Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Lipid Res ; 58(4): 636-648, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28188148

RESUMEN

The pathophysiology of human keratoconus (KC), a bilateral progressive corneal disease leading to protrusion of the cornea, stromal thinning, and scarring, is not well-understood. In this study, we investigated a novel sphingolipid (SPL) signaling pathway through which KC may be regulated. Using human corneal fibroblasts (HCFs) and human KC cells (HKCs), we examined the SPL pathway modulation. Both cell types were stimulated by the three transforming growth factor (TGF)-ß isoforms: TGF-ß1 (T1), TGF-ß2 (T2), and TGF-ß3 (T3). All samples were analyzed using lipidomics and real-time PCR. Our data showed that HKCs have increased levels of signaling SPLs, ceramide (Cer), and sphingosine 1-phosphate (S1P). Treatment with T1 reversed the increase in Cer in HKCs and treatment with T3 reversed the increase in S1P. S1P3 receptor mRNA levels were also significantly upregulated in HKCs, but were reduced to normal levels following T3 treatment. Furthermore, stimulation with Cer and S1P led to significant upregulation of fibrotic markers in HCFs, but not in HKCs. Additionally, stimulation with a Cer synthesis inhibitor (FTY720) led to significant downregulation of specific fibrotic markers in HKCs (TGF-ß1, collagen type III, and α smooth muscle actin) without an effect on healthy HCFs, suggesting a causative role of Cer and S1P in fibrogenesis. Overall, this study suggests an association of the SPL signaling pathway in KC disease and its relation with the TGF-ß pathway.


Asunto(s)
Ceramidas/genética , Queratocono/genética , Lisofosfolípidos/biosíntesis , Esfingolípidos/genética , Esfingosina/análogos & derivados , Línea Celular , Ceramidas/administración & dosificación , Córnea/metabolismo , Córnea/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/fisiología , Clorhidrato de Fingolimod/administración & dosificación , Humanos , Queratocono/patología , Lisofosfolípidos/administración & dosificación , ARN Mensajero/genética , Transducción de Señal , Esfingolípidos/aislamiento & purificación , Esfingolípidos/metabolismo , Esfingosina/administración & dosificación , Esfingosina/biosíntesis , Factor de Crecimiento Transformador beta1/administración & dosificación , Factor de Crecimiento Transformador beta2/administración & dosificación , Factor de Crecimiento Transformador beta3/administración & dosificación
2.
PLoS One ; 11(12): e0168845, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28005998

RESUMEN

PURPOSE: To establish an in vitro model that would mirror the in vivo corneal stromal environment in diabetes (DM) patients. METHODS: Human corneal fibroblasts from Healthy (HCFs), Type 1DM (T1DM) and Type 2DM (T2DM) donors were isolated and cultured for 4 weeks with Vitamin C stimulation in order to allow for extracellular matrix (ECM) secretion and assembly. RESULTS: Our data indicated altered cellular morphology, increased cellular migration, increased ECM assembly, and severe mitochondrial damage in both T1DM and T2DMs when compared to HCFs. Furthermore, we found significant downregulation of Collagen I and Collagen V expression in both T1DM and T2DMs. Furthermore, a significant up regulation of fibrotic markers was seen, including α-smooth muscle actin in T2DM and Collagen III in both T1DM and T2DMs. Metabolic analysis suggested impaired Glycolysis and Tricarboxylic acid cycle (TCA) pathway. CONCLUSION: DM has significant effects on physiological and clinical aspects of the human cornea. The benefits in developing and fully characterizing our 3D in vitro model are enormous and might provide clues for the development of novel therapeutics.


Asunto(s)
Enfermedades de la Córnea/patología , Sustancia Propia/patología , Complicaciones de la Diabetes/patología , Técnicas de Cultivo de Célula , Movimiento Celular , Proliferación Celular , Fibroblastos , Humanos , Técnicas In Vitro , Metabolómica , Microscopía Electrónica de Transmisión , Mitocondrias/ultraestructura
3.
Exp Eye Res ; 153: 90-100, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27742548

RESUMEN

Prolonged hyperglycemia during diabetes mellitus can cause severe ophthalmic complications affecting both the anterior and posterior ocular segments leading to impaired vision or blindness. Diabetes-induced corneal pathologies are associated with decreased wound healing capacity, corneal edema, and altered epithelial basement membrane. The mechanism by which diabetes modulates structure and function within the corneal stroma are unknown. In our study, we characterized the effects of diabetes on extracellular matrix, lipid transport, and cellular metabolism by defining the entire metabolome and lipidome of Type 1 and Type 2 human diabetic corneal stroma. Significant increases in Collagen I and III were found in diabetic corneas suggesting that diabetes promotes defects in matrix structure leading to scarring. Furthermore, increased lipid content, including sphingosine-1-phosphate and dihydrosphingosine, in diabetic corneas compared to healthy controls were measured suggesting altered lipid retention. Metabolomics analysis identified elevated tryptophan metabolites, independent of glucose metabolism, which correlated with upregulation of the Kynurenine pathway in diabetic corneas. We also found significant upregulation of novel biomarkers aminoadipic acid, D,L-pipecolic acid, and dihydroorotate. Our study links aberrant tryptophan metabolism to end-stage pathologies associated with diabetes indicating the potential of the Kynurenine pathway as a therapeutic target for inhibiting diabetes-associated defects in the eye.


Asunto(s)
Biomarcadores/metabolismo , Enfermedades de la Córnea/metabolismo , Sustancia Propia/metabolismo , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Lípidos/análisis , Metaboloma/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Colágeno Tipo III/metabolismo , Enfermedades de la Córnea/diagnóstico , Enfermedades de la Córnea/etiología , Sustancia Propia/patología , Femenino , Humanos , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Esfingosina/metabolismo
4.
Curr Eye Res ; 41(7): 900-7, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26430764

RESUMEN

PURPOSE: Keratoconus (KC) is a complex corneal dystrophy with multifactorial etiology. Previous studies have shown evidence of mitochondrial abnormalities in KC; however, the exact cause of these abnormalities remains unknown. The aim of this study was to identify if transforming growth factor-ß (TGF-ß) isoforms play a role in the regulation of mitochondrial proteins in human KC cells (HKC). MATERIALS AND METHODS: Human corneal fibroblasts (HCF) and HKC were isolated and cultured for 4 weeks in three different conditions: (a) CONTROL: MEM + 10%FBS, (b) MEM + 10%FBS + TGF-ß1 and (c) MEM + 10%FBS + TGF-ß3. All samples were processed for mitochondrial damage analysis using real-time PCR. RESULTS: We quantified and analyzed 84 mitochondrial and five housekeeping genes in HCFs and HKCs. Our data showed that when TGF-ß1 and/or TGF-ß3 were compared with control in HCFs, nine genes were significantly different; however, no genes were significantly regulated by the TGF-ß isoforms in HKCs. Significant differences were also seen in seven genes when HFCs were compared with HKCs, in all three conditions. CONCLUSIONS: Overall, our data support the growing consensus that mitochondrial dysfunction is a key player in KC disease. These in vitro data show clear links between mitochondrial function and TGF-ß isoforms, with TGF-ß1 severely disrupting KC-mitochondrial function, while TGF-ß3 maintained it, thus suggesting that TGF-ß may play a role in KC-disease treatment.


Asunto(s)
Queratocono/metabolismo , Mitocondrias/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células Cultivadas , Córnea/metabolismo , Córnea/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Queratocono/genética , Queratocono/patología , Ligandos , Mitocondrias/genética , Reacción en Cadena de la Polimerasa , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Cell Biochem Funct ; 33(5): 341-50, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26173740

RESUMEN

Corneal scarring is the result of a disease, infection or injury. The resulting scars cause significant loss of vision or even blindness. To-date, the most successful treatment is corneal transplantation, but it does not come without side effects. One of the corneal dystrophies that are correlated with corneal scarring is keratoconus (KC). The onset of the disease is still unknown; however, altered cellular metabolism has been linked to promoting the fibrotic phenotype and therefore scarring. We have previously shown that human keratoconus cells (HKCs) have altered metabolic activity when compared to normal human corneal fibroblasts (HCFs). In our current study, we present evidence that quercetin, a natural flavonoid, is a strong candidate for regulating metabolic activity of both HCFs and HKCs in vitro and therefore a potential therapeutic to target the altered cellular metabolism characteristic of HKCs. Targeted mass spectrometry-based metabolomics was performed on HCFs and HKCs with and without quercetin treatment in order to identify variations in metabolite flux. Overall, our study reveals a novel therapeutic target OF Quercetin on corneal stromal cell metabolism in both healthy and diseased states. Clearly, further studies are necessary in order to dissect the mechanism of action of quercetin.


Asunto(s)
Antioxidantes/farmacología , Queratocono/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Quercetina/farmacología , Diferenciación Celular/efectos de los fármacos , Córnea/citología , Metabolismo Energético/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Cultivo Primario de Células
6.
Mol Vis ; 21: 577-88, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26015770

RESUMEN

PURPOSE: Keratoconus (KC) is a corneal thinning disease of unknown etiology whose pathophysiology is correlated with the presence of a thin corneal stroma and altered extracellular matrix (ECM). Transforming growth factor-ß (TGF-ß) signaling is a key regulator of ECM secretion and assembly in multiple tissues, including the anterior segment of the eye, and it has been linked to KC. We have previously shown that human keratoconus cells (HKCs) have a myofibroblast phenotype and altered ECM assembly compared to normal human corneal fibroblasts (HCFs). Moreover, TGF-ß3 treatment promotes assembly of a more normal stromal ECM and modulates the fibrotic phenotype in HKCs. Herein, we identify alterations in TGF-ß signaling that contribute to the observed fibrotic phenotype in HKCs. METHODS: HCFs and HKCs were stimulated with TGF-ß1, TGF-ß2, or TGF-ß3 isoforms (0.1 ng/mL) in the presence of a stable vitamin C derivative (0.5 mM) for 4 weeks. All samples were examined using RT-PCR and western blotting to quantify changes in the expressions of key TGF-ß signaling molecules between HCFs and HKCs. RESULTS: We found a significant downregulation in the SMAD6 and SMAD7 expressions by HKCs when compared to HCFs (p≤0.05). Moreover, stimulation of HKCs with any of the three TGF-ß isoforms did not significantly alter the expressions of SMAD6 or SMAD7. HCFs also showed an upregulation in TGF-ßRI, TGF-ßRII, and TGF-ßRIII following TGF-ß3 treatment, whereas HKCs showed a significant two-fold downregulation. CONCLUSIONS: Overall, our data shows the decreased expressions of the regulatory SMADs SMAD6 and SMAD7 by HKCs contribute to the pathological ECM structure observed in KC, and TGF-ß3 may attenuate this mechanism by downregulating the expression of the key profibrotic receptor, TGF-ßRII. Our study suggests a significant role of altered regulation of TGF-ß signaling in KC progression and that it may enable novel therapeutic developments targeting TGF-ß receptor regulation.


Asunto(s)
Queratocono/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Córnea/metabolismo , Regulación hacia Abajo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Queratocono/genética , Miofibroblastos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteoglicanos/genética , Proteoglicanos/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteína smad6/genética , Proteína smad6/metabolismo , Proteína smad7/genética , Proteína smad7/metabolismo
7.
Curr Eye Res ; 40(12): 1204-10, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25426847

RESUMEN

PURPOSE: Diabetes mellitus (DM) is characterized by high blood sugar levels over a prolonged period. Long term complications include but not limited heart disease, stroke, kidney failure, and ocular damage. An estimated 382 million people are diagnosed with Type 2 DM accounting for 90% of the cases. Common corneal dysfunctions associated with DM result in impaired vision due to decreased wound healing, corneal edema, and altered epithelial basement membrane. Lipids play a fundamental role in tissue metabolism and disease states. We attempt to determine the role of sphingolipids (SPL) in human Type I and Type II diabetic corneas. MATERIALS AND METHODS: Cadaver corneas from healthy (non-diabetic/no ocular trauma), Type I (T1DM), and Type II diabetic (T2DM) donors were obtained and processed for lipidomics using LC-MS/MS. RESULTS: Our data show significant differences in the SPL composition between control, T1DM and T2DM corneas. Both T1DM and T2DM showed a 10-folddownregulation of sphingomyelin(SM), 5-fold up regulation of Ceramides (Cer) and 2-fold upregulation of monohexosylceramides (MHC). Differences were also seen in total amounts of SPL where Cer was increased by approximately 3 fold in both T1DM and T2DM where SM decreased by 50% in both T1DM and T2DM when compared to healthy controls. No differences were seen in MHC amounts. CONCLUSIONS: Overall, our data indicate major differences in SPL distribution in human diabetic corneas. Information on the sphingolipids role in cornea, corneal cell physiology, and diseases are very limitedwhich highlights the importance of these findings.


Asunto(s)
Enfermedades de la Córnea/metabolismo , Complicaciones de la Diabetes/metabolismo , Esfingolípidos/metabolismo , Ceramidas/metabolismo , Cerebrósidos/metabolismo , Cromatografía Liquida , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Regulación hacia Abajo , Humanos , Esfingomielinas/metabolismo , Espectrometría de Masas en Tándem , Donantes de Tejidos , Regulación hacia Arriba
8.
PLoS One ; 9(11): e113310, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25405607

RESUMEN

Keratoconus (KC) is a bilateral degenerative disease of the cornea characterized by corneal bulging, stromal thinning, and scarring. The etiology of the disease is unknown. In this study, we identified a new biomarker for KC that is present in vivo and in vitro. In vivo, tear samples were collected from age-matched controls with no eye disease (n = 36) and KC diagnosed subjects (n = 17). Samples were processed for proteomics using LC-MS/MS. In vitro, cells were isolated from controls (Human Corneal Fibroblasts-HCF) and KC subjects (Human Keratoconus Cells-HKC) and stimulated with a Vitamin C (VitC) derivative for 4 weeks, and with one of the three transforming growth factor-beta (TGF-ß) isoforms. Samples were analyzed using real-time PCR and Western Blots. By using proteomics analysis, the Gross cystic disease fluid protein-15 (GCDFP-15) or prolactin-inducible protein (PIP) was found to be the best independent biomarker able to discriminate between KC and controls. The intensity of GCDFP-15/PIP was significantly higher in healthy subjects compared to KC-diagnosed. Similar findings were seen in vitro, using a 3D culture model. All three TGF-ß isoforms significantly down-regulated the expression of GCDFP-15/PIP. Zinc-alpha-2-glycoprotein (AZGP1), a protein that binds to PIP, was identified by proteomics and cell culture to be highly regulated. In this study by different complementary techniques we confirmed the potential role of GCDFP-15/PIP as a novel biomarker for KC disease. It is likely that exploring the GCDFP-15/PIP-AZGP1 interactions will help better understand the mechanism of KC disease.


Asunto(s)
Biomarcadores/metabolismo , Proteínas Portadoras/metabolismo , Glicoproteínas/metabolismo , Queratocono/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Adipoquinas , Adulto , Ácido Ascórbico/farmacología , Western Blotting , Proteínas Portadoras/genética , Células Cultivadas , Cromatografía Liquida , Córnea/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Humanos , Queratocono/diagnóstico , Queratocono/genética , Proteínas de Transporte de Membrana , Proteoma/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Masas en Tándem , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta2/farmacología , Factor de Crecimiento Transformador beta3/farmacología , Vitaminas/farmacología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...