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 The entry describes physiological and pathological functions of PPARα in the eye. Results of clinical trials may be added in the future. + 1321 word(s) 1321 2020-11-10 06:49:45 |
2 I deleted the note on the tope of the Content. -27 word(s) 1294 2020-11-10 08:36:39 | |
3 format correct Meta information modification 1294 2020-11-10 08:55:16 | |
4 Configuration improvements and one more checking + 228 word(s) 1522 2020-11-10 10:09:22 |

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.
Tomita, Y.; Lee, D.; Tsubota, K.; Kurihara, T. PPARα and Ocular Dieseases. Encyclopedia. Available online: https://encyclopedia.pub/entry/2951 (accessed on 26 April 2024).
Tomita Y, Lee D, Tsubota K, Kurihara T. PPARα and Ocular Dieseases. Encyclopedia. Available at: https://encyclopedia.pub/entry/2951. Accessed April 26, 2024.
Tomita, Yohei, Deokho Lee, Kazuo Tsubota, Toshihide Kurihara. "PPARα and Ocular Dieseases" Encyclopedia, https://encyclopedia.pub/entry/2951 (accessed April 26, 2024).
Tomita, Y., Lee, D., Tsubota, K., & Kurihara, T. (2020, November 10). PPARα and Ocular Dieseases. In Encyclopedia. https://encyclopedia.pub/entry/2951
Tomita, Yohei, et al. "PPARα and Ocular Dieseases." Encyclopedia. Web. 10 November, 2020.
PPARα and Ocular Dieseases
Edit

Mounting evidence suggests that peroxisome proliferator-activator receptor alpha (PPARα) activation can be a  therapeutic target for various ocular diseases including diabetic retinopathy (DR). Here we describe functions of PPARα in the eye contributing to ocular homeostasis.

diabetic retinopathy PPARα SPPARMα HIF-1α

1. Introduction

Diabetic retinopathy (DR) is a complication of diabetes that affects the eyes in subjects with type 1 or type 2 diabetes mellitus [1]. DR develops because of a chronic abnormality of glycemic control [2]. In detail, DR progresses from an initial stage where high blood glucose levels damage the microvasculature [3]. Then, microvascular irregularities including hemorrhage, ischemia, and microaneurysms bring retinal neovascularization [3]. Abnormal vasculatures by retinal neovascularization lead to severe hypoxic conditions in the retina of the eye [3]. At the final stage, fibrovascular proliferation resulting in tractional retinal detachment by chronic severe hypoxic conditions causes vision loss [3]. However, recent evidence indicates that pathogenesis of DR contains far more complex mechanisms that are the involvement of multiple interlinked alterations via impairment of crosstalk between retinal neurons, glial cells, and vasculatures [4][5][6][7]. Emerging studies have demonstrated that neurons and glial cells in the central nervous system act as oxygen sensors and vascular regulators to interact with vascular cells for neurovascular homeostasis [8], and impairment of their crosstalk damages neurovascular homeostasis [8][9]. Although exact mechanisms have not been completely defined, possible major factors such as mitochondrial oxidative stress, inflammation, production of advanced glycation end products (AGEs), and activation of the protein kinase C (PKC) pathway have been proposed (along with the traditional concept of retinal neovascularization by hyperglycemia-induced microvascular irregularities) for the comprehensive pathogenesis of DR [10][11].

PPARα is from a nuclear receptor PPAR family (PPARα, PPARδ, and PPARγ), which regulates expressions of several genes affecting lipid and carbohydrate metabolism [12]. PPARα is named based on its ability to be activated by peroxisome proliferator chemicals, and it is the first member to be cloned among the PPAR isotypes [13]. PPARα is expressed in various types of cells in the skeletal muscle, heart, liver, brown adipose, kidney, intestinal mucosa, adrenal gland, eye, and most cell types present in the vasculature including endothelial cells, smooth muscle cells, monocytes, and macrophages [14][15][16][17][18][19][20][21]. PPARα activation was found to increase circulating levels of high-density lipoprotein cholesterol and decrease serum levels of triglycerides, free fatty acids and apolipoprotein, which improves an overall serum lipid profile and finally exerts positive effects on inflammation and insulin resistance [22][23][24]. Increasing evidence suggests that PPARα activation can be a strong therapeutic target for various types of diseases such as cardiovascular diseases [25], dyslipidemia [26], and diabetes and its complications including DR [22]. However, its molecular mechanisms are far from being elucidated. In this paper, we review the therapeutic effects of PPARα agonists as a promising approach for the treatment of DR.

2. Functions of PPARα in the Eye

Experimental evidence indicates that PPARα is expressed in various tissues in diabetic microvascular diseases—the retina as well as kidney and nerve [20][21]. PPARα has been spotlighted as its expression levels decreased in the retinas with diabetes [27][28]. Decreased PPARα expression has been found to contribute to retinal inflammation and neovascularization, and pharmacological activation of PPARα has been shown to exert therapeutic effects against various ocular degenerative disorders [28][29][30]. A previous study showed that more severe retinal acellular capillary formation and pericyte dropout were observed in PPARα−/− mice with diabetes, compared with those in diabetic wild-type mice [29]. Another study demonstrated that retinal neurodegeneration was exacerbated in PPARα−/− mice with diabetes in comparison with that in diabetic wild-type mice [31]. Multiple proteomics data indicated that several oxidative stress markers such as Gstm1 (glutathione-s-transferase m1), Prdx6 (peroxidase 6) and Txnrd1 (thioredoxin reductase 1) increased in diabetic retinas and that there were further increases in diabetic PPARα−/− retinas [31]. This implies that oxidative stress may become worsened by PPARα ablation in DR. PPARα activation increased retinal NADH (nicotinamide adenine dinucleotide + hydrogen) oxidation in diabetic mice, and treatment of fenofibric acid, an active metabolite of fenofibrate, reduced mitochondrial oxidative stress and cell death in retinal neuronal cell lines under 4-hydroxynonenal (4-HNE)-induced oxidative stress conditions [31]. This implies mitochondrial dysfunction by oxidative stress could be restored by PPARα activation.

In terms of an ischemic model other than the diabetic model, PPARα−/− mice with a laser-induced choroidal neovascularization (CNV) developed more severe CNV compared with wild-type CNV mice [32]. PPARα−/− mice with oxygen-induced retinopathy (OIR) also showed deleterious effects (such as increased retinal cell death and glial activation) in comparison with wild-type OIR mice [33]. Overexpression of PPARα using an adenovirus system attenuated increased endothelial progenitor cell circulation in OIR mice through inhibition of the hypoxia-inducible factor (HIF)-1α pathway, and mouse brain endothelial cells from PPARα−/− mice showed prominent activation of HIF-1α induced by hypoxia, compared with wild-type mouse brain endothelial cells [34]. This suggests anti-angiogenic effects of PPARα via HIF-1α inhibition, a novel protective molecular mechanism against retinal ischemic conditions.

Pharmacological PPARα activation by palmitoylethanolamide (PEA) reduced retinal neovascularization and fibrotic changes and suppressed glial activation in proliferative retinopathy and neovascular age-related macular degeneration mouse models [35]. In cardiovascular studies, PEA exerted direct vaso-relaxation of the bovine ophthalmic artery by using PPARα transcription factors, which suggests a role of PPARα on physiological vascular regulation [36]. This vaso-relaxing effect could increase supply of oxygen to the retina and prevent ischemic lesions, as observed in patients with ocular hypertension [37]. Another study demonstrated that administration of PEA showed enhancement of aqueous humor outflow facility and this effect appeared to be mediated partially by the involvement of PPARα [38]. Those studies imply PPARα modulation could also be a promising therapeutic target for glaucoma [39].

Even though there are not many reports available on roles of PPARα in the ocular surface, recent evidence suggests that PPARα may play a critical role in regulation of inflammatory processes in the ocular surface [40]. Fenofibrate ameliorated a severity of ocular surface squamous metaplasia, commonly seen in patients with long-term deficiency of tear film [41] and suppressed formation of tear film instability via inhibition of macrophages and downregulation of pro-inflammatory factors [40]. In the corneal epithelium of mice with dry eyes by sleep deprivation, downregulation in PPARα expression was detected [42], and fenofibrate increased PPARα expression in cultured corneal epithelium sheets and restored a microvilli morphology [42]. This implies PPARα activation could have potential for use as a preventive agent in patients with a high risk of the dry eye. Other cornea studies indicated therapeutic roles of PPARα agonists against corneal inflammation and neovascularization [43][44]. Corneal neovascularization is closely related to a reduction in corneal transparency which is important for a visual acuity [45][46]. In rat corneal alkali burn models, corneal neovascularization was seen and a topical injection of fenofibrate suppressed its neovascularization through upregulation of PPARα mRNA expression and suppression of Il-6, Il-1β, Vegf and Ang-2 mRNA expressions [43][44].

The above, taken together as accumulating evidence, supports the concept that it may be important to restore or boost PPARα expression for prevention of various ocular diseases. However, studies on downstream signaling effector molecules regarding PPARα activation need to be further unraveled.

3. Selective PPARα modulatorα (SPPARMα) 

Pemafibrate is a novel selective PPARα modulator (SPPARMα) and has higher potency and selectivity for the activation of PPARα than other PPARα agonists, especially fenofibrate [43][47][48][49]. Several studies have shown that pemafibrate has therapeutic effects on retinal diseases [50][51][52]. One report showed that oral administration of pemafibrate increased plasma fibroblast growth factor 21 (FGF21) levels in OIR and inhibited retinal neovascularization through inhibition of HIF-1α and Vegf expressions [51]. In addition, long-acting FGF21 inhibited HIF activity in a photoreceptor cell line under hypoxic conditions [51]. Another report demonstrated oral administration of pemafibrate increased serum FGF21 levels in a streptozotocin-induced diabetic mouse model and preserved retinal function through maintaining synaptophysin expression, which regulates synaptic vesicle endocytosis [50]. Additionally, long-acting FGF21 directly upregulated synaptophysin expression in differentiated neurons in vitro [50]. The other study reported that pemafibrate directly inhibited diabetes-induced vascular leukostasis and leakage in the rat retina through upregulation of THBD expression which encodes the glycoprotein thrombomodulin [52], and indicated that pemafibrate can increase thrombomodulin expression in human umbilical vein endothelial cells and human retinal microvascular endothelial cells in vitro [52]. Even though further studies are required to elucidate various mechanisms, pemafibrate could offer retinal protection through upregulation of blood FGF21 levels to work on the damaged retina with suppression of pathological neovascularization, maintenance of retinal function, modulation of systemic metabolisms such as triglyceride or blood glucose levels, or a direct increase in thrombomodulin expression in retinal endothelial cells to work on the damaged retina with suppression of vascular leakage, leukostasis and inflammation.

4. Conclusions

Diabetes mellitus is a complex metabolic disorder which is associated with insulin resistance, insulin signaling impairment, β-cell dysfunction, abnormal glucose and lipid metabolisms, inflammation and mitochondrial oxidative stress [53]. Likewise, development of DR has multiple interlinked alterations via dysfunctions of various cell types with enormous complex pathological mechanisms. Its development in turn cannot be prevented or protected by one therapeutic molecular target. Fortunately, PPARα targeting could be an alternative therapy to other present therapeutic agents in that it covers systemic enhancement of glucose and lipid metabolisms, anti-inflammation and anti-oxidative stress. Moreover, PPARα activation could ameliorate development of DR at an early stage through prevention of retinal vascular leakage[54][50][51]. This summary enables comprehensive understandings of protective roles of PPARα agonists against DR development. It can be useful for future studies on the protective effects of PPARα agonists against DR.

References

  1. Duh, E.J.; Sun, J.K.; Stitt, A.W. Diabetic retinopathy: Current understanding, mechanisms, and treatment strategies. JCI Insight 2017, 2, e93751.
  2. Chatziralli, I.P. The Role of Glycemic Control and Variability in Diabetic Retinopathy. Diabetes 2018, 9, 431–434.
  3. Wang, W.; Lo, A.C.Y. Diabetic Retinopathy: Pathophysiology and Treatments. Int. J. Mol. Sci. 2018, 19, 1816.
  4. Frank, R.N. Diabetic retinopathy. N. Engl. J. Med. 2004, 350, 48–58.
  5. Curtis, T.M.; Gardiner, T.A.; Stitt, A.W. Microvascular lesions of diabetic retinopathy: Clues towards understanding pathogenesis? Eye 2009, 23, 1496–1508.
  6. Barber, A.J.; Gardner, T.W.; Abcouwer, S.F. The significance of vascular and neural apoptosis to the pathology of diabetic retinopathy. Invest. Ophthalmol. Vis. Sci. 2011, 52, 1156–1163.
  7. Antonetti, D.A.; Barber, A.J.; Bronson, S.K.; Freeman, W.M.; Gardner, T.W.; Jefferson, L.S.; Kester, M.; Kimball, S.R.; Krady, J.K.; LaNoue, K.F.; et al. Diabetic retinopathy: Seeing beyond glucose-induced microvascular disease. Diabetes 2006, 55, 2401–2411.
  8. Hawkins, B.T.; Davis, T.P. The Blood-Brain Barrier/Neurovascular Unit in Health and Disease. Pharmacol. Rev. 2005, 57, 173.
  9. Yu, X.; Ji, C.; Shao, A. Neurovascular Unit Dysfunction and Neurodegenerative Disorders. Front. Neurosci. 2020, 14, 334.
  10. Lechner, J.; O’Leary, O.E.; Stitt, A.W. The pathology associated with diabetic retinopathy. Vis. Res. 2017, 139, 7–14.
  11. Robinson, R.; Barathi, V.A.; Chaurasia, S.S.; Wong, T.Y.; Kern, T.S. Update on animal models of diabetic retinopathy: From molecular approaches to mice and higher mammals. Dis. Models Mech. 2012, 5, 444–456.
  12. Tajoaek, P.; Petrovj, D.; Petrovi, M.G.N.; Kunej, T.J.P.R. Association of Peroxisome Proliferator-Activated Receptors (PPARs) with Diabetic Retinopathy in Human and Animal Models: Analysis of the Literature and Genome Browsers. PPAR Res. 2020, 2020, 1783564.
  13. Tyagi, S.; Gupta, P.; Saini, A.S.; Kaushal, C.; Sharma, S. The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. J. Adv. Pharm. Technol. Res. 2011, 2, 236–240.
  14. Yu, X.H.; Zheng, X.L.; Tang, C.K. Peroxisome Proliferator-Activated Receptor α in Lipid Metabolism and Atherosclerosis. Adv. Clin. Chem. 2015, 71, 171–203.
  15. Braissant, O.; Foufelle, F.; Scotto, C.; Dauça, M.; Wahli, W. Differential expression of peroxisome proliferator-activated receptors (PPARs): Tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat. Endocrinology 1996, 137, 354–366.
  16. Abbott, B.D. Review of the expression of peroxisome proliferator-activated receptors alpha (PPARα), beta (PPARβ), and gamma (PPARγ) in rodent and human development. Reprod. Toxicol. 2009, 27, 246–257.
  17. Auboeuf, D.; Rieusset, J.; Fajas, L.; Vallier, P.; Frering, V.; Riou, J.P.; Staels, B.; Auwerx, J.; Laville, M.; Vidal, H. Tissue distribution and quantification of the expression of mRNAs of peroxisome proliferator-activated receptors and liver X receptor-alpha in humans: No alteration in adipose tissue of obese and NIDDM patients. Diabetes 1997, 46, 1319–1327.
  18. Kliewer, S.A.; Forman, B.M.; Blumberg, B.; Ong, E.S.; Borgmeyer, U.; Mangelsdorf, D.J.; Umesono, K.; Evans, R.M. Differential expression and activation of a family of murine peroxisome proliferator-activated receptors. Proc. Natl. Acad. Sci. USA 1994, 91, 7355–7359.
  19. Touyz, R.M.; Schiffrin, E.L. Peroxisome proliferator-activated receptors in vascular biology-molecular mechanisms and clinical implications. Vasc. Pharmacol. 2006, 45, 19–28.
  20. Lefebvre, P.; Chinetti, G.; Fruchart, J.C.; Staels, B. Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J. Clin. Investig. 2006, 116, 571–580.
  21. Desouza, C.V.; Rentschler, L.; Fonseca, V. Peroxisome proliferator-activated receptors as stimulants of angiogenesis in cardiovascular disease and diabetes. Diabetes Metab. Syndr. Obes. 2009, 2, 165–172.
  22. Krauss, R.M. Lipids and Lipoproteins in Patients with Type 2 Diabetes. Diabetes Care 2004, 27, 1496.
  23. Katsiki, N.; Nikolic, D.; Montalto, G.; Banach, M.; Mikhailidis, D.P.; Rizzo, M. The role of fibrate treatment in dyslipidemia: An overview. Curr. Pharm. Des. 2013, 19, 3124–3131.
  24. Staels, B.; Dallongeville, J.; Auwerx, J.; Schoonjans, K.; Leitersdorf, E.; Fruchart, J.-C. Mechanism of Action of Fibrates on Lipid and Lipoprotein Metabolism. Circulation 1998, 98, 2088–2093.
  25. Han, L.; Shen, W.J.; Bittner, S.; Kraemer, F.B.; Azhar, S. PPARs: Regulators of metabolism and as therapeutic targets in cardiovascular disease. Part I: PPAR-α. Future Cardiol. 2017, 13, 259–278.
  26. Tenenbaum, A.; Fisman, E.Z. Fibrates are an essential part of modern anti-dyslipidemic arsenal: Spotlight on atherogenic dyslipidemia and residual risk reduction. Cardiovasc. Diabetol. 2012, 11, 125.
  27. Wang, F.; Gao, L.; Gong, B.; Hu, J.; Li, M.; Guan, Q.; Zhao, J. Tissue-specific expression of PPAR mRNAs in diabetic rats and divergent effects of cilostazol. Can. J. Physiol. Pharmacol. 2008, 86, 465–471.
  28. Hu, Y.; Chen, Y.; Ding, L.; He, X.; Takahashi, Y.; Gao, Y.; Shen, W.; Cheng, R.; Chen, Q.; Qi, X.; et al. Pathogenic role of diabetes-induced PPAR-α down-regulation in microvascular dysfunction. Proc. Natl. Acad. Sci. USA 2013, 110, 15401–15406.
  29. Ding, L.; Cheng, R.; Hu, Y.; Takahashi, Y.; Jenkins, A.J.; Keech, A.C.; Humphries, K.M.; Gu, X.; Elliott, M.H.; Xia, X.; et al. Peroxisome proliferator-activated receptor α protects capillary pericytes in the retina. Am. J. Pathol. 2014, 184, 2709–2720.
  30. Chen, Y.; Hu, Y.; Lin, M.; Jenkins, A.J.; Keech, A.C.; Mott, R.; Lyons, T.J.; Ma, J.-X. Therapeutic Effects of PPARα Agonists on Diabetic Retinopathy in Type 1 Diabetes Models. Diabetes 2013, 62, 261.
  31. Pearsall, E.A.; Cheng, R.; Matsuzaki, S.; Zhou, K.; Ding, L.; Ahn, B.; Kinter, M.; Humphries, K.M.; Quiambao, A.B.; Farjo, R.A.; et al. Neuroprotective effects of PPARα in retinopathy of type 1 diabetes. PLoS ONE 2019, 14, e0208399.
  32. Qiu, F.; Matlock, G.; Chen, Q.; Zhou, K.; Du, Y.; Wang, X.; Ma, J.X. Therapeutic Effects of PPARα Agonist on Ocular Neovascularization in Models Recapitulating Neovascular Age-Related Macular Degeneration. Invest. Ophthalmol. Vis. Sci. 2017, 58, 5065–5075.
  33. Moran, E.; Ding, L.; Wang, Z.; Cheng, R.; Chen, Q.; Moore, R.; Takahashi, Y.; Ma, J.X. Protective and antioxidant effects of PPARα in the ischemic retina. Invest. Ophthalmol. Vis. Sci. 2014, 55, 4568–4576.
  34. Wang, Z.; Moran, E.; Ding, L.; Cheng, R.; Xu, X.; Ma, J.X. PPARα regulates mobilization and homing of endothelial progenitor cells through the HIF-1α/SDF-1 pathway. Invest. Ophthalmol. Vis. Sci. 2014, 55, 3820–3832.
  35. Ye, S.; Chen, Q.; Jiang, N.; Liang, X.; Li, J.; Zong, R.; Huang, C.; Qiu, Y.; Ma, J.-X.; Liu, Z. PPARα-Dependent Effects of Palmitoylethanolamide Against Retinal Neovascularization and Fibrosis. Investig. Ophthalmol. Vis. Sci. 2020, 61, 15.
  36. Romano, M.R.; Lograno, M.D. Involvement of the peroxisome proliferator-activated receptor (PPAR) alpha in vascular response of endocannabinoids in the bovine ophthalmic artery. Eur. J. Pharmacol. 2012, 683, 197–203.
  37. Strobbe, E.; Cellini, M.; Campos, E.C. Effectiveness of palmitoylethanolamide on endothelial dysfunction in ocular hypertensive patients: A randomized, placebo-controlled cross-over study. Invest. Ophthalmol. Vis. Sci. 2013, 54, 968–973.
  38. Kumar, A.; Qiao, Z.; Kumar, P.; Song, Z.H. Effects of palmitoylethanolamide on aqueous humor outflow. Invest. Ophthalmol. Vis. Sci. 2012, 53, 4416–4425.
  39. Keppel Hesselink, J.M.; Costagliola, C.; Fakhry, J.; Kopsky, D.J. Palmitoylethanolamide, a Natural Retinoprotectant: Its Putative Relevance for the Treatment of Glaucoma and Diabetic Retinopathy. J. Ophthalmol. 2015, 2015, 430596.
  40. He, H.; Liang, M.; Li, L.; Luo, S.; Fang, X.; He, H.; Xiao, X.; Wu, H.; Lin, Z. PPAR-α Agonist Fenofibrate Suppressed the Formation of Ocular Surface Squamous Metaplasia Induced by Topical Benzalkonium Chloride. Invest. Ophthalmol. Vis. Sci. 2020, 61, 54.
  41. Li, W.; Hayashida, Y.; Chen, Y.T.; He, H.; Tseng, D.Y.; Alonso, M.; Chen, S.Y.; Xi, X.; Tseng, S.C. Air exposure induced squamous metaplasia of human limbal epithelium. Invest. Ophthalmol. Vis. Sci. 2008, 49, 154–162.
  42. Tang, L.; Wang, X.; Wu, J.; Li, S.M.; Zhang, Z.; Wu, S.; Su, T.; Lin, Z.; Chen, X.; Liao, X.; et al. Sleep Deprivation Induces Dry Eye Through Inhibition of PPARα Expression in Corneal Epithelium. Investig. Ophthalmol. Vis. Sci. 2018, 59, 5494–5508.
  43. Sana Raza-Iqbal; Toshiya Tanaka; Motonobu Anai; Takeshi Inagaki; Yoshihiro Matsumura; Kaori Ikeda; Akashi Taguchi; Frank J. Gonzalez; Juro Sakai; Tatsuhiko Kodama; et al. Transcriptome Analysis of K-877 (a Novel Selective PPARα Modulator (SPPARMα))-Regulated Genes in Primary Human Hepatocytes and the Mouse Liver. Journal of Atherosclerosis and Thrombosis 2015, 22, 754-772, 10.5551/jat.28720.
  44. Arima, T.; Uchiyama, M.; Nakano, Y.; Nagasaka, S.; Kang, D.; Shimizu, A.; Takahashi, H. Peroxisome proliferator-activated receptor alpha agonist suppresses neovascularization by reducing both vascular endothelial growth factor and angiopoietin-2 in corneal alkali burn. Sci. Rep. 2017, 7, 17763.
  45. Nakano, Y.; Arima, T.; Tobita, Y.; Uchiyama, M.; Shimizu, A.; Takahashi, H. Combination of Peroxisome Proliferator-Activated Receptor (PPAR) Alpha and Gamma Agonists Prevents Corneal Inflammation and Neovascularization in a Rat Alkali Burn Model. Int. J. Mol. Sci. 2020, 21, 5093.
  46. Sharma, N.; Kaur, M.; Agarwal, T.; Sangwan, V.S.; Vajpayee, R.B. Treatment of acute ocular chemical burns. Surv. Ophthalmol. 2018, 63, 214–235.
  47. Jean-Charles Fruchart; Pemafibrate (K-877), a novel selective peroxisome proliferator-activated receptor alpha modulator for management of atherogenic dyslipidaemia. Cardiovascular Diabetology 2017, 16, 1-12, 10.1186/s12933-017-0602-y.
  48. Jean-Charles Fruchart; Selective peroxisome proliferator-activated receptorα modulators (SPPARMα): The next generation of peroxisome proliferator-activated receptor α-agonists. Cardiovascular Diabetology 2013, 12, 82-82, 10.1186/1475-2840-12-82.
  49. Yusuke Sasaki; Sana Raza-Iqbal; Toshiya Tanaka; Kentaro Murakami; Motonobu Anai; Tsuyoshi Osawa; Yoshihiro Matsumura; Juro Sakai; Tatsuhiko Kodama; Gene Expression Profiles Induced by a Novel Selective Peroxisome Proliferator-Activated Receptor α Modulator (SPPARMα) Pemafibrate. International Journal of Molecular Sciences 2019, 20, 5682, 10.3390/ijms20225682.
  50. Yohei Tomita; Deokho Lee; Yukihiro Miwa; Xiaoyan Jiang; Kazuo Tsubota; Toshihide Kurihara; Toshihide Kurihara; Pemafibrate Protects Against Retinal Dysfunction in a Murine Model of Diabetic Retinopathy. International Journal of Molecular Sciences 2020, 21, 6243, 10.3390/ijms21176243.
  51. Yohei Tomita; Nobuhiro Ozawa; Yukihiro Miwa; Ayako Ishida; Masayuki Ohta; Kazuo Tsubota; Toshihide Kurihara; Pemafibrate Prevents Retinal Pathological Neovascularization by Increasing FGF21 Level in a Murine Oxygen-Induced Retinopathy Model. International Journal of Molecular Sciences 2019, 20, 5878, 10.3390/ijms20235878.
  52. Akira Shiono; Hiroki Sasaki; Reio Sekine; Yohei Abe; Yoshihiro Matsumura; Takeshi Inagaki; Toshiya Tanaka; Tatsuhiko Kodama; Hiroyuki Aburatani; Juro Sakai; et al.Hitoshi Takagi PPARα activation directly upregulates thrombomodulin in the diabetic retina. Scientific Reports 2020, 10, 10837, 10.1038/s41598-020-67579-1.
  53. Wagoner, M.D. Chemical injuries of the eye: Current concepts in pathophysiology and therapy. Surv. Ophthalmol. 1997, 41, 275–313.
  54. Wu, T.; Qiao, S.; Shi, C.; Wang, S.; Ji, G. Metabolomics window into diabetic complications. J. Diabetes Investig. 2018, 9, 244–255.
  55. Akira Shiono; Hiroki Sasaki; Reio Sekine; Yohei Abe; Yoshihiro Matsumura; Takeshi Inagaki; Toshiya Tanaka; Tatsuhiko Kodama; Hiroyuki Aburatani; Juro Sakai; et al.Hitoshi Takagi PPARα activation directly upregulates thrombomodulin in the diabetic retina. Scientific Reports 2020, 10, 10837, 10.1038/s41598-020-67579-1.
More
Information
Subjects: Others
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: 735
Revisions: 4 times (View History)
Update Date: 24 Nov 2020
1000/1000