Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Exp Med Biol ; 1415: 207-213, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37440035

RESUMO

Age-related macular degeneration (AMD) is the leading cause of visual impairment in the elderly in developed countries. It is a complex, multifactorial, progressive disease with diverse molecular pathways, including inflammation, regulating its pathogenesis. The myeloid marker CD68 is a protein highly expressed in circulating and tissue macrophages. Recent observations of immune markers in human AMD tissues have varied with some finding ectopic RPE cells in advanced AMD and others noting negligible numbers of CD68-positive cells. Additionally, animal models of retinal degeneration have shown upregulation of CD68, in a protective population of retinal microglia. Herein, we review the potential role of CD68 in regulating RPE health and inflammation in the sub-retinal space and discuss observations on its localization in a mouse model that presents with AMD-like features.


Assuntos
Degeneração Macular , Degeneração Retiniana , Camundongos , Animais , Humanos , Idoso , Epitélio Pigmentado da Retina/patologia , Retina/patologia , Degeneração Macular/patologia , Degeneração Retiniana/patologia , Inflamação/metabolismo
2.
J Ocul Pharmacol Ther ; 39(4): 290-299, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36944130

RESUMO

Purpose: Angiotensin-(1-12) [Ang-(1-12)] serves as a primary substrate to generate angiotensin II (Ang II) by angiotensin-converting enzyme and/or chymase suggests it may be an unrecognized source of Ang II-mediated microvascular complication in hypertension-mediated retinopathy. We investigated Ang-(1-12) expression and internalization in adult retinal pigment epithelial-19 (ARPE-19) cultured cells. We performed the internalization of Ang-(1-12) in ARPE-19 cells in the presence of a highly specific monoclonal antibody (mAb) developed against the C-terminal end of the Ang-(1-12) sequence. Methods: All experiments were performed in confluent ARPE-19 cells (passage 28-35). We employed high-performance liquid chromatography to purify radiolabeled, 125I-Ang-(1-12) and immuno-neutralization with Ang-(1-12) mAb to demonstrate Ang-(1-12)'s internalization in ARPE-19 cells. Internalization was also demonstrated by immunofluorescence (IF) method. Results: These procedures revealed internalization of an intact 125I-Ang-(1-12) in ARPE-19 cells. A significant reduction (∼53%, P < 0.0001) in 125I-Ang-(1-12) internalization was detected in APRE-19 cells in the presence of the mAb. IF staining experiments further confirms internalization of Ang-(1-12) into the cells from the extracellular culture medium. No endogenous expression was detected in the ARPE-19 cells. An increased intensity of IF staining was detected in cells exposed to 1.0 µM Ang-(1-12) compared with 0.1 µM. Furthermore, we found hydrolysis of Ang-(1-12) into Ang II by ARPE-19 cells' plasma membranes. Conclusions: Intact Ang-(1-12) peptide is internalized from the extracellular spaces in ARPE-19 cells and metabolized into Ang II. The finding that a selective mAb blocks cellular internalization of Ang-(1-12) suggests alternate therapeutic approaches to prevent/reduce the RPE cells Ang II burden.


Assuntos
Angiotensina II , Radioisótopos do Iodo , Angiotensina II/farmacologia , Angiotensina II/metabolismo , Pigmentos da Retina , Células Cultivadas
3.
Prog Retin Eye Res ; 94: 101130, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36220751

RESUMO

The functions and activities of nuclear receptors, the largest family of transcription factors in the human genome, have classically focused on their ability to act as steroid and hormone sensors in endocrine organs. However, they are responsible for a diverse array of physiological functions, including cellular homeostasis and metabolism, during development and aging. Though the eye is not a traditional endocrine organ, recent studies have revealed high expression levels of nuclear receptors in cells throughout the posterior pole. These findings have precipitated an interest in investigating the role of these transcription factors in the eye as a function of age and ocular disease, in particular age-related macular degeneration (AMD). As the leading cause of vision impairment in the elderly, identifying signaling pathways that may be targeted for AMD therapy is of great importance, given the lack of therapeutic options for over 85% of patients with this disease. Herein we review this relatively new field and recent findings supporting the hypothesis that the eye is a secondary endocrine organ, in which nuclear receptors serve as the bedrock for biological processes in cells vulnerable in AMD, including retinal pigment epithelial and choroidal endothelial cells, and discuss the therapeutic potential of targeting these receptors for AMD.


Assuntos
Células Endoteliais , Degeneração Macular , Humanos , Idoso , Células Endoteliais/metabolismo , Degeneração Macular/genética , Envelhecimento/fisiologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/uso terapêutico , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/uso terapêutico , Epitélio Pigmentado da Retina/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(28): e2202256119, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35867766

RESUMO

Phenotypic variations in the retinal pigment epithelial (RPE) layer are often a predecessor and driver of ocular degenerative diseases, such as age-related macular degeneration (AMD), the leading cause of vision loss in the elderly. We previously identified the orphan nuclear receptor-related 1 (NURR1), from a nuclear receptor atlas of human RPE cells, as a candidate transcription factor potentially involved in AMD development and progression. In the present study we characterized the expression of NURR1 as a function of age in RPE cells harvested from human donor eyes and in donor tissue from AMD patients. Mechanistically, we found an age-dependent shift in NURR1 dimerization from NURR1-RXRα heterodimers toward NURR1-NURR1 homodimers in primary human RPE cells. Additionally, overexpression and activation of NURR1 attenuated TNF-α-induced epithelial-to-mesenchymal transition (EMT) and migration, and modulated EMT-associated gene and protein expression in human RPE cells independent of age. In vivo, oral administration of IP7e, a potent NURR1 activator, ameliorated EMT in an experimental model of wet AMD and improved retinal function in a mouse model that presents with dry AMD features, impacting AMD phenotype, structure, and function of RPE cells, inhibiting accumulation of immune cells, and diminishing lipid accumulation. These results provide insight into the mechanisms of action of NURR1 in the aging eye, and demonstrate that the relative expression levels and activity of NURR1 is critical for both physiological and pathological functions of human RPE cells through RXRα-dependent regulation, and that targeting NURR1 may have therapeutic potential for AMD by modulating EMT, inflammation, and lipid homeostasis.


Assuntos
Transição Epitelial-Mesenquimal , Degeneração Macular , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares , Epitélio Pigmentado da Retina , Idoso , Animais , Humanos , Lipídeos , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Camundongos , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/química , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Fenótipo , Multimerização Proteica , Epitélio Pigmentado da Retina/metabolismo
5.
Lab Invest ; 102(10): 1132-1142, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35589984

RESUMO

Apolipoprotein B100 (apoB100) is the structural protein of cholesterol carriers including low-density lipoproteins. It is a constituent of sub-retinal pigment epithelial (sub-RPE) deposits and pro-atherogenic plaques, hallmarks of early dry age-related macular degeneration (AMD), an ocular neurodegenerative blinding disease, and cardiovascular disease, respectively. Herein, we characterized the retinal pathology of transgenic mice expressing mouse apoB100 in order to catalog their functional and morphological ocular phenotypes as a function of age and establish measurable endpoints for their use as a mouse model to test potential therapies. ApoB100 mice were found to exhibit an age-related decline in retinal function, as measured by electroretinogram (ERG) recordings of their scotopic a-wave, scotopic b-wave; and c-wave amplitudes. ApoB100 mice also displayed a buildup of the cholesterol carrier, apolipoprotein E (apoE) within and below the supporting extracellular matrix, Bruch's membrane (BrM), along with BrM thickening, and accumulation of thin diffuse electron-dense sub-RPE deposits, the severity of which increased with age. Moreover, the combination of apoB100 and advanced age were found to be associated with RPE morphological changes and the presence of sub-retinal immune cells as visualized in RPE-choroid flatmounts. Finally, aged apoB100 mice showed higher levels of circulating and ocular pro-inflammatory cytokines, supporting a link between age and increased local and systemic inflammation. Collectively, the data support the use of aged apoB100 mice as a platform to evaluate potential therapies for retinal degeneration, specifically drugs intended to target removal of lipids from Bruch's membrane and/or alleviate ocular inflammation.


Assuntos
Degeneração Macular , Degeneração Retiniana , Animais , Apolipoproteínas E , Colesterol/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Inflamação/metabolismo , Lipoproteínas LDL/metabolismo , Degeneração Macular/genética , Camundongos , Camundongos Transgênicos , Epitélio Pigmentado da Retina/metabolismo , Pigmentos da Retina/metabolismo
6.
J Ocul Pharmacol Ther ; 38(5): 359-371, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35446130

RESUMO

Purpose: The NLRP3 inflammasome, a cytoplasmic signal transduction complex that regulates inflammation, has been implicated in the pathogenesis of age-related macular degeneration (AMD), the leading cause of visual impairment in industrialized countries. We tested the therapeutic effect of anti-inflammatory gene therapy, delivered preventively, in Liver-X-Receptor alpha knockout (LXRα-/-) mice, which exhibit features of dry AMD. Methods:LXRα-/- mice were treated with an adeno-associated virus (AAV) vector that delivers a secretable and cell-penetrating form of the caspase activation and recruitment domain (CARD). A sGFP-FCS-TatCARD-AAV or sGFP-FCS (control) vector was delivered intravitreally to 3-5 month-old, LXRα-/- mice, who were then aged to 15-18 months (12-13 month treatment). Retinal function and morphology were assessed pre- and post-treatment. Results: TatCARD treated LXRα-/- mice did not show improvement in rod and cone photoreceptor function, measured by dark adapted a- and b-wave amplitudes, and rod-saturated b-wave amplitudes. We found a sex-dependent, significant therapeutic effect in c-wave amplitudes in the TatCARD treated mice, which exhibited maintenance of amplitudes in comparison to the significant decline recorded in the control treated group, indicating a therapeutic effect mediated in part through retinal pigment epithelial (RPE) cells. Additionally, the retinas of the TatCARD treated mice exhibited a significant decline in the concentration of interleukin-1 beta (IL-1ß) concomitant with modulation of several inflammatory cytokines in the retina and RPE-choroid tissues, as measured by ELISA and cytokine array, respectively. Conclusion: Collectively, these results support that anti-inflammatory gene constructs such as AAV-TatCARD may be considered for the treatment of inflammation in AMD and other ocular diseases of the posterior pole in which inflammation may play a role. Furthermore, our findings emphasize the need to carefully consider potential sex-different responses when assessing potential therapies in pre-clinical models.


Assuntos
Degeneração Macular , Pigmentos da Retina , Animais , Domínio de Ativação e Recrutamento de Caspases , Modelos Animais de Doenças , Terapia Genética , Inflamação/patologia , Degeneração Macular/genética , Degeneração Macular/patologia , Degeneração Macular/terapia , Camundongos , Epitélio Pigmentado da Retina/patologia
7.
Mod Pathol ; 35(2): 165-176, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34389792

RESUMO

A common clinical phenotype of several neurodegenerative and systemic disorders including Alzheimer's disease and atherosclerosis is the abnormal accumulation of extracellular material, which interferes with routine cellular functions. Similarly, patients with age-related macular degeneration (AMD), the leading cause of vision loss among the aged population, present with extracellular lipid- and protein-filled basal deposits in the back of the eye. While the exact mechanism of growth and formation of these deposits is poorly understood, much has been learned from investigating their composition, providing critical insights into AMD pathogenesis, prevention, and therapeutics. We identified human osteopontin (OPN), a phosphoprotein expressed in a variety of tissues in the body, as a newly discovered component of basal deposits in AMD patients, with a distinctive punctate staining pattern. OPN expression within these lesions, which are associated with AMD disease progression, were found to co-localize with abnormal calcium deposition. Additionally, OPN puncta colocalized with an AMD risk-associated complement pathway protein, but not with apolipoprotein E or vitronectin, two other well-established basal deposit components. Mechanistically, we found that retinal pigment epithelial cells, cells vulnerable in AMD, will secrete OPN into the extracellular space, under oxidative stress conditions, supporting OPN biosynthesis locally within the outer retina. Finally, we report that OPN levels in plasma of aged (non-AMD) human donors were significantly higher than levels in young (non-AMD) donors, but were not significantly different from donors with the different clinical subtypes of AMD. Collectively, our study defines the expression pattern of OPN in the posterior pole as a function of disease, and its local expression as a potential histopathologic biomarker of AMD.


Assuntos
Degeneração Macular , Osteopontina , Envelhecimento/patologia , Biomarcadores , Humanos , Degeneração Macular/genética , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Retina/metabolismo , Retina/patologia
8.
Int J Mol Sci ; 21(18)2020 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-32947781

RESUMO

The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, which senses environmental, dietary or metabolic signals to mount a transcriptional response, vital in health and disease. As environmental stimuli and metabolic products have been shown to impact the central nervous system (CNS), a burgeoning area of research has been on the role of the AHR in ocular and non-ocular neurodegenerative diseases. Herein, we summarize our current knowledge, of AHR-controlled cellular processes and their impact on regulating pathobiology of select ocular and neurodegenerative diseases. We catalogue animal models generated to study the role of the AHR in tissue homeostasis and disease pathogenesis. Finally, we discuss the potential of targeting the AHR pathway as a therapeutic strategy, in the context of the maladies of the eye and brain.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Oftalmopatias/metabolismo , Terapia de Alvo Molecular , Doenças Neurodegenerativas/metabolismo , Receptores de Hidrocarboneto Arílico/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/agonistas , Biotransformação , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/fisiologia , Modelos Animais de Doenças , Oftalmopatias/tratamento farmacológico , Oftalmopatias/genética , Proteínas do Olho/fisiologia , Regulação da Expressão Gênica , Sequências Hélice-Alça-Hélice , Humanos , Ligantes , Mamíferos/metabolismo , Esclerose Múltipla/tratamento farmacológico , Degeneração Neural , Proteínas do Tecido Nervoso/fisiologia , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/genética , Doença de Parkinson/metabolismo , Dibenzodioxinas Policloradas/farmacologia , Quinolonas/farmacologia , Quinolonas/uso terapêutico , Ratos , Receptores de Hidrocarboneto Arílico/agonistas , Transdução de Sinais/fisiologia , Transcrição Gênica , Xenobióticos/metabolismo
9.
JCI Insight ; 5(1)2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31829999

RESUMO

Effective treatments and animal models for the most prevalent neurodegenerative form of blindness in elderly people, called age-related macular degeneration (AMD), are lacking. Genome-wide association studies have identified lipid metabolism and inflammation as AMD-associated pathogenic pathways. Given liver X receptors (LXRs), encoded by the nuclear receptor subfamily 1 group H members 2 and 3 (NR1H3 and NR1H2), are master regulators of these pathways, herein we investigated the role of LXR in human and mouse eyes as a function of age and disease and tested the therapeutic potential of targeting LXR. We identified immunopositive LXR fragments in human extracellular early dry AMD lesions and a decrease in LXR expression within the retinal pigment epithelium (RPE) as a function of age. Aged mice lacking LXR presented with isoform-dependent ocular pathologies. Specifically, loss of the Nr1h3 isoform resulted in pathobiologies aligned with AMD, supported by compromised visual function, accumulation of native and oxidized lipids in the outer retina, and upregulation of ocular inflammatory cytokines, while absence of Nr1h2 was associated with ocular lipoidal degeneration. LXR activation not only ameliorated lipid accumulation and oxidant-induced injury in RPE cells but also decreased ocular inflammatory markers and lipid deposition in a mouse model, thereby providing translational support for pursuing LXR-active pharmaceuticals as potential therapies for dry AMD.


Assuntos
Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Degeneração Macular/genética , Degeneração Macular/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Animais , Modelos Animais de Doenças , Células Endoteliais , Feminino , Estudo de Associação Genômica Ampla , Humanos , Inflamação/metabolismo , Degeneração Macular/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Fenótipo , Retina/metabolismo , Retina/patologia , Epitélio Pigmentado da Retina , Transcriptoma , Adulto Jovem
10.
Adv Exp Med Biol ; 1185: 9-13, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31884581

RESUMO

Age-related macular degeneration (AMD) continues to be the leading cause of visual impairment for the elderly in developed countries. It is a complex, multifactorial, progressive disease with diverse molecular pathways regulating its pathogenesis. One of the cardinal features of the early clinical subtype of AMD is the accumulation of lipid- and protein-rich deposits within Bruch's membrane, called drusen, which can be visualized by fundus imaging. Currently, multiple in vitro and in vivo model systems exist, which can be used to help tease out mechanisms associated with different molecular pathways driving disease initiation and progression. Given the lack of treatments for patients suffering from the dry form of AMD, it is imperative to appreciate the different known morphological endpoints associated with the various pathogenic pathways, in order to derive further insights, for the ultimate purpose of disease modeling and development of effective therapeutic interventions.


Assuntos
Lâmina Basilar da Corioide/patologia , Degeneração Macular/patologia , Retina/patologia , Idoso , Humanos
11.
JCI Insight ; 3(17)2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30185655

RESUMO

Advanced age-related macular degeneration (AMD), the leading cause of blindness among people over 50 years of age, is characterized by atrophic neurodegeneration or pathologic angiogenesis. Early AMD is characterized by extracellular cholesterol-rich deposits underneath the retinal pigment epithelium (RPE) called drusen or in the subretinal space called subretinal drusenoid deposits (SDD) that drive disease progression. However, mechanisms of drusen and SDD biogenesis remain poorly understood. Although human AMD is characterized by abnormalities in cholesterol homeostasis and shares phenotypic features with atherosclerosis, it is unclear whether systemic immunity or local tissue metabolism regulates this homeostasis. Here, we demonstrate that targeted deletion of macrophage cholesterol ABC transporters A1 (ABCA1) and -G1 (ABCG1) leads to age-associated extracellular cholesterol-rich deposits underneath the neurosensory retina similar to SDD seen in early human AMD. These mice also develop impaired dark adaptation, a cardinal feature of RPE cell dysfunction seen in human AMD patients even before central vision is affected. Subretinal deposits in these mice progressively worsen with age, with concomitant accumulation of cholesterol metabolites including several oxysterols and cholesterol esters causing lipotoxicity that manifests as photoreceptor dysfunction and neurodegeneration. These findings suggest that impaired macrophage cholesterol transport initiates several key elements of early human AMD, demonstrating the importance of systemic immunity and aging in promoting disease manifestation. Polymorphisms in genes involved with cholesterol transport and homeostasis are associated with a significantly higher risk of developing AMD, thus making these studies translationally relevant by identifying potential targets for therapy.


Assuntos
Cegueira/induzido quimicamente , Cegueira/metabolismo , Colesterol/metabolismo , Degeneração Macular/induzido quimicamente , Degeneração Macular/metabolismo , Monócitos/metabolismo , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Cegueira/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Ésteres do Colesterol/metabolismo , Progressão da Doença , Deleção de Genes , Humanos , Imunidade Inata , Degeneração Macular/imunologia , Degeneração Macular/patologia , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Oxisteróis/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Receptores Acoplados a Proteínas G/metabolismo , Retina/anormalidades , Retina/metabolismo , Retina/patologia , Epitélio Pigmentado da Retina/anormalidades , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia
12.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1583-1595, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29481912

RESUMO

The aryl hydrocarbon receptor (AhR) is a ligand activated transcription factor, initially discovered for its role in regulating xenobiotic metabolism. There is extensive evidence supporting a multi-faceted role for AhR, modulating physiological pathways important in cell health and disease. Recently we demonstrated that the AhR plays a role in the pathogenesis of age-related macular degeneration (AMD), the leading cause of vision loss in the elderly. We found that loss of AhR exacerbates choroidal neovascular (CNV) lesion formation in a murine model. Herein we tested the therapeutic impact of AhR activation on CNV lesion formation and factors associated with aberrant neovascularization. We screened a panel of synthetic drugs and endogenous AhR ligands, assessed their ability to activate AhR in choroidal endothelial cells, and inhibit angiogenesis in vitro. Drugs with an anti-angiogenic profile were then administered to a murine model of CNV. Two compounds, leflunomide and flutamide, significantly inhibited CNV formation concurrent with positive modifying effects on angiogenesis, inflammation, extracellular matrix remodeling, and fibrosis. These results validate the role of the AhR pathway in regulating CNV pathogenesis, identify mechanisms of AhR-based therapies in the eye, and argue in favor of developing AhR as a drug target for the treatment of neovascular AMD.


Assuntos
Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/prevenção & controle , Células Endoteliais/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Linhagem Celular , Neovascularização de Coroide/genética , Neovascularização de Coroide/patologia , Células Endoteliais/patologia , Macaca mulatta , Degeneração Macular/genética , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Degeneração Macular/prevenção & controle , Camundongos , Camundongos Knockout , Receptores de Hidrocarboneto Arílico/genética
13.
Expert Opin Drug Discov ; 13(4): 359-377, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29382242

RESUMO

INTRODUCTION: The impact of vision debilitating diseases is a global public health concern, which will continue until effective preventative and management protocols are developed. Two retinal diseases responsible for the majority of vision loss in the working age adults and elderly populations are diabetic retinopathy (DR) and age-related macular degeneration (AMD), respectively. Model systems, which recapitulate aspects of human pathology, are valid experimental modalities that have contributed to the identification of signaling pathways involved in disease development and consequently potential therapies. Areas covered: The pathology of DR and AMD, which serve as the basis for designing appropriate models of disease, is discussed. The authors also review in vitro and in vivo models of DR and AMD and evaluate the utility of these models in exploratory and pre-clinical studies. Expert opinion: The complex nature of non-Mendelian diseases such as DR and AMD has made identification of effective therapeutic treatments challenging. However, the authors believe that while in vivo models are often criticized for not being a 'perfect' recapitulation of disease, they have been valuable experimentally when used with consideration of the strengths and limitations of the experimental model selected and have a place in the drug discovery process.


Assuntos
Retinopatia Diabética/tratamento farmacológico , Descoberta de Drogas/métodos , Degeneração Macular/tratamento farmacológico , Adulto , Idoso , Animais , Retinopatia Diabética/fisiopatologia , Modelos Animais de Doenças , Desenho de Fármacos , Humanos , Degeneração Macular/fisiopatologia
14.
Aging (Albany NY) ; 8(9): 1952-1978, 2016 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-27622388

RESUMO

Peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) is a nuclear receptor that regulates differentiation, inflammation, lipid metabolism, extracellular matrix remodeling, and angiogenesis in multiple tissues. These pathways are also central to the pathogenesis of age-related macular degeneration (AMD), the leading cause of vision loss globally. With the goal of identifying signaling pathways that may be important in the development of AMD, we investigated the impact of PPARß/δ activation on ocular tissues affected in the disease. PPARß/δ is expressed and can be activated in AMD vulnerable cells, including retinal pigment epithelial (RPE) and choroidal endothelial cells. Further, PPARß/δ knockdown modulates AMD-related pathways selectively. Specifically, genetic ablation of Pparß/δ in aged mice resulted in exacerbation of several phenotypic features of early dry AMD, but attenuation of experimentally induced choroidal neovascular (CNV) lesions. Antagonizing PPARß/δ in both in vitro angiogenesis assays and in the in vivo experimentally induced CNV model, inhibited angiogenesis and angiogenic pathways, while ligand activation of PPARß/δ, in vitro, decreased RPE lipid accumulation, characteristic of dry AMD. This study demonstrates for the first time, selective regulation of a nuclear receptor in the eye and establishes that selective targeting of PPARß/δ may be a suitable strategy for treatment of different clinical sub-types of AMD.


Assuntos
Degeneração Macular/metabolismo , Neovascularização Patológica/metabolismo , PPAR beta/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Idoso , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Eletrorretinografia , Feminino , Humanos , Macaca mulatta , Degeneração Macular/genética , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Neovascularização Patológica/genética , PPAR beta/agonistas , PPAR beta/antagonistas & inibidores , PPAR beta/genética , Fenótipo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sulfonas/farmacologia , Tiazóis/farmacologia , Tiofenos/farmacologia , Adulto Jovem
15.
J Biomol Screen ; 21(10): 1007-1018, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27455994

RESUMO

Collectively, retinal diseases, including age-related macular degeneration, retinitis pigmentosa, and diabetic retinopathy, result in severe vision impairment worldwide. The absence and/or limited availability of successful drug therapies for these blinding disorders necessitates further understanding their pathobiology and identifying new targetable signaling pathways. Nuclear receptors are transcription regulators of many key aspects of human physiology, as well as pathophysiology, with reported roles in development, aging, and disease. Some of the pathways regulated by nuclear receptors include, but are not limited to, angiogenesis, inflammation, and lipid metabolic dysregulation, mechanisms also important in the initiation and development of several retinal diseases. Herein, we present an overview of the biology of three diseases affecting the posterior eye, summarize a growing body of evidence that suggests direct or indirect involvement of nuclear receptors in disease progression, and discuss the therapeutic potential of targeting nuclear receptors for treatment.


Assuntos
Retinopatia Diabética/genética , Degeneração Macular/genética , Receptores Citoplasmáticos e Nucleares/genética , Doenças Retinianas/genética , Retinose Pigmentar/genética , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/patologia , Humanos , Degeneração Macular/tratamento farmacológico , Degeneração Macular/patologia , Terapia de Alvo Molecular , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Doenças Retinianas/tratamento farmacológico , Doenças Retinianas/patologia , Retinose Pigmentar/tratamento farmacológico , Retinose Pigmentar/patologia , Transdução de Sinais
16.
Adv Exp Med Biol ; 854: 45-51, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26427392

RESUMO

Age-related macular degeneration (AMD) is the leading cause of legal blindness and visual impairment in individuals over 60 years of age in the Western World. A common morphological denominator in all forms of AMD is the accumulation of microglia within the sub-retinal space, which is believed to be a contributing factor to AMD progression. However, the signaling pathway and molecular players regulating microglial recruitment have not been completely identified. Multiple in-vitro and in-vivo studies, to date, have highlighted the contributions of nuclear receptor ligands in the treatment of inflammation related disorders such as atherosclerosis and Alzheimer's disease. Given that inflammation and the immune response play a vital role in the initiation and progression of AMD, in this brief review we will highlight some of these studies with a particular focus on the lipid activated "adopted orphan" nuclear receptors, the liver x receptors (LXRs) and the peroxisome proliferator-activated receptors (PPARs). The results of these studies strongly support the rationale that treatment with LXR and PPAR ligands may ameliorate microglial activation in the sub-retinal space and ultimately slow down or reverse the progression of AMD.


Assuntos
Degeneração Macular/metabolismo , Microglia/metabolismo , Receptores Nucleares Órfãos/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Animais , Humanos , Inflamação/metabolismo , Receptores X do Fígado , Epitélio Pigmentado da Retina/metabolismo , Transdução de Sinais
17.
BMC Cancer ; 15: 427, 2015 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-26002762

RESUMO

BACKGROUND: Proper functional association between mural cells and endothelial cells (EC) causes EC of blood vessels to become quiescent. Mural cells on tumor vessels exhibit decreased attachment to EC, which allows vessels to be unstable and proliferative. The mechanisms by which tumors prevent proper association between mural cells and EC are not well understood. Since gap junctions (GJ) play an important role in cell-cell contact and communication, we investigated whether loss of GJ plays a role in tumor-induced mural cell dissociation. METHODS: Mural cell regulation of endothelial proliferation was assessed by direct co-culture assays of fluorescently labeled cells quantified by flow cytometry or plate reader. Gap junction function was assessed by parachute assay. Connexin 43 (Cx43) protein in mural cells exposed to conditioned media from cancer cells was assessed by Western and confocal microscopy; mRNA levels were assessed by quantitative real-time PCR. Expression vectors or siRNA were utilized to overexpress or knock down Cx43. Tumor growth and angiogenesis was assessed in mouse hosts deficient for Cx43. RESULTS: Using parachute dye transfer assay, we demonstrate that media conditioned by MDA-MB-231 breast cancer cells diminishes GJ communication between mural cells (vascular smooth muscle cells, vSMC) and EC. Both protein and mRNA of the GJ component Connexin 43 (Cx43) are downregulated in mural cells by tumor-conditioned media; media from non-tumorigenic MCF10A cells had no effect. Loss of GJ communication by Cx43 siRNA knockdown, treatment with blocking peptide, or exposure to tumor-conditioned media diminishes the ability of mural cells to inhibit EC proliferation in co-culture assays, while overexpression of Cx43 in vSMC restores GJ and endothelial inhibition. Breast tumor cells implanted into mice heterozygous for Cx43 show no changes in tumor growth, but exhibit significantly increased tumor vascularization determined by CD31 staining, along with decreased mural cell support detected by NG2 staining. CONCLUSIONS: Our data indicate that i) functional Cx43 is required for mural cell-induced endothelial quiescence, and ii) downregulation of Cx43 GJ by tumors frees endothelium to respond to angiogenic cues. These data define a novel and important role for maintained Cx43 function in regulation of vessel quiescence, and suggest its loss may contribute to pathological tumor angiogenesis.


Assuntos
Neoplasias da Mama/metabolismo , Conexina 43/metabolismo , Células Endoteliais/fisiologia , Endotélio Vascular/fisiopatologia , Junções Comunicantes/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Neoplasias da Mama/irrigação sanguínea , Comunicação Celular , Linhagem Celular Tumoral , Proliferação de Células , Conexina 43/antagonistas & inibidores , Conexina 43/genética , Meios de Cultivo Condicionados , Humanos , Camundongos , Neovascularização Patológica/fisiopatologia , RNA Mensageiro/metabolismo
18.
J Pathol ; 235(1): 101-12, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25186463

RESUMO

The aryl hydrocarbon receptor (AhR) is a heterodimeric transcriptional regulator with pleiotropic functions in xenobiotic metabolism and detoxification, vascular development and cancer. Herein, we report a previously undescribed role for the AhR signalling pathway in the pathogenesis of the wet, neovascular subtype of age-related macular degeneration (AMD), the leading cause of vision loss in the elderly in the Western world. Comparative analysis of gene expression profiles of aged AhR(-/-) and wild-type (wt) mice, using high-throughput RNA sequencing, revealed differential modulation of genes belonging to several AMD-related pathogenic pathways, including inflammation, angiogenesis and extracellular matrix regulation. To investigate AhR regulation of these pathways in wet AMD, we experimentally induced choroidal neovascular lesions in AhR(-/-) mice and found that they measured significantly larger in area and volume compared to age-matched wt mice. Furthermore, these lesions displayed a higher number of ionized calcium-binding adaptor molecule 1-positive (Iba1(+) ) microglial cells and a greater amount of collagen type IV deposition, events also seen in human wet AMD pathology specimens. Consistent with our in vivo observations, AhR knock-down was sufficient to increase choroidal endothelial cell migration and tube formation in vitro. Moreover, AhR knock-down caused an increase in collagen type IV production and secretion in both retinal pigment epithelial (RPE) and choroidal endothelial cell cultures, increased expression of angiogenic and inflammatory molecules, including vascular endothelial growth factor A (VEGFA) and chemokine (C-C motif) ligand 2 (CCL2) in RPE cells, and increased expression of secreted phosphoprotein 1 (SPP1) and transforming growth factor-ß1 (TGFß1) in choroidal endothelial cells. Collectively, our findings identify AhR as a regulator of multiple pathogenic pathways in experimentally induced choroidal neovascularization, findings that are consistent with a possible role of AhR in wet AMD. The data discussed in this paper have been deposited in NCBI's Gene Expression Omnibus; GEO Submission No. GSE56983, NCBI Tracking System No. 17021116.


Assuntos
Neovascularização de Coroide/genética , Regulação da Expressão Gênica/genética , Receptores de Hidrocarboneto Arílico/genética , Epitélio Pigmentado da Retina/metabolismo , Animais , Movimento Celular/genética , Células Cultivadas , Corioide , Neovascularização de Coroide/metabolismo , Humanos , Degeneração Macular/genética , Degeneração Macular/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Hidrocarboneto Arílico/metabolismo , Epitélio Pigmentado da Retina/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Cancer Cell ; 12(4): 381-94, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17936562

RESUMO

Dmp1 (Dmtf1) is activated by oncogenic Ras-Raf signaling and induces cell-cycle arrest in an Arf, p53-dependent fashion. The survival of K-ras(LA) mice was shortened by approximately 15 weeks in both Dmp1(+/-) and Dmp1(-/-) backgrounds, the lung tumors of which showed significantly decreased frequency of p53 mutations compared to Dmp1(+/+). Approximately 40% of K-ras(LA) lung tumors from Dmp1(+/+) mice lost one allele of the Dmp1 gene, suggesting the primary involvement of Dmp1 in K-ras-induced tumorigenesis. Loss of heterozygosity (LOH) of the hDMP1 gene was detectable in approximately 35% of human lung carcinomas, which was found in mutually exclusive fashion with LOH of INK4a/ARF or that of P53. Thus, DMP1 is a pivotal tumor suppressor for both human and murine lung cancers.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animais , Carcinoma Adenoescamoso/genética , Carcinoma Adenoescamoso/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Metilação de DNA , Humanos , Perda de Heterozigosidade , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Regiões Promotoras Genéticas , Fatores de Tempo , Fatores de Transcrição/metabolismo , Transfecção , Proteína Supressora de Tumor p53/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...