Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
Diabetes ; 70(7): 1549-1560, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33883214

RESUMEN

Diabetic retinopathy is one of the leading causes of vision loss and blindness. Extensive preclinical and clinical evidence exists for both vascular and neuronal pathology. However, the relationship of these changes in the neurovascular unit and impact on vision remains to be determined. Here, we investigate the role of tight junction protein occludin phosphorylation at S490 in modulating barrier properties and its impact on visual function. Conditional vascular expression of the phosphorylation-resistant Ser490 to Ala (S490A) form of occludin preserved tight junction organization and reduced vascular endothelial growth factor (VEGF)-induced permeability and edema formation after intraocular injection. In the retinas of streptozotocin-induced diabetic mice, endothelial-specific expression of the S490A form of occludin completely prevented diabetes-induced permeability to labeled dextran and inhibited leukostasis. Importantly, vascular-specific expression of the occludin mutant completely blocked the diabetes-induced decrease in visual acuity and contrast sensitivity. Together, these results reveal that occludin acts to regulate barrier properties downstream of VEGF in a phosphorylation-dependent manner and that loss of inner blood-retinal barrier integrity induced by diabetes contributes to vision loss.


Asunto(s)
Barrera Hematorretinal/fisiología , Diabetes Mellitus Experimental/fisiopatología , Retinopatía Diabética/fisiopatología , Ocludina/fisiología , Agudeza Visual , Animales , Leucostasis/prevención & control , Ratones , Ratones Endogámicos C57BL , Permeabilidad , Fosforilación , Estreptozocina , Factor A de Crecimiento Endotelial Vascular/fisiología
2.
J Ophthalmic Vis Res ; 15(4): 446-452, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33133434

RESUMEN

PURPOSE: To evaluate the penetration of carbon nanotubes (CNTs) throughout retinoblastoma in a transgenic mice model. METHODS: CNTs functionalized with fluorescein isothiocyanate and targeting ligands biotin (CTN-FITC-Bio, 0.5mg/ml), or folic acid (CNT-FITC-FA, 0.5mg/ml) were injected into the vitreous of one eye of LH BETA T AG transgenic mice. Other eye did not receive any injection and was used as control. Three mice were sacrificed at days 1, 2, and 3. Eyes were enucleated and stained with 4,6-diamidino-2-phenylindole. The sections were imaged by fluorescent microscope. The images were transformed into grey-scale in MATLAB for intensity analysis. Background intensity was normalized by marking squares outside the eyeball and using the mean intensity of these squares. Fluorescent intensity (FI) for each image was measured by calculating the intensity of a same-sized square within retinoblastoma. RESULTS: Nine eyes of nine mice were included in each CNT-FITC-Bio and CNT-FITC-FA groups. The mean FI in CNT-FITC-Bio was 52.08 ± 6.33, 53.62 ± 9.00, and 65.54 ± 5.14 in days 1, 2, and 3, respectively. The mean FI in CNT-FITC-FA was 50.28 ± 7.37, 59.21 ± 6.43, and 58.38 ± 2.32 on days 1, 2, and 3, respectively. FI was significantly higher in eyes injected with CNT-FITC-Bio and CNT-FITC-FA compared to the control eyes (P = 0.02). There was no difference in FI between eyes with CNT-FITC-Bio and CNT-FITC-FA, and FI remained stable on days 1-3 in CNT-FITC-Bio, CNT-FITC-FA, and control eyes (P > 0.05). CONCLUSION: We observed higher FI in eyes with CNT-FITC-Bio and CNT-FITC-FA compared to control eyes, showing penetration of CNTs throughout retinoblastoma. CNTs can be a carrier candidate for imaging or therapeutic purposes in retinoblastoma.

3.
Bioorg Med Chem ; 28(11): 115480, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32327351

RESUMEN

Studies demonstrate that small molecule targeting of atypical protein kinase C (aPKC) may provide an effective means to control vascular permeability, prevent edema, and reduce inflammation providing novel and important alternatives to anti-VEGF therapies for certain blinding eye diseases. Based on a literature tricyclic thieno[2,3-d]pyrimidine lead (1), an ATP-competitive inhibitor of the aPKC iota (ι) and aPKC zeta (ζ) isoforms, we have synthesized a small series of compounds in 1-2 steps from a readily available chloro intermediate. A single pyridine congener was also made using 2D NMR to assign regiochemistry. Within the parent pyrimidine series, a range of potencies was observed against aPKCζ whereas the pyridine congener was inactive. Selected compounds were also tested for their effect toward VEGF-induced permeability in BREC cells. The most potent of these (7l) was further assayed against the aPKCι isoform and showed a favorable selectivity profile against a panel of 31 kinases, including kinases from the AGC superfamily, with a focus on PKC isoforms and kinases previously shown to affect permeability. Further testing of 7l in a luciferase assay in HEK293 cells showed an ability to prevent TNF-α induced NFκB activation while not having any effect on cell survival. Intravitreal administration of 7l to the eye yielded a complete reduction in permeability in a test to determine whether the compound could block VEGF- and TNFα-induced permeability across the retinal vasculature in a rat model. The compound in mice displayed good microsomal stability and in plasma moderate exposure (AUC and Cmax), low clearance, a long half-life and high oral bioavailability. With IV dosing, higher levels were observed in the brain and eye relative to plasma, with highest levels in the eye by either IV or PO dosing. With a slow oral absorption profile, 7l accumulates in the eye to maintain a high concentration after dosing with higher levels than in plasma. Compound 7l may represent a class of aPKC inhibitors for further investigation.


Asunto(s)
Citocinas/antagonistas & inhibidores , Edema/tratamiento farmacológico , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Edema/inducido químicamente , Edema/metabolismo , Femenino , Células HEK293 , Humanos , Ratones , Estructura Molecular , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Pirimidinas/síntesis química , Pirimidinas/química , Ratas , Ratas Long-Evans , Relación Estructura-Actividad , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
J Biol Chem ; 295(14): 4647-4660, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32086377

RESUMEN

Vascular endothelial growth factor (VEGF) contributes to blood-retinal barrier (BRB) dysfunction in several blinding eye diseases, including diabetic retinopathy. Signaling via the secreted protein norrin through the frizzled class receptor 4 (FZD4)/LDL receptor-related protein 5-6 (LRP5-6)/tetraspanin 12 (TSPAN12) receptor complex is required for developmental vascularization and BRB formation. Here, we tested the hypothesis that norrin restores BRB properties after VEGF-induced vascular permeability in diabetic rats or in animals intravitreally injected with cytokines. Intravitreal co-injection of norrin with VEGF completely ablated VEGF-induced BRB permeability to Evans Blue-albumin. Likewise, 5-month diabetic rats exhibited increased permeability of FITC-albumin, and a single norrin injection restored BRB properties. These results were corroborated in vitro, where co-stimulation of norrin with VEGF or stimulation of norrin after VEGF exposure restored barrier properties, indicated by electrical resistance or 70-kDa RITC-dextran permeability in primary endothelial cell culture. Interestingly, VEGF promoted norrin signaling by increasing the FZD4 co-receptor TSPAN12 at cell membranes in an MAPK/ERK kinase (MEK)/ERK-dependent manner. Norrin signaling through ß-catenin was required for BRB restoration, but glycogen synthase kinase 3 α/ß (GSK-3α/ß) inhibition did not restore BRB properties. Moreover, levels of the tight junction protein claudin-5 were increased with norrin and VEGF or with VEGF alone, but both norrin and VEGF were required for enriched claudin-5 localization at the tight junction. These results suggest that VEGF simultaneously induces vascular permeability and promotes responsiveness to norrin. Norrin, in turn, restores tight junction complex organization and BRB properties in a ß-catenin-dependent manner.


Asunto(s)
Barrera Hematorretinal/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Proteínas del Ojo/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Barrera Hematorretinal/efectos de los fármacos , Bovinos , Claudina-5/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/patología , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Ratas , Ratas Long-Evans , Retina/metabolismo , Vasos Retinianos/citología , Vasos Retinianos/metabolismo , Transducción de Señal/efectos de los fármacos , Tetraspaninas/genética , Tetraspaninas/metabolismo , Regulación hacia Arriba/efectos de los fármacos , beta Catenina/antagonistas & inhibidores , beta Catenina/metabolismo
5.
J Immunol ; 202(4): 1057-1068, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30635390

RESUMEN

Autoimmune retinopathy (AIR) is a treatable condition that manifests in acute and progressive vision loss in patients. It has recently been determined that AIR is associated with an imbalance of TH1 versus regulatory T cell immunity toward the retinal protein, recoverin. This study describes a new murine model to understand the immunopathology of AIR and its association with T cell responses toward recoverin. Immunization of C57BL/6 mice with recoverin resulted in ocular inflammation including infiltration of CD4+ and CD8+ T lymphocytes, B cells, and CD11b+Ly6C+ inflammatory monocytes in the eyes. Production of IFN-γ and IL-17 from T cells was exacerbated in IL-10 knockout (KO) mice and kinetics of disease development was accelerated. Infiltration of T cells and inflammatory monocytes into the eyes dramatically increased in recoverin-immunized IL-10 KO mice. An immunodominant peptide of recoverin, AG-16, was capable of inducing disease in IL-10 KO mice and resulted in expansion of AG-16 tetramer-specific CD4+ T cells in lymphoid organs and eyes. Adoptive transfer of recoverin-stimulated cells into naive mice was sufficient to induce AIR, and immunization of B cell-deficient mice led to a milder form of the disease. This model supports the hypothesis that recoverin-specific T cell responses are major drivers of AIR pathogenesis and that IL-10 is an important factor in protection.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Ojo/inmunología , Interleucina-10/inmunología , Recoverina/inmunología , Enfermedades de la Retina/inmunología , Animales , Ojo/patología , Inflamación/inmunología , Interleucina-10/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Células TH1/inmunología , Células Th17/inmunología
6.
Am J Pathol ; 188(10): 2392-2405, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30220554

RESUMEN

Changes in permeability of retinal blood vessels contribute to macular edema and the pathophysiology of numerous ocular diseases, including diabetic retinopathy, retinal vein occlusions, and macular degeneration. Vascular endothelial growth factor (VEGF) induces retinal permeability and macular thickening in these diseases. However, inflammatory agents, such as tumor necrosis factor-α (TNF-α), also may drive vascular permeability, specifically in patients unresponsive to anti-VEGF therapy. Recent evidence suggests VEGF and TNF-α induce permeability through distinct mechanisms; however, both require the activation of atypical protein kinase C (aPKC). We provide evidence, using genetic mouse models and therapeutic intervention with small molecules, that inhibition of aPKC prevented or reduced vascular permeability in animal models of retinal inflammation. Expression of a kinase-dead aPKC transgene, driven by a vascular and hematopoietic restricted promoter, reduced retinal vascular permeability in an ischemia-reperfusion model of retinal injury. This effect was recapitulated with a small-molecule inhibitor of aPKC. Expression of the kinase-dead aPKC transgene dramatically reduced the expression of inflammatory factors and blocked the attraction of inflammatory monocytes and granulocytes after ischemic injury. Coinjection of VEGF with TNF-α was sufficient to induce permeability, edema, and retinal inflammation, and treatment with an aPKC inhibitor prevented VEGF/TNF-α-induced permeability. These data suggest that aPKC contributes to inflammation-driven retinal vascular pathology and may be an attractive target for therapeutic intervention.


Asunto(s)
Permeabilidad Capilar/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Vasos Retinianos/fisiología , Animales , Permeabilidad Capilar/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Papiledema/inducido químicamente , Papiledema/fisiopatología , Ratas Long-Evans , Proteínas Recombinantes , Daño por Reperfusión/fisiopatología , Retinitis/inducido químicamente , Retinitis/fisiopatología , Uniones Estrechas/química , Uniones Estrechas/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
7.
Invest Ophthalmol Vis Sci ; 57(6): 2584-92, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27163772

RESUMEN

PURPOSE: Inflammation associated with blood-retinal barrier (BRB) breakdown is a common feature of several retinal diseases. Therefore, the development of novel nonsteroidal anti-inflammatory approaches may provide important therapeutic options. Previous studies demonstrated that inhibition of dipeptidyl peptidase-IV, the enzyme responsible for the degradation of glucagon-like peptide-1 (GLP-1), led to insulin-independent prevention of diabetes-induced increases in BRB permeability, suggesting that incretin-based drugs may have beneficial pleiotropic effects in the retina. In the current study, the barrier protective and anti-inflammatory properties of exendin-4 (Ex-4), an analog of GLP-1, after ischemia-reperfusion (IR) injury were examined. METHODS: Ischemia-reperfusion injury was induced in rat retinas by increasing the intraocular pressure for 45 minutes followed by 48 hours of reperfusion. Rats were treated with Ex-4 prior to and following IR. Blood-retinal barrier permeability was assessed by Evans blue dye leakage. Retinal inflammatory gene expression and leukocytic infiltration were measured by qRT-PCR and immunofluorescence, respectively. A microglial cell line was used to determine the effects of Ex-4 on lipopolysaccharide (LPS)-induced inflammatory response. RESULTS: Exendin-4 dramatically reduced the BRB permeability induced by IR injury, which was associated with suppression of inflammatory gene expression. Moreover, in vitro studies showed that Ex-4 also reduced the inflammatory response to LPS and inhibited NF-κB activation. CONCLUSIONS: The present work suggests that Ex-4 can prevent IR injury-induced BRB breakdown and inflammation through inhibition of inflammatory cytokine production by activated microglia and may provide a novel option for therapeutic intervention in diseases involving retinal inflammation.


Asunto(s)
Barrera Hematorretinal/efectos de los fármacos , Péptido 1 Similar al Glucagón/análogos & derivados , Inflamación/prevención & control , Isquemia/prevención & control , Péptidos/farmacología , Daño por Reperfusión/complicaciones , Enfermedades de la Retina/prevención & control , Ponzoñas/farmacología , Animales , Bovinos , Células Cultivadas , Modelos Animales de Enfermedad , Exenatida , Immunoblotting , Inmunohistoquímica , Incretinas/farmacología , Inflamación/metabolismo , Isquemia/etiología , Isquemia/metabolismo , Masculino , Ratas , Ratas Long-Evans , Daño por Reperfusión/metabolismo , Enfermedades de la Retina/etiología , Enfermedades de la Retina/metabolismo
8.
J Neuroinflammation ; 10: 149, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24325836

RESUMEN

BACKGROUND: Many retinal diseases are associated with vascular dysfunction accompanied by neuroinflammation. We examined the ability of minocycline (Mino), a tetracycline derivative with anti-inflammatory and neuroprotective properties, to prevent vascular permeability and inflammation following retinal ischemia-reperfusion (IR) injury, a model of retinal neurodegeneration with breakdown of the blood-retinal barrier (BRB). METHODS: Male Sprague-Dawley rats were subjected to 45 min of pressure-induced retinal ischemia, with the contralateral eye serving as control. Rats were treated with Mino prior to and following IR. At 48 h after reperfusion, retinal gene expression, cellular inflammation, Evan's blue dye leakage, tight junction protein organization, caspase-3 activation, and DNA fragmentation were measured. Cellular inflammation was quantified by flow-cytometric evaluation of retinal tissue using the myeloid marker CD11b and leukocyte common antigen CD45 to differentiate and quantify CD11b+/CD45low microglia, CD11b+/CD45hi myeloid leukocytes and CD11bneg/CD45hi lymphocytes. Major histocompatibility complex class II (MHCII) immunoreactivity was used to determine the inflammatory state of these cells. RESULTS: Mino treatment significantly inhibited IR-induced retinal vascular permeability and disruption of tight junction organization. Retinal IR injury significantly altered mRNA expression for 21 of 25 inflammation- and gliosis-related genes examined. Of these, Mino treatment effectively attenuated IR-induced expression of lipocalin 2 (LCN2), serpin peptidase inhibitor clade A member 3 N (SERPINA3N), TNF receptor superfamily member 12A (TNFRSF12A), monocyte chemoattractant-1 (MCP-1, CCL2) and intercellular adhesion molecule-1 (ICAM-1). A marked increase in leukostasis of both myeloid leukocytes and lymphocytes was observed following IR. Mino treatment significantly reduced retinal leukocyte numbers following IR and was particularly effective in decreasing the appearance of MHCII+ inflammatory leukocytes. Surprisingly, Mino did not significantly inhibit retinal cell death in this model. CONCLUSIONS: IR induces a retinal neuroinflammation within hours of reperfusion characterized by inflammatory gene expression, leukocyte adhesion and invasion, and vascular permeability. Despite Mino significantly inhibiting these responses, it failed to block neurodegeneration.


Asunto(s)
Barrera Hematorretinal/efectos de los fármacos , Inflamación/patología , Minociclina/farmacología , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/patología , Retina/patología , Animales , Barrera Hematorretinal/patología , Permeabilidad Capilar/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Expresión Génica/efectos de los fármacos , Masculino , Degeneración Nerviosa/patología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/efectos de los fármacos
9.
Invest Ophthalmol Vis Sci ; 54(8): 5550-8, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23882687

RESUMEN

PURPOSE: To determine the utility of polychromatic angiography (PCA) in the assessment of VEGF-induced blood retinal barrier (BRB) dysfunction in rabbits. METHODS: Twenty-six eyes of 24 Dutch Belted rabbits were injected intravitreally with 1.25 µg (group A, n = 5), 10 µg (group C, n = 7), or 4 µg (group B, n = 6; group D, n = 4; and group E, n = 4) of VEGF on day 0. Groups D and E were also injected intravitreally with 1.25 µg and 12.5 µg bevacizumab, respectively, on day 2. On days 0, 2, 4, 7, 11, and 14, PCA was performed using a contrast agent mixture composed of fluorescein sodium, indocyanine green, PCM102, and PCM107 and imaged with a modified fundus camera. PCA scores were based on detected leaking fluorophores. RESULTS: On day 7, there was a statistically significant difference between PCA scores of group A (0.6 ± 0.89) and both groups B (2.67 ± 1.37, P = 0.0154) and C (3.33 ± 0.52, P = 0.00085). There was also a statistically significant difference between groups B and E (PCA score 0.75 ± 0.96, P = 0.032) on day 7. On day 11, there was statistically significant difference between group C (1.80 ± 1.1) and both groups A (0, P = 0.021) and B (0.33 ± 0.52, P = 0.037). CONCLUSIONS: A differential response to both increasing VEGF dose and administration of bevacizumab could be discerned using the PCA. PCA allowed stratification of VEGF-induced BRB dysfunction and inhibitory effects of bevacizumab therapy in the rabbit retina.


Asunto(s)
Barrera Hematorretinal/efectos de los fármacos , Angiografía con Fluoresceína/métodos , Retina/efectos de los fármacos , Enfermedades de la Retina/diagnóstico , Factor A de Crecimiento Endotelial Vascular/toxicidad , Animales , Modelos Animales de Enfermedad , Fondo de Ojo , Inyecciones Intravítreas , Masculino , Conejos , Retina/patología , Enfermedades de la Retina/inducido químicamente , Enfermedades de la Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/administración & dosificación
10.
Biochem J ; 446(3): 455-67, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22721706

RESUMEN

Pro-inflammatory cytokines and growth factors such as VEGF (vascular endothelial growth factor) contribute to the loss of the BRB (blood-retinal barrier) and subsequent macular oedema in various retinal pathologies. VEGF signalling requires PKCß [conventional PKC (protein kinase C)] activity; however, PKCß inhibition only partially prevents VEGF-induced endothelial permeability and does not affect pro-inflammatory cytokine-induced permeability, suggesting the involvement of alternative signalling pathways. In the present study, we provide evidence for the involvement of aPKC (atypical PKC) signalling in VEGF-induced endothelial permeability and identify a novel class of inhibitors of aPKC that prevent BRB breakdown in vivo. Genetic and pharmacological manipulations of aPKC isoforms were used to assess their contribution to endothelial permeability in culture. A chemical library was screened using an in vitro kinase assay to identify novel small-molecule inhibitors, and further medicinal chemistry was performed to delineate a novel pharmacophore. We demonstrate that aPKC isoforms are both sufficient and required for VEGF-induced endothelial permeability. Furthermore, these specific, potent, non-competitive, small-molecule inhibitors prevented VEGF-induced tight junction internalization and retinal endothelial permeability in response to VEGF in both primary culture and in rodent retina. The results of the present study suggest that aPKC inhibition with 2-amino-4-phenyl-thiophene derivatives may be developed to preserve the BRB in retinal diseases such as diabetic retinopathy or uveitis, and the BBB (blood-brain barrier) in the presence of brain tumours.


Asunto(s)
Barrera Hematorretinal/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Bovinos , Retinopatía Diabética/metabolismo , Humanos , Masculino , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Proteína Quinasa C/metabolismo , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Retina/citología , Retina/metabolismo , Vasos Retinianos/citología , Vasos Retinianos/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética
11.
Diabetes ; 59(11): 2872-82, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20693346

RESUMEN

OBJECTIVE: Tumor necrosis factor-α (TNF-α) and interleukin-1 beta (IL-1ß) are elevated in the vitreous of diabetic patients and in retinas of diabetic rats associated with increased retinal vascular permeability. However, the molecular mechanisms underlying retinal vascular permeability induced by these cytokines are poorly understood. In this study, the effects of IL-1ß and TNF-α on retinal endothelial cell permeability were compared and the molecular mechanisms by which TNF-α increases cell permeability were elucidated. RESEARCH DESIGN AND METHODS: Cytokine-induced retinal vascular permeability was measured in bovine retinal endothelial cells (BRECs) and rat retinas. Western blotting, quantitative real-time PCR, and immunocytochemistry were performed to determine tight junction protein expression and localization. RESULTS: IL-1ß and TNF-α increased BREC permeability, and TNF-α was more potent. TNF-α decreased the protein and mRNA content of the tight junction proteins ZO-1 and claudin-5 and altered the cellular localization of these tight junction proteins. Dexamethasone prevented TNF-α-induced cell permeability through glucocorticoid receptor transactivation and nuclear factor-kappaB (NF-κB) transrepression. Preventing NF-κB activation with an inhibitor κB kinase (IKK) chemical inhibitor or adenoviral overexpression of inhibitor κB alpha (IκBα) reduced TNF-α-stimulated permeability. Finally, inhibiting protein kinase C zeta (PKCζ) using both a peptide and a novel chemical inhibitor reduced NF-κB activation and completely prevented the alterations in the tight junction complex and cell permeability induced by TNF-α in cell culture and rat retinas. CONCLUSIONS: These results suggest that PKCζ may provide a specific therapeutic target for the prevention of vascular permeability in retinal diseases characterized by elevated TNF-α, including diabetic retinopathy.


Asunto(s)
Células Endoteliales/fisiología , FN-kappa B/metabolismo , Proteína Quinasa C/metabolismo , Retina/fisiología , Uniones Estrechas/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Bovinos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/fisiología , Dexametasona/farmacología , Retinopatía Diabética/tratamiento farmacológico , Humanos , Interleucina-1beta/farmacología , Masculino , FN-kappa B/uso terapéutico , Permeabilidad , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/uso terapéutico , Ratas , Ratas Sprague-Dawley , Enfermedades de la Retina/tratamiento farmacológico , Uniones Estrechas/efectos de los fármacos
12.
Invest Ophthalmol Vis Sci ; 51(11): 5920-33, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20554620

RESUMEN

PURPOSE: Using transient ischemia followed by reperfusion (IR) to model ischemic retinal disease, this study compares the effects of ischemic preconditioning (IPC) and therapies targeting vascular endothelial growth factor (VEGF) and tumor necrosis factor (TNF)-α on retinal apoptosis, vascular permeability, and mRNA expression. METHODS: Rats were subjected to 30 or 45 minutes of retinal ischemia followed by reperfusion for up to 48 hours. Neurodegeneration was quantified by caspase-3 (DEVDase) activity and by measuring nucleosomal DNA content (cell death ELISA). Vascular leakage was quantified by the Evans Blue dye method. A set of IR-responsive mRNAs was identified by whole-genome microarray and confirmed by RT-PCR analyses. VEGF protein was measured by Western blot analysis. IPC was accomplished with 10 minutes of ischemia 24 hours before IR. VEGF and TNFα signaling was inhibited by intravitreal injection of bevacizumab or etanercept, respectively. RESULTS: IR caused significant retinal cell apoptosis and vascular permeability after 4 and 48 hours. Whereas IR decreased VegfA mRNA, VEGF protein was significantly increased. IPC effectively inhibited neurodegeneration, bevacizumab effectively inhibited vascular permeability, and etanercept failed to affect either outcome. IPC significantly altered the IR responses of 15 of 33 IR-responsive mRNAs, whereas bevacizumab had no significant effect on these mRNAs. CONCLUSIONS: IR provides an acute model of ischemic retinopathy that includes neurodegeneration and VEGF-dependent vascular permeability and is amenable to rapid drug therapy testing. The distinct effects of IPC and bevacizumab demonstrate that the apoptotic and vascular responses to IR may be separated and that therapeutics targeting each pathologic endpoint may be warranted in treating ischemic retinal diseases.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Apoptosis , Permeabilidad Capilar/efectos de los fármacos , Precondicionamiento Isquémico , Daño por Reperfusión/prevención & control , Enfermedades de la Retina/prevención & control , Vasos Retinianos/patología , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Anticuerpos Monoclonales Humanizados , Bevacizumab , Barrera Hematorretinal , Western Blotting , Caspasa 3/metabolismo , Daño del ADN , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Inyecciones Intravítreas , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Vasos Retinianos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
J Gastrointest Surg ; 6(6): 876-82, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12504227

RESUMEN

L-Arginine uptake by the small intestine can play a pivotal role in regulating nitric oxide synthesis and immune functions in catabolic states. We previously showed that protein kinase C (PKC) activation stimulates intestinal brush-border membrane arginine transport. However, the signaling pathways implicated in this activation have not been studied. The purpose of this study was to investigate the intracellular signal transduction pathways involved in the protein kinase C stimulation of arginine transport across the apical membrane of intestinal epithelial Caco-2 cells. [3H]-L-arginine transport activity, Northern blot analysis of mRNA levels of the intestinal arginine transporter CAT1, and Western blot analysis of the mitogen-activated protein (MAP) kinases phospho-p44/42 activity and phospho-MEK1/2 were measured in cultured Caco-2 cells treated with phorbol ester (phorbol 12-myristate 13-acetate, TPA; 0 to 0.5 micromol/L), and the MEK1 inhibitor PD 98059 (0 to 50 micromol/L). Phorbol ester stimulated intestinal arginine transport activity. Arginine transporter gene CAT1 mRNA, phospho-p44/42, and phospho-MEK1/2 levels were stimulated in phorbol ester-treated cells, compared with the control group. Phorbol ester stimulation of arginine transport activity and transporter CAT1 mRNA levels was blocked by PD 98059. These data suggest that phorbol ester stimulates arginine transport in Caco-2 cells via signaling pathways that lead to increased transcription and/or stabilization of CAT1 mRNA. Protein kinase C and MAP kinases MEK1/2 and p44/42 are key intracellular regulators involved in this signal transduction cascade.


Asunto(s)
Arginina/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Quinasa C/farmacología , ARN Mensajero/análisis , Secuencia de Bases , Transporte Biológico/efectos de los fármacos , Northern Blotting , Western Blotting , Células CACO-2/efectos de los fármacos , Células CACO-2/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Valores de Referencia , Sensibilidad y Especificidad
14.
Am J Physiol Endocrinol Metab ; 283(3): E472-81, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12169440

RESUMEN

During sepsis, growth hormone (GH) resistance contributes to the catabolism of muscle protein. To determine the role of tumor necrosis factor (TNF) as a mediator of GH resistance, we examined the effects of a TNF antagonist [TNF-binding protein (TNFbp)] on the GH/insulin-like growth factor (IGF) I system during abdominal sepsis. To investigate potential mechanisms, the effects of TNF on the IGF-I response to GH and GH signaling were examined in cultured rat hepatocytes (CWSV-1). Three groups of rats were studied: Control, Sepsis, and Sepsis + TNFbp. Liver, gastrocnemius, and plasma were collected on day 5. In gastrocnemius, neither sepsis nor TNFbp altered the abundance of IGF-I mRNA. However, septic rats demonstrated an increase in circulating GH and a reduction in plasma IGF-I concentrations that was ameliorated by pretreatment with TNFbp. Liver from septic rats demonstrated a 50% reduction in GH receptor (GHR) and IGF-I mRNA on day 5 that was attenuated by TNFbp. However, the abundance of GHR protein was not different in liver from Control, Sepsis, or Sepsis + TNFbp rats. Consequently, a decreased amount of hepatic GHR does not explain the GH-resistant septic state. In CWSV-1 hepatocytes, TNF-alpha had no effect on GHR protein level but inhibited the induction of IGF-I mRNA by GH. Nuclear protein from TNF-treated hepatocytes demonstrated similar levels of phosphorylated signal transducer and activator of transcription-5 (STAT5) and DNA binding relative to controls 5 min after GH treatment. However, both of these parameters were decreased (vs. control) in TNF-treated cells 60 min after GH treatment. Collectively, these results suggest that TNF mediates hepatic GH resistance during sepsis by inhibiting the duration of signaling via the janus kinase-2/STAT5 pathway.


Asunto(s)
Infecciones Bacterianas/fisiopatología , Hormona del Crecimiento/metabolismo , Hígado/fisiología , Proteínas de la Leche , Proteínas Proto-Oncogénicas , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Infecciones Bacterianas/genética , Línea Celular Transformada , Proteínas de Unión al ADN/metabolismo , Resistencia a Medicamentos/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Janus Quinasa 2 , Masculino , Proteínas Tirosina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Necrosis Tumoral , Receptores Tipo I de Factores de Necrosis Tumoral , Factor de Transcripción STAT5 , Transducción de Señal , Transactivadores/metabolismo , Transcripción Genética , Receptores Señuelo del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/farmacología
15.
J Gastrointest Surg ; 6(3): 379-86, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12022990

RESUMEN

L-Arginine, which is intimately involved in cellular immune functions and nitric oxide biology, is transported by intestinal cells largely via transport System y(+). The gut epithelium is exposed to various luminal amino acids at any given time, and therefore the purpose of this study was to study the regulation of luminal arginine transport by other amino acids. System y(+) L-arginine transport activity was measured in Caco-2 monolayers exposed to various amino acids. L-arginine and/or other System y(+) substrates specifically upregulated System y(+) transport activity twofold after 1 hour, with a response noted as early as 5 minutes. Non-System y(+) substrates did not affect L-arginine absorption. Kinetic analysis indicated that L-arginine exposure increased both System y(+) K(m) and V(max). Neither cycloheximide nor actinomycin affected this stimulation, indicating that the regulation did not involve transcription or translation. The System y(+) substrate activation effect was reversible. L-arginine transport activity returned to baseline within 3 hours when cells were reincubated in amino acid-free media. These data indicate that System y(+) arginine transport activity is rapidly and reversibly activated by System y(+) substrates via a mechanism consistent with transmembrane stimulation. These findings identify a mechanism by which luminal nutrients regulate arginine uptake by the gut.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/fisiología , Arginina/metabolismo , Células CACO-2/metabolismo , Adaptación Fisiológica , Transporte Biológico , Células Cultivadas , Humanos
16.
J Gastrointest Surg ; 6(3): 410-7, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12022994

RESUMEN

Epidermal growth factor (EGF) in intestinal lumen regulates many gut epithelial cell functions. Influenced by growth factors at various differentiation stages, enterocytes execute the major task of absorbing nutrient amino acids. The purpose of this study was to investigate the effects of EGF on Na(+)-independent L-alanine transport in intestinal epithelial cells. Na(+)-independent [3H]-L-alanine transport was measured in the differentiating Caco-2 cells. In both the undifferentiated and differentiated states, L-alanine uptake occurred via a single saturable Na(+)-independent system L plus simple passive diffusion. System L activity decreased as the cells progressed from the undifferentiated to the differentiated state. Prolonged incubation with EGF (>30 hours) resulted in a 70% increase in system L activity in both undifferentiated and differentiated cells. EGF stimulated the system L V(max) without affecting K(m). System L activity stimulation was inhibited by chelerythrine chloride, cycloheximide, or actinomycin D. These data suggest that intestinal epithelial cell differentiation is associated with a decrease in system L transport capacity. EGF activates system L transport activity through a signaling mechanism involving protein kinase C, independent of cell differentiation state. Both cell differentiation and EGF regulation of system L activity occur via alteration of functional copies of the system L transporter.


Asunto(s)
Alanina/metabolismo , Sistema de Transporte de Aminoácidos L/fisiología , Células CACO-2/fisiología , Diferenciación Celular , Factor de Crecimiento Epidérmico/fisiología , Transporte Biológico , Senescencia Celular , Humanos , Absorción Intestinal/fisiología , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA