1
|
Jeltsch BM, Hanson JVM, Füglistaler J, Heyard R, Sisera L, Wehrle FM, Hagmann CF, Fauchère JC, Gerth-Kahlert C. The Effect of Perinatal High-Dose Erythropoietin on Retinal Structural and Vascular Characteristics in Children Born Preterm. Am J Ophthalmol 2024; 266:264-273. [PMID: 38880371 DOI: 10.1016/j.ajo.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024]
Abstract
PURPOSE To study the long-term effects of perinatal high-dose recombinant human erythropoietin (rhEPO) on macular structural and vascular development in preterm children. DESIGN Randomized, double-blind clinical trial follow-up plus cohort study. METHODS Setting: Department of Ophthalmology, University Hospital Zurich, Zurich, Switzerland. STUDY POPULATION extremely or very preterm born children aged 7-15 years from an ongoing neuropediatric study (EpoKids). These had been previously randomized to receive either high-dose rhEPO or placebo perinatally. INCLUSION CRITERIA participation in the EpoKids Study, written informed consent (IC). EXCLUSION CRITERIA previous ocular trauma or surgery; retinal or developmental disease unrelated to prematurity. Term-born children of comparable age were enrolled as a healthy control (HC) group. INCLUSION CRITERIA term birth, IC. EXCLUSION CRITERIA any ocular or visual abnormality, high refractive error. Examiners were blinded regarding intervention status until completion of all analyses. (Participants/guardians remain blinded). OBSERVATION PROCEDURES Spectral-domain OCT scans (Heidelberg Spectralis system) and OCTA imaging (Zeiss PlexElite 9000) were obtained. Ophthalmological and orthoptic examinations excluded ocular comorbidities. MAIN OUTCOME MEASURES OCT (central retinal thickness, CRT; total macular volume, TMV), superficial plexus OCTA (foveal avascular zone, FAZ; vessel density, VD; vessel length density, VLD) parameters and foveal hypoplasia grade according to published criteria. RESULTS Macular vessel density parameters (VD and VLD) were significantly lower (p =0.015, CI-95: 0.01 to 0.06 and p=0.015, CI-95: 0.74 to 3.64) in the EPO group (n= 52) when compared to placebo (n=35). No other significant differences were observed between the EPO and placebo group. When comparing the intervention subgroups to HC we found six significant differences in OCT and OCTA parameters (FAZ, VD, VLD and CRT comparing HC and EPO group; FAZ and CRT when comparing HC and placebo group). CONCLUSIONS Early high-dose rhEPO in infants born extremely or very preterm affects macular vessel density parameters compared to placebo. Premature birth (regardless of intervention status) affects retinal structure and vascular development. Our findings on macular vascular development do not contraindicate the administration of early high-dose EPO in preterm infants. For further understanding of the role of EPO on macular development and its clinical significance, future studies are needed.
Collapse
Affiliation(s)
- Brida M Jeltsch
- From the Department of Ophthalmology (B.M.J., J.V.M.H., L.S., C.G-K.), University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - James V M Hanson
- From the Department of Ophthalmology (B.M.J., J.V.M.H., L.S., C.G-K.), University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Jonas Füglistaler
- Department of Biostatistics, Epidemiology, Biostatistics and Prevention Institute (J.F., R.H.), University of Zurich, Zurich, Switzerland
| | - Rachel Heyard
- Department of Biostatistics, Epidemiology, Biostatistics and Prevention Institute (J.F., R.H.), University of Zurich, Zurich, Switzerland
| | - Lorena Sisera
- From the Department of Ophthalmology (B.M.J., J.V.M.H., L.S., C.G-K.), University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Flavia M Wehrle
- Department of Neonatology and Pediatric Intensive Care (F.M.W., C.F.H), University Children's Hospital Zurich, Zurich, Switzerland
| | - Cornelia F Hagmann
- Department of Neonatology and Pediatric Intensive Care (F.M.W., C.F.H), University Children's Hospital Zurich, Zurich, Switzerland
| | - Jean-Claude Fauchère
- Department of Neonatology (J-C.F.), University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Christina Gerth-Kahlert
- From the Department of Ophthalmology (B.M.J., J.V.M.H., L.S., C.G-K.), University Hospital Zurich and University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Systemic Cytokines in Retinopathy of Prematurity. J Pers Med 2023; 13:jpm13020291. [PMID: 36836525 PMCID: PMC9966226 DOI: 10.3390/jpm13020291] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Retinopathy of prematurity (ROP), a vasoproliferative vitreoretinal disorder, is the leading cause of childhood blindness worldwide. Although angiogenic pathways have been the main focus, cytokine-mediated inflammation is also involved in ROP etiology. Herein, we illustrate the characteristics and actions of all cytokines involved in ROP pathogenesis. The two-phase (vaso-obliteration followed by vasoproliferation) theory outlines the evaluation of cytokines in a time-dependent manner. Levels of cytokines may even differ between the blood and the vitreous. Data from animal models of oxygen-induced retinopathy are also valuable. Although conventional cryotherapy and laser photocoagulation are well established and anti-vascular endothelial growth factor agents are available, less destructive novel therapeutics that can precisely target the signaling pathways are required. Linking the cytokines involved in ROP to other maternal and neonatal diseases and conditions provides insights into the management of ROP. Suppressing disordered retinal angiogenesis via the modulation of hypoxia-inducible factor, supplementation of insulin-like growth factor (IGF)-1/IGF-binding protein 3 complex, erythropoietin, and its derivatives, polyunsaturated fatty acids, and inhibition of secretogranin III have attracted the attention of researchers. Recently, gut microbiota modulation, non-coding RNAs, and gene therapies have shown promise in regulating ROP. These emerging therapeutics can be used to treat preterm infants with ROP.
Collapse
|
3
|
Erythropoietin in Glaucoma: From Mechanism to Therapy. Int J Mol Sci 2023; 24:ijms24032985. [PMID: 36769310 PMCID: PMC9917746 DOI: 10.3390/ijms24032985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Glaucoma can cause irreversible vision loss and is the second leading cause of blindness worldwide. The disease mechanism is complex and various factors have been implicated in its pathogenesis, including ischemia, excessive oxidative stress, neurotropic factor deprivation, and neuron excitotoxicity. Erythropoietin (EPO) is a hormone that induces erythropoiesis in response to hypoxia. However, studies have shown that EPO also has neuroprotective effects and may be useful for rescuing apoptotic retinal ganglion cells in glaucoma. This article explores the relationship between EPO and glaucoma and summarizes preclinical experiments that have used EPO to treat glaucoma, with an aim to provide a different perspective from the current view that glaucoma is incurable.
Collapse
|
4
|
Lv W, Chen W, Huang S, Xu Y, Liang JJ, Zheng Y, Chen S, Chen SL, Ng TK, Chen H. Reduction of Laser-Induced Choroidal Neovascularization in Mice With Erythropoietin RNA Interference. Transl Vis Sci Technol 2022; 11:1. [PMID: 35913417 PMCID: PMC9351596 DOI: 10.1167/tvst.11.8.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE The purpose of this study was to evaluate the pathological involvement of erythropoietin (EPO) in experimental choroidal neovascularization (CNV) and its association with neovascular age-related macular degeneration (AMD) and polypoidal choroidal vasculopathy (PCV) in the Chinese population. METHODS Treatment effect of recombinant EPO protein were assessed by human umbilical vein endothelial cell (HUVEC) proliferation, migration, and tube formation, and ex vivo choroid-sprouting ability. The effect of intravitreal injection of Epo siRNA against neovascularization was evaluated in the laser-induced CNV mouse model. In addition, the association of EPO variants with neovascular AMD and PCV was determined. RESULTS Exogenous supplementation of EPO significantly enhanced the migration and tube formation of HUVECs and promoted ex vivo choroid sprouting in mouse retinal pigment epithelium (RPE)-choroid-sclera complex culture. In the experimental CNV mouse model, Epo expression was found to be significantly upregulated by 3.5-folds in RPE-choroid-sclera complex at day 10 after laser induction as compared to the baseline. Immunofluorescence analysis showed that Epo was mainly expressed around the vascular endothelial cells in the RPE-choroid-sclera complex. Intravitreal injection of siRNA targeting Epo reduced 40% Epo expression and 40% CNV lesion areas as compared to the scramble control. However, EPO variants were not associated with neovascular AMD nor PCV in the Chinese population. CONCLUSIONS This study revealed the promotion of human endothelial cell tube formation in vitro and choroid sprouting ex vivo by EPO, and the reduction of laser-induced CNV in vivo by Epo RNA interference. TRANSLATIONAL RELEVANCE Targeting EPO could be a potential additional treatment for CNV-related diseases.
Collapse
Affiliation(s)
- Wenjuan Lv
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Wen Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Shaofen Huang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Yanxuan Xu
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Jia-Jian Liang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Yuqian Zheng
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Shaowan Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Shao-Lang Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Ophthalmology and Visual Sciences, the Chinese University of Hong Kong, Hong Kong
| | - Haoyu Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| |
Collapse
|
5
|
Lai YF, Lin TY, Ho PK, Chen YH, Huang YC, Lu DW. Erythropoietin in Optic Neuropathies: Current Future Strategies for Optic Nerve Protection and Repair. Int J Mol Sci 2022; 23:ijms23137143. [PMID: 35806148 PMCID: PMC9267007 DOI: 10.3390/ijms23137143] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 02/05/2023] Open
Abstract
Erythropoietin (EPO) is known as a hormone for erythropoiesis in response to anemia and hypoxia. However, the effect of EPO is not only limited to hematopoietic tissue. Several studies have highlighted the neuroprotective function of EPO in extra-hematopoietic tissues, especially the retina. EPO could interact with its heterodimer receptor (EPOR/βcR) to exert its anti-apoptosis, anti-inflammation and anti-oxidation effects in preventing retinal ganglion cells death through different intracellular signaling pathways. In this review, we summarized the available pre-clinical studies of EPO in treating glaucomatous optic neuropathy, optic neuritis, non-arteritic anterior ischemic optic neuropathy and traumatic optic neuropathy. In addition, we explore the future strategies of EPO for optic nerve protection and repair, including advances in EPO derivates, and EPO deliveries. These strategies will lead to a new chapter in the treatment of optic neuropathy.
Collapse
Affiliation(s)
- Yi-Fen Lai
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-F.L.); (T.-Y.L.); (Y.-H.C.)
| | - Ting-Yi Lin
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-F.L.); (T.-Y.L.); (Y.-H.C.)
| | - Pin-Kuan Ho
- School of Dentistry, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Yi-Hao Chen
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-F.L.); (T.-Y.L.); (Y.-H.C.)
| | - Yu-Chuan Huang
- School of Pharmacy, National Defense Medical Center, Taipei 11490, Taiwan
- Department of Research and Development, National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence: (Y.-C.H.); (D.-W.L.); Tel.: +886-2-87923100 (Y.-C.H.); +886-2-87927163 (D.-W.L.)
| | - Da-Wen Lu
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-F.L.); (T.-Y.L.); (Y.-H.C.)
- Correspondence: (Y.-C.H.); (D.-W.L.); Tel.: +886-2-87923100 (Y.-C.H.); +886-2-87927163 (D.-W.L.)
| |
Collapse
|
6
|
Abd Rashid M, Chenshen L, Ee Hwan AK, AlSaeedy H, Mok PL, Subbiah SK, Isa HM, Yong TK, Nizam Harun MH, B S Mohamad SMS, Min Hwei AN, Luu CD, Catherine Bastion ML. Rescue of photoreceptor with human mesenchyme stem cell and human mesenchyme stem cell expressing erythropoietin in total degeneration of retina animal model. Indian J Ophthalmol 2022; 70:921-929. [PMID: 35225544 PMCID: PMC9114553 DOI: 10.4103/ijo.ijo_472_21] [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: 12/01/2022] Open
Abstract
Purpose: This study aimed to investigate the efficacy of human-derived umbilical cord mesenchymal stem cells (HDUMSC) and human-derived umbilical cord mesenchymal stem cells expressing erythropoietin (HDUMSC-EPO) to rescue total degenerated retina in a rat model. Methods: The study included four treatment groups, namely negative control using normal saline (HBSS) injection, positive control using sodium iodide 60 mg/kg (SI), SI treated with HDUMSC, and SI treated with HDUMSC-EPO given via subretinal and intravenous routes, to test the efficacy of retinal regeneration following SI-induced retinal degeneration. Retinal function in both phases was tested via electroretinography (ERG) and histological staining examining the outer nuclear layer (ONL). Results: There was a statistically significant result (P < 0.05) in the SI treated with HDUMSC-EPO only when comparing day 11 (mean = 23.6 μv), day 18 (mean = 25.2 μv), day 26 (mean = 26.3 μv), and day 32 (mean = 28.2 μv) to the b-wave ERG on day 4 rescue injection day (mean = 12.5 μv). The SI treated with HDUMSC-EPO showed significant improvement in b-wave ERG readings in the Sprague–Dawley (SD) rat but did not restore baseline readings prior to degeneration (day 0). Both treated groups’ ONL thicknesses did not show significant changes compared to the negative control group (HBSS) following rescue therapy. Conclusion: Total retinal degeneration following intravenous SI injection was observed at 60 mg/kg. SI treated with HDUMSC and HDUMSC-EPO showed no regenerative potential compared to baseline in SI-induced total retina degeneration on ERG or histology, whereas SI treated with HDUMSC-EPO group showed a substantial increase in b-wave ERG amplitude over time.
Collapse
Affiliation(s)
- Munirah Abd Rashid
- Department of Ophthalmology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM) Medical Centre, Sakaka, Al-Jawf Province, Saudi Arabia
| | - Lam Chenshen
- Department of Ophthalmology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM) Medical Centre, Sakaka, Al-Jawf Province, Saudi Arabia
| | - Avin Koh Ee Hwan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Al-Jawf Province, Saudi Arabia; Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hiba AlSaeedy
- Department of Medical Microbiology, Universiti Putra Malaysia, Serdang, Malaysia
| | - Pooi Ling Mok
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Al-Jawf Province, Saudi Arabia; Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Suresh Kumar Subbiah
- Department of Medical Microbiology, Universiti Putra Malaysia, Serdang, Malaysia; Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| | - Hazlita Md Isa
- Department of Ophthalmology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM) Medical Centre, Sakaka, Al-Jawf Province, Saudi Arabia
| | - Then Kong Yong
- Department of Ophthalmology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM) Medical Centre; Brighton Healthcare, UKM Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Mohd Hairul Nizam Harun
- Department of Ophthalmology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM) Medical Centre, Sakaka, Al-Jawf Province, Saudi Arabia
| | - Syed Mohamed Suhail B S Mohamad
- Department of Ophthalmology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM) Medical Centre, Sakaka, Al-Jawf Province, Saudi Arabia
| | - Angela Ng Min Hwei
- Tissue Engineering Centre, UKM Medical Centre, Cheras, Kuala Lumpur,, Malaysia
| | - Chi D Luu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital; Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Mae-Lynn Catherine Bastion
- Department of Ophthalmology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM) Medical Centre, Sakaka, Al-Jawf Province, Saudi Arabia
| |
Collapse
|
7
|
Maiese K. A Common Link in Neurovascular Regenerative Pathways: Protein Kinase B (Akt). Curr Neurovasc Res 2022; 19:1-4. [PMID: 35139797 DOI: 10.2174/1567202619666220209111655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
8
|
Nadeem MS, Kazmi I, Ullah I, Muhammad K, Anwar F. Allicin, an Antioxidant and Neuroprotective Agent, Ameliorates Cognitive Impairment. Antioxidants (Basel) 2021; 11:87. [PMID: 35052591 PMCID: PMC8772758 DOI: 10.3390/antiox11010087] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/26/2021] [Accepted: 12/29/2021] [Indexed: 02/08/2023] Open
Abstract
Allicin (diallylthiosulfinate) is a defense molecule produced by cellular contents of garlic (Allium sativum L.). On tissue damage, the non-proteinogenic amino acid alliin (S-allylcysteine sulfoxide) is converted to allicin in an enzyme-mediated process catalysed by alliinase. Allicin is hydrophobic in nature, can efficiently cross the cellular membranes and behaves as a reactive sulfur species (RSS) inside the cells. It is physiologically active molecule with the ability to oxidise the thiol groups of glutathione and between cysteine residues in proteins. Allicin has shown anticancer, antimicrobial, antioxidant properties and also serves as an efficient therapeutic agent against cardiovascular diseases. In this context, the present review describes allicin as an antioxidant, and neuroprotective molecule that can ameliorate the cognitive abilities in case of neurodegenerative and neuropsychological disorders. As an antioxidant, allicin fights the reactive oxygen species (ROS) by downregulation of NOX (NADPH oxidizing) enzymes, it can directly interact to reduce the cellular levels of different types of ROS produced by a variety of peroxidases. Most of the neuroprotective actions of allicin are mediated via redox-dependent pathways. Allicin inhibits neuroinflammation by suppressing the ROS production, inhibition of TLR4/MyD88/NF-κB, P38 and JNK pathways. As an inhibitor of cholinesterase and (AChE) and butyrylcholinesterase (BuChE) it can be applied to manage the Alzheimer's disease, helps to maintain the balance of neurotransmitters in case of autism spectrum disorder (ASD) and attention deficit hyperactive syndrome (ADHD). In case of acute traumatic spinal cord injury (SCI) allicin protects neuron damage by regulating inflammation, apoptosis and promoting the expression levels of Nrf2 (nuclear factor erythroid 2-related factor 2). Metal induced neurodegeneration can also be attenuated and cognitive abilities of patients suffering from neurological diseases can be ameliorates by allicin administration.
Collapse
Affiliation(s)
- Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; or
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; or
| | - Inam Ullah
- Department of Biotechnology and Genetic Engineering, Hazara University, Mansehra 21300, Pakistan; (I.U.); (K.M.)
| | - Khushi Muhammad
- Department of Biotechnology and Genetic Engineering, Hazara University, Mansehra 21300, Pakistan; (I.U.); (K.M.)
| | - Firoz Anwar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; or
| |
Collapse
|
9
|
Chuang CJ, Wang M, Yeh JH, Chen TC, Tsou SC, Lee YJ, Chang YY, Lin HW. The Protective Effects of α-Mangostin Attenuate Sodium Iodate-Induced Cytotoxicity and Oxidative Injury via Mediating SIRT-3 Inactivation via the PI3K/AKT/PGC-1 α Pathway. Antioxidants (Basel) 2021; 10:1870. [PMID: 34942973 PMCID: PMC8698330 DOI: 10.3390/antiox10121870] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 11/16/2022] Open
Abstract
It is well known that age-related macular degeneration (AMD) is an irreversible neurodegenerative disease that can cause blindness in the elderly. Oxidative stress-induced retinal pigment epithelial (RPE) cell damage is a part of the pathogenesis of AMD. In this study, we evaluated the protective effect and mechanisms of alpha-mangostin (α-mangostin, α-MG) against NaIO3-induced reactive oxygen species (ROS)-dependent toxicity, which activates apoptosis in vivo and in vitro. MTT assay and flow cytometry demonstrated that the pretreatment of ARPE-19 cells with α-MG (0, 3.75, 7.5, and 15 μM) significantly increased cell viability and reduced apoptosis from NaIO3-induced oxidative stress in a concentration-dependent manner, which was achieved by the inhibition of Bax, cleaved PARP-1, cleaved caspase-3 protein expression, and enhancement of Bcl-2 protein. Furthermore, pre-incubation of ARPE-19 cells with α-MG markedly inhibited the intracellular ROS and extracellular H2O2 generation via blocking of the abnormal enzyme activities of superoxide dismutase (SOD), the downregulated levels of catalase (CAT), and the endogenous antioxidant, glutathione (GSH), which were regulated by decreasing PI3K-AKT-PGC-1α-STRT-3 signaling in ARPE-19 cells. In addition, our in vivo results indicated that α-MG improved retinal deformation and increased the thickness of both the outer nuclear layer and inner nuclear layer by inhibiting the expression of cleaved caspase-3 protein. Taken together, our results suggest that α-MG effectively protects human ARPE-19 cells from NaIO3-induced oxidative damage via antiapoptotic and antioxidant effects.
Collapse
Affiliation(s)
- Chen-Ju Chuang
- Emergency Department, Kaohsiung Municipal United Hospital, Kaohsiung 80457, Taiwan;
| | - Meilin Wang
- Department of Microbiology and Immunology, School of Medicine, Chung Shan Medical University and Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan;
| | - Jui-Hsuan Yeh
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (J.-H.Y.); (T.-C.C.)
| | - Tzu-Chun Chen
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (J.-H.Y.); (T.-C.C.)
| | - Shang-Chun Tsou
- Department of Nutrition, Chung Shan Medical University, Taichung 40201, Taiwan;
| | - Yi-Ju Lee
- Department of Pathology, Chung-Shan Medical University, Chung-Shan Medical University Hospital, Taichung 40201, Taiwan;
| | - Yuan-Yen Chang
- Department of Microbiology and Immunology, School of Medicine, Chung Shan Medical University and Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan;
| | - Hui-Wen Lin
- Department of Optometry, Asia University, Taichung 41354, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
10
|
Feizi S, Alemzadeh-Ansari M, Karimian F, Esfandiari H. Use of erythropoietin in ophthalmology: a review. Surv Ophthalmol 2021; 67:427-439. [PMID: 34157346 DOI: 10.1016/j.survophthal.2021.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022]
Abstract
Erythropoietin (EPO) is a glycoprotein hormone that regulates hematopoiesis in the human body. The presence of EPO and its receptors in different tissues indicates that this hormone has extramedullary effects in other tissues, including the eye. We focus on the biological roles of this hormone in the development and normal physiologic functions of the eye. Furthermore, we explore the role of EPO in the management of different ocular diseases - including diabetic retinopathy, retinopathy of prematurity, inherited retinal degeneration, branch and central retinal vein occlusion, retinal detachment, traumatic optic neuropathy, optic neuritis, methanol optic neuropathy, nonarteritic anterior ischemic optic neuropathy, glaucoma, and scleral necrosis.
Collapse
Affiliation(s)
- Sepehr Feizi
- Ophthalmic Research Center, Department of Ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Farid Karimian
- Ophthalmic Research Center, Department of Ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Esfandiari
- Department of Ophthalmology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
11
|
Erythropoietin Gene Therapy Delays Retinal Degeneration Resulting from Oxidative Stress in the Retinal Pigment Epithelium. Antioxidants (Basel) 2021; 10:antiox10060842. [PMID: 34070383 PMCID: PMC8229633 DOI: 10.3390/antiox10060842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 02/08/2023] Open
Abstract
Erythropoietin (EPO) plays an important role in erythropoiesis by its action in blocking apoptosis of progenitor cells and protects both photoreceptors and retinal ganglion cells from induced or inherited degeneration. A modified form of EPO, EPO-R76E has attenuated erythropoietic activity but is effective in inhibiting apoptosis, oxidative stress, and inflammation in several models of retinal degeneration. In this study, we used recombinant Adeno Associated Virus (AAV) to provide long-term sustained delivery of EPO-R76E and demonstrated its effects in a mouse model of dry-AMD in which retinal degeneration is induced by oxidative stress in the retinal pigment epithelial (RPE) cells. Experimental vector AAV-EPO-R76E and control vector AAV-GFP were packaged into serotype-1 (AAV1) to enable RPE selective expression. RPE oxidative stress-mediated retinal degeneration was induced by exon specific deletion of the protective enzyme MnSOD (encoded by Sod2) by cre/lox mechanism. Experimental mice received subretinal injection of AAV-EPO-R76E in the right eye and AAV-GFP in the left eye. Western blotting of RPE/choroid protein samples from AAV-EPO-R76E injected eyes showed RPE specific EPO expression. Retinal function was monitored by electroretinography (ERG). EPO-R76E over-expression in RPE delayed the retinal degeneration as measured by light microscopy in RPE specific Sod2 knockout mice. Delivery of EPO-R76E vector can be used as a tool to prevent retinal degeneration induced by RPE oxidative stress, which is implicated as a potential cause of Age-Related Macular Degeneration.
Collapse
|
12
|
Koh AEH, Subbiah SK, Farhana A, Alam MK, Mok PL. Mitigation of Sodium Iodate-Induced Cytotoxicity in Retinal Pigment Epithelial Cells in vitro by Transgenic Erythropoietin-Expressing Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:652065. [PMID: 33937251 PMCID: PMC8082501 DOI: 10.3389/fcell.2021.652065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/15/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSC) have shown promise in restoring the vision of patients in clinical trials. However, this therapeutic effect is not observed in every treated patient and is possibly due to the inefficacies of cell delivery and high cell death following transplantation. Utilizing erythropoietin can significantly enhance the regenerative properties of MSCs and hence improve retinal neuron survivability in oxidative stress. Hence, this study aimed to investigate the efficacy of conditioned medium (CM) obtained from transgenic human erythropoietin-expressing MSCs (MSC EPO ) in protecting human retinal pigment epithelial cells from sodium iodate (NaIO3)-induced cell death. Human MSC and MSC EPO were first cultured to obtain conditioned media (CM). The IC50 of NaIO3 in the ARPE-19 culture was then determined by an MTT assay. After that, the efficacy of both MSC-CM and MSC-CM EPO in ARPE-19 cell survival were compared at 24 and 48 h after NaIO3 treatment with MTT. The treatment effects on mitochondrial membrane potential was then measured by a JC-1 flow cytometric assay. The MTT results indicated a corresponding increase in cell survivability (5-58%) in the ARPE-19 cell cultures. In comparison to MSC-CM, the use of conditioned medium collected from the MSC-CM EPO further enhanced the rate of ARPE-19 survivability at 24 h (P < 0.05) and 48 h (P < 0.05) in the presence of NaIO3. Furthermore, more than 90% were found viable with the JC-1 assay after MSC-CM EPO treatment, showing a positive implication on the mitochondrial dynamics of ARPE-19. The MSC-CM EPO provided an enhanced mitigating effect against NaIO3-induced ARPE-19 cell death over that of MSC-CM alone during the early phase of the treatment, and it may act as a future therapy in treating retinal degenerative diseases.
Collapse
Affiliation(s)
- Avin Ee-Hwan Koh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, Seri Kembangan, Malaysia
| | - Suresh Kumar Subbiah
- Department of Medical Microbiology and Parasitology, Universiti Putra Malaysia, UPM, Seri Kembangan, Malaysia.,Genetics and Regenerative Medicine Research Group, Universiti Putra Malaysia, UPM, Seri Kembangan, Malaysia.,Department of Biotechnology, Bharath Institute of Higher Education and Research, Chennai, India
| | - Aisha Farhana
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | | | - Pooi Ling Mok
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, Seri Kembangan, Malaysia.,Genetics and Regenerative Medicine Research Group, Universiti Putra Malaysia, UPM, Seri Kembangan, Malaysia.,Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| |
Collapse
|
13
|
Cytoprotective effects of erythropoietin: What about the lung? Biomed Pharmacother 2021; 139:111547. [PMID: 33831836 DOI: 10.1016/j.biopha.2021.111547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Erythropoietin (Epo) is a pleiotropic cytokine, essential for erythropoiesis. Epo and its receptor (Epo-R) are produced by several tissues and it is now admitted that Epo displays other physiological functions than red blood cell synthesis. Indeed, Epo provides cytoprotective effects, which consist in prevention or fight against pathological processes. This perspective article reviews the various protective effects of Epo in several organs and tries to give a proof of concept about its effects in the lung. The tissue-protective effects of Epo could be a promising approach to limit the symptoms of acute and chronic lung diseases.
Collapse
|
14
|
Lai TT, Yang CM, Yang CH. Astaxanthin Protects Retinal Photoreceptor Cells against High Glucose-Induced Oxidative Stress by Induction of Antioxidant Enzymes via the PI3K/Akt/Nrf2 Pathway. Antioxidants (Basel) 2020; 9:antiox9080729. [PMID: 32785112 PMCID: PMC7465141 DOI: 10.3390/antiox9080729] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetic retinopathy (DR) is a major microvascular complication that can lead to severe visual impairment in patients with diabetes. The elevated oxidative stress and increased reactive oxygen species (ROS) production induced by hyperglycemia have been reported to play an important role in the complex pathogenesis of DR. Astaxanthin (AST), a natural carotenoid derivative, has been recently recognized as a strong free radical scavenger and might, therefore, be beneficial in different diseases, including DR. In this study, we evaluated the potential role of AST as an antioxidative and antiapoptotic agent in protecting retinal cells and also investigated the involvement of the PI3K/Akt/Nrf2 pathway in AST-mediated effects. We treated high glucose-cultured mouse photoreceptor cells (661W) with different concentrations of AST and analyzed ROS production and cell apoptosis in the different regimens. Moreover, we also analyzed the expression of PI3K, Akt, Nrf2, and Phase II enzymes after AST treatment. Our results showed that AST dose-dependently reduced ROS production and attenuated 661W cell apoptosis in a high glucose environment. Importantly, its protective effect was abolished by treatment with PI3K or Nrf2 inhibitors, indicating the involvement of the PI3K/Akt/Nrf2 pathway. These results suggest AST as a nutritional supplement that could benefit patients with DR.
Collapse
Affiliation(s)
- Tso-Ting Lai
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 100, Taiwan; (T.-T.L.); (C.-M.Y.)
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chung-May Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 100, Taiwan; (T.-T.L.); (C.-M.Y.)
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 100, Taiwan; (T.-T.L.); (C.-M.Y.)
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Correspondence: ; Tel.: +886-2-2312-3456 (ext. 62131); Fax: +886-2-2393-4420
| |
Collapse
|
15
|
Wang SV, Kulldorff M, Poor S, Rice DS, Banks A, Li N, Lii J, Gagne JJ. Screening Medications for Association with Progression to Wet Age-Related Macular Degeneration. Ophthalmology 2020; 128:248-255. [PMID: 32777229 DOI: 10.1016/j.ophtha.2020.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 11/27/2022] Open
Abstract
PURPOSE There is an urgent need for treatments that prevent or delay development to advanced age-related macular degeneration (AMD). Drugs already on the market for other conditions could affect progression to neovascular AMD (nAMD). If identified, these drugs could provide insights for drug development targets. The objective of this study was to use a novel data mining method that can simultaneously evaluate thousands of correlated hypotheses, while adjusting for multiple testing, to screen for associations between drugs and delayed progression to nAMD. DESIGN We applied a nested case-control study to administrative insurance claims data to identify cases with nAMD and risk-set sampled controls that were 1:4 variable ratio matched on age, gender, and recent healthcare use. PARTICIPANTS The study population included cases with nAMD and risk set matched controls. METHODS We used a tree-based scanning method to evaluate associations between hierarchical classifications of drugs that patients were exposed to within 6 months, 7 to 24 months, or ever before their index date. The index date was the date of first nAMD diagnosis in cases. Risk-set sampled controls were assigned the same index date as the case to which they were matched. The study was implemented using Medicare data from New Jersey and Pennsylvania, and national data from IBM MarketScan Research Database. We set an a priori threshold for statistical alerting at P ≤ 0.01 and focused on associations with large magnitude (relative risks ≥ 2.0). MAIN OUTCOME MEASURES Progression to nAMD. RESULTS Of approximately 4000 generic drugs and drug classes evaluated, the method detected 19 distinct drug exposures with statistically significant, large relative risks indicating that cases were less frequently exposed than controls. These included (1) drugs with prior evidence for a causal relationship (e.g., megestrol); (2) drugs without prior evidence for a causal relationship, but potentially worth further exploration (e.g., donepezil, epoetin alfa); (3) drugs with alternative biologic explanations for the association (e.g., sevelamer); and (4) drugs that may have resulted in statistical alerts due to their correlation with drugs that alerted for other reasons. CONCLUSIONS This exploratory drug-screening study identified several potential targets for follow-up studies to further evaluate and determine if they may prevent or delay progression to advanced AMD.
Collapse
Affiliation(s)
- Shirley V Wang
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Martin Kulldorff
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Stephen Poor
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Dennis S Rice
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Angela Banks
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Ning Li
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Joyce Lii
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Joshua J Gagne
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Pakdel F, Sanjari MS, Naderi A, Pirmarzdashti N, Haghighi A, Kashkouli MB. Erythropoietin in Treatment of Methanol Optic Neuropathy. J Neuroophthalmol 2019; 38:167-171. [PMID: 29300238 DOI: 10.1097/wno.0000000000000614] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Methanol poisoning can cause an optic neuropathy that is usually severe and irreversible and often occurs after ingestion of illicit or homemade alcoholic beverages. In this study, we evaluated the potential neuroprotective effect of erythropoietin (EPO) on visual acuity (VA) in patients with methanol optic neuropathy. METHODS In a prospective, noncomparative interventional case series, consecutive patients with methanol optic neuropathy after alcoholic beverage ingestion were included. All patients initially received systemic therapy including metabolic stabilization and detoxification. Treatment with intravenous recombinant human EPO consisted of 20,000 units/day for 3 successive days. Depending on clinical response, some patients received a second course of EPO. VA, funduscopy, and spectral domain optical coherence tomography were assessed during the study. Main outcome measure was VA. RESULTS Thirty-two eyes of 16 patients with methanol optic neuropathy were included. Mean age was 34.2 years (±13.3 years). The mean time interval between methanol ingestion and treatment with intravenous EPO was 9.1 days (±5.56 days). Mean follow-up after treatment was 7.5 months (±5.88 months). Median VA in the better eye of each patient before treatment was light perception (range: 3.90-0.60 logMAR). Median last acuity after treatment in the best eye was 1.00 logMAR (range: 3.90-0.00 logMAR). VA significantly increased in the last follow-up examination (P < 0.0001). Age and time to EPO treatment after methanol ingestion were not significantly related to final VA. No ocular or systemic complications occurred in our patient cohort. CONCLUSIONS Intravenous EPO appears to improve VA in patients with methanol optic neuropathy and may represent a promising treatment for this disorder.
Collapse
Affiliation(s)
- Farzad Pakdel
- Department of Ophthalmology (FP, MSS, MBK), Eye Research Center, Tehran University of Medical Sciences, Rassoul Akram Hospital, Tehran, Iran; Department of Ophthalmology (FP), Eye Research Center, Farabi Hospital, Tehran University of Medical Sciences, Tehran, Iran; Eye Research Center (AN, NP), Farabi Hospital, Tehran University of Medical Sciences; and Department of Internal Medicine (AH), Rassoul Akram Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
17
|
He Y, Wang X, Liu X, Ji Z, Ren Y. Decreased uncoupling protein 2 expression in aging retinal pigment epithelial cells. Int J Ophthalmol 2019; 12:375-380. [PMID: 30918803 DOI: 10.18240/ijo.2019.03.04] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 12/17/2018] [Indexed: 11/23/2022] Open
Abstract
AIM To analyze the expression of uncoupling protein 2 (UCP2) in retinal pigment epithelium (RPE) cells at the different human age, further explore the possible new target of RPE cells protection. METHODS Adult retinal pigment epithelial-19 (ARPE-19) cells and the primary RPE cells at the different age (9-20y, 50-55y, 60-70y, >70y) were cultured and harvested. The expression of UCP2 in these cells was detected by reverse transcription-polymerase chain reaction (RT-PCR), Western blot and confocal microscopy. RESULTS Cells from the donors more than 60y are larger and more fibroblastic in appearance compared to ARPE-19 cells and those primary cultures obtained from the younger individuals by using phase-contrast micrographs. Results of RT-PCR, Western blot and confocal microscopy all showed that UCP2 was highly expressed in ARPE-19 cells and in the younger primary cultured human RPE cells at the age of 9-20y and 50-55y, whereas lower expression of UCP2 was measured in the older primary cultured human RPE cells at the age more than 60y. CONCLUSION Expression of UCP2 gene is decreased in aged RPE cells, promoting the lower ability of anti-oxidation in these cells. It is indicated that UCP2 gene might be a new target for protecting the cells from oxidative stress damage.
Collapse
Affiliation(s)
- Yuan He
- Department of Ophthalmology, the Second Affiliated Hospital of Xi'an Medical University; Ocular immunology and Inflammation Institute; Shaanxi Provincial Clinical Research Center for Ophthalmology, Xi'an 710038, Shaanxi Province, China
| | - Xia Wang
- Department of Ophthalmology, the Second Affiliated Hospital of Xi'an Medical University; Ocular immunology and Inflammation Institute; Shaanxi Provincial Clinical Research Center for Ophthalmology, Xi'an 710038, Shaanxi Province, China
| | - Xu Liu
- Department of Ophthalmology, the Second Affiliated Hospital of Xi'an Medical University; Ocular immunology and Inflammation Institute; Shaanxi Provincial Clinical Research Center for Ophthalmology, Xi'an 710038, Shaanxi Province, China
| | - Zhi Ji
- Department of Ophthalmology, the Second Affiliated Hospital of Xi'an Medical University; Ocular immunology and Inflammation Institute; Shaanxi Provincial Clinical Research Center for Ophthalmology, Xi'an 710038, Shaanxi Province, China
| | - Yuan Ren
- Department of Ophthalmology, the Second Affiliated Hospital of Xi'an Medical University; Ocular immunology and Inflammation Institute; Shaanxi Provincial Clinical Research Center for Ophthalmology, Xi'an 710038, Shaanxi Province, China
| |
Collapse
|
18
|
Aouiss A, Anka Idrissi D, Kabine M, Zaid Y. Update of inflammatory proliferative retinopathy: Ischemia, hypoxia and angiogenesis. Curr Res Transl Med 2019; 67:62-71. [PMID: 30685380 DOI: 10.1016/j.retram.2019.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/19/2018] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Abstract
Diabetic retinopathy (DR) and retinopathy of prematurity (ROP) present two examples of proliferative retinopathy, characterized by the same stages of progression; ischemia of the retinal vessels, leads to hypoxia and to correct the problem there is the setting up of uncontrolled angiogenesis, which subsequently causes blindness or even detachment of the retina. The difference is the following; that DR initiated by the metabolic complications that are due to hyperglycemia, and ROP is induced by overexposure of the neonatal retina to oxygen. In this review, we will demonstrate the physiopathological mechanism of the two forms of proliferative retinopathy DR and ROP, in particular the role of the CD40/CD40L axis and IL-1 on vascular complications and onset of inflammation of the retina, the implications of their effects on the onset of pathogenic angiogenesis, thus understanding the link between platelets and retinal ischemia. In addition, what are the therapeutic targets that could slow its progression?
Collapse
Affiliation(s)
- A Aouiss
- Laboratory of Health and Environment, Department of Biology, Faculty of Sciences Ain Chock, University of Hassan II, Casablanca, Morocco.
| | - D Anka Idrissi
- Laboratory of Health and Environment, Department of Biology, Faculty of Sciences Ain Chock, University of Hassan II, Casablanca, Morocco
| | - M Kabine
- Laboratory of Health and Environment, Department of Biology, Faculty of Sciences Ain Chock, University of Hassan II, Casablanca, Morocco
| | - Y Zaid
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Montreal, H1T1C8, Quebec, Canada
| |
Collapse
|
19
|
Shirley Ding SL, Kumar S, Ali Khan MS, Ling Mok P. Human Mesenchymal Stem Cells Expressing Erythropoietin Enhance Survivability of Retinal Neurons Against Oxidative Stress: An In Vitro Study. Front Cell Neurosci 2018; 12:190. [PMID: 30108483 PMCID: PMC6079241 DOI: 10.3389/fncel.2018.00190] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 06/13/2018] [Indexed: 12/15/2022] Open
Abstract
Retinal degeneration is a prominent feature in ocular disorders. In exploring possible treatments, Mesenchymal Stem Cells (MSCs) have been recognized to yield therapeutic role for retinal degenerative diseases. Studies have also displayed that erythropoietin (EPO) administration into degenerative retina models confers significant neuroprotective actions in limiting pathological cell death. In this study, we aimed to use MSCs to deliver EPO and to evaluate the ability of EPO to rescue retinal neurons from dying upon reactive oxidative stress induction. We derived human MSCs from Wharton's jelly (hWJMSCs) of the umbilical cord and cells were transduced with lentivirus particles encoding EPO and a reporter gene of green fluorescent protein (GFP). The supernatants of both transduced and non-transduced cells were collected and used as a pre-conditioning medium for Y79 retinoblastoma cells (retinal neuron cell line) following exposure to glutamate induction. Retinal cells exposed to glutamate showed reduced mitochondrial depolarization and enhanced improvement in cell viability when incubated with pre-conditioned media of transduced cells. Our results established a proof-of-concept that MSCs could be used as a candidate for the delivery of EPO therapeutic gene in the treatment of retinal degenerations.
Collapse
Affiliation(s)
- Suet Lee Shirley Ding
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Seri Kembangan, Malaysia
| | - Suresh Kumar
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Seri Kembangan, Malaysia
- Genetics and Regenerative Medicine Research Centre, Universiti Putra Malaysia, Seri Kembangan, Malaysia
| | - Mohammed Safwan Ali Khan
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
- Department of Pharmacology, Anwarul Uloom College of Pharmacy affiliated to Jawaharlal Nehru Technological University-Hyderabad, Hyderabad, India
| | - Pooi Ling Mok
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Seri Kembangan, Malaysia
- Genetics and Regenerative Medicine Research Centre, Universiti Putra Malaysia, Seri Kembangan, Malaysia
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| |
Collapse
|
20
|
Chen L, Liu M, Luan Y, Liu Y, Zhang Z, Ma B, Liu X, Liu Y. BMP‑6 protects retinal pigment epithelial cells from oxidative stress‑induced injury by inhibiting the MAPK signaling pathways. Int J Mol Med 2018; 42:1096-1105. [PMID: 29767257 DOI: 10.3892/ijmm.2018.3675] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 05/10/2018] [Indexed: 11/06/2022] Open
Abstract
Worldwide, neovascular age‑related macular degeneration (nAMD) is one of the most common causes of blindness in the elderly. In particular, degeneration of retinal pigment epithelial (RPE) cells represents the main pathological process in the development of nAMD, and oxidative stress serves a major role. The present study aimed to investigate the association between bone morphogenetic protein 6 (BMP‑6) and nAMD. BMP‑6 concentration was significantly reduced in patients with wet nAMD compared with in the control group. Furthermore, the present study investigated the protective effects of BMP‑6 on RPE cells following oxidative stress‑induced injury. Cell Counting Kit‑8 assay and terminal deoxynucleotidyl transferase dUTP nick‑end labeling staining demonstrated that BMP‑6 increased RPE cell viability, which was decreased following treatment with hydrogen peroxide (H2O2), and reduced H2O2‑induced apoptosis. In addition, western blotting revealed that BMP‑6 reversed the decrease in pro‑caspase‑3 levels and the dysregulation of the B‑cell lymphoma 2 (Bcl‑2)/Bcl‑2‑associated X protein (Bax) balance caused by H2O2. In addition, alterations in c‑Jun N‑terminal protein kinase (JNK) and p38 mitogen‑activated protein kinase (MAPK) expression were examined, and pretreatment with BMP‑6 was demonstrated to reduce H2O2‑induced activation of JNK and p38 MAPK. Conversely, the effects of BMP‑6 were attenuated by its inhibitor noggin. In conclusion, the present study demonstrated that BMP‑6 may protect RPE cells from oxidative stress injury to a certain extent, which may be associated with alterations in the MAPK signaling pathway. However, the specific mechanism of action underlying this effect requires further investigation. Overall, the present study laid a foundation for exploring novel nAMD treatment methods.
Collapse
Affiliation(s)
- Li Chen
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ming Liu
- Department of Ophthalmology, The First Hospital of Xi'an, Xi'an, Shaanxi 710002, P.R. China
| | - Yan Luan
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Yingfei Liu
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Zhichao Zhang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Bo Ma
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xuan Liu
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yong Liu
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
21
|
de Carvalho LA, Fleming R, Sant’Anna M, Guimarães R, Dantas AM, Morizot-Leite E, Cavalcante LA, Allodi S. Neuroprotective effects of erythropoietin on rat retinas subjected to oligemia. Clinics (Sao Paulo) 2018; 73:e161. [PMID: 29694605 PMCID: PMC5890171 DOI: 10.6061/clinics/2018/e161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 11/27/2017] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Erythropoietin may have neuroprotective potential after ischemia of the central nervous system. Here, we conducted a study to characterize the protective effects of erythropoietin on retinal ganglion cells and gliotic reactions in an experimentally induced oligemia model. METHODS Rats were subjected to global oligemia by bilateral common carotid artery occlusion and then received either vehicle or erythropoietin via intravitreal injection after 48 h; they were euthanized one week after the injection. The densities of retinal ganglion cells and contents of glial fibrillary acidic protein (astrocytes/Müller cells) and cluster of differentiation 68 clone ED1 (microglia/macrophages), assessed by fluorescence intensity, were evaluated in frozen retinal sections by immunofluorescence and epifluorescence microscopy. RESULTS Retinal ganglion cells were nearly undetectable one week after oligemia compared with the sham controls; however, these cells were partially preserved in erythropoietin-treated retinas. The contents of glial fibrillary acidic protein and cluster of differentiation 68 clone ED1, markers for reactive gliosis, were significantly higher in retinas after bilateral common carotid artery occlusion than those in both sham and erythropoietin-treated retinas. CONCLUSIONS The number of partially preserved retinal ganglion cells in the erythropoietin-treated group suggests that erythropoietin exerts a neuroprotective effect on oligemic/ischemic retinas. This effect could be related to the down-modulation of glial reactivity, usually observed in hypoxic conditions, clinically observed during glaucoma or retinal artery occlusion conditions. Therefore, glial reactivity may enhance neurodegeneration in hypoxic conditions, like normal-tension glaucoma and retinal ischemia, and erythropoietin is thus a candidate to be clinically applied after the detection of decreased retinal blood flow.
Collapse
Affiliation(s)
- Litia Alves de Carvalho
- Programa de Neurobiologia, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
- Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
- Corresponding authors. E-mails: /
| | - Renata Fleming
- Programa de Neurobiologia, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
- Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Moysés Sant’Anna
- Programa de Neurobiologia, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
| | - Roberta Guimarães
- Programa de Neurobiologia, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
| | - Adalmir Morterá Dantas
- Hospital Universitário Clementino Fraga Filho, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
| | | | - Leny A. Cavalcante
- Programa de Neurobiologia, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
| | - Silvana Allodi
- Programa de Neurobiologia, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
- Corresponding authors. E-mails: /
| |
Collapse
|
22
|
Rjiba-Touati K, Amara I, Bousabbeh M, Salem IB, Azzebi A, Guedri Y, Achour A, Bacha H, Abid S. Recombinant human erythropoietin prevents etoposide- and methotrexate-induced toxicity in kidney and liver tissues via the regulation of oxidative damage and genotoxicity in Wistar rats. Hum Exp Toxicol 2017; 37:848-858. [PMID: 29069929 DOI: 10.1177/0960327117733553] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Etoposide (ETO) and methotrexate (MTX) are two effective chemotherapeutic drugs. However, the clinical use of these drugs is limited by its toxicity in normal tissues, especially in kidney and in liver tissues. Recombinant human erythropoietin (rhEPO), erythropoietin hormone, has also been shown to exert tissue protective effects. The purpose of this study was to explore the protective effect of rhEPO against oxidative stress and genotoxicity induced by ETO and MTX in vivo. Adult male Wistar rats were divided into 10 groups (6 animals each): control group, rhEPO alone group, ETO alone group, MTX alone group and rhEPO + ETO/MTX groups. In rhEPO + ETO/MTX groups, three doses of pretreatment with rhEPO were performed: 1000, 3000 and 6000 IU/kg. Our results showed that rhEPO pretreatment protects liver and kidney tissues against oxidative stress induced by the anticancer drugs. The glycoprotein decreased malondialdehyde (MDA) levels, reduced catalase activity and ameliorated glutathione depletion. Furthermore, we showed that rhEPO administration prevented drug-induced DNA damage accessed by comet test. Altogether, our results suggested a protective role of rhEPO, especially at 3000 IU/kg, against ETO- and MTX-induced oxidative stress and genotoxicity in vivo.
Collapse
Affiliation(s)
- K Rjiba-Touati
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - I Amara
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - M Bousabbeh
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - I Ben Salem
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - A Azzebi
- 2 Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, Sousse, Tunisia
| | - Y Guedri
- 2 Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, Sousse, Tunisia
| | - A Achour
- 2 Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, Sousse, Tunisia
| | - H Bacha
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - S Abid
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| |
Collapse
|
23
|
Cheng L, Yu H, Yan N, Lai K, Xiang M. Hypoxia-Inducible Factor-1α Target Genes Contribute to Retinal Neuroprotection. Front Cell Neurosci 2017; 11:20. [PMID: 28289375 PMCID: PMC5326762 DOI: 10.3389/fncel.2017.00020] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 01/23/2017] [Indexed: 02/05/2023] Open
Abstract
Hypoxia-inducible factor (HIF) is a transcription factor that facilitates cellular adaptation to hypoxia and ischemia. Long-standing evidence suggests that one isotype of HIF, HIF-1α, is involved in the pathogenesis of various solid tumors and cardiac diseases. However, the role of HIF-1α in retina remains poorly understood. HIF-1α has been recognized as neuroprotective in cerebral ischemia in the past two decades. Additionally, an increasing number of studies has shown that HIF-1α and its target genes contribute to retinal neuroprotection. This review will focus on recent advances in the studies of HIF-1α and its target genes that contribute to retinal neuroprotection. A thorough understanding of the function of HIF-1α and its target genes may lead to identification of novel therapeutic targets for treating degenerative retinal diseases including glaucoma, age-related macular degeneration, diabetic retinopathy, and retinal vein occlusions.
Collapse
Affiliation(s)
- Lin Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University Guangzhou, China
| | - Honghua Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China; Department of Ophthalmology, General Hospital of Guangzhou Military Command of PLAGuangzhou, China
| | - Naihong Yan
- Department of Ophthalmology and Ophthalmic Laboratories, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University Chengdu, China
| | - Kunbei Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University Guangzhou, China
| | - Mengqing Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China; Center for Advanced Biotechnology and Medicine and Department of Pediatrics, Rutgers University-Robert Wood Johnson Medical SchoolPiscataway, NJ, USA
| |
Collapse
|
24
|
Du L, Chen J, Xing YQ. Eupatilin prevents H 2O 2-induced oxidative stress and apoptosis in human retinal pigment epithelial cells. Biomed Pharmacother 2016; 85:136-140. [PMID: 27930977 DOI: 10.1016/j.biopha.2016.11.108] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 11/22/2016] [Accepted: 11/27/2016] [Indexed: 12/31/2022] Open
Abstract
Eupatilin, a pharmacologically active flavone derived from the Artemisia plant species, is known to possess anti-oxidant activity. However, the effects of eupatilin on oxidative stress-induced retinal damage in retinal pigment epithelium (RPE) cells and the potential mechanisms involved have not been explored. Therefore, the aim of this study was to investigate the effects of eupatilin on oxidative stress-induced retinal damage in RPE cells. Our results showed that eupatilin significantly attenuated H2O2-induced cell injury and ROS production in ARPE-19 cells. In addition, eupatilin pretreatment greatly upregulated Bcl-2 expression, downregulated Bax expression, as well as suppressed caspase-3 activity in ARPE-19 cells exposed to H2O2. Furthermore, eupatilin pretreatment markedly enhanced phosphorylation levels of PI3K and Akt in ARPE-19 cells exposed to H2O2. In conclusion, our data showed that eupatilin protected against H2O2-induced oxidative stress and apoptosis through the activation of PI3K/Akt signaling pathway in ARPE-19 cells. Thus, eupatilin may be useful for the prevention or treatment of proliferative vitreoretinopathy (PVR).
Collapse
Affiliation(s)
- Lei Du
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Jia Chen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yi-Qiao Xing
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
25
|
Role of recombinant human erythropoietin loading chitosan-tripolyphosphate nanoparticles in busulfan-induced genotoxicity: Analysis of DNA fragmentation via comet assay in cultured HepG2 cells. Toxicol In Vitro 2016; 36:46-52. [DOI: 10.1016/j.tiv.2016.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/25/2016] [Accepted: 07/05/2016] [Indexed: 02/07/2023]
|
26
|
Chen S, Ren H, Mei Z, Zhuo H, Yang H, Ge Z. Exploring the Biocompatibility of Zwitterionic Copolymers for Controlling Macrophage Phagocytosis of Bacteria. Macromol Biosci 2016; 16:1714-1722. [DOI: 10.1002/mabi.201600306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/22/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Shaojun Chen
- Guangdong Research Center for Interfacial Engineering of Functional Materials; Shenzhen Key Laboratory of Polymer Science and Technology; Nanshan District Key Lab for Biopolymers and Safety Evaluation; College of Materials Science and Engineering; Shenzhen University; Shenzhen 518060 China
| | - Huanhuan Ren
- Guangdong Research Center for Interfacial Engineering of Functional Materials; Shenzhen Key Laboratory of Polymer Science and Technology; Nanshan District Key Lab for Biopolymers and Safety Evaluation; College of Materials Science and Engineering; Shenzhen University; Shenzhen 518060 China
| | - Zhankui Mei
- Guangdong Research Center for Interfacial Engineering of Functional Materials; Shenzhen Key Laboratory of Polymer Science and Technology; Nanshan District Key Lab for Biopolymers and Safety Evaluation; College of Materials Science and Engineering; Shenzhen University; Shenzhen 518060 China
| | - Haitao Zhuo
- College of Chemistry and Environmental Engineering; Shenzhen University; Shenzhen 518060 China
| | - Haipeng Yang
- Guangdong Research Center for Interfacial Engineering of Functional Materials; Shenzhen Key Laboratory of Polymer Science and Technology; Nanshan District Key Lab for Biopolymers and Safety Evaluation; College of Materials Science and Engineering; Shenzhen University; Shenzhen 518060 China
| | - Zaochuan Ge
- Guangdong Research Center for Interfacial Engineering of Functional Materials; Shenzhen Key Laboratory of Polymer Science and Technology; Nanshan District Key Lab for Biopolymers and Safety Evaluation; College of Materials Science and Engineering; Shenzhen University; Shenzhen 518060 China
| |
Collapse
|
27
|
Shirley Ding SL, Leow SN, Munisvaradass R, Koh EH, Bastion MLC, Then KY, Kumar S, Mok PL. Revisiting the role of erythropoietin for treatment of ocular disorders. Eye (Lond) 2016; 30:1293-1309. [PMID: 27285322 DOI: 10.1038/eye.2016.94] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 03/23/2016] [Indexed: 12/15/2022] Open
Abstract
Erythropoietin (EPO) is a glycoprotein hormone conventionally thought to be responsible only in producing red blood cells in our body. However, with the discovery of the presence of EPO and EPO receptors in the retinal layers, the EPO seems to have physiological roles in the eye. In this review, we revisit the role of EPO in the eye. We look into the biological role of EPO in the development of the eye and the physiologic roles that it has. Apart from that, we seek to understand the mechanisms and pathways of EPO that contributes to the therapeutic and pathological conditions of the various ocular disorders such as diabetic retinopathy, retinopathy of prematurity, glaucoma, age-related macular degeneration, optic neuritis, and retinal detachment. With these understandings, we discuss the clinical applications of EPO for treatment of ocular disorders, modes of administration, EPO formulations, current clinical trials, and its future directions.
Collapse
Affiliation(s)
- S L Shirley Ding
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - S N Leow
- Department of Ophthalmology, Hospital Sultanah Aminah, Johor Bahru, Malaysia
| | - R Munisvaradass
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - E H Koh
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - M L C Bastion
- Department of Ophthalmology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - K Y Then
- Department of Ophthalmology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - S Kumar
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia.,Department of Medical Microbiology and Parasitology, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - P L Mok
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia.,Genetics and Regenerative Medicine Research Centre, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| |
Collapse
|
28
|
Kast B, Schori C, Grimm C. Hypoxic preconditioning protects photoreceptors against light damage independently of hypoxia inducible transcription factors in rods. Exp Eye Res 2016; 146:60-71. [DOI: 10.1016/j.exer.2015.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/15/2015] [Accepted: 12/17/2015] [Indexed: 12/17/2022]
|
29
|
Erythropoietin in ophthalmology: A literature review. J Curr Ophthalmol 2016; 28:5-11. [PMID: 27239595 PMCID: PMC4881220 DOI: 10.1016/j.joco.2016.01.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 01/27/2016] [Accepted: 01/27/2016] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To review the current literature on ocular application of erythropoietin (EPO). METHODS A comprehensive search was performed on Pubmed and Scopus databases. All selected articles were reviewed thoroughly by the authors to review current applications of the EPO in ocular diseases. RESULTS Various aspects of administration of EPO for different ischemic, traumatic, vascular, and degenerative disorders have been explained. The articles are generally preclinical with few small studies reporting clinical outcomes. CONCLUSION EPO has been used for the treatment of different ophthalmic conditions with promising results. Further studies are needed to elaborate the role of EPO in management of ocular diseases.
Collapse
|
30
|
TU GERILE, ZHANG YUFENG, WEI WEI, LI LANGEN, ZHANG YANMEI, YANG JIA, XING YIQIAO. Allicin attenuates H2O2-induced cytotoxicity in retinal pigmented epithelial cells by regulating the levels of reactive oxygen species. Mol Med Rep 2016; 13:2320-6. [DOI: 10.3892/mmr.2016.4797] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 12/10/2015] [Indexed: 11/06/2022] Open
|
31
|
Abstract
Globally, greater than 30 million individuals are afflicted with disorders of the nervous system accompanied by tens of thousands of new cases annually with limited, if any, treatment options. Erythropoietin (EPO) offers an exciting and novel therapeutic strategy to address both acute and chronic neurodegenerative disorders. EPO governs a number of critical protective and regenerative mechanisms that can impact apoptotic and autophagic programmed cell death pathways through protein kinase B (Akt), sirtuins, mammalian forkhead transcription factors, and wingless signaling. Translation of the cytoprotective pathways of EPO into clinically effective treatments for some neurodegenerative disorders has been promising, but additional work is necessary. In particular, development of new treatments with erythropoiesis-stimulating agents such as EPO brings several important challenges that involve detrimental vascular outcomes and tumorigenesis. Future work that can effectively and safely harness the complexity of the signaling pathways of EPO will be vital for the fruitful treatment of disorders of the nervous system.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101
| |
Collapse
|
32
|
Maiese K. Erythropoietin and diabetes mellitus. World J Diabetes 2015; 6:1259-1273. [PMID: 26516410 PMCID: PMC4620106 DOI: 10.4239/wjd.v6.i14.1259] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/25/2015] [Accepted: 09/28/2015] [Indexed: 02/05/2023] Open
Abstract
Erythropoietin (EPO) is a 30.4 kDa growth factor and cytokine that governs cell proliferation, immune modulation, metabolic homeostasis, vascular function, and cytoprotection. EPO is under investigation for the treatment of variety of diseases, but appears especially suited for the treatment of disorders of metabolism that include diabetes mellitus (DM). DM and the complications of this disease impact a significant portion of the global population leading to disability and death with currently limited therapeutic options. In addition to its utility for the treatment of anemia, EPO can improve cardiac function, reduce fatigue, and improve cognition in patients with DM as well as regulate cellular energy metabolism, obesity, tissue repair and regeneration, apoptosis, and autophagy in experimental models of DM. Yet, EPO can have adverse effects that involve the vasculature system and unchecked cellular proliferation. Critical to the cytoprotective capacity and the potential for a positive clinical outcome with EPO are the control of signal transduction pathways that include protein kinase B, the mechanistic target of rapamycin, Wnt signaling, mammalian forkhead transcription factors of the O class, silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae), and AMP activated protein kinase. Therapeutic strategies that can specifically target and control EPO and its signaling pathways hold great promise for the development of new and effective clinical treatments for DM and the complications of this disorder.
Collapse
|
33
|
Makarev E, Cantor C, Zhavoronkov A, Buzdin A, Aliper A, Csoka AB. Pathway activation profiling reveals new insights into age-related macular degeneration and provides avenues for therapeutic interventions. Aging (Albany NY) 2015; 6:1064-75. [PMID: 25543336 PMCID: PMC4298366 DOI: 10.18632/aging.100711] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Age-related macular degeneration (AMD) is a major cause of blindness in older people and is caused by loss of the central region of the retinal pigment epithelium (RPE). Conventional methods of gene expression analysis have yielded important insights into AMD pathogenesis, but the precise molecular pathway alterations are still poorly understood. Therefore we developed a new software program, “AMD Medicine”, and discovered differential pathway activation profiles in samples of human RPE/choroid from AMD patients and controls. We identified 29 pathways in RPE-choroid AMD phenotypes: 27 pathways were activated in AMD compared to controls, and 2 pathways were activated in controls compared to AMD. In AMD, we identified a graded activation of pathways related to wound response, complement cascade, and cell survival. Also, there was downregulation of two pathways responsible for apoptosis. Furthermore, significant activation of pro-mitotic pathways is consistent with dedifferentiation and cell proliferation events, which occur early in the pathogenesis of AMD. Significantly, we discovered new global pathway activation signatures of AMD involved in the cell-based inflammatory response: IL-2, STAT3, and ERK. The ultimate aim of our research is to achieve a better understanding of signaling pathways involved in AMD pathology, which will eventually lead to better treatments.
Collapse
Affiliation(s)
- Evgeny Makarev
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Charles Cantor
- Boston University, Boston, MA 02215, USA. Retrotope, Inc, Los Altos Hills, CA 94022, USA
| | - Alex Zhavoronkov
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA. The Biogerontology Research Foundation, London, UK
| | - Anton Buzdin
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA. Pathway Pharmaceutivals, Ltd, Hong Kong
| | - Alexander Aliper
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anotonei Benjamin Csoka
- Vision Genomics, LLC, Washington, DC 20010, USA. Epigenetics Laboratory, Dept. of Anatomy, Howard University, Washington, DC 20059, USA
| |
Collapse
|
34
|
LI LANGEN, WEI WEI, ZHANG YUFENG, TU GERILE, ZHANG YANMEI, YANG JIA, XING YIQIAO. SirT1 and STAT3 protect retinal pigmented epithelium cells against oxidative stress. Mol Med Rep 2015; 12:2231-8. [DOI: 10.3892/mmr.2015.3570] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 02/19/2015] [Indexed: 11/06/2022] Open
|
35
|
Luo W, Hu L, Wang F. The Protective Effect of Erythropoietin on the Retina. Ophthalmic Res 2015; 53:74-81. [DOI: 10.1159/000369885] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/11/2014] [Indexed: 11/19/2022]
|
36
|
Vitamin C protected human retinal pigmented epithelium from oxidant injury depending on regulating SIRT1. ScientificWorldJournal 2014; 2014:750634. [PMID: 25147862 PMCID: PMC4132313 DOI: 10.1155/2014/750634] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/11/2014] [Accepted: 06/30/2014] [Indexed: 11/18/2022] Open
Abstract
The purpose was to investigate the protective effects of Vitamin C (Vit C) and the regulatory mechanism between Vit C and sirtuin 1 (SIRT1) in PREs during oxidative stress as Vit C and SIRT1 exerted famous effects as antioxidants. We found that moderate Vit C (100 µM) prevented ARPE-19 cells from damages induced by H2O2, including increasing viability, reducing apoptosis, and attenuating intracellular ROS levels. But lower and higher concentration of Vit C had no effects. Further results indicated that Vit C caused the dysregulation of some stress responses factors (SIRT1, p53 and FOXO3) in ARPE-19 cells response to H2O2. Moreover we found that SIRT1 activator resveratrol (SRV) stimulated significantly the protective effects of moderate Vit C, provided the property of antioxidative stress for the lower and higher concentration of Vit C in ARPE-19 cells as well. Consistently, nicotinamide (NA) relieved the protective functions of moderate Vit C. Interestingly, data also revealed the dysregulation of p53 and FOXO3 was dependent on the regulation of SIRT1 rather than Vit C. Summarily, the protective effect of Vit C against oxidative stress was involved in regulation of SIRT1. It suggested that combined application of Vit C and RSV might be a promising therapeutic method for AMD.
Collapse
|
37
|
Hu ZX, Chen CL, Yang JS, Zhou ZL, Song ZM, Wang ZY. PI3K-mediated glioprotective effect of epidermal growth factor under oxidative stress conditions. Int J Ophthalmol 2014; 7:413-20. [PMID: 24967183 DOI: 10.3980/j.issn.2222-3959.2014.03.05] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 02/21/2014] [Indexed: 11/02/2022] Open
Abstract
AIM To determine the effects of epidermal growth factor (EGF) on the proliferation and migration of Müller cell line Moorfields/Institute of Ophthalmology-Müller 1 (MIO-M1), and its related molecular mechanisms under normal and oxidative stress conditions. METHODS Müller cells were cultured with different concentrations of EGF in the presence or absence of varied amounts of H2O2 and glucose oxidase (GO) which induced oxidative stress. The proliferation and migration of Müller cells were examined by 5-Bromo-2-deoxyUridine (BrdU), MTT assay, Transwell assay and scratch wound healing assays. The cell viability was determined with the MTT assay. The secretion of EGF by Müller cells was evaluated by ELISA. Western blot was performed to detect the activation of extracellular regulated protein kinases (ERK)1/2 and Akt signal pathways. RESULTS EGF stimulated the proliferation and migration of Müller cells in a concentration-dependent manner in vitro. Under oxidative damage condition, 2h of pretreatment with 10-100 ng/mL EGF can mostly inhibit 50% lethal dose of 0.08 mmol/L H2O2-induced cell damage. The Western blot results showed that after Müller cells were exposed to varying EGF for 24h, Akt and ERK1/2 were phosphorylated in a dose-dependent manner. In the presence of the LY294002, the potent PI3K inhibitor, the p-Akt was significantly attenuated. CONCLUSION EGF may induce the proliferation and migration of human Müller cells through the Akt and the ERK1/2 signal pathways, and induce PI3K-mediated glioprotective effect under oxidative stress.
Collapse
Affiliation(s)
- Zhi-Xiang Hu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical College, Wenzhou 325027, Zhejiang Province, China
| | - Chun-Li Chen
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical College, Wenzhou 325027, Zhejiang Province, China
| | - Jia-Song Yang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical College, Wenzhou 325027, Zhejiang Province, China
| | - Zhong-Lou Zhou
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical College, Wenzhou 325027, Zhejiang Province, China
| | - Zong-Ming Song
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical College, Wenzhou 325027, Zhejiang Province, China
| | - Zhao-Yang Wang
- Department of Ophthalmology, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang Province, China
| |
Collapse
|
38
|
Shin HJ, Kim HC, Moon JW. Aqueous levels of erythropoietin in acute retinal vein occlusion with macular edema. Int J Ophthalmol 2014; 7:501-6. [PMID: 24967199 DOI: 10.3980/j.issn.2222-3959.2014.03.21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 08/12/2013] [Indexed: 11/02/2022] Open
Abstract
AIM To investigate the aqueous erythropoietin (EPO) levels and associated factors in patients with acute retinal vein occlusion (RVO). METHODS The aqueous EPO level was measured in patients with macular edema (ME) secondary to acute branched retinal vein occlusion (BRVO) or central retinal vein occlusion (CRVO). Aqueous fluid from cataract patients served as the control. We also evaluated whether aqueous level of EPO was associated with factors such as serum EPO level, non-perfusion area, central macular thickness (CMT), and arterio-venous (AV) transit time. RESULTS Twenty-seven RVO patients (16 BRVO, 11 CRVO) and 9 control subjects were enrolled in the study. The aqueous EPO level (mU/mL) was higher in RVO (68.2±54.3) than that in the control subjects (12.9±5.9). More specifically, the aqueous EPO level was higher in CRVO (118.9±52.1) than that in BRVO (33.3±10.8). However, no differences were found in serum EPO levels among three groups. CMT in RVO patients had a positive correlation with the aqueous EPO level (r=0.66). Also, in terms of non-perfusion area, the aqueous EPO levels were more elevated in the ischemic subgroup than in the non-ischemic subgroup in both BRVO and CRVO. CONCLUSION Aqueous EPO levels are elevated in patients with macular edema secondary to recent onset RVO. Patients with CRVO have higher EPO levels than those with BRVO. The aqueous EPO level in RVO has a positive correlation with CMT and is associated with non-perfusion area. These results suggest that the aqueous EPO level could be associated with retinal ischemia and may be involved in the pathogenesis of macular edema secondary to RVO.
Collapse
Affiliation(s)
- Hyun Jin Shin
- Department of Ophthalmology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 143-729, Korea
| | - Hyung Chan Kim
- Department of Ophthalmology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 143-729, Korea
| | | |
Collapse
|
39
|
Kim YC, Mungunsukh O, McCart EA, Roehrich PJ, Yee DK, Day RM. Mechanism of erythropoietin regulation by angiotensin II. Mol Pharmacol 2014; 85:898-908. [PMID: 24695083 DOI: 10.1124/mol.113.091157] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Erythropoietin (EPO) is the primary regulator of red blood cell development. Although hypoxic regulation of EPO has been extensively studied, the mechanism(s) for basal regulation of EPO are not well understood. In vivo studies in healthy human volunteers and animal models indicated that angiotensin II (Ang II) and angiotensin converting enzyme inhibitors regulated blood EPO levels. In the current study, we found that Ang II induced EPO expression in situ in murine kidney slices and in 786-O kidney cells in culture as determined by reverse transcription polymerase chain reaction. We further investigated the signaling mechanism of Ang II regulation of EPO in 786-O cells. Pharmacological inhibitors of Ang II type 1 receptor (AT1R) and extracellular signal-regulated kinase 1/2 (ERK1/2) suppressed Ang II transcriptional activation of EPO. Inhibitors of AT2R or Src homology 2 domain-containing tyrosine phosphatase had no effect. Coimmunoprecipiation experiments demonstrated that p21Ras was constitutively bound to the AT1R; this association was increased by Ang II but was reduced by the AT1R inhibitor telmisartan. Transmembrane domain (TM) 2 of AT1R is important for G protein-dependent ERK1/2 activation, and mutant D74E in TM2 blocked Ang II activation of ERK1/2. Ang II signaling induced the nuclear translocation of the Egr-1 transcription factor, and overexpression of dominant-negative Egr-1 blocked EPO promoter activation by Ang II. These data identify a novel pathway for basal regulation of EPO via AT1R-mediated Egr-1 activation by p21Ras-mitogen-activated protein kinase/ERK kinase-ERK1/2. Our current data suggest that Ang II, in addition to regulating blood volume and pressure, may be a master regulator of erythropoiesis.
Collapse
Affiliation(s)
- Yong-Chul Kim
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland (Y.-C.K., O.M., E.A.M., P.J.R., R.M.D.); and Department of Animal Biology, University of Pennsylvania, Philadelphia, Pennsylvania (D.K.Y.)
| | | | | | | | | | | |
Collapse
|
40
|
Bujarborua D, Borooah S, Dhillon B. The stress response as a target for treatment of central serous chorioretinopathy. EXPERT REVIEW OF OPHTHALMOLOGY 2013. [DOI: 10.1586/17469899.2013.837298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
41
|
Rjiba-Touati K, Ayed-Boussema I, Soualeh N, Achour A, Bacha H, Abid S. Antioxidant and antigenotoxic role of recombinant human erythropoeitin against alkylating agents: Cisplatin and mitomycin C in cultured Vero cells. Exp Biol Med (Maywood) 2013; 238:943-50. [DOI: 10.1177/1535370213494643] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Cisplatin (CDDP) and mitomycin C (MMC), two alkylating agents used against various solid tumours, are a common source of acute kidney injury. Thus, strategies for minimizing CDDP and MMC toxicity are of a clinical interest. In this study, we aimed to investigate the protective role of recombinant human erythropoietin (rhEPO) against oxidative stress and genotoxicity induced by CDDP and MMC in cultured Vero cells. Three types of treatments were performed: (i) cells were treated with rhEPO 24 h before exposure to CDDP/MMC (pre-treatment), (ii) cells were treated with rhEPO and CDDP/MMC simultaneously (co-treatment), (iii) cells were treated with rhEPO 24 h after exposure to CDDP/MMC (post-treatment). Our results showed that rhEPO decreased the reactive oxygen species levels, the malondialdehyde levels and ameliorated glutathione (reduced and oxidized glutathione) modulation induced by CDDP and MMC in cultured Vero cells. Furthermore, rhEPO administration prevented alkylating agents-induced DNA damage accessed by comet test. Altogether, our results suggested a protective role of rhEPO, against CDDP- and MMC-induced oxidative stress and genotoxicity, especially in pre-treatment condition.
Collapse
Affiliation(s)
- Karima Rjiba-Touati
- Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, 5019 Monastir, Tunisia
| | - Imen Ayed-Boussema
- Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, 5019 Monastir, Tunisia
| | - Nidhal Soualeh
- Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, 5019 Monastir, Tunisia
| | - Abdellatif Achour
- Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, 4021 Sousse, Tunisia
| | - Hassen Bacha
- Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, 5019 Monastir, Tunisia
| | - Salwa Abid
- Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, 5019 Monastir, Tunisia
| |
Collapse
|
42
|
Alternative route for erythropoietin ocular administration. Graefes Arch Clin Exp Ophthalmol 2013; 251:2051-6. [PMID: 23702929 DOI: 10.1007/s00417-013-2367-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 04/02/2013] [Accepted: 04/22/2013] [Indexed: 02/04/2023] Open
Abstract
PURPOSE This study aimed to find an alternative route for erythropoietin (EPO) ocular administration because of its neuroprotective and neuroregenerative known properties. Ocular penetration of EPO after subconjunctival injection was assessed, and potential side-effects on the haematocrit for a 28-day period were also evaluated. METHODS Wistar Hannover female albino rats (n = 42) divided into seven groups of six were used. One group (n = 6) served as control. Six groups (n = 36) received 1,000 UI of EPO through the subconjunctival route in one of the eyes. According to the group, animals were humanely killed at 12 h (n = 6), 24 h (n = 6), 36 h (n = 6), 48 h (n = 6), and 60 h (n = 6), after EPO administration, in a total of 30 animals. Enucleation of both eyes was performed, and EPO protein distribution in the rat's retina was analyzed by immunohistochemistry. Another group of animals (n = 6) was used to collect blood samples and perform haematocrit analysis at 0, 7, 14, 21, and 28 days after unilateral EPO subconjunctival administration. RESULTS The evaluation of EPO expression in the animals' retinas after subconjunctival administration yielded a strong immunostaining signal. Among the retina's layers, EPO expression was more evident in the RGC layer 24 h after the administration, and was still present on that layer till the end of the study (60 h). When administered subconjunctivally EPO reached several neuronal cells, in all retinal layers. The subconjunctival EPO administration did not cause significant changes in the haematocrit values over a 28-day period. CONCLUSION In this study, it was demonstrated that EPO reached the retinal ganglion cell layers when administered subconjunctivally. EPO reached the retina 24 h after the subconjunctival administration, and was still present 60 h after the administration. Furthermore, it was also proved that EPO subconjunctival administration did not cause any haematopoietic significant side-effects. The subconjunctival route was shown to be a promising alternative for EPO ocular delivery.
Collapse
|
43
|
Hamidi G, Arabpour Z, Shabrang M, Rashidi B, Alaei H, Sharifi MR, Salami M, Reisi P. Erythropoietin improves spatial learning and memory in streptozotocin model of dementia. PATHOPHYSIOLOGY 2013; 20:153-8. [DOI: 10.1016/j.pathophys.2013.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 01/20/2013] [Indexed: 12/12/2022] Open
|
44
|
Wenker SD, Chamorro ME, Vittori DC, Nesse AB. Protective action of erythropoietin on neuronal damage induced by activated microglia. FEBS J 2013; 280:1630-42. [PMID: 23384249 DOI: 10.1111/febs.12172] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 12/22/2012] [Accepted: 01/22/2013] [Indexed: 11/30/2022]
Abstract
Inflammation is a physiological defense response, but may also represent a potential pathological process in neurological diseases. In this regard, microglia have a crucial role in either progression or amelioration of degenerative neuronal damage. Because of the role of hypoxia in pro-inflammatory mechanisms in the nervous system, and the potential anti-inflammatory protective effect of erythropoietin (Epo), we focused our investigation on the role of this factor on activation of microglia and neuroprotection. Activation of microglial cells (EOC-2) was achieved by chemical hypoxia induced by cobalt chloride (CoCl2 ) and characterized by increased levels of nitrite, tumor necrosis factor-α and reactive oxygen species production, as well as up-regulation of inducible nitric oxide synthase expression. Under these conditions, cell proliferation data and proliferating cell nuclear antigen (PCNA) staining demonstrated a mitogenic effect of chemical hypoxia. Even though pre-treatment with Epo did not prevent nitrite production, inducible nitric oxide synthase protein expression or tumor necrosis factor-α secretion, it prevented the oxidative stress induced by CoCl2 as well as cell proliferation. Neuronal cells (SH-SY5Y) cultured in the presence of conditioned medium from activated EOC-2 cells or macrophages (RAW 264.7) developed significant apoptosis, an effect that was abolished by Epo via Epo/Epo receptor activation. The results show that even though Epo did not exert a direct anti-inflammatory effect on microglia activation, it did increase the resistance of neurons to subsequent damage from pro-inflammatory agents. In addition to its anti-apoptotic ability, the Epo antioxidant effect may have an indirect influence on neuronal survival by modulation of the pro-inflammatory environment.
Collapse
Affiliation(s)
- Shirley D Wenker
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, IQUIBICEN-CONICET (Instituto de Química Biológica, Facultad de Ciencias Exactas y Naturales-Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | | | | | | |
Collapse
|
45
|
|
46
|
Photoreceptor degeneration in mice: adeno-associated viral vector-mediated delivery of erythropoietin. Methods Mol Biol 2013; 982:237-63. [PMID: 23456874 DOI: 10.1007/978-1-62703-308-4_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The exogenous delivery of erythropoietin (EPO) and EPO derivatives (EPO-Ds) represents a valuable strategy to protect the retina from degeneration. In this chapter we describe a method to deliver EPO and the EPO derivative S100E in the light-damage model of induced retinal degeneration using adeno--associated viral (AAV) vectors and to evaluate the functional and morphological protection of the retina from light damage.
Collapse
|
47
|
Maiese K, Chong ZZ, Shang YC, Wang S. Erythropoietin: new directions for the nervous system. Int J Mol Sci 2012; 13:11102-11129. [PMID: 23109841 PMCID: PMC3472733 DOI: 10.3390/ijms130911102] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/16/2012] [Accepted: 08/30/2012] [Indexed: 12/14/2022] Open
Abstract
New treatment strategies with erythropoietin (EPO) offer exciting opportunities to prevent the onset and progression of neurodegenerative disorders that currently lack effective therapy and can progress to devastating disability in patients. EPO and its receptor are present in multiple systems of the body and can impact disease progression in the nervous, vascular, and immune systems that ultimately affect disorders such as Alzheimer's disease, Parkinson's disease, retinal injury, stroke, and demyelinating disease. EPO relies upon wingless signaling with Wnt1 and an intimate relationship with the pathways of phosphoinositide 3-kinase (PI 3-K), protein kinase B (Akt), and mammalian target of rapamycin (mTOR). Modulation of these pathways by EPO can govern the apoptotic cascade to control β-catenin, glycogen synthase kinase-3β, mitochondrial permeability, cytochrome c release, and caspase activation. Yet, EPO and each of these downstream pathways require precise biological modulation to avert complications associated with the vascular system, tumorigenesis, and progression of nervous system disorders. Further understanding of the intimate and complex relationship of EPO and the signaling pathways of Wnt, PI 3-K, Akt, and mTOR are critical for the effective clinical translation of these cell pathways into robust treatments for neurodegenerative disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- Cancer Institute of New Jersey, New Brunswick, New Jersey 08901, USA
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| | - Zhao Zhong Chong
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| | - Yan Chen Shang
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| | - Shaohui Wang
- Laboratory of Cellular and Molecular Signaling, Cancer Center, F 1220, New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (Y.C.S.); (S.W.)
- New Jersey Health Sciences University, Newark, New Jersey 07101, USA
| |
Collapse
|
48
|
Hwang JW, Kim EK, Lee SJ, Kim YS, Moon SH, Jeon BT, Sung SH, Kim ET, Park PJ. Antioxidant activity and protective effect of anthocyanin oligomers on H₂O₂-triggered G2/M arrest in retinal cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:4282-4288. [PMID: 22380882 DOI: 10.1021/jf205321j] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
In this study, the free-radical-scavenging properties of anthocyanin oligomers for 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical, alkyl radical, and hydroxyl radical were evaluated using electron spin resonance (ESR) spectroscopy. The DPPH radical, alkyl radical, and hydroxyl radical scavenging activity of anthocyanin oligomers increased in a dose-dependent manner, with the 50% inhibitory concentration (IC₅₀) value of 13.0, 14.0, and 448.0 μg/mL, respectively. The inhibitory effect of anthocyanin oligomers on lipid peroxidation was examined with ferric thiocyanate (FTC) and thiobarbituric acid (TBA). The inhibitory activity of anthocyanin oligomers was found to be comparable to that of vitamin E. In addition, anthocyanin oligomers enhanced the activities of superoxide dismutase (SOD, EC 1.15.1.1), catalase (CAT, EC 1.11.1.6), glutathione peroxidase (GPx, EC 1.11.1.9), and glutathione-S-transferase (GST, EC 2.5.1.18) in ARPE-19 cells. In addition, anthocyanin oligomers inhibited the H₂O₂-induced G2/M phase arrest in ARPE-19 cells. Taken together, the present results demonstrate that anthocyanin oligomers have high antioxidative activity.
Collapse
Affiliation(s)
- Jin-Woo Hwang
- Department of Biotechnology, School of Medicine, Konkuk University, Chungju 380-701, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Basile LA, Ellefson D, Gluzman-Poltorak Z, Junes-Gill K, Mar V, Mendonca S, Miller JD, Tom J, Trinh A, Gallaher TK. HemaMax™, a recombinant human interleukin-12, is a potent mitigator of acute radiation injury in mice and non-human primates. PLoS One 2012; 7:e30434. [PMID: 22383962 PMCID: PMC3286478 DOI: 10.1371/journal.pone.0030434] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 12/18/2011] [Indexed: 01/10/2023] Open
Abstract
HemaMax, a recombinant human interleukin-12 (IL-12), is under development to address an unmet medical need for effective treatments against acute radiation syndrome due to radiological terrorism or accident when administered at least 24 hours after radiation exposure. This study investigated pharmacokinetics, pharmacodynamics, and efficacy of m-HemaMax (recombinant murine IL-12), and HemaMax to increase survival after total body irradiation (TBI) in mice and rhesus monkeys, respectively, with no supportive care. In mice, m-HemaMax at an optimal 20 ng/mouse dose significantly increased percent survival and survival time when administered 24 hours after TBI between 8–9 Gy (p<0.05 Pearson's chi-square test). This survival benefit was accompanied by increases in plasma interferon-γ (IFN-γ) and erythropoietin levels, recovery of femoral bone hematopoiesis characterized with the presence of IL-12 receptor β2 subunit–expressing myeloid progenitors, megakaryocytes, and osteoblasts. Mitigation of jejunal radiation damage was also examined. At allometrically equivalent doses, HemaMax showed similar pharmacokinetics in rhesus monkeys compared to m-HemaMax in mice, but more robustly increased plasma IFN-γ levels. HemaMax also increased plasma erythropoietin, IL-15, IL-18, and neopterin levels. At non-human primate doses pharmacologically equivalent to murine doses, HemaMax (100 ng/Kg and 250 ng/Kg) administered at 24 hours after TBI (6.7 Gy/LD50/30) significantly increased percent survival of HemaMax groups compared to vehicle (p<0.05 Pearson's chi-square test). This survival benefit was accompanied by a significantly higher leukocyte (neutrophils and lymphocytes), thrombocyte, and reticulocyte counts during nadir (days 12–14) and significantly less weight loss at day 12 compared to vehicle. These findings indicate successful interspecies dose conversion and provide proof of concept that HemaMax increases survival in irradiated rhesus monkeys by promoting hematopoiesis and recovery of immune functions and possibly gastrointestinal functions, likely through a network of interactions involving dendritic cells, osteoblasts, and soluble factors such as IL-12, IFN-γ, and cytoprotectant erythropoietin.
Collapse
Affiliation(s)
- Lena A Basile
- Neumedicines, Inc, Pasadena, California, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Newman AM, Gallo NB, Hancox LS, Miller NJ, Radeke CM, Maloney MA, Cooper JB, Hageman GS, Anderson DH, Johnson LV, Radeke MJ. Systems-level analysis of age-related macular degeneration reveals global biomarkers and phenotype-specific functional networks. Genome Med 2012; 4:16. [PMID: 22364233 PMCID: PMC3372225 DOI: 10.1186/gm315] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 01/18/2012] [Accepted: 02/24/2012] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED Please see related commentary: http://www.biomedcentral.com/1741-7015/10/21/abstract BACKGROUND Age-related macular degeneration (AMD) is a leading cause of blindness that affects the central region of the retinal pigmented epithelium (RPE), choroid, and neural retina. Initially characterized by an accumulation of sub-RPE deposits, AMD leads to progressive retinal degeneration, and in advanced cases, irreversible vision loss. Although genetic analysis, animal models, and cell culture systems have yielded important insights into AMD, the molecular pathways underlying AMD's onset and progression remain poorly delineated. We sought to better understand the molecular underpinnings of this devastating disease by performing the first comparative transcriptome analysis of AMD and normal human donor eyes. METHODS RPE-choroid and retina tissue samples were obtained from a common cohort of 31 normal, 26 AMD, and 11 potential pre-AMD human donor eyes. Transcriptome profiles were generated for macular and extramacular regions, and statistical and bioinformatic methods were employed to identify disease-associated gene signatures and functionally enriched protein association networks. Selected genes of high significance were validated using an independent donor cohort. RESULTS We identified over 50 annotated genes enriched in cell-mediated immune responses that are globally over-expressed in RPE-choroid AMD phenotypes. Using a machine learning model and a second donor cohort, we show that the top 20 global genes are predictive of AMD clinical diagnosis. We also discovered functionally enriched gene sets in the RPE-choroid that delineate the advanced AMD phenotypes, neovascular AMD and geographic atrophy. Moreover, we identified a graded increase of transcript levels in the retina related to wound response, complement cascade, and neurogenesis that strongly correlates with decreased levels of phototransduction transcripts and increased AMD severity. Based on our findings, we assembled protein-protein interactomes that highlight functional networks likely to be involved in AMD pathogenesis. CONCLUSIONS We discovered new global biomarkers and gene expression signatures of AMD. These results are consistent with a model whereby cell-based inflammatory responses represent a central feature of AMD etiology, and depending on genetics, environment, or stochastic factors, may give rise to the advanced AMD phenotypes characterized by angiogenesis and/or cell death. Genes regulating these immunological activities, along with numerous other genes identified here, represent promising new targets for AMD-directed therapeutics and diagnostics.
Collapse
Affiliation(s)
- Aaron M Newman
- Center for the Study of Macular Degeneration, Neuroscience Research Institute,
Biological Sciences 2 Building, University of California, Santa Barbara, CA
93106-5060, USA
- Current address: Institute for Stem Cell Biology and Regenerative Medicine,
Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305,
USA
| | - Natasha B Gallo
- Center for the Study of Macular Degeneration, Neuroscience Research Institute,
Biological Sciences 2 Building, University of California, Santa Barbara, CA
93106-5060, USA
| | - Lisa S Hancox
- Department of Ophthalmology and Visual Sciences, University of Iowa, 200 Hawkins
Drive Iowa City, IA 52242-1109, USA
| | - Norma J Miller
- Department of Ophthalmology and Visual Sciences, John A Moran Eye Center,
University of Utah, 65 Mario Capecchi Drive, Salt Lake City, UT 84132-5230,
USA
| | - Carolyn M Radeke
- Center for the Study of Macular Degeneration, Neuroscience Research Institute,
Biological Sciences 2 Building, University of California, Santa Barbara, CA
93106-5060, USA
| | - Michelle A Maloney
- Center for the Study of Macular Degeneration, Neuroscience Research Institute,
Biological Sciences 2 Building, University of California, Santa Barbara, CA
93106-5060, USA
| | - James B Cooper
- Molecular, Cellular, and Developmental Biology Department, Life Sciences Building,
University of California, Santa Barbara, CA 93106-9610, USA
| | - Gregory S Hageman
- Department of Ophthalmology and Visual Sciences, John A Moran Eye Center,
University of Utah, 65 Mario Capecchi Drive, Salt Lake City, UT 84132-5230,
USA
| | - Don H Anderson
- Center for the Study of Macular Degeneration, Neuroscience Research Institute,
Biological Sciences 2 Building, University of California, Santa Barbara, CA
93106-5060, USA
| | - Lincoln V Johnson
- Center for the Study of Macular Degeneration, Neuroscience Research Institute,
Biological Sciences 2 Building, University of California, Santa Barbara, CA
93106-5060, USA
| | - Monte J Radeke
- Center for the Study of Macular Degeneration, Neuroscience Research Institute,
Biological Sciences 2 Building, University of California, Santa Barbara, CA
93106-5060, USA
| |
Collapse
|