Characterization and contribution of RPE senescence to Age-related macular degeneration in Tnfrsf10 knock out mice

Background Retinal pigment epithelial cells (RPE) play vital role in the pathogenesis of age-related macular degeneration (AMD). Our laboratory has shown that RPE cellular senescence contributed to the pathophysiology of experimental AMD, and SASP members are involved in this process. Recently, we presented confirmatory evidence to earlier GWAS studies that dysregulation of tumor necrosis factor receptor superfamily 10A (TNFRSF10A) dysregulation leads to AMD development and is linked to RPE dysfunction. This study aims to investigate the contribution of RPE senescence to AMD pathophysiology using TNFRSF10A silenced human RPE (hRPE) cells and Tnfrsf10 KO mice. Methods Sub-confluent primary hRPE cells and TNFRSF10A silenced hRPE were exposed to stress-induced premature senescence with H2O2 (500 μM, 48h), and senescence-associated markers (βgal, p16, and p21) were analyzed by RT-PCR and WB analysis. The effect of H2O2-induced senescence in non-silenced and silenced hRPE on OXPHOS and glycolysis was determined using Seahorse XF96 analyzer. Male C57BL/6J Tnfrsf10 KO (Tnfrsf10-/-) mice were used to study the regulation of senescence by TNFRSF10A in vivo. Expression of p16 and p21 in control and KO mice of varying ages were determined by RT-PCR, WB, and immunostaining analysis. Results The senescence-associated p16 and p21 showed a significant (p < 0.01) upregulation with H2O2 induction at the gene (1.8- and 3-fold) and protein (3.2- and 4-fold) levels in hRPE cells. The protein expression of p16 and p21 was further significantly increased by co-treatment with siRNA (p < 0.05 vs. H2O2). Mitochondrial oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) (pmol/min/total DNA) increased with senescence induction by H2O2 for 48h in control RPE, and knockdown of TNFRSF10A caused a further increase in OCR and ECAR. In addition, co-treatment with PKC activator significantly improved all parameters. Similarly, in vivo studies showed upregulation of p16 and p21 by RT-PCR, WB, and immunostaining analysis in RPE/choroid of Tnfrsf10 KO mice. When subjected to examination across distinct age groups, namely young (1-3 months), middle (6-9 months), and old (12-15 months) mice, a discernible age-related elevation in the expression of p16 and p21 was observed. Conclusions Our findings suggest that TNRSF10A is a regulator of regulates in RPE senescence. Further work on elucidating pathways of senescence will facilitate the development of new therapeutic targets for AMD.

[1]  S. Reddy,et al.  Mechanisms of RPE senescence and potential role of αB crystallin peptide as a senolytic agent in experimental AMD. , 2022, Experimental eye research.

[2]  D. Koya,et al.  Autophagy in metabolic disease and ageing , 2021, Nature Reviews Endocrinology.

[3]  A. Dick,et al.  Cellular senescence in the aging retina and developments of senotherapies for age-related macular degeneration , 2021, Journal of Neuroinflammation.

[4]  A. Salminen,et al.  Mechanisms of mitochondrial dysfunction and their impact on age-related macular degeneration , 2020, Progress in retinal and eye research.

[5]  D. Hinton,et al.  The Emerging Role of Senescence in Ocular Disease , 2020, Oxidative medicine and cellular longevity.

[6]  Jongjin Park,et al.  Oxidative stress-mediated TXNIP loss causes RPE dysfunction , 2019, Experimental & Molecular Medicine.

[7]  M. Bollati-Fogolín,et al.  Mitofusins modulate the increase in mitochondrial length, bioenergetics and secretory phenotype in therapy-induced senescent melanoma cells , 2019, The Biochemical journal.

[8]  L. Ferrucci,et al.  A proteomic atlas of senescence-associated secretomes for aging biomarker development , 2019, bioRxiv.

[9]  A. Hoffman,et al.  Mitochondrial peptides modulate mitochondrial function during cellular senescence , 2018, Aging.

[10]  K. Kaarniranta,et al.  Mitochondrial quality control in AMD: does mitophagy play a pivotal role? , 2018, Cellular and Molecular Life Sciences.

[11]  J. Hoek,et al.  The path from mitochondrial ROS to aging runs through the mitochondrial permeability transition pore , 2017, Aging cell.

[12]  Lanjuan Li,et al.  Drp1-Dependent Mitochondrial Fission Plays Critical Roles in Physiological and Pathological Progresses in Mammals , 2017, International journal of molecular sciences.

[13]  C. Winborn,et al.  Genomic regulation of senescence and innate immunity signaling in the retinal pigment epithelium , 2015, Mammalian Genome.

[14]  C. Kukat,et al.  mtDNA makes a U-turn for the mitochondrial nucleoid. , 2013, Trends in cell biology.

[15]  Gabriëlle H S Buitendijk,et al.  Seven New Loci Associated with Age-Related Macular Degeneration , 2013, Nature Genetics.

[16]  J. Campisi,et al.  Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. , 2013, The Journal of clinical investigation.

[17]  S. Jarrett,et al.  Consequences of oxidative stress in age-related macular degeneration. , 2012, Molecular aspects of medicine.

[18]  J. Handa How does the macula protect itself from oxidative stress? , 2012, Molecular aspects of medicine.

[19]  J. Campisi,et al.  The senescence-associated secretory phenotype: the dark side of tumor suppression. , 2010, Annual review of pathology.

[20]  J. Shay,et al.  BRAFE600-associated senescence-like cell cycle arrest of human naevi , 2005, Nature.

[21]  M. Boulton,et al.  RPE lipofuscin and its role in retinal pathobiology. , 2005, Experimental eye research.

[22]  Z. Werb,et al.  Regulation of matrix biology by matrix metalloproteinases. , 2004, Current opinion in cell biology.

[23]  B. Ames,et al.  Oxidants Are a Major Contributor to Aging a , 1992, Annals of the New York Academy of Sciences.

[24]  K. Elgass,et al.  Recent advances into the understanding of mitochondrial fission. , 2013, Biochimica et biophysica acta.

[25]  A. Ramé [Age-related macular degeneration]. , 2006, Revue de l'infirmiere.

[26]  J. Handa,et al.  Beta-galactosidase histochemistry and telomere loss in senescent retinal pigment epithelial cells. , 1999, Investigative ophthalmology & visual science.

[27]  J. Handa,et al.  Senescence-associated beta-galactosidase histochemistry for the primate eye. , 1999, Investigative ophthalmology & visual science.

[28]  J. Weiter,et al.  CELL LOSS IN THE AGING RETINA , 1989 .