Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Graefes Arch Clin Exp Ophthalmol ; 259(1): 93-100, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32816099

RESUMEN

PURPOSE: We have previously shown that the chemokine CCL2 plays an important role in monocyte trafficking into the retina and alteration of the BRB in an animal model of diabetic retinopathy. In this study, we examined the effect of pharmacologically targeting the chemokine pathway to reduce the increased retinal vascular permeability in this model. METHODS: C57BL/6 J mice were made diabetic using streptozotocin. After 4 months of diabetes, mice (n = 10) were treated by intraperitoneal injections of TAK-779 (dual CCR2/CCR5 inhibitor) (30 mg/kg) daily for 2 weeks. Retinal vascular permeability and protein expression were done using western blot. The SDF-1 levels were measured by ELISA. Immune cell infiltration in the retinas was measured using flow cytometry. RESULTS: The dual inhibitor significantly decreased retinal vascular permeability in diabetic animals. There was a significant reduction in macrophage/microglia infiltration in the retinas of treated animals. Levels of SDF-1 and ICAM-1 were significantly reduced and the tight junction protein ZO-1 level was increased, and phospho-VE-Cad was significantly reduced with drug treatment. CONCLUSIONS: A chemokine receptor inhibitor (CCR2/CCR5) can reduce retinal vascular permeability in diabetic animals. Targeting the chemokine pathway pharmacologically may be used as a novel therapeutic strategy in management of diabetic macular edema.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética , Edema Macular , Animales , Barrera Hematorretinal , Permeabilidad Capilar , Diabetes Mellitus Experimental/tratamiento farmacológico , Retinopatía Diabética/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Receptores CCR2 , Receptores de Quimiocina , Retina , Vasos Retinianos
2.
Teach Learn Med ; 33(2): 139-153, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33289589

RESUMEN

Phenomenon: Because of its importance in residency selection, the United States Medical Licensing Examination Step 1 occupies a critical position in medical education, stimulating national debate about appropriate score use, equitable selection criteria, and the goals of undergraduate medical education. Yet, student perspectives on these issues and their implications for engagement with health systems science-related curricular content are relatively underexplored. Approach: We conducted an online survey of medical students at 19 American allopathic medical schools from March-July, 2019. Survey items were designed to elicit student opinions on the Step 1 examination and the impact of the examination on their engagement with new, non-test curricular content related to health systems science. Findings: A total of 2856 students participated in the survey, representing 23.5% of those invited. While 87% of students agreed that doing well on the Step 1 exam was their top priority, 56% disagreed that studying for Step 1 had a positive impact on engagement in the medical school curriculum. Eighty-two percent of students disagreed that Step 1 scores should be the top item residency programs use to offer interviews. When asked whether Step 1 results should be reported pass/fail with no numeric score, 55% of students agreed, while 33% disagreed. The majority of medical students agreed that health systems science topics were important but disagreed that studying for Step 1 helped learn this content. Students reported being more motivated to study a topic if it was on the exam, part of a course grade, prioritized by residency program directors, or if it would make them a better physician in the future. Insights: These results confirm the primacy of the United States Medical Licensing Examination Step 1 exam in preclinical medical education and demonstrate the need to balance the objectives of medical licensure and residency selection with the goals of the broader medical profession. The survey responses suggest several potential solutions to increase student engagement in health systems science curricula which may be especially important after Step 1 examination results are reported as pass/fail.


Asunto(s)
Educación de Pregrado en Medicina , Internado y Residencia , Estudiantes de Medicina , Actitud , Evaluación Educacional , Humanos , Licencia Médica , Estados Unidos
3.
Exp Eye Res ; 195: 108043, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32376470

RESUMEN

Selective pericyte loss, the histological hallmark of early diabetic retinopathy (DR), enhances the breakdown of the blood-retinal barrier (BRB) in diabetes. However, the role of pericytes on BRB alteration in diabetes and the signaling pathways involved in their effects are currently unknown. To understand the role of diabetes-induced molecular alteration of pericytes, we performed transcriptomic analysis of sorted retinal pericytes from mice model of diabetes. Retinal tissue from non-diabetic and diabetic (duration 3 months) mouse eyes (n = 10 in each group) were used to isolate pericytes through fluorescent activated cell sorting (FACS) using pericyte specific fluorescent antibodies, PDGFRb-APC. For RNA sequencing and qPCR analysis, a cDNA library was generated using template switching oligo and the resulting libraries were sequenced using paired-end Illumina sequencing. Molecular functional pathways were analyzed using differentially expressed genes (DEGs). Differential expression analysis revealed 217 genes significantly upregulated and 495 genes downregulated, in pericytes isolated from diabetic animals. These analyses revealed a core set of differentially expressed genes that could potentially contribute to the pericyte dysfunction in diabetes and highlighted the pattern of functional connectivity between key candidate genes and blood retinal barrier alteration mechanisms. The top up-regulated gene list included: Ext2, B3gat3, Gpc6, Pip5k1c and Pten and down-regulated genes included: Notch3, Xbp1, Gpc4, Atp1a2 and AKT3. Out of these genes, we further validated one of the down regulated genes, Notch 3 and its role in BRB alteration in diabetic retinopathy. We confirmed the downregulation of Notch3 expression in human retinal pericytes exposed to Advanced Glycation End-products (AGEs) treatment mimicking the chronic hyperglycemia effect. Exploration of pericyte-conditioned media demonstrated that loss of NOTCH3 in pericyte led to increased permeability of endothelial cell monolayers. Collectively, we identify a role for NOTCH3 in pericyte dysfunction in diabetes. Further validation of other DEGs to identify cell specific molecular change through whole transcriptomic approach in diabetic retina will provide novel insight into the pathogenesis of DR and novel therapeutic targets.


Asunto(s)
Barrera Hematorretinal/metabolismo , Diabetes Mellitus Experimental , Retinopatía Diabética/genética , Células Endoteliales/metabolismo , Retina/metabolismo , Transcriptoma/genética , Animales , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Células Endoteliales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Retina/patología , Transducción de Señal
4.
FASEB J ; 32(5): 2539-2548, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29263022

RESUMEN

Inflammation plays an important role in the pathogenesis of diabetic retinopathy. We have previously demonstrated the effect of cathepsin D (CD) on the mechanical disruption of retinal endothelial cell junctions and increased vasopermeability, as well as increased levels of CD in retinas of diabetic mice. Here, we have also examined the effect of CD on endothelial-pericyte interactions, as well as the effect of dipeptidyl peptidase-4 (DPP4) inhibitor on CD in endothelial-pericyte interactions in vitro and in vivo. Cocultured cells that were treated with pro-CD demonstrated a significant decrease in the expression of platelet-derived growth factor receptor-ß, a tyrosine kinase receptor that is required for pericyte cell survival; N-cadherin, the key adherens junction protein between endothelium and pericytes; and increases in the vessel destabilizing agent, angiopoietin-2. The effect was reversed in cells that were treated with DPP4 inhibitor along with pro-CD. With pro-CD treatment, there was a significant increase in the phosphorylation of the downstream signaling protein, PKC-α, and Ca2+/calmodulin-dependent protein kinase II in endothelial cells and pericytes, which disrupts adherens junction structure and function, and this was significantly reduced with DPP4 inhibitor treatment. Increased CD levels, vasopermeability, and alteration in junctional-related proteins were observed in the retinas of diabetic rats, which were significantly changed with DPP4 inhibitor treatment. Thus, DPP4 inhibitors may be used as potential adjuvant therapeutic agents to treat increased vascular leakage observed in patients with diabetic macular edema.-Monickaraj, F., McGuire, P., Das, A. Cathepsin D plays a role in endothelial-pericyte interactions during alteration of the blood-retinal barrier in diabetic retinopathy.


Asunto(s)
Barrera Hematorretinal/enzimología , Catepsina D/metabolismo , Comunicación Celular , Retinopatía Diabética/enzimología , Células Endoteliales/enzimología , Pericitos/enzimología , Angiopoyetina 2/metabolismo , Animales , Barrera Hematorretinal/patología , Cadherinas/metabolismo , Señalización del Calcio/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Catepsina D/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Retinopatía Diabética/patología , Células Endoteliales/patología , Masculino , Proteínas del Tejido Nervioso/metabolismo , Pericitos/patología , Ratas , Ratas Sprague-Dawley , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
5.
FASEB J ; 30(4): 1670-82, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26718887

RESUMEN

Inflammation plays an important role in the pathogenesis of diabetic retinopathy (DR). We have previously reported increased monocyte (Mono) trafficking into the retinas of diabetic animals. In this study, we have examined the effect of activated Monos on retinal endothelial cells (ECs). The U937 Mϕ-conditioned medium (CM) significantly decreased the transendothelial resistance of EC monolayers as measured by electric cell-substrate impedance sensing (P= 0.007). The CM was fractioned, and the effective fraction (30-100 kDa) was analyzed by liquid chromatography-mass spectrometry, and cathepsin D (CD) was identified as a major secreted product. Immunoprecipitated CD resulted in decreased resistance in ECs (P= 0.006). The specificity of CD in mediating alterations of the EC barrier was confirmed using small interfering RNA. The decreased resistance correlated with a significantly increased gap between ECs. CD altered the Ras homolog gene family, member A/Rho-associated kinase pathway with increased stress actin filament formation in the EC layer. Increased CD levels were found in the retinas of diabetic mice (3-fold) and serum samples of patients with diabetic macular edema (1.6-fold) measured by Western blot and ELISA. These findings suggest an important role for Mϕ-derived CD in altering the blood-retinal barrier and reveal a potential therapeutic target in the treatment of DR.-Monickaraj, F., McGuire, P. G., Nitta, C. F., Ghosh, K., Das, A. Cathepsin D: an Mϕ-derived factor mediating increased endothelial cell permeability with implications for alteration of the blood-retinal barrier in diabetic retinopathy.


Asunto(s)
Barrera Hematorretinal/metabolismo , Catepsina D/metabolismo , Retinopatía Diabética/metabolismo , Células Endoteliales/metabolismo , Adulto , Anciano , Animales , Western Blotting , Permeabilidad Capilar , Catepsina D/sangre , Catepsina D/genética , Permeabilidad de la Membrana Celular , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/sangre , Ensayo de Inmunoadsorción Enzimática , Humanos , Macrófagos/enzimología , Edema Macular/sangre , Edema Macular/metabolismo , Masculino , Ratones Endogámicos C57BL , Microscopía Confocal , Persona de Mediana Edad , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células U937 , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
6.
FASEB J ; 30(2): 601-11, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26443820

RESUMEN

Endothelial activation is a hallmark of the high-glucose (HG)-induced retinal inflammation associated with diabetic retinopathy (DR). However, precisely how HG induces retinal endothelial activation is not fully understood. We hypothesized that HG-induced up-regulation of lysyl oxidase (LOX), a collagen-cross-linking enzyme, in retinal capillary endothelial cells (ECs) enhances subendothelial basement membrane (BM) stiffness, which, in turn, promotes retinal EC activation. Diabetic C57BL/6 mice exhibiting a 70 and 50% increase in retinal intercellular adhesion molecule (ICAM)-1 expression and leukocyte accumulation, respectively, demonstrated a 2-fold increase in the levels of BM collagen IV and LOX, key determinants of capillary BM stiffness. Using atomic force microscopy, we confirmed that HG significantly enhances LOX-dependent subendothelial matrix stiffness in vitro, which correlated with an ∼2.5-fold increase in endothelial ICAM-1 expression, a 4-fold greater monocyte-EC adhesion, and an ∼2-fold alteration in endothelial NO (decrease) and NF-κB activation (increase). Inhibition of LOX-dependent subendothelial matrix stiffening alone suppressed HG-induced retinal EC activation. Finally, using synthetic matrices of tunable stiffness, we demonstrated that subendothelial matrix stiffening is necessary and sufficient to promote EC activation. These findings implicate BM stiffening as a critical determinant of HG-induced retinal EC activation and provide a rationale for examining BM stiffness and underlying mechanotransduction pathways as therapeutic targets for diabetic retinopathy.


Asunto(s)
Membrana Basal/patología , Diabetes Mellitus Experimental/complicaciones , Retinopatía Diabética/inducido químicamente , Endotelio/patología , Retina/patología , Animales , Línea Celular , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/fisiología , Haplorrinos , Humanos , Ratones , Ratones Endogámicos C57BL , Monocitos , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Proteína-Lisina 6-Oxidasa/genética , Proteína-Lisina 6-Oxidasa/metabolismo
7.
Ophthalmology ; 122(7): 1375-94, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25935789

RESUMEN

Diabetic macular edema (DME) is the major cause of vision loss in diabetic persons. Alteration of the blood-retinal barrier is the hallmark of this disease, characterized by pericyte loss and endothelial cell-cell junction breakdown. Recent animal and clinical studies strongly indicate that DME is an inflammatory disease. Multiple cytokines and chemokines are involved in the pathogenesis of DME, with multiple cellular involvement affecting the neurovascular unit. With the introduction of anti-vascular endothelial growth factor (VEGF) agents, the treatment of DME has been revolutionized, and the indication for laser therapy has been limited. However, the response to anti-VEGF drugs in DME is not as robust as in proliferative diabetic retinopathy, and many patients with DME do not show complete resolution of fluid despite multiple intravitreal injections. Potential novel therapies targeting molecules other than VEGF and using new drug-delivery systems currently are being developed and evaluated in clinical trials.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/fisiopatología , Edema Macular/tratamiento farmacológico , Edema Macular/fisiopatología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Humanos , Factores de Riesgo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
8.
Blood ; 117(20): 5541-50, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21460248

RESUMEN

In this study we investigated the role of CB1 receptor signaling in angiogenesis and the therapeutic exploitation of CB1 inactivation as an antiangiogenic strategy. We started from the observation that CB1 receptor expression is induced during angiogenesis and that the endocannabinoid anandamide stimulated bFGF-induced angiogenesis in the nanomolar physiologic range. To define the functional involvement of CB1 receptor signaling during angiogenesis, 2 different strategies have been carried out: siRNA-mediated knockdown and pharmacologic antagonism of CB1 receptors. CB1 receptors inactivation resulted in the inhibition of bFGF-induced endothelial proliferation, migration, and capillary-like tube formation, through prosurvival and migratory pathways involving ERK, Akt, FAK, JNK, Rho, and MMP-2. To corroborate the potential therapeutic exploitation of CB1 blockade as an antiangiogenic strategy, we performed in vivo assays founding that CB1 blockade was able to inhibit bFGF-induced neovascular growth in the rabbit cornea assay. A relevant finding was the ability to reduce ocular pathologic neo-vascularization in mouse oxygen-induced retinopathy. These results demonstrate that CB1 signaling participates to the proliferative response elicited by proangiogenic growth factors in angiogenesis and that for this reason CB1 receptor could represent a novel target for the treatment of diseases where excessive neoangiogenesis is the underlying pathology.


Asunto(s)
Neovascularización Fisiológica , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/deficiencia , Animales , Secuencia de Bases , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Córnea/irrigación sanguínea , Córnea/efectos de los fármacos , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Recién Nacido , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , ARN Interferente Pequeño/genética , Conejos , Receptor Cannabinoide CB1/genética , Retinopatía de la Prematuridad/tratamiento farmacológico , Transducción de Señal , Quinasas Asociadas a rho/metabolismo
9.
J Surg Res ; 183(2): 963-75, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23498341

RESUMEN

BACKGROUND: The controlled recruitment of monocytes from the circulation to the site of injury and their differentiation into tissue macrophages are critical events in the reconstitution of tissue integrity. Subsets of monocytes/macrophages have been implicated in the pathogenesis of atherosclerosis and tumor vascularity; however, the significance of monocyte heterogeneity in physiologic neovascularization is just emerging. MATERIALS AND METHODS: A cranial-based, peninsular-shaped myocutaneous flap was surgically created on the dorsum of wild-type mice (C57BL6) and populations of mice with genetic deletion of subset-specific chemokine ligand-receptor axes important in monocyte trafficking and function (CCL2(-/-) and CX3CR1(-/-)) (n=36 total; 12 mice per group, nine with flap and three unoperated controls). Planimetric analysis of digital photographic images was utilized to determine flap surface viability in wild-type and knockout mice. Real-time myocutaneous flap perfusion and functional revascularization was determined by laser speckle contrast imaging. Image analysis of CD-31 immunostained sections confirmed flap microvascular density and anatomy. Macrophage quantification and localization in flap tissues was determined by F4/80 gene and protein expression. Quantitative reverse transcription-polymerase chain reaction was performed on nonoperative back skin and postoperative flap tissue specimens to determine local gene expression. RESULTS: Myocutaneous flaps created on wild type and CX3CR1(-/-) mice were engrafted to the recipient site, resulting in viability. In contrast, distal full thickness cutaneous necrosis and resultant flap dehiscence was evident by d 10 in CCL2(-/-) mice. Over 10 d, laser speckle contrast imaging documented immediate graded flap ischemia in all three groups of mice, functional flap revascularization in wild type and CX3CR1(-/-) mice, and lack of distal flap reperfusion in CCL2(-/-) mice. Immunostaining of serial histologic specimens confirmed marked increases in microvascular density and number of macrophages in wild type mice, intermediate increases in CX3CR1(-/-) mice, and no significant change in vessel count or macrophage quantity in CCL2(-/-) mice over the study interval. Finally, quantitative reverse transcriptase polymerase chain reaction demonstrated that the loss of function of chemokine ligand and receptor genes influenced the transcription of local genes involved in monocyte chemotaxis and wound angiogenesis. CONCLUSIONS: In a graded-ischemia wound healing model, monocyte recruitment was severely impaired in CCL2(-/-) mice, resulting in failure of flap revascularization and concomitant cutaneous necrosis. Analysis of CX3CR1-deficient mice revealed adequate monocyte recruitment and revascularization for flap survival; however, the myeloid cell response and magnitude of neovascularization were dampened compared with wild type mice.


Asunto(s)
Monocitos/fisiología , Neovascularización Fisiológica/fisiología , Piel/irrigación sanguínea , Cicatrización de Heridas/fisiología , Animales , Receptor 1 de Quimiocinas CX3C , Quimiocina CCL2/deficiencia , Quimiocina CCL2/genética , Quimiocina CCL2/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación/fisiología , Modelos Animales , Monocitos/patología , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Receptores de Quimiocina/fisiología , Piel/patología , Colgajos Quirúrgicos/irrigación sanguínea
10.
Arterioscler Thromb Vasc Biol ; 31(12): e107-15, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21940944

RESUMEN

OBJECTIVE: The mechanisms that regulate the physical interaction of pericytes and endothelial cells and the effects of these interactions on interendothelial cell junctions are not well understood. We determined the extent to which vascular pericytes could regulate pericyte-endothelial adhesion and the consequences that this disruption might have on the function of the endothelial barrier. METHODS AND RESULTS: Human retinal microvascular endothelial cells were cocultured with pericytes, and the effect on the monolayer resistance of endothelial cells and expression of the cell junction molecules N-cadherin and VE-cadherin were measured. The molecules responsible for the effect of pericytes or pericyte-conditioned media on the endothelial resistance and cell junction molecules were further analyzed. Our results indicate that pericytes increase the barrier properties of endothelial cell monolayers. This barrier function is maintained through the secretion of pericyte-derived sphingosine 1-phosphate. Sphingosine 1-phosphate aids in maintenance of microvascular stability by upregulating the expression of N-cadherin and VE-cadherin, and downregulating the expression of angiopoietin 2. CONCLUSIONS: Under normal circumstances, the retinal vascular pericytes maintain pericyte-endothelial contacts and vascular barrier function through the secretion of sphingosine 1-phosphate. Alteration of pericyte-derived sphingosine 1-phosphate production may be an important mechanism in the development of diseases characterized by vascular dysfunction and increased permeability.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Endotelio Vascular/citología , Uniones Intercelulares/fisiología , Lisofosfolípidos/metabolismo , Pericitos/citología , Pericitos/metabolismo , Vasos Retinianos/citología , Esfingosina/análogos & derivados , Angiopoyetina 2/metabolismo , Permeabilidad Capilar/fisiología , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Regulación hacia Abajo/fisiología , Endotelio Vascular/fisiología , Humanos , Vasos Retinianos/fisiología , Esfingosina/metabolismo , Regulación hacia Arriba/fisiología
11.
J Surg Res ; 164(1): e201-12, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20863524

RESUMEN

BACKGROUND: The delivery of proangiogenic agents in clinical trials of wound healing has produced equivocal results, the lack of real-time assessment of vascular growth is a major weakness in monitoring the efficacy of therapeutic angiogenesis, and surgical solutions fall short in addressing the deficiency in microvascular blood supply to ischemic wounds. Therefore, elucidation of the mechanisms involved in ischemia-induced blood vessel growth has potential diagnostic and therapeutic implications in wound healing. MATERIALS AND METHODS: Three surgical models of wound ischemia, a cranial-based myocutaneous flap, an identical flap with underlying silicone sheeting to prevent engraftment, and a complete incisional flap without circulation were created on C57BL6 transgenic mice. Laser speckle contrast imaging was utilized to study the pattern of ischemia and return of revascularization. Simultaneous analysis of wound histology and microvascular density provided correlation of wound perfusion and morphology. RESULTS: Creation of the peninsular-shaped flap produced a gradient of ischemia. Laser speckle contrast imaging accurately predicted the spatial and temporal pattern of ischemia, the return of functional revascularization, and the importance of engraftment in distal flap perfusion and survival. Histologic analysis demonstrated engraftment resulted in flap revascularization by new blood vessel growth from the recipient bed and dilatation of pre-existing flap vasculature. CONCLUSIONS: Further research utilizing this model of graded wound ischemia and the technology of laser speckle perfusion imaging will allow monitoring of the real-time restitution of blood flow for correlation with molecular biomarkers of revascularization in an attempt to gain further understanding of wound microvascular biology.


Asunto(s)
Isquemia/diagnóstico , Rayos Láser , Angioscopía Microscópica/instrumentación , Angioscopía Microscópica/métodos , Colgajos Quirúrgicos/irrigación sanguínea , Animales , Capilares/fisiología , Dextranos/farmacocinética , Modelos Animales de Enfermedad , Femenino , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Proteínas Fluorescentes Verdes/genética , Isquemia/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Fisiológica/fisiología , Perfusión/métodos
12.
J Clin Med ; 9(1)2020 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-31947513

RESUMEN

Although there is strong clinical evidence that the control of blood glucose, blood pressure, and lipid level can prevent and slow down the progression of diabetic retinopathy (DR) as shown by landmark clinical trials, it has been shown that these factors only account for 10% of the risk for developing this disease. This suggests that other factors, such as genetics, may play a role in the development and progression of DR. Clinical evidence shows that some diabetics, despite the long duration of their diabetes (25 years or more) do not show any sign of DR or show minimal non-proliferative diabetic retinopathy (NPDR). Similarly, not all diabetics develop proliferative diabetic retinopathy (PDR). So far, linkage analysis, candidate gene studies, and genome-wide association studies (GWAS) have not produced any statistically significant results. We recently initiated a genomics study, the Diabetic Retinopathy Genetics (DRGen) Study, to examine the contribution of rare and common variants in the development of different phenotypes of DR, as well as their responsiveness to anti-VEGF treatment in diabetic macular edema (DME). Our preliminary findings reveal a novel set of genetic variants involved in the angiogenesis and inflammatory pathways that contribute to DR progression or protection. Further investigation of variants can help to develop novel biomarkers and lead to new therapeutic targets in DR.

13.
Front Oncol ; 10: 965, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32626657

RESUMEN

Colorectal cancer (CRC) is a leading cause of cancer death in the United States. Artemisinin derivatives, including the dihydroartemisinin (DHA) monomers, are widely used as clinical agents for the treatment of malaria. Numerous studies demonstrate that these molecules also display antineoplastic activity with minimal toxicity. Of interest, dimeric DHA molecules are more active than their monomeric counterparts. Our previous data showed that the DHA dimer, NSC735847, was a potent inducer of death in different cancer cell types. However, the mechanism of action and activity of NSC735847 in colon cancer cells was not explored. The present study investigated the anticancer activity of NSC735847 and four structurally similar analog in human tumorigenic (HT-29 and HCT-116) and non-tumorigenic (FHC) colon cell lines. NSC735847 was more cytotoxic toward tumorigenic than non-tumorigenic colonocytes. In addition, NSC735847 exhibited greater cytotoxicity and tumor selectivity than the NSC735847 derivatives. To gain insight into mechanisms of NSC735847 activity, the requirement for endoplasmic reticulum (ER) stress and oxidative stress was tested. The data show that ER stress played a key role in the cytotoxicity of NSC735847 while oxidative stress had little impact on cell fate. In addition, it was observed that the cytotoxic activity of NSC735847 required the presence of heme, but not iron. The activity of NSC735847 was then compared to clinically utilized CRC therapeutics. NSC735847 was cytotoxic toward colon tumor cells at lower concentrations than oxaliplatin (OX). In addition, cell death was achieved at lower concentrations in colon cancer cells that were co-treated with folinic acid (Fol), 5-FU (F), and NSC735847 (FolFNSC), than Fol, F, and OX (FolFOX). The selective activity of NSC735847 and its ability to induce cytotoxicity at low concentrations suggest that NSC735847 may be an alternative for oxaliplatin in the FolFOX regimen for patients who are unable to tolerate its adverse effects.

14.
FASEB J ; 22(9): 3310-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18559877

RESUMEN

One of the major complications of diabetes is the alteration of the blood-retinal barrier, leading to retinal edema and consequent vision loss. The aim of this study was to evaluate the role of the urokinase plasminogen activator (uPA)/uPA receptor (uPAR) system in the regulation of retinal vascular permeability. Biochemical, molecular, and histological techniques were used to examine the role of uPA and uPAR in the regulation of retinal vascular permeability in diabetic rats and cultured retinal endothelial cells. The increased retinal vascular permeability in diabetic rats was associated with a decrease in vascular endothelial (VE) -cadherin expression in retinal vessels. Treatment with the uPA/uPAR-inhibiting peptide (A6) was shown to reduce diabetes-induced permeability and the loss of VE-cadherin. The increased permeability of cultured cells in response to advanced glycation end products (AGEs) was significantly inhibited with A6. Treatment of endothelial cells with specific matrix metalloproteinases or AGEs resulted in loss of VE-cadherin from the cell surface, which could be inhibited by A6. uPA/uPAR physically interacts with AGEs/receptor for advanced glycation end products on the cell surface and regulates its activity. uPA and its receptor uPAR play important roles in the alteration of the blood-retinal barrier through proteolytic degradation of VE-cadherin. The ability of A6 to block retinal vascular permeability in diabetes suggests a potential therapeutic approach for the treatment of diabetic macular edema.


Asunto(s)
Barrera Hematorretinal/efectos de los fármacos , Diabetes Mellitus Experimental/fisiopatología , Fragmentos de Péptidos/farmacología , Receptores de Superficie Celular/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Animales , Antígenos CD/biosíntesis , Cadherinas/biosíntesis , Permeabilidad Capilar/efectos de los fármacos , Bovinos , Células Cultivadas , Endotelio Vascular/citología , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Ratas , Receptores de Superficie Celular/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Retina/citología , Retina/metabolismo , Regulación hacia Arriba , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Activador de Plasminógeno de Tipo Uroquinasa/farmacología
15.
Invest Ophthalmol Vis Sci ; 48(4): 1793-800, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17389513

RESUMEN

PURPOSE: The purpose of this study was to determine the role of hepatocyte growth factor (HGF) and c-Met in the initiation and development of retinal neovascularization and to determine whether inhibition of this system can suppress the extent of angiogenesis in an animal model. METHODS: Retinal tissues from animals with oxygen-induced neovascularization were analyzed for HGF and c-Met expression and localization. The effect of HGF on the migratory and invasive behavior of isolated retinal endothelial cells was quantitated, and the role of the extracellular proteinase urokinase in facilitating this process was determined. Mice were treated with intraocular injections of anti-c-Met antibody, and the extent of neovascularization was quantitated. RESULTS: HGF and c-Met were upregulated in the retinas of mice with hypoxia-induced retinal neovascularization. HGF was active, as evidenced by the increased presence of the phosphorylated form of c-Met in the tissues. c-Met was localized to various cell types in the retina, including vascular cells, and HGF was produced by cells in the ganglion and inner nuclear layers. HGF stimulated the secretion of urokinase and its receptor, uPAR, in isolated retinal endothelial cells. HGF increased the migratory and invasive capacity of these cells, which could be inhibited by the disruption of urokinase/uPAR interactions with the A6 peptide. Inhibition of c-Met activation in vivo resulted in a 70% decrease in retinal angiogenesis and a 40% decrease in urokinase activity in the retina. CONCLUSIONS: These studies suggest that HGF may play an important role in the initial stages of retinal angiogenesis by stimulating a migratory phenotype in endothelial cells mediated by increased urokinase activity.


Asunto(s)
Factor de Crecimiento de Hepatocito/fisiología , Neovascularización Retiniana/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Western Blotting , Bovinos , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Técnica del Anticuerpo Fluorescente Indirecta , Factor de Crecimiento de Hepatocito/farmacología , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Oxígeno/toxicidad , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/fisiología , ARN Mensajero/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Células Ganglionares de la Retina/metabolismo , Neovascularización Retiniana/inducido químicamente , Neovascularización Retiniana/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Organismos Libres de Patógenos Específicos , Regulación hacia Arriba
16.
Vision Res ; 139: 221-227, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28993218

RESUMEN

The leading cause of major vision loss in diabetic persons is diabetic macular edema (DME). The hallmark feature of diabetic retinopathy is the alteration of the blood-retinal barrier (BRB). Inflammation plays a crucial role in DME with involvement of several chemokines and cytokines including vascular endothelial growth factor (VEGF). VEGF is a potent cytokine and vaso-permeability factor that has been targeted in multiple, large clinical trials. Multiple anti-VEGF drugs are widely used in the treatment of diabetic macular edema (DME) as the first line of treatment, and have been shown to be effective in vision improvement and prevention of vision loss. However, many DME patients do not show complete response to anti-VEGF drugs despite multiple intravitreal injections with these drugs. Also, the effect seems to be transient in those responders, and many patients do not show complete resolution of fluid. This article summarizes the mechanisms other than VEGF, and how these novel factors can be targeted as promising therapies of DME.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Edema Macular/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Barrera Hematorretinal/fisiología , Retinopatía Diabética/fisiopatología , Humanos , Inyecciones Intravítreas , Edema Macular/fisiopatología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
17.
Semin Ophthalmol ; 21(1): 23-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16517441

RESUMEN

Cell migration is a critical step in the angiogenesis cascade that involves proteolysis of the basement membrane and extracellular matrix around existing blood vessels. The urokinase plasminogen activator (uPA) system has been involved in cellular invasion, angiogenesis and tumor growth. Similar expression of urokinase and its receptor (uPAR) is seen in both retinal and choroidal neovascularization. Significant inhibition of choroidal neovascularization (CNV) has been observed when cell surface associated uPA-uPAR activity is prevented with a specific inhibitor of this proteinase system. As the current treatments of CNV are not optimal, the urokinase-uPAR system appears to be an attractive target for alternative pharamacological therapy for CNV.


Asunto(s)
Proteínas Sanguíneas/uso terapéutico , Neovascularización Coroidal/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Animales , Movimiento Celular/efectos de los fármacos , Neovascularización Coroidal/enzimología , Neovascularización Coroidal/patología , Humanos , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
18.
Indian J Ophthalmol ; 64(1): 4-13, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26953018

RESUMEN

The blood-retinal barrier (BRB) alteration is the hallmark feature of diabetic retinopathy. Vascular endothelial growth factor (VEGF) is a potent vasopermeability factor that has been implicated in the pathogenesis of BRB alteration. Inflammation also plays a crucial role in this process with involvement of several chemokines and cytokines. Multiple anti-VEGF drugs are widely used as in the treatment of diabetic macular edema (DME) as well as proliferative diabetic retinopathy. Several clinical trials have proved the beneficial effects of these drugs in improvement of vision and prevention of vision loss. However, the response to anti-VEGF drugs in DME is not complete in a significant number of patients. The effect seems transient in this latter group, and many patients do not show complete resolution of fluid. Potential novel therapies targeting molecules beyond VEGF are being developed and examined in clinical trials.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Barrera Hematorretinal , Retinopatía Diabética/etiología , Retinopatía Diabética/fisiopatología , Humanos , Preparaciones Farmacéuticas , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
19.
Diabetes Metab Syndr Obes ; 9: 325-336, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27757044

RESUMEN

BACKGROUND: Murine models of diabetes and obesity have provided insight into the pathogenesis of impaired epithelialization of excisional skin wounds. However, knowledge of postischemic myocutaneous revascularization in these models is limited. MATERIALS AND METHODS: A myocutaneous flap was created on the dorsum of wild type (C57BL/6), genetically obese and diabetic (ob/ob, db/db), complementary heterozygous (ob+/ob-, db+/db-), and diet-induced obese (DIO) mice (n=48 total; five operative mice per strain and three unoperated mice per strain as controls). Flap perfusion was documented by laser speckle contrast imaging. Local gene expression in control and postoperative flap tissue specimens was determined by quantitative reverse transcription polymerase chain reaction (RT-PCR). Image analysis of immunochemically stained histologic sections confirmed microvascular density and macrophage presence. RESULTS: Day 10 planimetric analysis revealed mean flap surface area necrosis values of 10.8%, 12.9%, 9.9%, 0.4%, 1.4%, and 23.0% for wild type, db+/db-, ob+/ob-, db/db, ob/ob, and DIO flaps, respectively. Over 10 days, laser speckle imaging documented increased perfusion at all time points with revascularization to supranormal perfusion in db/db and ob/ob flaps. In contrast, wild type, heterozygous, and DIO flaps displayed expected graded ischemia with failure of perfusion to return to baseline values. RT-PCR demonstrated statistically significant differences in angiogenic gene expression between lean and obese mice at baseline (unoperated) and at day 10. CONCLUSION: Unexpected increased baseline skin perfusion and augmented myocutaneous revascularization accompanied by a control proangiogenic transcriptional signature in genetically obese mice compared to DIO and lean mice are reported. In future research, laser speckle imaging has been planned to be utilized in order to correlate spatiotemporal wound reperfusion with changes in cell recruitment and gene expression to better understand the differences in wound microvascular biology in lean and obese states.

20.
Prog Retin Eye Res ; 22(6): 721-48, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14575722

RESUMEN

Retinal angiogenesis and choroidal angiogenesis are major causes of vision loss, and the pathogenesis of this angiogenesis process is still uncertain. However, several key steps of the angiogenic cascade have been elucidated. In retinal angiogenesis, hypoxia is the initial stimulus that causes up regulation of growth factors, integrins and proteinases, which result in endothelial cell proliferation and migration that are critical steps in this process. Once the endothelial tube is formed from the existing blood vessels, maturation starts with recruitment of mural cell precursors and formation of the basement membrane. Normally, there is a tight balance between angiogenic factors and endogenous angiogenesis inhibitors that help to keep the angiogenic process under control. Although the steps of choroidal angiogenesis seem to be similar to those of retinal angiogenesis, there are some major differences between these two processes. Several anti-angiogenic approaches are being developed in animal models to prevent ocular angiogenesis by blocking the key steps of the angiogenic cascade. Based on these pre-clinical studies, several anti-angiogenic clinical trials are ongoing in patients with diabetic retinopathy and age-related macular degeneration. This review discusses the pathogenesis of retinal and choroidal angiogenesis, and alternative pharmacological approaches to inhibit angiogenesis in ocular diseases.


Asunto(s)
Coroides/irrigación sanguínea , Neovascularización Patológica/prevención & control , Neovascularización Patológica/fisiopatología , Vasos Retinianos , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA