Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 149(4): 847-59, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22541070

RESUMEN

Alu RNA accumulation due to DICER1 deficiency in the retinal pigmented epithelium (RPE) is implicated in geographic atrophy (GA), an advanced form of age-related macular degeneration that causes blindness in millions of individuals. The mechanism of Alu RNA-induced cytotoxicity is unknown. Here we show that DICER1 deficit or Alu RNA exposure activates the NLRP3 inflammasome and triggers TLR-independent MyD88 signaling via IL18 in the RPE. Genetic or pharmacological inhibition of inflammasome components (NLRP3, Pycard, Caspase-1), MyD88, or IL18 prevents RPE degeneration induced by DICER1 loss or Alu RNA exposure. These findings, coupled with our observation that human GA RPE contains elevated amounts of NLRP3, PYCARD, and IL18 and evidence of increased Caspase-1 and MyD88 activation, provide a rationale for targeting this pathway in GA. Our findings also reveal a function of the inflammasome outside the immune system and an immunomodulatory action of mobile elements.


Asunto(s)
Elementos Alu , ARN Helicasas DEAD-box/metabolismo , Atrofia Geográfica/inmunología , Atrofia Geográfica/patología , Inflamasomas/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Ribonucleasa III/metabolismo , Animales , Proteínas Portadoras/metabolismo , Atrofia Geográfica/metabolismo , Humanos , Inflamasomas/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Epitelio Pigmentado de la Retina/patología , Receptores Toll-Like/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35197297

RESUMEN

Muller glia (MG) play a central role in reactive gliosis, a stress response associated with rare and common retinal degenerative diseases, including age-related macular degeneration (AMD). The posttranslational modification citrullination​ targeting glial fibrillary acidic protein (GFAP) in MG was initially discovered in a panocular chemical injury model. Here, we report in the paradigms of retinal laser injury, a genetic model of spontaneous retinal degeneration (JR5558 mice) and human wet-AMD tissues that MG citrullination is broadly conserved. After laser injury, GFAP polymers that accumulate in reactive MG are citrullinated in MG endfeet and glial cell processes. The enzyme responsible for citrullination, peptidyl arginine deiminase-4 (PAD4), localizes to endfeet and associates with GFAP polymers. Glial cell-specific PAD4 deficiency attenuates retinal hypercitrullination in injured retinas, indicating PAD4 requirement for MG citrullination. In retinas of 1-mo-old JR5558 mice, hypercitrullinated GFAP and PAD4 accumulate in MG endfeet/cell processes in a lesion-specific manner. Finally, we show that human donor maculae from patients with wet-AMD also feature the canonical endfeet localization of hypercitrullinated GFAP. Thus, we propose that endfeet are a "citrullination bunker" that initiates and sustains citrullination in retinal degeneration.


Asunto(s)
Citrulinación , Gliosis/metabolismo , Neuroglía/metabolismo , Degeneración Retiniana/metabolismo , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Macular Húmeda/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34620711

RESUMEN

The atrophic form of age-related macular degeneration (dry AMD) affects nearly 200 million people worldwide. There is no Food and Drug Administration (FDA)-approved therapy for this disease, which is the leading cause of irreversible blindness among people over 50 y of age. Vision loss in dry AMD results from degeneration of the retinal pigmented epithelium (RPE). RPE cell death is driven in part by accumulation of Alu RNAs, which are noncoding transcripts of a human retrotransposon. Alu RNA induces RPE degeneration by activating the NLRP3-ASC inflammasome. We report that fluoxetine, an FDA-approved drug for treating clinical depression, binds NLRP3 in silico, in vitro, and in vivo and inhibits activation of the NLRP3-ASC inflammasome and inflammatory cytokine release in RPE cells and macrophages, two critical cell types in dry AMD. We also demonstrate that fluoxetine, unlike several other antidepressant drugs, reduces Alu RNA-induced RPE degeneration in mice. Finally, by analyzing two health insurance databases comprising more than 100 million Americans, we report a reduced hazard of developing dry AMD among patients with depression who were treated with fluoxetine. Collectively, these studies identify fluoxetine as a potential drug-repurposing candidate for dry AMD.


Asunto(s)
Antidepresivos de Segunda Generación/farmacología , Reposicionamiento de Medicamentos/métodos , Fluoxetina/farmacología , Degeneración Macular/tratamiento farmacológico , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Epitelio Pigmentado de la Retina/efectos de los fármacos , Elementos Alu/genética , Animales , Ceguera/patología , Ceguera/prevención & control , Línea Celular , Citocinas/metabolismo , Depresión/tratamiento farmacológico , Modelos Animales de Enfermedad , Inflamasomas/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , ARN/genética , Retina/patología , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/patología
4.
Proc Natl Acad Sci U S A ; 117(5): 2579-2587, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31964819

RESUMEN

Degeneration of the retinal pigmented epithelium (RPE) and aberrant blood vessel growth in the eye are advanced-stage processes in blinding diseases such as age-related macular degeneration (AMD), which affect hundreds of millions of people worldwide. Loss of the RNase DICER1, an essential factor in micro-RNA biogenesis, is implicated in RPE atrophy. However, the functional implications of DICER1 loss in choroidal and retinal neovascularization are unknown. Here, we report that two independent hypomorphic mouse strains, as well as a separate model of postnatal RPE-specific DICER1 ablation, all presented with spontaneous RPE degeneration and choroidal and retinal neovascularization. DICER1 hypomorphic mice lacking critical inflammasome components or the innate immune adaptor MyD88 developed less severe RPE atrophy and pathological neovascularization. DICER1 abundance was also reduced in retinas of the JR5558 mouse model of spontaneous choroidal neovascularization. Finally, adenoassociated vector-mediated gene delivery of a truncated DICER1 variant (OptiDicer) reduced spontaneous choroidal neovascularization in JR5558 mice. Collectively, these findings significantly expand the repertoire of DICER1 in preserving retinal homeostasis by preventing both RPE degeneration and pathological neovascularization.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/irrigación sanguínea , Ribonucleasa III/metabolismo , Animales , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Neovascularización Coroidal/fisiopatología , ARN Helicasas DEAD-box/genética , Humanos , Degeneración Macular/genética , Degeneración Macular/patología , Degeneración Macular/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/parasitología , Neovascularización Retiniana/fisiopatología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Ribonucleasa III/genética
5.
Nature ; 471(7338): 325-30, 2011 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-21297615

RESUMEN

Geographic atrophy (GA), an untreatable advanced form of age-related macular degeneration, results from retinal pigmented epithelium (RPE) cell degeneration. Here we show that the microRNA (miRNA)-processing enzyme DICER1 is reduced in the RPE of humans with GA, and that conditional ablation of Dicer1, but not seven other miRNA-processing enzymes, induces RPE degeneration in mice. DICER1 knockdown induces accumulation of Alu RNA in human RPE cells and Alu-like B1 and B2 RNAs in mouse RPE. Alu RNA is increased in the RPE of humans with GA, and this pathogenic RNA induces human RPE cytotoxicity and RPE degeneration in mice. Antisense oligonucleotides targeting Alu/B1/B2 RNAs prevent DICER1 depletion-induced RPE degeneration despite global miRNA downregulation. DICER1 degrades Alu RNA, and this digested Alu RNA cannot induce RPE degeneration in mice. These findings reveal a miRNA-independent cell survival function for DICER1 involving retrotransposon transcript degradation, show that Alu RNA can directly cause human pathology, and identify new targets for a major cause of blindness.


Asunto(s)
Elementos Alu/genética , ARN Helicasas DEAD-box/deficiencia , Degeneración Macular/genética , Degeneración Macular/patología , ARN/genética , ARN/metabolismo , Ribonucleasa III/deficiencia , Animales , Muerte Celular , Supervivencia Celular , Células Cultivadas , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , MicroARNs/metabolismo , Datos de Secuencia Molecular , Oligonucleótidos Antisentido , Fenotipo , Epitelio Pigmentado de la Retina/enzimología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Ribonucleasa III/genética , Ribonucleasa III/metabolismo
6.
Proc Natl Acad Sci U S A ; 111(45): 16082-7, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25349431

RESUMEN

Geographic atrophy, an advanced form of age-related macular degeneration (AMD) characterized by death of the retinal pigmented epithelium (RPE), causes untreatable blindness in millions worldwide. The RPE of human eyes with geographic atrophy accumulates toxic Alu RNA in response to a deficit in the enzyme DICER1, which in turn leads to activation of the NLRP3 inflammasome and elaboration of IL-18. Despite these recent insights, it is still unclear how RPE cells die during the course of the disease. In this study, we implicate the involvement of Caspase-8 as a critical mediator of RPE degeneration. Here we show that DICER1 deficiency, Alu RNA accumulation, and IL-18 up-regulation lead to RPE cell death via activation of Caspase-8 through a Fas ligand-dependent mechanism. Coupled with our observation of increased Caspase-8 expression in the RPE of human eyes with geographic atrophy, our findings provide a rationale for targeting this apoptotic pathway in this disease.


Asunto(s)
Elementos Alu , Apoptosis , Caspasa 8/metabolismo , ARN Helicasas DEAD-box/metabolismo , Proteínas del Ojo/metabolismo , Degeneración Macular/metabolismo , ARN/metabolismo , Ribonucleasa III/metabolismo , Animales , Caspasa 8/genética , ARN Helicasas DEAD-box/genética , Proteínas del Ojo/genética , Humanos , Interleucina-18/genética , Interleucina-18/metabolismo , Degeneración Macular/patología , Ratones , Ratones Noqueados , ARN/genética , Ribonucleasa III/genética , Regulación hacia Arriba/genética
7.
Proc Natl Acad Sci U S A ; 109(34): 13781-6, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22869729

RESUMEN

Deficient expression of the RNase III DICER1, which leads to the accumulation of cytotoxic Alu RNA, has been implicated in degeneration of the retinal pigmented epithelium (RPE) in geographic atrophy (GA), a late stage of age-related macular degeneration that causes blindness in millions of people worldwide. Here we show increased extracellular-signal-regulated kinase (ERK) 1/2 phosphorylation in the RPE of human eyes with GA and that RPE degeneration in mouse eyes and in human cell culture induced by DICER1 depletion or Alu RNA exposure is mediated via ERK1/2 signaling. Alu RNA overexpression or DICER1 knockdown increases ERK1/2 phosphorylation in the RPE in mice and in human cell culture. Alu RNA-induced RPE degeneration in mice is rescued by intravitreous administration of PD98059, an inhibitor of the ERK1/2-activating kinase MEK1, but not by inhibitors of other MAP kinases such as p38 or JNK. These findings reveal a previously unrecognized function of ERK1/2 in the pathogenesis of GA and provide a mechanistic basis for evaluation of ERK1/2 inhibition in treatment of this disease.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Degeneración Macular/enzimología , Degeneración Macular/terapia , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Animales , ARN Helicasas DEAD-box/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Humanos , Ratones , Fosforilación , Epitelio Pigmentado de la Retina/metabolismo , Ribonucleasa III/metabolismo , Transducción de Señal
8.
Mol Ther ; 20(1): 101-8, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21988875

RESUMEN

The discovery of sequence-specific gene silencing by endogenous double-stranded RNAs (dsRNA) has propelled synthetic short-interfering RNAs (siRNAs) to the forefront of targeted pharmaceutical engineering. The first clinical trials utilized 21-nucleotide (nt) siRNAs for the treatment of neovascular age-related macular degeneration (AMD). Surprisingly, these compounds were not formulated for cell permeation, which is required for bona fide RNA interference (RNAi). We showed that these "naked" siRNAs suppress neovascularization in mice not via RNAi but via sequence-independent activation of cell surface Toll-like receptor-3 (TLR3). Here, we demonstrate that noninternalized siRNAs induce retinal degeneration in mice by activating surface TLR3 on retinal pigmented epithelial cells. Cholesterol conjugated siRNAs capable of cell permeation and triggering RNAi also induce the same phenotype. Retinal degeneration was not observed after treatment with siRNAs shorter than 21-nts. Other cytosolic dsRNA sensors are not critical to this response. TLR3 activation triggers caspase-3-mediated apoptotic death of the retinal pigment epithelium (RPE) via nuclear translocation of interferon regulatory factor-3. While this unexpected adverse effect of siRNAs has implications for future clinical trials, these findings also introduce a new preclinical model of geographic atrophy (GA), a late stage of dry AMD that causes blindness in millions worldwide.


Asunto(s)
Factor 3 Regulador del Interferón/metabolismo , ARN Interferente Pequeño/toxicidad , Degeneración Retiniana/inducido químicamente , Receptor Toll-Like 3/metabolismo , Animales , Caspasa 3/metabolismo , Muerte Celular/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , ARN Interferente Pequeño/metabolismo , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal
10.
Sci Rep ; 13(1): 22666, 2023 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-38114564

RESUMEN

Evidence from histopathology and clinical imaging suggest that choroidal anatomy and hemodynamic perfusion are among the earliest changes in retinal diseases such as age-related macular degeneration (AMD). However, how inner choroidal anatomy affects hemodynamic perfusion is not well understood. Therefore, we sought to understand the influences of choroidal microvascular architecture on the spatial distribution of hemodynamic parameters in choriocapillaris from human donor eyes using image-based computational hemodynamic (ICH) simulations. We subjected image-based inner choroid reconstructions from eight human donor eyes to ICH simulation using a kinetic-based volumetric lattice Boltzmann method to compute hemodynamic distributions of velocity, pressure, and endothelial shear stress. Here, we demonstrate that anatomic parameters, including arteriolar and venular arrangements and intercapillary pillar density and distribution exert profound influences on inner choroidal hemodynamic characteristics. Reductions in capillary, arteriolar, and venular density not only reduce the overall blood velocity within choriocapillaris, but also substantially increase its spatial heterogeneity. These first-ever findings improve understanding of how choroidal anatomy affects hemodynamics and may contribute to pathogenesis of retinal diseases such as AMD.


Asunto(s)
Coroides , Degeneración Macular , Humanos , Coroides/irrigación sanguínea , Degeneración Macular/patología , Hemodinámica , Perfusión
11.
Invest Ophthalmol Vis Sci ; 64(5): 3, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37129905

RESUMEN

Purpose: Rhegmatogenous retinal detachment (RRD) is a vision-threatening event that benefits from surgical intervention. While awaiting surgical reattachment, irreversible hypoxic and inflammatory damage to the retina often occurs. An interim therapy protecting photoreceptors could improve functional outcomes. We sought to determine whether Kamuvudine-9 (K-9), a derivative of nucleoside reverse transcriptase inhibitors (NRTIs) that inhibits inflammasome activation, and the NRTIs lamivudine (3TC) and azidothymidine (AZT) could protect the retina following RRD. Methods: RRD was induced in mice via subretinal injection (SRI) of 1% carboxymethylcellulose (CMC). To simulate outcomes following the clinical management of RRD, we determined the optimal conditions by which SRI of CMC induced spontaneous retinal reattachment (SRR) occurs over 10 days (RRD/SRR). K-9, 3TC, or AZT was administered via intraperitoneal injection. Inflammasome activation pathways were monitored by abundance of cleaved caspase-1, IL-18, and cleaved caspase-8, and photoreceptor death was assessed by TUNEL staining. Retinal function was assessed by full-field scotopic electroretinography. Results: RRD induced retinal inflammasome activation and photoreceptor death in mice. Systemic administration of K-9, 3TC, or AZT inhibited retinal inflammasome activation and photoreceptor death. In the RRD/SRR model, K-9 protected retinal electrical function during the time of RRD and induced an improvement following retinal reattachment. Conclusions: K-9 and NRTIs exhibit anti-inflammatory and neuroprotective activities in experimental RRD. Given its capacity to protect photoreceptor function during the period of RRD and enhance retinal function following reattachment, K-9 shows promise as a retinal neuroprotectant and warrants study in RRD. Further, this novel RRD/SRR model may facilitate experimental evaluation of functional outcomes relevant to RRD.


Asunto(s)
Desprendimiento de Retina , Animales , Ratones , Desprendimiento de Retina/cirugía , Inflamasomas , Agudeza Visual , Retina , Estudios Retrospectivos , Vitrectomía
12.
medRxiv ; 2023 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-36993694

RESUMEN

Innate immune signaling through the NLRP3 inflammasome has been implicated in the pathogenesis of Alzheimer's disease (AD), the most prevalent form of dementia. We previously demonstrated that nucleoside reverse transcriptase inhibitors (NRTIs), drugs approved to treat HIV and hepatitis B infections, also inhibit inflammasome activation. Here we report that in humans, NRTI exposure was associated with a significantly lower incidence of AD in two of the largest health insurance databases in the United States. Treatment of aged 5xFAD mice (a mouse model of amyloid-ß deposition that expresses five mutations found in familial AD) with Kamuvudine-9 (K-9), an NRTI-derivative with enhanced safety profile, reduced Aß deposition and reversed their cognitive deficit by improving their spatial memory and learning performance to that of young wild-type mice. These findings support the concept that inflammasome inhibition could benefit AD and provide a rationale for prospective clinical testing of NRTIs or K-9 in AD.

13.
Circ Res ; 106(11): 1703-11, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20378855

RESUMEN

RATIONALE: The extracellular matrix protein fibronectin (FN) is focally deposited in regions of atherosclerosis, where it contributes to inflammatory signaling. OBJECTIVE: To elucidate the mechanism by which FN deposition is regulated by local shear stress patterns, its dependence on platelet-endothelial cell adhesion molecule (PECAM)-1 mechanotransduction and the role this pathway plays in sustaining an atheroprone/proinflammatory phenotype. METHODS AND RESULTS: Human endothelial cells were exposed in vitro to atheroprone or atheroprotective shear stress patterns derived from human carotid arteries. Onset of atheroprotective flow induced a transient increase in FN deposition, whereas atheroprone flow caused a steady increase in FN expression and integrin activation over time, leading to a significant and sustained increase in FN deposition relative to atheroprotective conditions. Comparing FN staining in ApoE(-/-) and ApoE(-/-)PECAM(-/-) mice showed that PECAM-1 was essential for FN accumulation in atheroprone regions of the aortic arch. In vitro, small interfering RNA against PECAM-1 blocked the induction of FN and the activation of nuclear factor (NF)-kappaB by atheroprone flow, which was rescued by the addition of exogenous FN. Additionally, blocking NF-kappaB activation attenuated the flow-induced FN expression. Small interfering RNA against FN significantly reduced NF-kappaB activity, which was rescued by the addition of exogenous FN. CONCLUSIONS: These results indicate that FN gene expression and assembly into matrix fibrils is induced by atheroprone fluid shear stress. This effect is mediated at least in part by the transcription factor NF-kappaB. Additionally, because FN promotes activation of NF-kappaB, atheroprone shear stress creates a positive feedback to maintain inflammation.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Endotelio Vascular/metabolismo , Fibronectinas/metabolismo , Hemodinámica , Inflamación/metabolismo , Mecanotransducción Celular , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/fisiopatología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/fisiopatología , Retroalimentación Fisiológica , Fibronectinas/genética , Humanos , Inflamación/genética , Inflamación/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Flujo Pulsátil , Interferencia de ARN , Flujo Sanguíneo Regional , Estrés Mecánico , Factores de Tiempo , Transfección , Regulación hacia Arriba
14.
Arterioscler Thromb Vasc Biol ; 31(7): 1625-33, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21527747

RESUMEN

OBJECTIVE: The goal of this study was to assess the activity of ß-catenin/T-cell-specific transcription factor (TCF) signaling in atherosclerosis development and its regulation of fibronectin in vascular endothelium. METHODS AND RESULTS: Histological staining identified preferential nuclear localization of ß-catenin in the endothelium of atheroprone aorta before and during lesion development. Transgenic reporter studies revealed that increased levels of TCF transcriptional activity in endothelium correlated anatomically with ß-catenin nuclear localization and fibronectin deposition. Exposure of endothelial cells to human-derived atheroprone shear stress induced nuclear localization of ß-catenin, transcriptional activation of TCF, and expression of fibronectin. Activation of fibronectin expression required ß-catenin, TCF, and the transcriptional coactivator CRBP-binding protein. Finally, we identified platelet endothelial cell adhesion molecule-1 as a critical regulator of constitutive ß-catenin and glycogen synthase kinase-3ß activities. CONCLUSIONS: These data reveal novel constitutive activation of the endothelial ß-catenin/TCF signaling pathway in atherosclerosis and regulation of fibronectin through hemodynamic shear stress.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Fibronectinas/metabolismo , Hemodinámica , Inflamación/metabolismo , Transducción de Señal , Factores de Transcripción TCF/metabolismo , beta Catenina/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Núcleo Celular/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Genes Reporteros , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Inflamación/genética , Inflamación/patología , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Interferencia de ARN , Estrés Mecánico , Factores de Transcripción TCF/genética , Factores de Tiempo , Activación Transcripcional , Transfección , beta Catenina/genética
15.
Transl Vis Sci Technol ; 11(3): 13, 2022 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-35275207

RESUMEN

Purpose: Subretinal injection (SRI) in mice is widely used in retinal research, yet the learning curve (LC) of this surgically challenging technique is unknown. Methods: To evaluate the LC for SRI in a murine model, we analyzed training data from three clinically trained ophthalmic surgeons from 2018 to 2020. Successful SRI was defined as either the absence of retinal pigment epithelium (RPE) degeneration after phosphate buffered saline injection or the presence of RPE degeneration after Alu RNA injection. Multivariable survival-time regression models were used to evaluate the association between surgeon experience and success rate, with adjustment for injection agents, and to calculate an approximate case number to achieve a 95% success rate. Cumulative sum (CUSUM) analyses were performed and plotted individually to monitor each surgeon's simultaneous performance. Results: Despite prior microsurgery experience, the combined average success rate of the first 50 cases in mice was only 27%. The predicted SRI success rate did not reach a plateau above 95% until approximately 364 prior cases. Using the 364 training cases as a cutoff point, the predicted probability of success for cases 1 to 364 was 65.38%, and for cases 365 to 455 it was 99.32% (P < 0.0001). CUSUM analysis showed an initial upward slope and then remained within the decision intervals with an acceptable success rate set at 95% in the late stage. Conclusions: This study demonstrates the complexity and substantial LC for successful SRI in mice with high confidence. A systematic training system could improve the reliability and reproducibility of SRI-related experiments and improve the interpretation of experimental results using this technique. Translational Relevance: Our prediction model and monitor system allow objective quantification of technical proficiency in the field of subretinal drug delivery and gene therapy for the first time, to the best of our knowledge.


Asunto(s)
Oftalmólogos , Cirujanos , Animales , Humanos , Curva de Aprendizaje , Ratones , Tempo Operativo , Reproducibilidad de los Resultados , Cirujanos/educación
16.
Nat Protoc ; 17(6): 1468-1485, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35418688

RESUMEN

Subretinal injection (SRI) is a widely used technique in retinal research and can be used to deliver nucleic acids, small molecules, macromolecules, viruses, cells or biomaterials such as nanobeads. Here we describe how to undertake SRI of mice. This protocol was adapted from a technique initially described for larger animals. Although SRI is a common procedure in eye research laboratories, there is no published guidance on the best practices for determining what constitutes a 'successful' SRI. Optimal injections are required for reproducibility of the procedure and, when carried out suboptimally, can lead to erroneous conclusions. To address this issue, we propose a standardized protocol for SRI with 'procedure success' defined by follow-up examination of the retina and the retinal pigmented epithelium rather than solely via intraoperative endpoints. This protocol takes 7-14 d to complete, depending on the reagent delivered. We have found, by instituting a standardized training program, that trained ophthalmologists achieve reliable proficiency in this technique after ~350 practice injections. This technique can be used to gain insights into retinal physiology and disease pathogenesis and to test the efficacy of experimental compounds in the retina or retinal pigmented epithelium.


Asunto(s)
Retina , Epitelio Pigmentado de la Retina , Animales , Inyecciones , Ratones , Reproducibilidad de los Resultados , Retina/patología
17.
Nat Commun ; 12(1): 6207, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34707113

RESUMEN

Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), produced by cyclic GMP-AMP synthase (cGAS), stimulates the production of type I interferons (IFN). Here we show that cGAMP activates DNA damage response (DDR) signaling independently of its canonical IFN pathways. Loss of cGAS dampens DDR signaling induced by genotoxic insults. Mechanistically, cGAS activates DDR in a STING-TBK1-dependent manner, wherein TBK1 stimulates the autophosphorylation of the DDR kinase ATM, with the consequent activation of the CHK2-p53-p21 signal transduction pathway and the induction of G1 cell cycle arrest. Despite its stimulatory activity on ATM, cGAMP suppresses homology-directed repair (HDR) through the inhibition of polyADP-ribosylation (PARylation), in which cGAMP reduces cellular levels of NAD+; meanwhile, restoring NAD+ levels abrogates cGAMP-mediated suppression of PARylation and HDR. Finally, we show that cGAMP also activates DDR signaling in invertebrate species lacking IFN (Crassostrea virginica and Nematostella vectensis), suggesting that the genome surveillance mechanism of cGAS predates metazoan interferon-based immunity.


Asunto(s)
Daño del ADN , Nucleótidos Cíclicos/metabolismo , Transducción de Señal , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Crassostrea/genética , Crassostrea/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Nucleotidiltransferasas/metabolismo , Fosforilación , Poli ADP Ribosilación , Proteínas Serina-Treonina Quinasas/metabolismo , Reparación del ADN por Recombinación , Anémonas de Mar/genética , Anémonas de Mar/metabolismo
18.
Signal Transduct Target Ther ; 6(1): 149, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33850097

RESUMEN

Nonfibrillar amyloid-ß oligomers (AßOs) are a major component of drusen, the sub-retinal pigmented epithelium (RPE) extracellular deposits characteristic of age-related macular degeneration (AMD), a common cause of global blindness. We report that AßOs induce RPE degeneration, a clinical hallmark of geographic atrophy (GA), a vision-threatening late stage of AMD that is currently untreatable. We demonstrate that AßOs induce activation of the NLRP3 inflammasome in the mouse RPE in vivo and that RPE expression of the purinergic ATP receptor P2RX7, an upstream mediator of NLRP3 inflammasome activation, is required for AßO-induced RPE degeneration. Two classes of small molecule inflammasome inhibitors-nucleoside reverse transcriptase inhibitors (NRTIs) and their antiretrovirally inert modified analog Kamuvudines-both inhibit AßOs-induced RPE degeneration. These findings crystallize the importance of P2RX7 and NLRP3 in a disease-relevant model of AMD and identify inflammasome inhibitors as potential treatments for GA.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Degeneración Macular/tratamiento farmacológico , Epitelio Pigmentado de la Retina/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología , Péptidos beta-Amiloides/genética , Animales , Modelos Animales de Enfermedad , Humanos , Degeneración Macular/genética , Degeneración Macular/metabolismo , Masculino , Ratones , Ratones Noqueados
19.
Sci Immunol ; 6(66): eabi4493, 2021 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-34860583

RESUMEN

Detection of microbial products by multiprotein complexes known as inflammasomes is pivotal to host defense against pathogens. Nucleotide-binding domain leucine-rich repeat (NLR) CARD domain containing 4 (NLRC4) forms an inflammasome in response to bacterial products; this requires their detection by NLR family apoptosis inhibitory proteins (NAIPs), with which NLRC4 physically associates. However, the mechanisms underlying sterile NLRC4 inflammasome activation, which is implicated in chronic noninfectious diseases, remain unknown. Here, we report that endogenous short interspersed nuclear element (SINE) RNAs, which promote atrophic macular degeneration (AMD) and systemic lupus erythematosus (SLE), induce NLRC4 inflammasome activation independent of NAIPs. We identify DDX17, a DExD/H box RNA helicase, as the sensor of SINE RNAs that licenses assembly of an inflammasome comprising NLRC4, NLR pyrin domain­containing protein 3, and apoptosis-associated speck-like protein­containing CARD and induces caspase-1 activation and cytokine release. Inhibiting DDX17-mediated NLRC4 inflammasome activation decreased interleukin-18 release in peripheral blood mononuclear cells of patients with SLE and prevented retinal degeneration in an animal model of AMD. Our findings uncover a previously unrecognized noncanonical NLRC4 inflammasome activated by endogenous retrotransposons and provide potential therapeutic targets for SINE RNA­driven diseases.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas de Unión al Calcio/inmunología , ARN Helicasas DEAD-box/inmunología , Inflamasomas/inmunología , ARN/inmunología , Retroelementos/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas de Unión al Calcio/deficiencia , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
20.
Sci Adv ; 7(40): eabj3658, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34586848

RESUMEN

Long interspersed nuclear element-1 (L1)­mediated reverse transcription (RT) of Alu RNA into cytoplasmic Alu complementary DNA (cDNA) has been implicated in retinal pigmented epithelium (RPE) degeneration. The mechanism of Alu cDNA­induced cytotoxicity and its relevance to human disease are unknown. Here we report that Alu cDNA is highly enriched in the RPE of human eyes with geographic atrophy, an untreatable form of age-related macular degeneration. We demonstrate that the DNA sensor cGAS engages Alu cDNA to induce cytosolic mitochondrial DNA escape, which amplifies cGAS activation, triggering RPE degeneration via the inflammasome. The L1-extinct rice rat was resistant to Alu RNA­induced Alu cDNA synthesis and RPE degeneration, which were enabled upon L1-RT overexpression. Nucleoside RT inhibitors (NRTIs), which inhibit both L1-RT and inflammasome activity, and NRTI derivatives (Kamuvudines) that inhibit inflammasome, but not RT, both block Alu cDNA toxicity, identifying inflammasome activation as the terminal effector of RPE degeneration.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA