Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-36834668

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of blindness in elderly people, with limited treatment options available for most patients. AMD involves the death of retinal pigment epithelium (RPE) and photoreceptor cells, with mitochondria dysfunction being a critical early event. In the current study, we utilized our unique resource of human donor RPE graded for AMD presence and severity to investigate proteome-wide dysregulation involved in early AMD. Organelle-enriched fractions of RPE were isolated from donors with early AMD (n = 45) and healthy age-matched controls (n = 32) and were analyzed by UHR-IonStar, an integrated proteomics platform enabling reliable and in-depth proteomic quantification in large cohorts. A total of 5941 proteins were quantified with excellent analytical reproducibility, and with further informatics analysis, many biological functions and pathways were found to be significantly dysregulated in donor RPE samples with early AMD. Several of these directly pinpointed changes in mitochondrial functions, e.g., translation, ATP metabolic process, lipid homeostasis, and oxidative stress. These novel findings highlighted the value of our proteomics investigation by allowing a better understanding of the molecular mechanisms underlying early AMD onset and facilitating both treatment development and biomarker discovery.


Asunto(s)
Degeneración Macular , Epitelio Pigmentado de la Retina , Humanos , Anciano , Epitelio Pigmentado de la Retina/metabolismo , Proteómica , Reproducibilidad de los Resultados , Degeneración Macular/metabolismo , Estrés Oxidativo
2.
Exp Eye Res ; 178: 247-254, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30114413

RESUMEN

Oxidative modification of proteins can perturb their structure and function, often compromising cellular viability. Such modifications include lipid-derived adducts (e.g., 4-hydroxynonenal (HNE) and carboxyethylpyrrole (CEP)) as well as nitrotyrosine (NTyr). We compared the retinal proteome and levels of such modifications in the AY9944-treated rat model of Smith-Lemli-Opitz syndrome (SLOS), in comparison to age-matched controls. Retinas harvested at 3 months of age were either subjected to proteomic analysis or to immuno-slot blot analysis, the latter probing blots with antibodies raised against HNE, CEP, and NTyr, followed by quantitative densitometry. HNE modification of retinal proteins was markedly (>9-fold) higher in AY9944-treated rats compared to controls, whereas CEP modification was only modestly (≤2-fold) greater, and NTyr modification was minimal and exhibited no difference as a function of AY9944 treatment. Anti-HNE immunoreactivity was greatest in the plexiform and ganglion cell layers, but also present in the RPE, choroid, and photoreceptor outer segment layer in AY9944-treated rats; control retinas showed minimal HNE labeling. 1D-PAGE/Western blot analysis of rod outer segment (ROS) membranes revealed HNE modification of both opsin and ß-transducin. Proteomic analysis revealed the differential expression of several retinal proteins as a consequence of AY9944 treatment. Upregulated proteins included those involved in chaperone/protein folding, oxidative and cellular stress responses, transcriptional regulation, and energy production. ßA3/A1 Crystallin, which has a role in regulation of lysosomal acidification, was down-regulated. Hence, oxidative modification of retinal proteins occurs in the SLOS rat model, in addition to the previously described oxidation of lipids. The results are discussed in the context of the histological and physiological changes that occur in the retina in the SLOS rat model.


Asunto(s)
Modelos Animales de Enfermedad , Opsinas/metabolismo , Estrés Oxidativo , Retina/metabolismo , Síndrome de Smith-Lemli-Opitz/metabolismo , Transducina/metabolismo , Aldehídos/metabolismo , Animales , Western Blotting , Electroforesis en Gel de Poliacrilamida , Inhibidores Enzimáticos/farmacología , Femenino , Embarazo , Proteómica , Ratas , Ratas Sprague-Dawley , Diclorhidrato de trans-1,4-Bis(2-clorobenzaminometil)ciclohexano/farmacología
3.
Exp Eye Res ; 186: 107686, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31158383

RESUMEN

The blood retinal barrier (BRB) closely regulates the retinal microenvironment. Its compromise leads to the accumulation of retinal fluid containing potentially harmful plasma components. While eyes with non-exudative age-related macular degeneration (AMD) were previously felt to have an intact BRB, we propose that the BRB in non-exudative AMD eyes may be subclinically compromised, allowing entry of retina-toxic plasma proteins. We test this hypothesis by measuring retinal levels of abundant plasma proteins that should not cross the intact BRB. Two cohorts of frozen, post mortem neurosensory retinas were studied by Western analysis. One cohort from Alabama had 4 normal controls and 4 eyes with various forms of AMD. Another cohort from Minnesota had 5 intermediate AMD eyes and 5 normals. Both cohorts were age/post mortem interval (PMI) matched. The non-exudative AMD retinas in the Alabama cohort had significantly higher levels of albumin and complement component 9 (C9) than normal controls. The positive control exudative AMD donor retina had higher levels of all but one serum protein. In both macular and peripheral neurosensory retina samples, intermediate AMD retinas in the Minnesota cohort had significantly higher levels of albumin, fibrinogen, IgG, and C9 than controls. Our results suggest that there may be moderate subclinical BRB leakage in non-exudative AMD. Potentially harmful plasma components including complement or iron could enter the neurosensory retina in AMD patients prior to advanced disease. Thus, therapies aiming to stabilize the BRB might have a role in the management of non-exudative AMD.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Atrofia Geográfica/sangre , Retina/metabolismo , Anciano , Anciano de 80 o más Años , Barrera Hematorretinal/fisiología , Western Blotting , Complemento C9/metabolismo , Exudados y Transudados , Femenino , Fibrinógeno/metabolismo , Humanos , Inmunoglobulina G/metabolismo , Masculino , Albúmina Sérica/metabolismo
4.
J Neurosci ; 35(18): 7304-11, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25948278

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of blindness among older adults in the developed world. Although the pathological mechanisms have not been definitively elucidated, evidence suggests a key role for mitochondrial (mt) dysfunction. The current study used our unique collection of human retinal samples graded for the donor's stage of AMD to address fundamental questions about mtDNA damage in the retina. To evaluate the distribution of mtDNA damage in the diseased retina, damage in the retinal pigment epithelium (RPE) and neural retina from individual donors were compared. To directly test a long-held belief that the macula is selectively damaged with AMD, RPE mtDNA damage was measured in the macula and peripheral sections from individual donors. Small segments of the entire mt genome were examined to determine whether specific regions are preferentially damaged. Our results show that mtDNA damage is limited to the RPE, equivalent mtDNA damage is found in the macular and peripheral RPE, and sites of damage are localized to regions of the mt genome that may impact mt function. These results provide a scientific basis for targeting the RPE mitochondria with therapies that protect and enhance mt function as a strategy for combating AMD.


Asunto(s)
Envejecimiento/metabolismo , Sistemas de Liberación de Medicamentos , Marcación de Gen , Degeneración Macular/metabolismo , Mitocondrias/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Anciano , Anciano de 80 o más Años , Envejecimiento/patología , ADN Mitocondrial/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , Femenino , Marcación de Gen/tendencias , Humanos , Degeneración Macular/patología , Masculino , Persona de Mediana Edad , Mitocondrias/patología , Epitelio Pigmentado de la Retina/patología
5.
Exp Eye Res ; 145: 269-277, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26854823

RESUMEN

Age-related macular degeneration (AMD) is a major cause of blindness among the elderly in the developed world. Genetic analysis of AMD has identified 34 high-risk loci associated with AMD. The genes at these high risk loci belong to diverse biological pathways, suggesting different mechanisms leading to AMD pathogenesis. Thus, therapies targeting a single pathway for all AMD patients will likely not be universally effective. Recent evidence suggests defects in mitochondria (mt) of the retinal pigment epithelium (RPE) may constitute a key pathogenic event in some AMD patients. The purpose of this study is to determine if individuals with a specific genetic background have a greater propensity for mtDNA damage. We used human eyebank tissues from 76 donors with AMD and 42 age-matched controls to determine the extent of mtDNA damage in the RPE that was harvested from the macula using a long extension polymerase chain reaction assay. Genotype analyses were performed for ten common AMD-associated nuclear risk alleles (ARMS2, TNFRSF10A, CFH, C2, C3, APOE, CETP, LIPC, VEGF and COL10A1) and mtDNA haplogroups. Sufficient samples were available for genotype association with mtDNA damage for TNFRSF10A, CFH, CETP, VEGFA, and COL10A1. Our results show that AMD donors carrying the high risk allele for CFH (C) had significantly more mtDNA damage compared with donors having the wild-type genetic profile. The data from an additional 39 donors (12 controls and 27 AMD) genotyped for CFH alleles further supported these findings. Taken together, these studies provide the rationale for a more personalized approach for treating AMD by uncovering a significant correlation between the CFH high risk allele and accelerated mtDNA damage. Patients harboring this genetic risk factor may benefit from therapies that stabilize and protect the mt in the RPE.


Asunto(s)
Factor H de Complemento/genética , Daño del ADN/fisiología , ADN Mitocondrial , Degeneración Macular/genética , Epitelio Pigmentado de la Retina , Anciano , Anciano de 80 o más Años , Alelos , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Degeneración Macular/metabolismo , Degeneración Macular/fisiopatología , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa
6.
Oxid Med Cell Longev ; 2022: 6009787, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36439688

RESUMEN

Age-related macular degeneration (AMD), the leading cause of blindness in elderly populations, involves the loss of central vision due to progressive dysfunction of the retinal pigment epithelium (RPE) and subsequent loss of light-sensing photoreceptors. While age is a key risk factor, not every aged individual develops AMD. Thus, the critical question is what specific cellular changes tip the balance from healthy aging to disease. To distinguish between changes associated with aging and AMD, we compared the RPE proteome in human eye bank tissue from nondiseased donors during aging (n = 50, 29-91 years) and in donors with AMD (n = 36) compared to age-matched donors without disease (n = 28). Proteins from RPE cells were separated on two-dimensional gels, analyzed for content, and identified using mass spectrometry. A total of 58 proteins displayed significantly altered content with either aging or AMD. Proteins involved in metabolism, protein turnover, stress response, and cell death were altered with both aging and AMD. However, the direction of change was predominantly opposite. With aging, we detected an overall decrease in metabolism and reductions in stress-associated proteins, proteases, and chaperones. With AMD, we observed upregulation of metabolic proteins involved in glycolysis, TCA, and fatty acid metabolism, with a concurrent decline in oxidative phosphorylation, suggesting a reprogramming of energy utilization. Additionally, we detected upregulation of proteins involved in the stress response and protein turnover. Predicted upstream regulators also showed divergent results, with inhibition of inflammation and immune response with aging and activation of these processes with AMD. Our results support the idea that AMD is not simply advanced aging but rather the culmination of perturbed protein homeostasis, defective bioenergetics, and increased oxidative stress within the aging RPE, exacerbated by environmental factors and the genetic background of an individual.


Asunto(s)
Degeneración Macular , Epitelio Pigmentado de la Retina , Anciano , Humanos , Epitelio Pigmentado de la Retina/metabolismo , Degeneración Macular/metabolismo , Envejecimiento , Estrés Oxidativo , Fosforilación Oxidativa
7.
Antioxidants (Basel) ; 11(4)2022 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-35453289

RESUMEN

Primary cultures of retinal pigment epithelium (RPE) from human adult donors (haRPE) and induced pluripotent stem cell derived-RPE (iPSC-RPE) are valuable model systems for gaining mechanistic insight and for testing potential therapies for age-related macular degeneration (AMD). This study evaluated the treatment response of haRPE and iPSC-RPE to oxidative stress and potential therapeutics addressing mitochondrial defects. haRPE and iSPC-RPE were derived from donors with or without AMD. Mitochondrial function was measured after treatment with menadione, AICAR, or trehalose and the response to treatment was compared between cell models and by disease status. In a subset of samples, haRPE and iPSC-RPE were generated from the same human donor to make a side-by-side comparison of the two cell models' response to treatment. Disease-specific responses to all three treatments was observed in the haRPE. In contrast, iPSC-RPE had a similar response to all treatments irrespective of disease status. Analysis of haRPE and iPSC-RPE generated from the same human donor showed a similar response for donors without AMD, but there were significant differences in treatment response between cell models generated from AMD donors. These results support the use of iPSC-RPE and haRPE when investigating AMD mechanisms and new therapeutics but indicates that attention to experimental conditions is required.

8.
Cells ; 11(13)2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35805159

RESUMEN

Age-related macular degeneration (AMD), the leading cause of blindness in the elderly, is characterized by the death of retinal pigment epithelium (RPE) and photoreceptors. One of the risk factors associated with developing AMD is the single nucleotide polymorphism (SNP) found within the gene encoding complement factor H (CFH). Part of the innate immune system, CFH inhibits alternative complement pathway activation. Multi-protein complexes called inflammasomes also play a role in the innate immune response. Previous studies reported that inflammasome activation may contribute to AMD pathology. In this study, we used primary human adult RPE cell cultures from multiple donors, with and without AMD, that were genotyped for the Y402H CFH risk allele. We found complement and inflammasome-related genes and proteins at basal levels in RPE tissue and cell cultures. Additionally, treatment with rotenone, bafilomycin A, and ATP led to inflammasome activation. Overall, the response to priming and activation was similar, irrespective of disease state or CFH genotype. While these data show that the inflammasome is present and active in RPE, our results suggest that inflammasome activation may not contribute to early AMD pathology.


Asunto(s)
Degeneración Macular , Epitelio Pigmentado de la Retina , Anciano , Genotipo , Humanos , Inflamasomas/metabolismo , Degeneración Macular/metabolismo , Polimorfismo de Nucleótido Simple , Epitelio Pigmentado de la Retina/metabolismo
9.
Pharmaceuticals (Basel) ; 15(1)2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-35056119

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of blindness in the elderly. No universally effective treatments exist for atrophic or "dry" AMD, which results from loss of the retinal pigment epithelium (RPE) and photoreceptors and accounts for ≈80% of all AMD patients. Prior studies provide evidence for the involvement of mitochondrial dysfunction in AMD pathology. This study used induced pluripotent stem cell (iPSC) RPE derived from five AMD patients to test the efficacy of three drugs (AICAR (5-Aminoimidazole-4-carboxamide ribonucleotide), Metformin, trehalose) that target key processes in maintaining optimal mitochondrial function. The patient iPSC-RPE lines were used in a proof-of-concept drug screen, utilizing an analysis of RPE mitochondrial function following acute and extended drug exposure. Results show considerable variability in drug response across patient cell lines, supporting the need for a personalized medicine approach for treating AMD. Furthermore, our results demonstrate the feasibility of using iPSC-RPE from AMD patients to develop a personalized drug treatment regime and provide a roadmap for the future clinical management of AMD.

10.
Cell Rep ; 39(11): 110942, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35705048

RESUMEN

Age-related macular degeneration (AMD), the leading cause of irreversible blindness among Americans over 50, is characterized by dysfunction and death of retinal pigment epithelial (RPE) cells. The RPE accumulates iron in AMD, and iron overload triggers RPE cell death in vitro and in vivo. However, the mechanism of RPE iron accumulation in AMD is unknown. We show that high-fat-diet-induced obesity, a risk factor for AMD, drives systemic and local inflammatory circuits upregulating interleukin-1ß (IL-1ß). IL-1ß upregulates RPE iron importers and downregulates iron exporters, causing iron accumulation, oxidative stress, and dysfunction. We term this maladaptive, chronic activation of a nutritional immunity pathway the cellular iron sequestration response (CISR). RNA sequencing (RNA-seq) analysis of choroid and retina from human donors revealed that hallmarks of this pathway are present in AMD microglia and macrophages. Together, these data suggest that inflamed adipose tissue, through the CISR, can lead to RPE pathology in AMD.


Asunto(s)
Degeneración Macular , Epitelio Pigmentado de la Retina , Tejido Adiposo/metabolismo , Humanos , Hierro/metabolismo , Degeneración Macular/metabolismo , Estrés Oxidativo , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo
11.
PLoS One ; 16(10): e0258803, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34665838

RESUMEN

PURPOSE: To determine whether age-related macular degeneration (AMD) severity or the frequency of retinal pigment epithelium mitochondrial DNA lesions differ in human donor eyes that have undergone cataract surgery compared to phakic eyes. METHODS: Eyes from human donors aged ≥ 55 years were obtained from the Minnesota Lions Eye Bank. Cataract surgery status was obtained from history provided to Eye Bank personnel by family members at the time of tissue procurement. Donor eyes were graded for AMD severity using the Minnesota Grading System. Quantitative PCR was performed on DNA isolated from macular punches of retinal pigment epithelium to quantitate the frequency of mitochondrial DNA lesions in the donor tissue. Univariable and multivariable analyses were performed to evaluate for associations between (1) cataract surgery and AMD severity and (2) cataract surgery and mitochondrial DNA lesion frequency. RESULTS: A total of 157 subjects qualified for study inclusion. Multivariable analysis with age, sex, smoking status, and cataract surgery status showed that only age was associated with AMD grade. Multivariable analysis with age, sex, smoking status, and cataract surgery status showed that none of these factors were associated with retinal pigment epithelium mitochondrial DNA lesion frequency. CONCLUSIONS: In this study of human donor eyes, neither retinal pigment epithelium mitochondrial DNA damage nor the stage of AMD severity are independently associated with cataract surgery after adjusting for other AMD risk factors. These new pathologic and molecular findings provide evidence against a relationship between cataract surgery and AMD progression and support the idea that cataract surgery is safe in the setting of AMD.


Asunto(s)
Extracción de Catarata/estadística & datos numéricos , Daño del ADN , ADN Mitocondrial/genética , Degeneración Macular/genética , Anciano , Anciano de 80 o más Años , Bancos de Muestras Biológicas , Extracción de Catarata/efectos adversos , Progresión de la Enfermedad , Femenino , Humanos , Degeneración Macular/etiología , Masculino , Persona de Mediana Edad , Análisis Multivariante , Epitelio Pigmentado de la Retina/química , Donantes de Tejidos
12.
Cells ; 10(4)2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33918210

RESUMEN

Age-related macular degeneration (AMD), the leading cause of vision loss in the elderly, is characterized by loss of the retinal pigment epithelium (RPE). While the disease mechanism remains unclear, prior studies have linked AMD with RPE mitochondrial defects and genetic polymorphisms in the complement pathway. This study used RPE generated from induced pluripotent stem cells (iPSC-RPE), which were derived from human donors with or without AMD and genotyped for the complement factor H (CFH) AMD high-risk allele (rs1061170, Y402H) to investigate whether donor disease state or genotype had a detrimental effect on mitochondrial function and inflammation. Results show that cells derived from donors with AMD display decreased mitochondrial function under conditions of stress and elevated expression of inflammatory markers compared to iPSC-RPE from individuals without AMD. A more pronounced reduction in mitochondrial function and increased inflammatory markers was observed in CFH high-risk cells, irrespective of disease state. These results provide evidence for a previously unrecognized link between CFH and mitochondrial function that could contribute to RPE loss in AMD patients harboring the CFH high-risk genotype.


Asunto(s)
Factor H de Complemento/genética , Células Madre Pluripotentes Inducidas/patología , Degeneración Macular/genética , Mitocondrias/patología , Polimorfismo Genético , Epitelio Pigmentado de la Retina/patología , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Línea Celular , Proteínas del Sistema Complemento/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Inflamación/patología , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , Riesgo , Donantes de Tejidos
13.
J Neurochem ; 113(6): 1481-90, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20345760

RESUMEN

Our previous work demonstrated that immunoproteasome is up-regulated in the retina and brain in response to injury that does not involve an inflammatory response (J. Neurochem. 2008; 106:158). These results suggest additional non-immune functions for the immunoproteasome in the cellular stress response pathway. The present study further investigates the potential involvement of the immunoproteasome in responding to the chronic stress of aging or oxidant exposure in the retina and cultured retinal pigment epithelial (RPE) cells from knock-out mice missing either one (lmp7(-/-)) or two (lmp7(-/-)/mecl-1(-/-)) immunoproteasome subunits. We show that aging and chronic oxidative stress up-regulates immunoproteasome in the retina and RPE from wild-type mice. No up-regulation of LMP2 was observed in retinas or RPE lacking MECL-1 and/or LMP7, suggesting that the full complement of immunoproteasome subunits is required to achieve maximal up-regulation in response to stress. We also show that RPE deficient in immunoproteasome are more susceptible to oxidation-induced cell death, supporting a role for immunoproteasome in protecting from oxidative stress. These results provide key mechanistic insight into novel aspects of proteasome biology and are an important first step in identifying alternative roles for retinal immunoproteasome that are unrelated to its role in the immune response.


Asunto(s)
Cisteína Endopeptidasas/deficiencia , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/genética , Estrés Oxidativo/fisiología , Receptores Citoplasmáticos y Nucleares/deficiencia , Epitelio Pigmentado de la Retina/citología , Regulación hacia Arriba/fisiología , Envejecimiento , Análisis de Varianza , Animales , Células Cultivadas , Cisteína Endopeptidasas/genética , Células Epiteliales/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidantes/farmacología , Complejo de la Endopetidasa Proteasomal , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
14.
PLoS One ; 15(4): e0231212, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32275682

RESUMEN

Two major proteolytic systems, the proteasome and the autophagy pathway, are key components of the proteostasis network. The immunoproteasome, a proteasome subtype, and autophagy are upregulated under stress conditions, forming a coordinated unit designed to minimize the effect of cell stress. We investigated how genetic ablation of the LMP2 immunoproteasome subunit affects autophagy in retinal pigment epithelium (RPE) from WT and LMP2 knockout mice. We monitored autophagy regulation by measuring LC3, phosphorylation of AKT (S473), and phosphorylation of S6, a downstream readout of AKT (mTOR) pathway activation. We also evaluated transcription factor EB (TFEB) nuclear translocation, a transcription factor that controls expression of autophagy and lysosome genes. WT and LMP2 KO cells were monitored after treatment with EBSS to stimulate autophagy, insulin to stimulate AKT, or an AKT inhibitor (trehalose or MK-2206). Under basal conditions, we observed hyper-phosphorylation of AKT and S6, as well as lower nuclear-TFEB content in LMP2 KO RPE compared with WT. AKT inhibitors MK-2206 and trehalose significantly inhibited AKT phosphorylation and stimulated nuclear translocation of TFEB. Starvation and AKT inhibition upregulated autophagy, albeit to a lesser extent in LMP2 KO RPE. These data support the idea that AKT hyper-activation is an underlying cause of defective autophagy regulation in LMP2 KO RPE, revealing a unique link between two proteolytic systems and a previously unknown function in autophagy regulation by the immunoproteasome.


Asunto(s)
Autofagia , Complejo de la Endopetidasa Proteasomal/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Retina/citología , Animales , Autofagia/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Células Cultivadas , Cisteína Endopeptidasas/metabolismo , Activación Enzimática/efectos de los fármacos , Células HEK293 , Humanos , Insulina/farmacología , Ratones Noqueados , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Epitelio Pigmentado de la Retina/citología , Transducción de Señal/efectos de los fármacos
15.
Oxid Med Cell Longev ; 2019: 5174957, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31485293

RESUMEN

Age-related macular degeneration (AMD) involves the loss of retinal pigment epithelium (RPE) and photoreceptors and is one of the leading causes of blindness in the elderly. Oxidative damage to proteins, lipids, and DNA has been associated with RPE dysfunction and AMD. In this study, we evaluated oxidative stress in AMD and the efficacy of antioxidant, N-acetyl-L-cysteine (NAC), in protecting RPE from oxidative damage. To test this idea, primary cultures of RPE from human donors with AMD (n = 32) or without AMD (No AMD, n = 21) were examined for expression of NADPH oxidase (NOX) genes, a source of reactive oxygen species (ROS). Additionally, the cells were pretreated with NAC for 2 hours and then treated with either hydrogen peroxide (H2O2) or tert-butyl hydroperoxide (t-BHP) to induce cellular oxidation. Twenty-four hours after treatment, ROS production, cell survival, the content of glutathione (GSH) and adenosine triphosphate (ATP), and cellular bioenergetics were measured. We found increased expression of p22phox, a NOX regulator, in AMD cells compared to No AMD cells (p = 0.02). In both AMD and No AMD cells, NAC pretreatment reduced t-BHP-induced ROS production and protected from H2O2-induced cell death and ATP depletion. In the absence of oxidation, NAC treatment improved mitochondrial function in both groups (p < 0.01). Conversely, the protective response exhibited by NAC was disease-dependent for some parameters. In the absence of oxidation, NAC significantly reduced ROS production (p < 0.001) and increased GSH content (p = 0.02) only in RPE from AMD donors. Additionally, NAC-mediated protection from H2O2-induced GSH depletion (p = 0.04) and mitochondrial dysfunction (p < 0.05) was more pronounced in AMD cells compared with No AMD cells. These results demonstrate the therapeutic benefit of NAC by mitigating oxidative damage in RPE. Additionally, the favorable outcomes observed for AMD RPE support NAC's relevance and the potential therapeutic value in treating AMD.


Asunto(s)
Acetilcisteína/uso terapéutico , Células Epiteliales/metabolismo , Degeneración Macular/genética , Epitelio Pigmentado de la Retina/metabolismo , Acetilcisteína/farmacología , Humanos , Degeneración Macular/patología
16.
Nat Genet ; 51(6): 1067, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31068672

RESUMEN

In the version of this article initially published, in Supplementary Data 5, the logFC, FC, P value and adjusted P value for advanced AMD versus control (DE 4/1) without age correction did not correspond to the correct gene IDs. The errors have been corrected in the HTML version of the article.

17.
Nat Genet ; 51(4): 606-610, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30742112

RESUMEN

Genome-wide association studies (GWAS) have identified genetic variants at 34 loci contributing to age-related macular degeneration (AMD)1-3. We generated transcriptional profiles of postmortem retinas from 453 controls and cases at distinct stages of AMD and integrated retinal transcriptomes, covering 13,662 protein-coding and 1,462 noncoding genes, with genotypes at more than 9 million common SNPs for expression quantitative trait loci (eQTL) analysis of a tissue not included in Genotype-Tissue Expression (GTEx) and other large datasets4,5. Cis-eQTL analysis identified 10,474 genes under genetic regulation, including 4,541 eQTLs detected only in the retina. Integrated analysis of AMD-GWAS with eQTLs ascertained likely target genes at six reported loci. Using transcriptome-wide association analysis (TWAS), we identified three additional genes, RLBP1, HIC1 and PARP12, after Bonferroni correction. Our studies expand the genetic landscape of AMD and establish the Eye Genotype Expression (EyeGEx) database as a resource for post-GWAS interpretation of multifactorial ocular traits.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Degeneración Macular/genética , Sitios de Carácter Cuantitativo/genética , Transcriptoma/genética , Estudios de Casos y Controles , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Estudio de Asociación del Genoma Completo/métodos , Genotipo , Humanos , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Retina/fisiopatología
18.
J Neurochem ; 106(1): 158-69, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18346202

RESUMEN

It is well known that immunoproteasome generates peptides for MHC Class I occupancy and recognition by cytotoxic T lymphocytes (CTL). The present study focused on evidence for alternative roles for immunoproteasome. Retina and brain were analyzed for expression of immunoproteasome subunits using immunohistochemistry and western blotting under normal conditions and after injury/stress induced by CTL attack on glia (brain) or neurons (retina). Normal retina expressed substantial levels of immunoproteasome in glia, neurons, and retinal pigment epithelium. The basal level of immunoproteasome in retina was two-fold higher than in brain; CTL-induced retinal injury further up-regulated immunoproteasome expression. Immunoproteasome up-regulation was also observed in injured brain and corresponded with expression in Purkinje cells, microglia, astrocytes, and oligodendrocytes. These results suggest that the normal environment of the retina is sufficiently challenging to require on-going expression of immunoproteasome. Further, immunoproteasome up-regulation with retinal and brain injury implies a role in neuronal protection and/or repair of damage.


Asunto(s)
Encéfalo/inmunología , Encefalitis/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Retina/inmunología , Retinitis/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Encéfalo/fisiopatología , Comunicación Celular/inmunología , Encefalitis/fisiopatología , Ratones , Ratones Transgénicos , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/fisiopatología , Regeneración Nerviosa/inmunología , Neuroglía/inmunología , Neuroglía/patología , Plasticidad Neuronal/inmunología , Neuronas/inmunología , Neuronas/patología , Epitelio Pigmentado Ocular/inmunología , Epitelio Pigmentado Ocular/fisiopatología , Subunidades de Proteína/inmunología , Recuperación de la Función/inmunología , Retina/fisiopatología , Retinitis/fisiopatología , Regulación hacia Arriba/inmunología
19.
PLoS One ; 12(3): e0173575, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28282420

RESUMEN

Fidelity in pluripotent stem cell differentiation protocols is necessary for the therapeutic and commercial use of cells derived from embryonic and induced pluripotent stem cells. Recent advances in stem cell technology, especially the widespread availability of a range of chemically defined media, substrates and differentiation components, now allow the design and implementation of fully defined derivation and differentiation protocols intended for replication across multiple research and manufacturing locations. In this report we present an application of these criteria to the generation of retinal pigmented epithelium from iPSCs derived from the conjunctiva of donors with and without age related macular degeneration. Primary conjunctival cells from human donors aged 70-85 years were reprogrammed to derive multiple iPSC lines that were differentiated into functional RPE using a rapid and defined differentiation protocol. The combination of defined iPSC derivation and culture with a defined RPE differentiation protocol, reproducibly generated functional RPE from each donor without requiring protocol adjustments for each individual. This successful validation of a standardized, iPSC derivation and RPE differentiation process demonstrates a practical approach for applications requiring the cost-effective generation of RPE from multiple individuals such as drug testing, population studies or for therapies requiring patient-specific RPE derivations. In addition, conjunctival cells are identified as a practical source of somatic cells for deriving iPSCs from elderly individuals.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Donantes de Tejidos , Anciano , Anciano de 80 o más Años , Células Cultivadas , Femenino , Humanos , Células Madre Pluripotentes Inducidas/patología , Degeneración Macular/patología , Degeneración Macular/terapia , Masculino , Epitelio Pigmentado de la Retina/patología
20.
Redox Biol ; 13: 255-265, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28600982

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of blindness among older adults. It has been suggested that mitochondrial defects in the retinal pigment epithelium (RPE) underlies AMD pathology. To test this idea, we developed primary cultures of RPE to ask whether RPE from donors with AMD differ in their metabolic profile compared with healthy age-matched donors. Analysis of gene expression, protein content, and RPE function showed that these cultured cells replicated many of the cardinal features of RPE in vivo. Using the Seahorse Extracellular Flux Analyzer to measure bioenergetics, we observed RPE from donors with AMD exhibited reduced mitochondrial and glycolytic function compared with healthy donors. RPE from AMD donors were also more resistant to oxidative inactivation of these two energy-producing pathways and were less susceptible to oxidation-induced cell death compared with cells from healthy donors. Investigation of the potential mechanism responsible for differences in bioenergetics and resistance to oxidative stress showed RPE from AMD donors had increased PGC1α protein as well as differential expression of multiple genes in response to an oxidative challenge. Based on our data, we propose that cultured RPE from donors phenotyped for the presence or absence of AMD provides an excellent model system for studying "AMD in a dish". Our results are consistent with the ideas that (i) a bioenergetics crisis in the RPE contributes to AMD pathology, and (ii) the diseased environment in vivo causes changes in the cellular profile that are retained in vitro.


Asunto(s)
Degeneración Macular/metabolismo , Estrés Oxidativo , Epitelio Pigmentado de la Retina/metabolismo , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Células Cultivadas , Células Epiteliales/metabolismo , Femenino , Glucólisis , Humanos , Degeneración Macular/patología , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Fosforilación Oxidativa , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Epitelio Pigmentado de la Retina/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA