PPARγ modulates refractive development and form deprivation myopia in Guinea pigs

https://doi.org/10.1016/j.exer.2020.108332Get rights and content

Highlights

  • PPARγ agonism and antagonism have opposing effect on refractive development and FDM.

  • PPARγ agonism reverses FD-induced declines of choroidal thickness and choroidal blood perfusion.

  • PPARγ agonism reverses FD-induced increases of scleral Hif-1α expression.

  • PPARγ agonism reverses FD-induced declines scleral collagen type 1 expression.

Abstract

Form deprivation myopia (FDM) is characterized by loss of choroidal thickness (ChT), reduced choroidal blood perfusion (ChBP), and consequently scleral hypoxia. In some tissues, changes in levels of peroxisome proliferator-activated receptor γ (PPARγ) expression modulate hypoxia-induced pathological responses. We determined if PPARγ modulates FDM through changes in ChT, ChBP, scleral hypoxia-inducible transcription factor (HIF-1α) that in turn regulate scleral collagen type 1 (COL1) expression levels in guinea pigs. Myopia was induced by occluding one eye, while the fellow eye served as control. They received daily peribulbar injections of either the PPARγ antagonist GW9662, or the GW1929 agonist, with or without ocular occlusion for 4 weeks. Ocular refraction and biometric parameters were estimated at baseline, 2 and 4 weeks post-treatment. ChT and ChBP were measured at the 2- and 4-week time points. Western blot analysis determined the expression levels of scleral HIF-1α and COL1. GW9662 induced a myopic shift in unoccluded eyes. Conversely, GW1929 inhibited FDM progression without affecting the refraction in unoccluded eyes. GW9662 reduced both ChT and ChBP in unoccluded eyes, while GW1929 inhibited their declines in occluded eyes. Scleral HIF-1α expression rose in GW9662-treated unoccluded eyes whereas GW1929 reduced HIF-1α upregulation in occluded eyes. GW9662 downregulated scleral COL1 expression in unoccluded eyes, while GW1929 reduced their decreases in occluded eyes. Therefore, PPARγ modulates collagen expression levels and FDM through an inverse relationship between changes in PPARγ and HIF-1α expression levels.

Introduction

Globally, myopia has become the second most common ocular disorder, which in severe cases can lead to blindness (Bourne et al., 2013). By 2050, it is predicted that the number of people afflicted with myopia and high myopia (≥6.00 diopters [D]) will increase to 49.8% and 9.8% of the world population, respectively(Holden et al., 2016). In Asian countries, myopia prevalence is already approaching crisis levels in some cohorts (Chen et al., 2018; Koh et al., 2014; Lam et al., 2012). Even though myopia is a non-life-threatening disorder, the visual pathology associated with severe cases can lead to cataract, glaucoma, retinal detachment, and myopic macular degeneration (Iwase et al., 2006; Yip et al., 2011). Therefore, a pressing need exists to clarify the underlying pathogenic mechanisms and identify novel targets for selectively improving therapeutic management of this condition.

Mammalian myopia develops from excessive ocular elongation and scleral extracellular matrix (ECM) remodeling that render the sclera more extensible, particularly at the posterior pole (Funata and Tokoro, 1990; McBrien et al., 2001; Phillips et al., 2000). These ECM changes are largely due to declines in the content of collagen I (COL1) that account for about 90% of the dry weight of the mammalian sclera (Rada et al., 2006; Yang et al., 2009).

We recently showed that the development of form deprivation myopia (FDM) is associated with upregulation of the scleral hypoxia-inducible transcription factor (HIF-1α) (Wu et al., 2018). Upon cessation of form deprivation (FD), HIF-1α reverts back to levels similar to those in the fellow unoccluded eyes (Wu et al., 2018). This association between changes in HIF-1α expression levels and myopia progression supports the notion that this transcription factor contributes to the control of genetic events inducing ECM remodeling and myopia progression. Other recent studies provided additional support for the conclusion that hypoxia triggers myopia progression. For example, during FDM and lens-induced myopia, hypoxia likely develops due to declines in both choroidal thickness (ChT) and choroidal blood perfusion (ChBP) whereas cessation of FD reversed these choroidal changes in guinea pigs (Zhang et al., 2019). These changes are not species specific because an inverse association between declines in choroidal blood flow and increased myopia progression was also identified in chicks after 14 days of FD (Shih et al., 1993). These findings are consistent with other reports that showed declines in choroidal blood flow occur in highly myopic humans (Dimitrova et al., 2002; Shih et al., 1991; Yang and Koh, 2015). In order to improve therapeutic management of myopia, it is relevant to identify targets controlling scleral hypoxia whose modulation can selectively inhibit responses underlying this sight compromising disease.

Peroxisome proliferator-activated receptor (PPAR) subtypes are ligand-activated transcription factors belonging to the nuclear hormone receptor superfamily with many diverse regulatory functions (Lakatos et al., 2007). The PPAR family includes three subtypes, PPARα, PPARβ, and PPARγ. PPARγ is widely viewed as an essential regulator of lipid metabolic processes (Wang et al., 2018) that may also contribute to myopia (Cordain et al., 2002; Lim et al., 2010). There is recent mounting evidence suggesting that hypoxia inhibits PPARγ expression and activation in different tissues. For example, hypoxic activation of ERK1/2 and NF-κB stimulates Nox4-mediated H2O2 generation, which in turn reduces PPARγ expression and activity (Lu et al., 2013). PPARγ has been shown to be inhibited by hypoxic stress during adipocyte differentiation (Yun et al., 2002). On the other hand, PPARγ upregulation inhibits hypoxia-induced proliferation of arterial smooth muscle cells (Yang et al., 2018). Such effects by changes in PPARγ expression are thought to stem from its interactions with inhibits signaling pathway mediators at upstream sites in a signaling pathway mediating control of responses to hypoxia. However, a few other studies reported an opposite effect of hypoxia on PPARγ expression. For example, long-term hypoxia induced instead increases in PPARγ mRNA in fetal sheep adipose tissue (Myers et al., 2008), and it stimulated differentiation of C2C12 and G8 murine myogenic cell lines (Itoigawa et al., 2010). Also, hypoxia caused PPARγ mRNA and protein levels to rise above those in normoxic HepG2 cells and promote carcinogenesis. (Zhao et al., 2014).

While studies carried out in other tissues highlight a possible relationship between the effects of hypoxia on PPARγ expression levels, their interaction during myopia development is unknown. We aimed to determine if PPARγ agonism and antagonism affect parameters modulating scleral hypoxia and collagen expression levels, which are reflective of changes in ECM remodeling during myopia progression.

Section snippets

Animals and experimental design

Animal treatment and care was approved by the Animal Care and Ethics Committee at Wenzhou Medical University (Wenzhou, China). The research protocols were conducted according to the Association for Research in Ophthalmology Statement for the Use of Animals in Ophthalmic and Vision Research. Three-week-old pigmented guinea pigs were reared in 12-h light (400–500 lux) and dark (0 lux) cycles and had ad libitum access to food and water. Guinea pigs were randomly assigned to unoccluded and FD

GW9662 promoted myopia progression in unoccluded eyes

Vehicle (DMSO) injections did not alter interocular differences of refraction, ACD, LT, VCD, or AL compared to their control groups (normal and FDM, all P values > 0.05, 2-way repeated ANOVA, Supplementary Tables S3–4) in either unoccluded eyes or form deprived eyes, which was consistent with our earlier study (Pan et al., 2018).

For all baseline results, none of the interocular differences were significantly different from each other among the three vehicle and GW9662 groups (all P

Discussion

This is the first study to identify the role of PPARγ in regulation of myopia. We found that PPARγ moderated refraction and ocular growth in normal and vision obstructed environments. Specifically, PPARγ antagonism induced axial elongation and a myopic shift during normal ocular growth. While FD increased myopia development, GW1929 reduced this myopic shift and retarded ocular elongation during FD. PPARγ agonism and antagonism had opposing effects on ocular refraction under two different visual

Declaration of competing interest

The authors declare no competing financial interests.

Acknowledgments

This project is supported by National Natural Science Foundation of China under Grant 81830027, 81670886, 81470659 and 81422007, Zhejiang Provincial Natural Science Foundation of China under Grant No. LQ20H120001, CAMS Innovation Fund for Medical Sciences under Grant 2019-I2M-5–048 and Grant Y20180711 from the Scientific Bureau of Wenzhou City, China.

References (60)

  • J. Wallman et al.

    Moving the retina: choroidal modulation of refractive state

    Vis. Res.

    (1995)
  • J. Wallman et al.

    Homeostasis of eye growth and the question of myopia

    Neuron

    (2004)
  • Y. Yang et al.

    Myopia: a collagen disease?

    Med. Hypotheses

    (2009)
  • Z. Yun et al.

    Inhibition of PPAR gamma 2 gene expression by the HIF-1-regulated gene DEC1/Stra13: a mechanism for regulation of adipogenesis by hypoxia

    Dev. Cell

    (2002)
  • Y.Z. Zhao et al.

    Hypoxia induces peroxisome proliferator-activated receptor gamma expression via HIF-1-dependent mechanisms in HepG2 cell line

    Arch. Biochem. Biophys.

    (2014)
  • X. Zhou et al.

    Normal development of refractive state and ocular dimensions in Guinea pigs

    Vis. Res.

    (2006)
  • R.R. Bourne et al.

    Causes of vision loss worldwide, 1990-2010: a systematic analysis

    Lancet Glob Health

    (2013)
  • J. Chhablani et al.

    Clinical applications of choroidal imaging technologies

    Indian J. Ophthalmol.

    (2015)
  • L. Cordain et al.

    An evolutionary analysis of the aetiology and pathogenesis of juvenile-onset myopia

    Acta Ophthalmol. Scand.

    (2002)
  • G. Dimitrova et al.

    Retrobulbar circulation in myopic patients with or without myopic choroidal neovascularisation

    Br. J. Ophthalmol.

    (2002)
  • P. Dromparis et al.

    Pioglitazone inhibits HIF-1alpha-dependent angiogenesis in rats by paracrine and direct effects on endothelial cells

    J. Mol. Med. (Berl.)

    (2014)
  • E. Duval et al.

    Hypoxia inducible factor 1 alpha down-regulates type i collagen through Sp3 transcription factor in human chondrocytes

    IUBMB Life

    (2016)
  • E. Duval et al.

    Hypoxia-inducible factor 1alpha inhibits the fibroblast-like markers type I and type III collagen during hypoxia-induced chondrocyte redifferentiation: hypoxia not only induces type II collagen and aggrecan, but it also inhibits type I and type III collagen in the hypoxia-inducible factor 1alpha-dependent redifferentiation of chondrocytes

    Arthritis Rheum.

    (2009)
  • H.M. El-Gowelli et al.

    PPARgamma dependence of cyclosporine-isoprenaline renovascular interaction: roles of nitric oxide synthase and heme oxygenase

    J. Cardiovasc. Pharmacol.

    (2011)
  • R.H. Fan et al.

    [Hypoxia induced by CoCl2 influencing the expression and the activity of matrix metalloproteinase-2 in rat hepatic stellate cells]

    Chinese journal of hepatology

    (2007)
  • M. Funata et al.

    Scleral change in experimentally myopic monkeys

    Graefes Arch. Clin. Exp. Ophthalmol.

    (1990)
  • L. Guo et al.

    Gene expression signatures in tree shrew sclera in response to three myopiagenic conditions

    Invest. Ophthalmol. Vis. Sci.

    (2013)
  • L. Guo et al.

    Scleral gene expression during recovery from myopia compared with expression during myopia development in tree shrew

    Mol. Vis.

    (2014)
  • M. Iglarz et al.

    Peroxisome proliferator-activated receptor-alpha and receptor-gamma activators prevent cardiac fibrosis in mineralocorticoid-dependent hypertension

    Hypertension

    (2003)
  • A. Iwase et al.

    Prevalence and causes of low vision and blindness in a Japanese adult population: the Tajimi Study

    Ophthalmology

    (2006)
  • Cited by (12)

    • Myopia: Histology, clinical features, and potential implications for the etiology of axial elongation

      2023, Progress in Retinal and Eye Research
      Citation Excerpt :

      Similarly, given that the principles of the process of axial elongation remain unknown, we focused on only one possible explanatory mechanism underlying axial elongation, without describing in detail other potential explanations of how axial elongation is associated with certain biomarkers and messenger molecules. That this study neglected other hypotheses, such as those on the role of the choroid and sclera in myopization, does not indicate that these hypotheses are not valid (Barathi and Beuerman, 2011; Guggenheim and McBrien, 1996; Guo, L et al., 2014; Gwiazda et al., 1993; Hung et al., 2018; Jiang, L et al., 2018; Markov et al., 2018; Mutti et al., 2006, 2007, 2011; Nickla and Wallman, 2010; Pan, M et al., 2021; Rymer and Wildsoet, 2005; Siegwart and Norton, 2002; Troilo and Wallman, 1991; Troilo et al., 2019; Wang, KK et al., 2017; Wallman et al., 1978, 1995; Wang, M et al., 2018; Wildsoet and Wallman, 1995; Wong, CW et al., 2017; Wu et al., 2018; Ye et al., 2020; Zhang and Wildsoet, 2015; Zhou, F et al., 2021; Zhou, X et al., 2021). Second, it has remained elusive whether anatomical features of myopic eyes are effect of or are caused by the process of axial elongation.

    • Candidate pathways for retina to scleral signaling in refractive eye growth

      2022, Experimental Eye Research
      Citation Excerpt :

      In support of this hypothesis, recent studies have found scleral Hif-1α to be correlated to myopigenesis. Hif-1α protein levels in the sclera, but not the retina, were associated with FDM in mice and guinea pigs and LIM in guinea pigs (Pan et al., 2021; Wu et al., 2018). Additionally, analysis of human gene databases revealed a moderate association with scleral Hif-1α signaling pathway in individuals with high myopia (Wu et al., 2018; Zhao et al., 2020).

    View all citing articles on Scopus
    View full text