1
|
Dubey SK, Dubey R, Prajapati SC, Jung K, Mohan K, Liu X, Roney J, Tian W, Abney J, Giarmarco MM, Hernandez AG, Liu J, Kleinman ME. Histone deficiency and hypoacetylation in the aging retinal pigment epithelium. Aging Cell 2024; 23:e14108. [PMID: 38408164 PMCID: PMC11113634 DOI: 10.1111/acel.14108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
Histones serve as a major carrier of epigenetic information in the form of post-translational modifications which are vital for controlling gene expression, maintaining cell identity, and ensuring proper cellular function. Loss of histones in the aging genome can drastically impact the epigenetic landscape of the cell leading to altered chromatin structure and changes in gene expression profiles. In this study, we investigated the impact of age-related changes on histone levels and histone acetylation in the retinal pigment epithelium (RPE) and retina of mice. We observed a global reduction of histones H1, H2A, H2B, H3, and H4 in aged RPE/choroid but not in the neural retina. Transcriptomic analyses revealed significant downregulation of histones in aged RPE/choroid including crucial elements of the histone locus body (HLB) complex involved in histone pre-mRNA processing. Knockdown of HINFP, a key HLB component, in human RPE cells induced histone loss, senescence, and the upregulation of senescence-associated secretory phenotype (SASP) markers. Replicative senescence and chronological aging in human RPE cells similarly resulted in progressive histone loss and acquisition of the SASP. Immunostaining of human retina sections revealed histone loss in RPE with age. Acetyl-histone profiling in aged mouse RPE/choroid revealed a specific molecular signature with loss of global acetyl-histone levels, including H3K14ac, H3K56ac, and H4K16ac marks. These findings strongly demonstrate histone loss as a unique feature of RPE aging and provide critical insights into the potential mechanisms linking histone dynamics, cellular senescence, and aging.
Collapse
Affiliation(s)
- Sushil K. Dubey
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Rashmi Dubey
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Subhash C. Prajapati
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Kyungsik Jung
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Kabhilan Mohan
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Xinan Liu
- Department of Computer ScienceUniversity of KentuckyLexingtonKentuckyUSA
| | - Jacob Roney
- Department of Ophthalmology and Visual SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | - Wenjian Tian
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Jennifer Abney
- Department of Ophthalmology and Visual SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | | | - Alvaro G. Hernandez
- Roy J. Carver Biotechnology CenterUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Jinze Liu
- Department of BiostatisticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Mark E. Kleinman
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| |
Collapse
|
2
|
Banerjee M, Al-Eryani L, Srivastava S, Rai SN, Pan J, Kalbfleisch TS, States JC. Delineating the Effects of Passaging and Exposure in a Longitudinal Study of Arsenic-Induced Squamous Cell Carcinoma in a HaCaT Cell Line Model. Toxicol Sci 2021; 185:184-196. [PMID: 34730829 DOI: 10.1093/toxsci/kfab129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is a major deleterious health effect of chronic arsenic (iAs) exposure. The molecular mechanism of arsenic-induced cSCC remains poorly understood. We recently demonstrated that chronic iAs exposure leads to temporally regulated genome-wide changes in profiles of differentially expressed mRNAs and miRNAs at each stage of carcinogenesis (7, 19 and 28 weeks) employing a well-established passage-matched HaCaT cell line model of arsenic-induced cSCC. Here, we performed longitudinal differential expression analysis (miRNA and mRNA) between the different time points (7 vs. 19 weeks and 19 vs. 28 weeks) within unexposed and exposed groups, coupled to expression pairing and pathway analyses to differentiate the relative effects of long-term passaging and chronic iAs exposure. Data showed that 66-105 miRNA [p < 0.05; log2(Fold Change)>I1I] and 2826-4079 mRNA [p < 0.001; log2(Fold Change)>I1I] molecules were differentially expressed depending on the longitudinal comparison. Several mRNA molecules differentially expressed as a function of time, independent of iAs exposure were being targeted by miRNA molecules which were also differentially expressed in a time dependent manner. Distinct pathways were predicted to be modulated as a function of time or iAs exposure. Some pathways were also modulated both by time and exposure. Thus, the HaCaT model can distinguish between the effects of passaging and chronic iAs exposure individually and corroborate our previously published data on effects of iAs exposure compared to unexposed passage matched HaCaT cells. In addition, this work provides a template for cell line based longitudinal chronic exposure studies to follow for optimal efficacy.
Collapse
Affiliation(s)
- Mayukh Banerjee
- Department of Pharmacology and Toxicology, University of Louisville, USA Louisville, KY
| | - Laila Al-Eryani
- Department of Pharmacology and Toxicology, University of Louisville, USA Louisville, KY
| | - Sudhir Srivastava
- Biostatistics and Bioinformatics Facility, James Graham Brown Cancer Center, University of Louisville, USA Louisville, KY.,Centre for Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, India New Delhi, 110012
| | - Shesh N Rai
- Biostatistics and Bioinformatics Facility, James Graham Brown Cancer Center, University of Louisville, USA Louisville, KY.,Department of Bioinformatics and Biostatistics, University of Louisville, USA Louisville, KY
| | - Jianmin Pan
- Biostatistics and Bioinformatics Facility, James Graham Brown Cancer Center, University of Louisville, USA Louisville, KY
| | - Theodore S Kalbfleisch
- Department of Biochemistry and Molecular Genetics, University of Louisville, USA Louisville, KY
| | - J Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, USA Louisville, KY
| |
Collapse
|
3
|
Gao J, Cui JZ, Wang A, Chen HHR, Fong A, Matsubara JA. The reduction of XIAP is associated with inflammasome activation in RPE: implications for AMD pathogenesis. J Neuroinflammation 2019; 16:171. [PMID: 31438981 PMCID: PMC6706877 DOI: 10.1186/s12974-019-1558-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/14/2019] [Indexed: 02/06/2023] Open
Abstract
Background Age-related macular degeneration (AMD) is a multifactorial chronic disease of the eye. Several candidate pathways have been hypothesized to play a role in AMD pathogenesis. Our work and those of others suggests inflammasome activity as a mechanism associated with retinal pigment epithelial (RPE) cell demise. X-linked inhibitor of apoptosis protein (XIAP), an anti-apoptosis factor, has recently been shown to regulate inflammasome activity in non-ocular cells. The purpose of this study is to characterize XIAP’s regulatory role in RPE. Methods Protein lysates of eye tissues from rats (vinpocetine- or aurin tricarboxylic acid complex-treated, ATAC, vs naïve) and mice (wild type vs Caspase-4−/−) were utilized to analyze XIAP protein levels. Immunohistochemistry was used to detect NLRP3 levels in the RPE layer. In vitro inflammasome activation on RPE cells was achieved with L-leucyl-L-leucine methyl ester (Leu-Leu-OMe) stimulation. Levels of XIAP mRNA and 18S RNA were quantified by RT-PCR. Cell culture supernatants were tested directly for secreted IL-1β by ELISA or concentrated for the detection of secreted IL-18 by western blot. Protein lysates from RPE in cell culture were collected for the measurement of cleaved caspase-1 p20, XIAP, and GAPDH. Data are presented as Mean ± SD. p < 0.05 is considered statistically significant. Results The XIAP protein level was significantly increased when the inflammasome was inhibited at the “activation” step by ATAC, but not the “priming” step, in vivo. Concomitantly, NLRP3 immunoreactivity was lower in the RPE layer of animals fed with ATAC. In mice where caspase-1 cleavage was impaired by the genetic deficiency in caspase-4, the XIAP protein level increased in eye tissues. In RPE cell culture, Leu-Leu-OMe stimulation led to caspase-1 cleavage, cytokine secretion, and XIAP reduction, which can be abolished by Z-YVAD-FMK. When XIAP siRNA was given as a pre-treatment to RPE in vitro, Leu-Leu-OMe induced IL-1β/IL-18 secretion was enhanced, whereas overexpressing XIAP reduced IL-1β secretion under inflammasome activation, both compared to controls cells. Conclusions Together, these data suggest XIAP-mediated inhibition of inflammasome activity in RPE may provide insights into the biological consequences of inflammasome activation in RPE and reveals the caspase-1/XIAP/IL-1β/IL-18 axis as a target for broader applications in AMD biology and treatment design. Electronic supplementary material The online version of this article (10.1186/s12974-019-1558-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jiangyuan Gao
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada
| | - Jing Z Cui
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada
| | - Aikun Wang
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada
| | - Hao Hang Rachel Chen
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada
| | - Alison Fong
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada.
| |
Collapse
|
4
|
Yang J, Menges S, Gu P, Tongbai R, Samuel M, Prather RS, Klassen H. Porcine Neural Progenitor Cells Derived from Tissue at Different Gestational Ages Can Be Distinguished by Global Transcriptome. Cell Transplant 2017; 26:1582-1595. [PMID: 29113465 PMCID: PMC5524599 DOI: 10.1177/0963689717723015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The impact of gestational age on mammalian neural progenitor cells is potentially important for both an understanding of neural development and the selection of donor cells for novel cell-based treatment strategies. In terms of the latter, it can be problematic to rely entirely on rodent models in which the gestational period is significantly shorter and the brain much smaller than is the case in humans. Here, we analyzed pig brain progenitor cells (pBPCs) harvested at 2 different gestational ages (E45 and E60) using gene expression profiles, obtained by microarray analysis and quantitative polymerase chain reaction (qPCR), across time in culture. Comparison of the global transcriptome of pBPCs from age-matched transgenic green flourescent protein (GFP)-expressing fetuses versus non-GFP-expressing fetuses did not reveal significant differences between the 2 cell types, whereas comparison between E45 and E60 pBPCs did show separation between the data sets by principle component analysis. Further examination by qPCR showed evidence of relative downregulation of proliferation markers and upregulation of glial markers in the gestationally older (E60) cells. Additional comparisons were made. This study provides evidence of age-related changes in the gene expression of cultured fetal porcine neural progenitors that are potentially relevant to the role of these cells during development and as donor cells for transplantation studies.
Collapse
Affiliation(s)
- Jing Yang
- 1 Stem Cell Research Center, University of California, Irvine, CA, USA.,2 Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - Steven Menges
- 1 Stem Cell Research Center, University of California, Irvine, CA, USA.,2 Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - Ping Gu
- 1 Stem Cell Research Center, University of California, Irvine, CA, USA.,2 Gavin Herbert Eye Institute, University of California, Irvine, CA, USA.,3 Present Address: Department of Ophthalmology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ronald Tongbai
- 1 Stem Cell Research Center, University of California, Irvine, CA, USA.,4 Present Address: Huntington Beach Eye Consultants, Huntington Beach, CA, USA
| | - Melissa Samuel
- 5 National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Randall S Prather
- 5 National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Henry Klassen
- 1 Stem Cell Research Center, University of California, Irvine, CA, USA.,2 Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| |
Collapse
|
5
|
Sachdeva MM, Eliott D. Stem Cell-Based Therapy for Diseases of the Retinal Pigment Epithelium: From Bench to Bedside. Semin Ophthalmol 2016; 31:25-9. [PMID: 26959126 DOI: 10.3109/08820538.2015.1115253] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Age-related macular degeneration (AMD) represents a leading cause of blindness in the elderly, and Stargardt's macular dystrophy (SMD) is the most common form of juvenile-onset macular degeneration. Dry AMD and SMD share an underlying pathophysiology, namely dysfunction and ultimately loss of the retinal pigment epithelium (RPE), suggesting that RPE transplantation may offer a potential treatment strategy for both patient populations. Stem cells have emerged as a promising source of replacement RPE. During the past 15 years, extraordinary strides have been made in the identification, characterization, and differentiation of stem cells. Recently, this large body of basic science and preclinical research has been translated to patient care with the publication of results from Phase 1/2 trials demonstrating safety of transplantation of human embryonic stem cell (hESC)-derived RPE into patients with AMD and SMD. While significant challenges remain before dry AMD and SMD become treatable diseases, the goal has become more tangible.
Collapse
Affiliation(s)
- Mira M Sachdeva
- a Harvard Medical School, Massachusetts Eye and Ear Infirmary , Boston , Massachusetts , USA
| | - Dean Eliott
- a Harvard Medical School, Massachusetts Eye and Ear Infirmary , Boston , Massachusetts , USA
| |
Collapse
|
6
|
Ardeljan D, Chan CC. Aging is not a disease: distinguishing age-related macular degeneration from aging. Prog Retin Eye Res 2013; 37:68-89. [PMID: 23933169 PMCID: PMC3830684 DOI: 10.1016/j.preteyeres.2013.07.003] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 07/29/2013] [Accepted: 07/30/2013] [Indexed: 12/18/2022]
Abstract
Age-related macular degeneration (AMD) is a disease of the outer retina, characterized most significantly by atrophy of photoreceptors and retinal pigment epithelium accompanied with or without choroidal neovascularization. Development of AMD has been recognized as contingent on environmental and genetic risk factors, the strongest being advanced age. In this review, we highlight pathogenic changes that destabilize ocular homeostasis and promote AMD development. With normal aging, photoreceptors are steadily lost, Bruch's membrane thickens, the choroid thins, and hard drusen may form in the periphery. In AMD, many of these changes are exacerbated in addition to the development of disease-specific factors such as soft macular drusen. Para-inflammation, which can be thought of as an intermediate between basal and robust levels of inflammation, develops within the retina in an attempt to maintain ocular homeostasis, reflected by increased expression of the anti-inflammatory cytokine IL-10 coupled with shifts in macrophage plasticity from the pro-inflammatory M1 to the anti-inflammatory M2 polarization. In AMD, imbalances in the M1 and M2 populations together with activation of retinal microglia are observed and potentially contribute to tissue degeneration. Nonetheless, the retina persists in a state of chronic inflammation and increased expression of certain cytokines and inflammasomes is observed. Since not everyone develops AMD, the vital question to ask is how the body establishes a balance between normal age-related changes and the pathological phenotypes in AMD.
Collapse
Affiliation(s)
- Daniel Ardeljan
- Immunopathology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | |
Collapse
|
7
|
Biological effects of cigarette smoke in cultured human retinal pigment epithelial cells. PLoS One 2012; 7:e48501. [PMID: 23155386 PMCID: PMC3498276 DOI: 10.1371/journal.pone.0048501] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 09/26/2012] [Indexed: 12/24/2022] Open
Abstract
The goal of the present study was to determine whether treatment with cigarette smoke extract (CSE) induces cell loss, cellular senescence, and extracellular matrix (ECM) synthesis in primary human retinal pigment epithelial (RPE) cells. Primary cultured human RPE cells were exposed to 2, 4, 8, and 12% of CSE concentration for 24 hours. Cell loss was detected by cell viability assay. Lipid peroxidation was assessed by loss of cis-parinaric acid (PNA) fluorescence. Senescence-associated ß-galactosidase (SA-ß-Gal) activity was detected by histochemical staining. Expression of apolipoprotein J (Apo J), connective tissue growth factor (CTGF), fibronectin, and laminin were examined by real-time PCR, western blot, or ELISA experiments. The results showed that exposure of cells to 12% of CSE concentration induced cell death, while treatment of cells with 2, 4, and 8% CSE increased lipid peroxidation. Exposure to 8% of CSE markedly increased the number of SA-ß-Gal positive cells to up to 82%, and the mRNA expression of Apo J, CTGF, and fibronectin by approximately 3–4 fold. Treatment with 8% of CSE also increased the protein expression of Apo J and CTGF and the secretion of fibronectin and laminin. Thus, treatment with CSE can induce cell loss, senescent changes, and ECM synthesis in primary human RPE cells. It may be speculated that cigarette smoke could be involved in cellular events in RPE cells as seen in age-related macular degeneration.
Collapse
|
8
|
Nguyen LTH, Liao S, Ramakrishna S, Chan CK. The role of nanofibrous structure in osteogenic differentiation of human mesenchymal stem cells with serial passage. Nanomedicine (Lond) 2011; 6:961-74. [DOI: 10.2217/nnm.11.26] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Using scaffolds with autologous stem cells is a golden strategy for the treatment of bone defects. In this strategy, human mesenchymal stem cells (hMSCs) have often been isolated and expanded in vitro on a plastic surface to obtain a sufficient cell number before seeding on a suitable scaffold. Materials & Methods: Investigating the influence of serial passages (from passage two to passage eight) on the abilities of proliferation and osteogenic differentiation of hMSCs on 24-well tissue culture polystyrene plates and poly L-lactic acid electrospun nanofibrous scaffolds was performed to determine how prolonged culture affected these cellular abilities and how the nanofibrous scaffolds supported the osteogenic differentiation potential of hMSCs. Results & Conclusion: Serial passage caused adverse changes in hMSCs characteristics, which were indicated by the decline in both proliferation and osteogenic differentiation abilities. Interestingly, the poly L-lactic acid nanofibrous scaffolds showed a significant support in recovering the osteogenic abilities of hMSCs, which had been severely affected by prolonged culture.
Collapse
Affiliation(s)
| | - Susan Liao
- Nanyang Technological University, 637551 Singapore
| | - Seeram Ramakrishna
- National University of Singapore, 117576 Singapore
- Institute of Materials Research & Engineering, 117602 Singapore
| | - Casey K Chan
- National University of Singapore, 117576 Singapore
| |
Collapse
|
9
|
Gerona G, López D, Palmero M, Maneu V. AntioxidantN-Acetyl-Cysteine Protects Retinal Pigmented Epithelial Cells from Long-Term Hypoxia Changes in Gene Expression. J Ocul Pharmacol Ther 2010; 26:309-14. [DOI: 10.1089/jop.2009.0101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Guillermo Gerona
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, San Vicente del Raspeig, Spain
| | - Damián López
- Departamento de Sistemas Informáticos y Computación, Universidad Politécnica de Valencia, Valencia, Spain
| | - Mercedes Palmero
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, San Vicente del Raspeig, Spain
| | - Victoria Maneu
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, San Vicente del Raspeig, Spain
- Instituto Teófilo Hernando de I + D del Medicamento, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
10
|
Kurji KH, Cui JZ, Lin T, Harriman D, Prasad SS, Kojic L, Matsubara JA. Microarray analysis identifies changes in inflammatory gene expression in response to amyloid-beta stimulation of cultured human retinal pigment epithelial cells. Invest Ophthalmol Vis Sci 2009; 51:1151-63. [PMID: 19797223 DOI: 10.1167/iovs.09-3622] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Age-related macular degeneration (AMD) is a common cause of irreversible vision loss in the elderly. The hypothesis was that in vitro stimulation of RPE cells with Abeta(1-40), a constituent of drusen, promotes changes in gene expression and cellular pathways associated with the pathogenesis of AMD, including oxidative stress, inflammation, and angiogenesis. METHODS Confluent human RPE cells were stimulated with Abeta(1-40), or the reverse peptide Abeta(40-1), and genome wide changes in gene expression were studied with gene microarrays. Selected genes were verified by qRT-PCR and ELISA. Pathway analysis with gene set enrichment analysis (GSEA) and ingenuity revealed top functional pathways in RPE after Abeta(1-40) stimulation. RESULTS RPE cells stimulated with Abeta(1-40) (0.3 microM) for 24 hours resulted in 63 upregulated and 22 downregulated previously known genes. The upregulated genes were predominantly in inflammatory and immune response categories, but other categories were also represented, including apoptosis, cell signaling, cell proliferation, and signal transduction. Categories of downregulated genes included immune response, transporters, metabolic functions and transcription factors. ELISA confirmed that secreted levels of IL-8 were two times higher than control levels. GSEA and ingenuity analysis confirmed that the top affected pathways in RPE cells after Abeta(1-40) stimulation were inflammation and immune response related. Surprisingly, few angiogenic pathways were activated at the doses and exposure times studied. CONCLUSIONS Abeta(1-40) promotes RPE gene expression changes in pathways associated with immune response, inflammation, and cytokine and interferon signaling pathways. Results may relate to in vivo mechanisms associated with the pathogenesis of AMD.
Collapse
Affiliation(s)
- Khaliq H Kurji
- Department of Ophthalmology and Visual Sciences, University of British Columbia, British Columbia, Canada
| | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Para-inflammation is a tissue adaptive response to noxious stress or malfunction and has characteristics that are intermediate between basal and inflammatory states (Medzhitov, 2008). The physiological purpose of para-inflammation is to restore tissue functionality and homeostasis. Para-inflammation may become chronic or turn into inflammation if tissue stress or malfunction persists for a sustained period. Chronic para-inflammation contributes to the initiation and progression of many human diseases including obesity, type 2 diabetes, atherosclerosis, and age-related neurodegenerative diseases. Evidence from our studies and the studies of some others suggests that para-inflammation also exists in the aging retina in physiological conditions and might contribute to age-related retinal pathologies. The purpose of this review is to introduce the notion of "para-inflammation" as a state between frank, overt destructive inflammation and the non-inflammatory removal of dead or dying cells by apoptosis, to the retinal community. In diabetes and atherosclerosis, leukocytes particularly monocytes and vascular endothelial cells are constantly under noxious stress due to glycaemic and/or lipidaemic dysregulation. These blood-borne stresses trigger para-inflammatory responses in leukocytes and endothelial cells by up-regulating the expression of adhesion molecules or releasing cytokines/chemokines, which in turn cause abnormal leukocyte-endothelial interactions and ultimately vascular damage. In the aging retina, on the other hand, oxidized lipoproteins and free radicals are considered to be major causes of tissue stress and serve as local triggers for retinal para-inflammation. Microarray analysis has revealed the up-regulation of a large number of inflammatory genes, including genes involved in complement activation and inflammatory cytokine/chemokine production, in the aging retina. Para-inflammatory responses in the neuroretina of aged mice are characterized by microglial activation and subretinal migration, and breakdown of blood-retinal barrier. At the retinal/choroidal interface para-inflammation is manifested by complement activation in Bruch's membrane and RPE cells, and microglia accumulation in subretinal space. With age, para-inflammatory changes have also been observed in the choroidal tissue, evidenced by 1) increased thickness of choroid; 2) increased number of CD45(+)CRIg(+) macrophages; 3) morphological abnormalities in choroidal melanocytes; and 4) fibrosis in choroidal tissue. An increased knowledge of contribution of retinal para-inflammation to various pathological conditions is essential for the better understanding of the pathogenesis of various age-related retinal diseases including diabetic retinopathy, glaucoma and age-related macular degeneration.
Collapse
Affiliation(s)
- Heping Xu
- Immunology and Infection, Division of Applied Medicine, University of Aberdeen School of Medicine, Foresterhill, UK.
| | | | | |
Collapse
|
12
|
An RPE cell line as a useful in vitro model for studying retinoic acid receptor beta: expression and affinity. Biosci Rep 2009; 28:327-34. [PMID: 18673301 DOI: 10.1042/bsr20080103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Retinoids mediate their biological effect by interacting with specific nuclear receptors. Of the several known RAR (retinoic acid receptor) subtypes, RAR-beta is of particular interest, since its expression is silenced in many cancers and it is believed to be a tumour suppressor. Specific ligands of RAR-beta can potentially be used in anti-cancer therapy. In the present study, we have investigated the feasibility of using HRPE cells (human retinal pigment epithelial cells) as an experimental model for characterizing RAR-beta-ligand interaction. RT-PCR (reverse transcription-PCR) and Western blot analyses show that HRPE cells specifically express only RAR-beta and none of the other receptor subtypes. In addition, we show that the expression of RAR-beta increases with increasing passage number of the cells. Interestingly, the increase in RAR-beta expression is not associated with telomere shortening, a typical biomarker of cellular senescence. In the present study, we also describe a protocol for characterizing RAR-beta-ligand interactions using nuclear extract from late passage HRPE cells as a source of endogenous RAR-beta. Using [(3)H]CD367 as the ligand, RAR-beta in HRPE cells showed an affinity of 9.6 +/- 0.6 nM and a B(max) of 780 +/- 14 fmol/mg of protein. We have confirmed the feasibility of using this assay to detect the interaction of ligands with RAR-beta by investigating the ability of certain flavonoids to inhibit the binding of [(3)H]CD367 to nuclear extracts from HRPE cells. The inhibition constant of the flavonoids for RAR-beta was between approx. 1-30 microM, showing that the flavonoids interact with RAR-beta with low affinity.
Collapse
|
13
|
Shitama T, Hayashi H, Noge S, Uchio E, Oshima K, Haniu H, Takemori N, Komori N, Matsumoto H. Proteome Profiling of Vitreoretinal Diseases by Cluster Analysis. Proteomics Clin Appl 2008; 2:1265-1280. [PMID: 19081814 DOI: 10.1002/prca.200800017] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Vitreous samples collected in retinopathic surgeries have diverse properties, making proteomics analysis difficult. We report a cluster analysis to evade this difficulty. Vitreous and subretinal fluid samples were collected from 60 patients during surgical operation of non-proliferative diabetic retinopathy, proliferative diabetic retinopathy, proliferative vitreoretinopathy, and rhegmatogenous retinal detachment. For controls we collected vitreous fluid from patients of idiopathic macular hole, epiretinal, and from a healthy postmortem donor. Proteins from these samples were subjected to quantitative proteomics using two-dimensional gel electrophoresis. We selected 105 proteins robustly expressed among ca 400 protein spots and subjected them to permutation test. By using permutation test analysis we observed unique variations in the expression of some of these proteins in vitreoretinal diseases when compared to the control and to each other: 1) the levels of inflammation-associate proteins such as AAT, APOA4, ALB, and TF were significantly higher in all four types of vitreoretinal diseases, and 2) each vitreoretinal disease elevates a unique set of proteins which can be interpreted based on the pathology of retinopathy. Our protocol will be effective for the study of protein expression in other types of clinical samples of diverse property.
Collapse
Affiliation(s)
- Tomomi Shitama
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, P.O. Box 26901, Oklahoma City, OK73190, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Glotin AL, Debacq-Chainiaux F, Brossas JY, Faussat AM, Tréton J, Zubielewicz A, Toussaint O, Mascarelli F. Prematurely senescent ARPE-19 cells display features of age-related macular degeneration. Free Radic Biol Med 2008; 44:1348-61. [PMID: 18226607 DOI: 10.1016/j.freeradbiomed.2007.12.023] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Revised: 12/07/2007] [Accepted: 12/12/2007] [Indexed: 10/22/2022]
Abstract
The etiology of age-related macular degeneration (AMD), the leading cause of blindness in the developed world, remains poorly understood, but may be related to cumulative oxidative stress. The prime target of the disease is the retinal pigmented epithelium (RPE). To study the molecular mechanisms underlying RPE degeneration, we investigated whether repetitive oxidative stress induced premature senescence in RPE cells from the human ARPE-19 cell line. After exposure to 8 mM tert-butylhydroperoxide (tert-BHP) for 1 h daily for 5 days, the cells showed four well-known senescence biomarkers: hypertrophy, senescence-associated beta-galactosidase activity, growth arrest, and cell cycle arrest in G1. A specific low-density array followed by qRT-PCR validation allowed us to identify 36 senescence-associated genes differentially expressed in the prematurely senescent cells. Functional analysis demonstrated that premature senescence induced amyloid beta secretion, resistance to acute stress by tert-BHP and amyloid beta, and defects in adhesion and transepithelial permeability. Coculture assays with choroidal endothelial cells showed the proangiogenic properties of the senescent RPE cells. These results demonstrate that chronic oxidative stress induces premature senescence in RPE cells that modifies the transcriptome and substantially alters cell processes involved in the pathophysiology of AMD. Oxidative stress-induced premature senescence may represent an in vitro model for screening therapeutics against AMD and other retinal degeneration disorders.
Collapse
Affiliation(s)
- Anne-Lise Glotin
- Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, Paris F-75006, France
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Othman M, Lu C, Klueber K, Winstead W, Roisen F. Clonal analysis of adult human olfactory neurosphere forming cells. Biotech Histochem 2006; 80:189-200. [PMID: 16720519 DOI: 10.1080/10520290500469777] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Olfactory neuroepithelium (ONe) is unique because it contains progenitor cells capable of mitotic division that replace damaged or lost neurons throughout life. We isolated populations of ONe progenitors from adult cadavers and patients undergoing nasal sinus surgery that were heterogeneous and consisted of neuronal and glial progenitors. Progenitor lines have been obtained from these cultures that continue to divide and form nestin positive neurospheres. In the present study, we used clonal and population analyses to probe the self-renewal and multipotency of the neurosphere forming cells (NSFCs). NSFCs plated at the single cell level produced additional neurospheres; dissociation of these spheres resulted in mitotically active cells that continued to divide and produce spheres as long as they were subcultured. The mitotic activity of clonal NSFCs was assessed using bromodeoxyuridine (BrdU) incorporation. Lineage restriction of the clonal cultures was determined using a variety of antibodies that were characteristic of different levels of neuronal commitment: ss-tubulin isotype III, neural cell adhesion molecule (NCAM) and microtubule associated protein (MAP2), or glial restriction: astrocytes, glial fibrillary acidic protein (GFAP); and oligodendrocytes, galactocerebroside (GalC). Furthermore, nestin expression, a marker indicative of progenitor nature, decreased in defined medium compared to serum-containing medium. Therefore, adult human ONe-derived neural progenitors retain their capacity for self-renewal, can be clonally expanded, and offer multipotent lineage restriction. Therefore, they are a unique source of progenitors for future cell replacement strategies in the treatment of neurotrauma and neurodegenerative diseases.
Collapse
Affiliation(s)
- M Othman
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, Kentucky 40292, USA
| | | | | | | | | |
Collapse
|