.; Hammes, H.P.; Schmidt, M.; Wieland, T.; et al. Glucosamine protects against neuronal but not vascular damage in experimental diabetic retinopathy. Mol. Metab. 2021, 54, 101333.
  15. Kaneto, H.; Xu, G.; Song, K.H.; Suzuma, K.; Bonner-Weir, S.; Sharma, A.; Weir, G.C. Activation of the hexosamine pathway leads to deterioration of pancreatic beta-cell function through the induction of oxidative stress. J. Biol. Chem. 2001, 276, 31099–31104.
  16. Koya, D.; King, G.L. Protein kinase C activation and the development of diabetic complications. Diabetes 1998, 47, 859–866.
  17. Dimitropoulos, A.; Rosado, C.J.; Thomas, M.C. Dicarbonyl-mediated AGEing and diabetic kidney disease. J. Nephrol. 2020, 33, 909–915.
  18. Portilla, D.; Dai, G.; Peters, J.M.; Gonzalez, F.J.; Crew, M.D.; Proia, A.D. Etomoxir-induced PPARalpha-modulated enzymes protect during acute renal failure. Am. J. Physiol. Ren. Physiol. 2000, 278, F667–F675.
  19. Keogh, R.J.; Dunlop, M.E.; Larkins, R.G. Effect of inhibition of aldose reductase on glucose flux, diacylglycerol formation, protein kinase C, and phospholipase A2 activation. Metabolism 1997, 46, 41–47.
  20. Xia, P.; Kramer, R.M.; King, G.L. Identification of the mechanism for the inhibition of Na+,K(+)-adenosine triphosphatase by hyperglycemia involving activation of protein kinase C and cytosolic phospholipase A2. J. Clin. Investig. 1995, 96, 733–740.
  21. Hempel, A.; Maasch, C.; Heintze, U.; Lindschau, C.; Dietz, R.; Luft, F.C.; Haller, H. High glucose concentrations increase endothelial cell permeability via activation of protein kinase C alpha. Circ. Res. 1997, 81, 363–371.
  22. Ishii, H.; Jirousek, M.R.; Koya, D.; Takagi, C.; Xia, P.; Clermont, A.; Bursell, S.E.; Kern, T.S.; Ballas, L.M.; Heath, W.F.; et al. Amelioration of vascular dysfunctions in diabetic rats by an oral PKC beta inhibitor. Science 1996, 272, 728–731.
  23. Williams, B.; Gallacher, B.; Patel, H.; Orme, C. Glucose-induced protein kinase C activation regulates vascular permeability factor mRNA expression and peptide production by human vascular smooth muscle cells in vitro. Diabetes 1997, 46, 1497–1503.
  24. Titchenell, P.M.; Antonetti, D.A. Using the past to inform the future: Anti-VEGF therapy as a road map to develop novel therapies for diabetic retinopathy. Diabetes 2013, 62, 1808–1815.
  25. Stitt, A.W.; Li, Y.M.; Gardiner, T.A.; Bucala, R.; Archer, D.B.; Vlassara, H. Advanced glycation end products (AGEs) co-localize with AGE receptors in the retinal vasculature of diabetic and of AGE-infused rats. Am. J. Pathol. 1997, 150, 523–531.
  26. Wu, M.Y.; Yiang, G.T.; Lai, T.T.; Li, C.J. The Oxidative Stress and Mitochondrial Dysfunction during the Pathogenesis of Diabetic Retinopathy. Oxidative Med. Cell. Longev. 2018, 2018, 3420187.
  27. Yamagishi, S.; Nakamura, K.; Matsui, T.; Inagaki, Y.; Takenaka, K.; Jinnouchi, Y.; Yoshida, Y.; Matsuura, T.; Narama, I.; Motomiya, Y.; et al. Pigment epithelium-derived factor inhibits advanced glycation end product-induced retinal vascular hyperpermeability by blocking reactive oxygen species-mediated vascular endothelial growth factor expression. J. Biol. Chem. 2006, 281, 20213–20220.
  28. Chen, L.; Cui, Y.; Li, B.; Weng, J.; Wang, W.; Zhang, S.; Huang, X.; Guo, X.; Huang, Q. Advanced glycation end products induce immature angiogenesis in in vivo and ex vivo mouse models. Am. J. Physiol. Heart Circ. Physiol. 2020, 318, H519–H533.
  29. Takino, J.I.; Sato, T.; Kanetaka, T.; Okihara, K.; Nagamine, K.; Takeuchi, M.; Hori, T. RasGRP2 inhibits glyceraldehyde-derived toxic advanced glycation end-products from inducing permeability in vascular endothelial cells. Sci. Rep. 2021, 11, 2959.
  30. Photocoagulation treatment of proliferative diabetic retinopathy. Clinical application of Diabetic Retinopathy Study (DRS) findings, DRS Report Number 8. The Diabetic Retinopathy Study Research Group. Ophthalmology 1981, 88, 583–600.
  31. Ying, L.; Shen, Y.; Zhang, Y.; Wang, Y.; Liu, Y.; Yin, J.; Wang, Y.; Yin, J.; Zhu, W.; Bao, Y.; et al. Association of advanced glycation end products with diabetic retinopathy in type 2 diabetes mellitus. Diabetes Res. Clin. Pract. 2021, 177, 108880.
  32. Hammes, H.P.; Martin, S.; Federlin, K.; Geisen, K.; Brownlee, M. Aminoguanidine treatment inhibits the development of experimental diabetic retinopathy. Proc. Natl. Acad. Sci. USA 1991, 88, 11555–11558.
  33. Maisonpierre, P.C.; Suri, C.; Jones, P.F.; Bartunkova, S.; Wiegand, S.J.; Radziejewski, C.; Compton, D.; McClain, J.; Aldrich, T.H.; Papadopoulos, N.; et al. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 1997, 277, 55–60.
  34. Athyros, V.G.; Doumas, M.; Imprialos, K.P.; Stavropoulos, K.; Georgianou, E.; Katsimardou, A.; Karagiannis, A. Diabetes and lipid metabolism. Hormones 2018, 17, 61–67.
  35. Busik, J.V. Lipid metabolism dysregulation in diabetic retinopathy. J. Lipid Res. 2021, 62, 100017.
  36. Kumar, B.; Kowluru, A.; Kowluru, R.A. Lipotoxicity augments glucotoxicity-induced mitochondrial damage in the development of diabetic retinopathy. Investig. Ophthalmol. Vis. Sci. 2015, 56, 2985–2992.
  37. Fritzen, A.M.; Lundsgaard, A.M.; Kiens, B. Tuning fatty acid oxidation in skeletal muscle with dietary fat and exercise. Nat. Rev. Endocrinol. 2020, 16, 683–696.
  38. Eid, S.; Sas, K.M.; Abcouwer, S.F.; Feldman, E.L.; Gardner, T.W.; Pennathur, S.; Fort, P.E. New insights into the mechanisms of diabetic complications: Role of lipids and lipid metabolism. Diabetologia 2019, 62, 1539–1549.
  39. Obrosova, I.G. Increased sorbitol pathway activity generates oxidative stress in tissue sites for diabetic complications. Antioxid. Redox Signal. 2005, 7, 1543–1552.
  40. Tserng, K.Y.; Griffin, R. Quantitation and molecular species determination of diacylglycerols, phosphatidylcholines, ceramides, and sphingomyelins with gas chromatography. Anal. Biochem. 2003, 323, 84–93.
  41. Fugio, L.B.; Coeli-Lacchini, F.B.; Leopoldino, A.M. Sphingolipids and Mitochondrial Dynamic. Cells 2020, 9, 581.
  42. Castro, B.M.; Prieto, M.; Silva, L.C. Ceramide: A simple sphingolipid with unique biophysical properties. Prog. Lipid Res. 2014, 54, 53–67.
  43. van Blitterswijk, W.J.; van der Luit, A.H.; Veldman, R.J.; Verheij, M.; Borst, J. Ceramide: Second messenger or modulator of membrane structure and dynamics? Biochem. J. 2003, 369, 199–211.
  44. Wang, A.; Li, C.; Liao, J.; Dong, M.; Xiao, Z.; Lei, M. Ceramide mediates inhibition of the Akt/eNOS pathway by high levels of glucose in human vascular endothelial cells. J. Pediatr. Endocrinol. Metab. 2013, 26, 31–38.
  45. Levitsky, Y.; Hammer, S.S.; Fisher, K.P.; Huang, C.; Gentles, T.L.; Pegouske, D.J.; Xi, C.; Lydic, T.A.; Busik, J.V.; Proshlyakov, D.A. Mitochondrial Ceramide Effects on the Retinal Pigment Epithelium in Diabetes. Int J. Mol. Sci. 2020, 21, 3830.
  46. Chouchani, E.T.; Pell, V.R.; Gaude, E.; Aksentijević, D.; Sundier, S.Y.; Robb, E.L.; Logan, A.; Nadtochiy, S.M.; Ord, E.N.J.; Smith, A.C.; et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature 2014, 515, 431–435.
  47. Chipuk, J.E.; McStay, G.P.; Bharti, A.; Kuwana, T.; Clarke, C.J.; Siskind, L.J.; Obeid, L.M.; Green, D.R. Sphingolipid metabolism cooperates with BAK and BAX to promote the mitochondrial pathway of apoptosis. Cell 2012, 148, 988–1000.
  48. Colombini, M. Ceramide channels and mitochondrial outer membrane permeability. J. Bioenerget. Biomembr. 2017, 49, 57–64.
  49. Alfarhan, M.; Jafari, E.; Narayanan, S.P. Acrolein: A Potential Mediator of Oxidative Damage in Diabetic Retinopathy. Biomolecules 2020, 10, 1579.
  50. Zhang, X.; Lai, Y.; McCance, D.R.; Uchida, K.; McDonald, D.M.; Gardiner, T.A.; Stitt, A.W.; Curtis, T.M. Evaluation of N (epsilon)-(3-formyl-3,4-dehydropiperidino)lysine as a novel biomarker for the severity of diabetic retinopathy. Diabetologia 2008, 51, 1723–1730.
  51. Noda, K.; Murata, M.; Saito, W.; Kanda, A.; Ishida, S. Accumulation of Acrolein-conjugated Protein in the Vitreous Fluid of Proliferative Diabetic Retinopathy. Investig. Ophthalmol. Vis. Sci. 2017, 58, 2514.
  52. Yong, P.H.; Zong, H.; Medina, R.J.; Limb, G.A.; Uchida, K.; Stitt, A.W.; Curtis, T.M. Evidence supporting a role for N-(3-formyl-3,4-dehydropiperidino)lysine accumulation in Muller glia dysfunction and death in diabetic retinopathy. Mol. Vis. 2010, 16, 2524–2538.
  53. Alfarhan, M.; Liu, F.; Shan, S.; Pichavaram, P.; Somanath, P.R.; Narayanan, S.P. Pharmacological Inhibition of Spermine Oxidase Suppresses Excitotoxicity Induced Neuroinflammation in Mouse Retina. Int. J. Mol. Sci. 2022, 23, 2133.
  54. Stevens, J.F.; Maier, C.S. Acrolein: Sources, metabolism, and biomolecular interactions relevant to human health and disease. Mol. Nutr. Food Res. 2008, 52, 7–25.
  55. Goncalves, L.C.P.; Massari, J.; Licciardi, S.; Prado, F.M.; Linares, E.; Klassen, A.; Tavares, M.F.M.; Augusto, O.; Di Mascio, P.; Bechara, E.J.H. Singlet oxygen generation by the reaction of acrolein with peroxynitrite via a 2-hydroxyvinyl radical intermediate. Free Radic. Biol. Med. 2020, 152, 83–90.
  56. Jia, L.; Liu, Z.; Sun, L.; Miller, S.S.; Ames, B.N.; Cotman, C.W.; Liu, J. Acrolein, a toxicant in cigarette smoke, causes oxidative damage and mitochondrial dysfunction in RPE cells: Protection by (R)-alpha-lipoic acid. Investig. Ophthalmol. Vis. Sci. 2007, 48, 339–348.
  57. Liu, Z.; Sun, L.; Zhu, L.; Jia, X.; Li, X.; Jia, H.; Wang, Y.; Weber, P.; Long, J.; Liu, J. Hydroxytyrosol protects retinal pigment epithelial cells from acrolein-induced oxidative stress and mitochondrial dysfunction. J. NeuroChem. 2007, 103, 2690–2700.
  58. Fu, D.; Wu, M.; Zhang, J.; Du, M.; Yang, S.; Hammad, S.M.; Wilson, K.; Chen, J.; Lyons, T.J. Mechanisms of modified LDL-induced pericyte loss and retinal injury in diabetic retinopathy. Diabetologia 2012, 55, 3128–3140.
  59. Wu, W.; Jing, D.; Huang, X.; Yang, W.; Shao, Z. Drp1-mediated mitochondrial fission is involved in oxidized low-density lipoprotein-induced AF cella poptosis. J. Orthop. Res. 2021, 39, 1496–1504.
  60. Ling, C.; Ronn, T. Epigenetics in Human Obesity and Type 2 Diabetes. Cell Metab. 2019, 29, 1028–1044.
  61. Hall, E.; Dayeh, T.; Kirkpatrick, C.L.; Wollheim, C.B.; Dekker Nitert, M.; Ling, C. DNA methylation of the glucagon-like peptide 1 receptor (GLP1R) in human pancreatic islets. BMC Med. Genet. 2013, 14, 76.
  62. Ling, C.; Del Guerra, S.; Lupi, R.; Ronn, T.; Granhall, C.; Luthman, H.; Masiello, P.; Marchetti, P.; Groop, L.; Del Prato, S. Epigenetic regulation of PPARGC1A in human type 2 diabetic islets and effect on insulin secretion. Diabetologia 2008, 51, 615–622.
  63. Yang, B.T.; Dayeh, T.A.; Kirkpatrick, C.L.; Taneera, J.; Kumar, R.; Groop, L.; Wollheim, C.B.; Nitert, M.D.; Ling, C. Insulin promoter DNA methylation correlates negatively with insulin gene expression and positively with HbA(1c) levels in human pancreatic islets. Diabetologia 2011, 54, 360–367.
  64. Yang, B.T.; Dayeh, T.A.; Volkov, P.A.; Kirkpatrick, C.L.; Malmgren, S.; Jing, X.; Renstrom, E.; Wollheim, C.B.; Nitert, M.D.; Ling, C. Increased DNA methylation and decreased expression of PDX-1 in pancreatic islets from patients with type 2 diabetes. Mol. Endocrinol. 2012, 26, 1203–1212.
  65. Abderrahmani, A.; Yengo, L.; Caiazzo, R.; Canouil, M.; Cauchi, S.; Raverdy, V.; Plaisance, V.; Pawlowski, V.; Lobbens, S.; Maillet, J.; et al. Increased Hepatic PDGF-AA Signaling Mediates Liver Insulin Resistance in Obesity-Associated Type 2 Diabetes. Diabetes 2018, 67, 1310–1321.
  66. Nilsson, E.; Jansson, P.A.; Perfilyev, A.; Volkov, P.; Pedersen, M.; Svensson, M.K.; Poulsen, P.; Ribel-Madsen, R.; Pedersen, N.L.; Almgren, P.; et al. Altered DNA methylation and differential expression of genes influencing metabolism and inflammation in adipose tissue from subjects with type 2 diabetes. Diabetes 2014, 63, 2962–2976.
  67. Nilsson, E.; Matte, A.; Perfilyev, A.; de Mello, V.D.; Kakela, P.; Pihlajamaki, J.; Ling, C. Epigenetic Alterations in Human Liver From Subjects With Type 2 Diabetes in Parallel With Reduced Folate Levels. J. Clin. Endocrinol. Metab. 2015, 100, E1491–E1501.
  68. Nitert, M.D.; Dayeh, T.; Volkov, P.; Elgzyri, T.; Hall, E.; Nilsson, E.; Yang, B.T.; Lang, S.; Parikh, H.; Wessman, Y.; et al. Impact of an exercise intervention on DNA methylation in skeletal muscle from first-degree relatives of patients with type 2 diabetes. Diabetes 2012, 61, 3322–3332.
  69. Kirchner, H.; Sinha, I.; Gao, H.; Ruby, M.A.; Schonke, M.; Lindvall, J.M.; Barres, R.; Krook, A.; Naslund, E.; Dahlman-Wright, K.; et al. Altered DNA methylation of glycolytic and lipogenic genes in liver from obese and type 2 diabetic patients. Mol. Metab. 2016, 5, 171–183.
  70. Ribel-Madsen, R.; Fraga, M.F.; Jacobsen, S.; Bork-Jensen, J.; Lara, E.; Calvanese, V.; Fernandez, A.F.; Friedrichsen, M.; Vind, B.F.; Hojlund, K.; et al. Genome-wide analysis of DNA methylation differences in muscle and fat from monozygotic twins discordant for type 2 diabetes. PLoS ONE 2012, 7, e51302.
  71. Hall, E.; Dekker Nitert, M.; Volkov, P.; Malmgren, S.; Mulder, H.; Bacos, K.; Ling, C. The effects of high glucose exposure on global gene expression and DNA methylation in human pancreatic islets. Mol. Cell. Endocrinol. 2018, 472, 57–67.
  72. Kowluru, R.A. Retinopathy in a Diet-Induced Type 2 Diabetic Rat Model and Role of Epigenetic Modifications. Diabetes 2020, 69, 689–698.
  73. Sahajpal, N.; Kowluru, A.; Kowluru, R.A. The Regulatory Role of Rac1, a Small Molecular Weight GTPase, in the Development of Diabetic Retinopathy. J. Clin. Med. 2019, 8, 965.
  74. Hu, C.; Wu, Z.; Huang, Z.; Hao, X.; Wang, S.; Deng, J.; Yin, Y.; Tan, C. Nox2 impairs VEGF-A-induced angiogenesis in placenta via mitochondrial ROS-STAT3 pathway. Redox Biol. 2021, 45, 102051.
  75. Zhong, Q.; Mishra, M.; Kowluru, R.A. Transcription factor Nrf2-mediated antioxidant defense system in the development of diabetic retinopathy. Investig. Ophthalmol. Vis. Sci. 2013, 54, 3941–3948.
  76. Mishra, M.; Zhong, Q.; Kowluru, R.A. Epigenetic modifications of Nrf2-mediated glutamate-cysteine ligase: Implications for the development of diabetic retinopathy and the metabolic memory phenomenon associated with its continued progression. Free Radic. Biol. Med. 2014, 75, 129–139.
  77. Mishra, M.; Zhong, Q.; Kowluru, R.A. Epigenetic modifications of Keap1 regulate its interaction with the protective factor Nrf2 in the development of diabetic retinopathy. Investig. Ophthalmol. Vis. Sci. 2014, 55, 7256–7265.
  78. Zhong, Q.; Kowluru, R.A. Epigenetic modification of Sod2 in the development of diabetic retinopathy and in the metabolic memory: Role of histone methylation. Investig. Ophthalmol. Vis. Sci. 2013, 54, 244–250.
  79. Kowluru, R.A.; Kowluru, A.; Mishra, M.; Kumar, B. Oxidative stress and epigenetic modifications in the pathogenesis of diabetic retinopathy. Prog. Retin. Eye Res. 2015, 48, 40–61.
  80. Kowluru, R.A.; Mishra, M. Oxidative stress, mitochondrial damage and diabetic retinopathy. Biochim. Biophys. Acta 2015, 1852, 2474–2483.
  81. Kowluru, R.A.; Mohammad, G. Epigenetics and Mitochondrial Stability in the Metabolic Memory Phenomenon Associated with Continued Progression of Diabetic Retinopathy. Sci. Rep. 2020, 10, 6655.
  82. D’Aquila, P.; De Rango, F.; Guarasci, F.; Mandalà, M.; Corsonello, A.; Bellizzi, D.; Passarino, G. Multi-Tissue DNA Methylation Remodeling at Mitochondrial Quality Control Genes According to Diet in Rat Aging Models. Nutrients 2020, 12, 460.
  83. Mohammad, G.; Radhakrishnan, R.; Kowluru, R.A. Epigenetic Modifications Compromise Mitochondrial DNA Quality Control in the Development of Diabetic Retinopathy. Investig. Ophthalmol. Vis. Sci. 2019, 60, 3943–3951.
  84. Rashid, S.; Freitas, M.O.; Cucchi, D.; Bridge, G.; Yao, Z.; Gay, L.; Williams, M.; Wang, J.; Suraweera, N.; Silver, A.; et al. MLH1 deficiency leads to deregulated mitochondrial metabolism. Cell Death Dis. 2019, 10, 795.
  85. Stoccoro, A.; Coppede, F. Mitochondrial DNA Methylation and Human Diseases. Int. J. Mol. Sci. 2021, 22, 4594.
  86. Liu, B.; Du, Q.; Chen, L.; Fu, G.; Li, S.; Fu, L.; Zhang, X.; Ma, C.; Bin, C. CpG methylation patterns of human mitochondrial DNA. Sci. Rep. 2016, 6, 23421.
  87. Shock, L.S.; Thakkar, P.V.; Peterson, E.J.; Moran, R.G.; Taylor, S.M. DNA methyltransferase 1, cytosine methylation, and cytosine hydroxymethylation in mammalian mitochondria. Proc. Natl. Acad. Sci. USA 2011, 108, 3630–3635.
  88. Mishra, M.; Kowluru, R.A. Epigenetic Modification of Mitochondrial DNA in the Development of Diabetic Retinopathy. Investig Ophthalmol. Vis. Sci. 2015, 56, 5133–5142.
  89. Mishra, M.; Kowluru, R.A. DNA Methylation-a Potential Source of Mitochondria DNA Base Mismatch in the Development of Diabetic Retinopathy. Mol. Neurobiol. 2019, 56, 88–101.
  90. Kowluru, R.A.; Mohammad, G.; dos Santos, J.M.; Zhong, Q. Abrogation of MMP-9 gene protects against the development of retinopathy in diabetic mice by preventing mitochondrial damage. Diabetes 2011, 60, 3023–3033.
  91. Kowluru, R.A.; Santos, J.M.; Zhong, Q. Sirt1, a negative regulator of matrix metalloproteinase-9 in diabetic retinopathy. Investig. Ophthalmol. Vis. Sci. 2014, 55, 5653–5660.
  92. Zhong, Q.; Kowluru, R.A. Regulation of matrix metalloproteinase-9 by epigenetic modifications and the development of diabetic retinopathy. Diabetes 2013, 62, 2559–2568.
  93. Kowluru, R.A.; Shan, Y.; Mishra, M. Dynamic DNA methylation of matrix metalloproteinase-9 in the development of diabetic retinopathy. Lab. Investig. 2016, 96, 1040–1049.
  94. Onishi, M.; Yamano, K.; Sato, M.; Matsuda, N.; Okamoto, K. Molecular mechanisms and physiological functions of mitophagy. EMBO J. 2021, 40, e104705.
  95. Quinn, P.M.J.; Moreira, P.I.; Ambrosio, A.F.; Alves, C.H. PINK1/PARKIN signalling in neurodegeneration and neuroinflammation. Acta Neuropathol. Commun. 2020, 8, 189.
  96. Yao, R.Q.; Ren, C.; Xia, Z.F.; Yao, Y.M. Organelle-specific autophagy in inflammatory diseases: A potential therapeutic target underlying the quality control of multiple organelles. Autophagy 2021, 17, 385–401.
  97. Hombrebueno, J.R.; Cairns, L.; Dutton, L.R.; Lyons, T.J.; Brazil, D.P.; Moynagh, P.; Curtis, T.M.; Xu, H. Uncoupled turnover disrupts mitochondrial quality control in diabetic retinopathy. JCI Insight 2019, 4, e129760.
  98. Devi, T.S.; Somayajulu, M.; Kowluru, R.A.; Singh, L.P. TXNIP regulates mitophagy in retinal Muller cells under high-glucose conditions: Implications for diabetic retinopathy. Cell Death Dis 2017, 8, e2777.
  99. Zhou, P.; Xie, W.; Meng, X.; Zhai, Y.; Dong, X.; Zhang, X.; Sun, G.; Sun, X. Notoginsenoside R1 Ameliorates Diabetic Retinopathy through PINK1-Dependent Activation of Mitophagy. Cells 2019, 8, 213.
  100. Taki, K.; Horie, T.; Kida, T.; Mimura, M.; Ikeda, T.; Oku, H. Impairment of Autophagy Causes Superoxide Formation and Caspase Activation in 661 W Cells, a Cell Line for Cone Photoreceptors, under Hyperglycemic Conditions. Int. J. Mol. Sci. 2020, 21, 4240.
  101. Zhang, M.Y.; Zhu, L.; Zheng, X.; Xie, T.H.; Wang, W.; Zou, J.; Li, Y.; Li, H.Y.; Cai, J.; Gu, S.; et al. TGR5 Activation Ameliorates Mitochondrial Homeostasis via Regulating the PKCdelta/Drp1-HK2 Signaling in Diabetic Retinopathy. Front. Cell Dev. Biol. 2021, 9, 759421.
  102. Huang, L.; Yao, T.; Chen, J.; Zhang, Z.; Yang, W.; Gao, X.; Dan, Y.; He, Y. Effect of Sirt3 on retinal pigment epithelial cells in high glucose through Foxo3a/PINK1-Parkin pathway mediated mitophagy. Exp. Eye Res. 2022, 218, 109015.
  103. Qiao, S.; Dennis, M.; Song, X.; Vadysirisack, D.D.; Salunke, D.; Nash, Z.; Yang, Z.; Liesa, M.; Yoshioka, J.; Matsuzawa, S.; et al. A REDD1/TXNIP pro-oxidant complex regulates ATG4B activity to control stress-induced autophagy and sustain exercise capacity. Nat. Commun. 2015, 6, 7014.
  104. Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709.
  105. Sharma, I.; Yadav, K.S.; Mugale, M.N. Redoxisome and diabetic retinopathy: Pathophysiology and therapeutic interventions. Pharm. Res. 2022, 182, 106292.
  106. Mohammad, G.; Kowluru, R.A. Mitochondria fission and diabetic retinopathy: Role of Drp1. Investig. Ophthalmol. Vis. Sci. 2021, 62, 2248.
  107. Zhang, M.Y.; Zhu, L.; Bao, X.; Xie, T.H.; Cai, J.; Zou, J.; Wang, W.; Gu, S.; Li, Y.; Li, H.Y.; et al. Inhibition of Drp1 ameliorates diabetic retinopathy by regulating mitochondrial homeostasis. Exp. Eye Res. 2022, 220, 109095.
  108. Kang, R.; Livesey, K.M.; Zeh, H.J.; Loze, M.T.; Tang, D. HMGB1: A novel Beclin 1-binding protein active in autophagy. Autophagy 2010, 6, 1209–1211.
  109. Li, X.; Zhang, M.; Zhou, H. The morphological features and mitochondrial oxidative stress mechanism of the retinal neurons apoptosis in early diabetic rats. J. Diabetes Res. 2014, 2014, 678123.
  110. de Souza, B.M.; Brondani, L.A.; Bouças, A.P.; Sortica, D.A.; Kramer, C.K.; Canani, L.H.; Leitão, C.B.; Crispim, D. Associations between UCP1 -3826A/G, UCP2 -866G/A, Ala55Val and Ins/Del, and UCP3 -55C/T polymorphisms and susceptibility to type 2 diabetes mellitus: Case-control study and meta-analysis. PLoS ONE 2013, 8, e54259.
  111. Kowluru, R.A.; Mishra, M.; Kumar, B. Diabetic retinopathy and transcriptional regulation of a small molecular weight G-Protein, Rac1. Exp. Eye Res. 2016, 147, 72–77.
  112. Madsen-Bouterse, S.A.; Mohammad, G.; Kanwar, M.; Kowluru, R.A. Role of mitochondrial DNA damage in the development of diabetic retinopathy, and the metabolic memory phenomenon associated with its progression. Antioxid. Redox Signal. 2010, 13, 797–805.
  113. Mohammad, G.; Kowluru, R.A. Novel role of mitochondrial matrix metalloproteinase-2 in the development of diabetic retinopathy. Investig. Ophthalmol. Vis. Sci. 2011, 52, 3832–3841.
  114. Kowluru, R.A. Mitochondria damage in the pathogenesis of diabetic retinopathy and in the metabolic memory associated with its continued progression. Curr. Med. Chem. 2013, 20, 3226–3233.
  115. Mustapha, N.M.; Tarr, J.M.; Kohner, E.M.; Chibber, R. NADPH Oxidase versus Mitochondria-Derived ROS in Glucose-Induced Apoptosis of Pericytes in Early Diabetic Retinopathy. J. Ophthalmol. 2010, 2010, 746978.
  116. Ghanian, Z.; Mehrvar, S.; Jamali, N.; Sheibani, N.; Ranji, M. Time-lapse microscopy of oxidative stress demonstrates metabolic sensitivity of retinal pericytes under high glucose condition. J. Biophotonics 2018, 11, e201700289.
  117. Santiago, A.R.; Cristovao, A.J.; Santos, P.F.; Carvalho, C.M.; Ambrosio, A.F. High glucose induces caspase-independent cell death in retinal neural cells. Neurobiol. Dis. 2007, 25, 464–472.
  118. Kim, Y.; Lee, J.H.; Park, J.E.; Cho, J.; Yi, H.; Kim, V.N. PKR is activated by cellular dsRNAs during mitosis and acts as a mitotic regulator. Genes Dev. 2014, 28, 1310–1322.
  119. Silva, V.A.O.; Andre, N.D.; Sousa, T.A.E.; Alves, V.M.; Kettelhut, I.D.C.; De Lucca, F.L. Nuclear PKR in retinal neurons in the early stage of diabetic retinopathy in streptozotocininduced diabetic rats. Mol. Med. Rep. 2021, 24, 614.
  120. Zhai, J.; Li, Z.; Zhang, H.; Ma, L.; Ma, Z.; Zhang, Y.; Zou, J.; Li, M.; Ma, L.; Wang, X.; et al. Berberine protects against diabetic retinopathy by inhibiting cell apoptosis via deactivation of the NFkappaB signaling pathway. Mol. Med. Rep. 2020, 22, 4227–4235.
  121. Mishra, M.; Kowluru, R.A. Role of PARP-1 as a novel transcriptional regulator of MMP-9 in diabetic retinopathy. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1761–1769.
  122. Mohammad, G.; Kowluru, R.A. Matrix metalloproteinase-2 in the development of diabetic retinopathy and mitochondrial dysfunction. Lab. Investig. 2010, 90, 1365–1372.
  123. Liu, G.; Wu, F.; Wu, H.; Wang, Y.; Jiang, X.; Hu, P.; Tong, X. Inactivation of cysteine 674 in the sarcoplasmic/endoplasmic reticulum calcium ATPase 2 causes retinopathy in the mouse. Exp. Eye Res. 2021, 207, 108559.
  124. Li, J.; Wang, P.; Yu, S.; Zheng, Z.; Xu, X. Calcium entry mediates hyperglycemia-induced apoptosis through Ca(2+)/calmodulin-dependent kinase II in retinal capillary endothelial cells. Mol. Vis. 2012, 18, 2371–2379.
  125. Roy, S.; Kim, D.; Sankaramoorthy, A. Mitochondrial Structural Changes in the Pathogenesis of Diabetic Retinopathy. J. Clin. Med. 2019, 8, 1363.
  126. Xie, J.; Cui, Y.; Chen, X.; Yu, H.; Chen, J.; Huang, T.; He, M.; Zhang, L.; Meng, Q. VDAC1 regulates mitophagy in NLRP3 inflammasome activation in retinal capillary endothelial cells under high-glucose conditions. Exp. Eye Res. 2021, 209, 108640.
  127. Kim, D.; Roy, S. Effects of Diabetes on Mitochondrial Morphology and Its Implications in Diabetic Retinopathy. Investig. Ophthalmol. Vis. Sci. 2020, 61, 10.
  128. Trudeau, K.; Muto, T.; Roy, S. Downregulation of mitochondrial connexin 43 by high glucose triggers mitochondrial shape change and cytochrome C release in retinal endothelial cells. Investig. Ophthalmol. Vis. Sci. 2012, 53, 6675–6681.
  129. Duraisamy, A.J.; Mohammad, G.; Kowluru, R.A. Mitochondrial fusion and maintenance of mitochondrial homeostasis in diabetic retinopathy. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 1617–1626.
  130. Wu, H.; Li, G.; Chen, W.; Luo, W.; Yang, Z.; You, Z.; Zou, Y. Drp1 knockdown represses apoptosis of rat retinal endothelial cells by inhibiting mitophagy. Acta HistoChem. 2022, 124, 151837.
  131. Kim, D.; Sankaramoorthy, A.; Roy, S. Downregulation of Drp1 and Fis1 Inhibits Mitochondrial Fission and Prevents High Glucose-Induced Apoptosis in Retinal Endothelial Cells. Cells 2020, 9, 1662.
More
Information
Subjects: Ophthalmology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 319
Revisions: 3 times (View History)
Update Date: 29 Nov 2022
1000/1000
Video Production Service