Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Clin Invest ; 133(13)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37227777

RESUMEN

Many patients with diabetic eye disease respond inadequately to anti-VEGF therapies, implicating additional vasoactive mediators in its pathogenesis. We demonstrate that levels of angiogenic proteins regulated by HIF-1 and -2 remain elevated in the eyes of people with diabetes despite treatment with anti-VEGF therapy. Conversely, by inhibiting HIFs, we normalized the expression of multiple vasoactive mediators in mouse models of diabetic eye disease. Accumulation of HIFs and HIF-regulated vasoactive mediators in hyperglycemic animals was observed in the absence of tissue hypoxia, suggesting that targeting HIFs may be an effective early treatment for diabetic retinopathy. However, while the HIF inhibitor acriflavine prevented retinal vascular hyperpermeability in diabetic mice for several months following a single intraocular injection, accumulation of acriflavine in the retina resulted in retinal toxicity over time, raising concerns for its use in patients. Conversely, 32-134D, a recently developed HIF inhibitor structurally unrelated to acriflavine, was not toxic to the retina, yet effectively inhibited HIF accumulation and normalized HIF-regulated gene expression in mice and in human retinal organoids. Intraocular administration of 32-134D prevented retinal neovascularization and vascular hyperpermeability in mice. These results provide the foundation for clinical studies assessing 32-134D for the treatment of patients with diabetic eye disease.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética , Neovascularización Retiniana , Humanos , Ratones , Animales , Acriflavina/metabolismo , Acriflavina/farmacología , Acriflavina/uso terapéutico , Diabetes Mellitus Experimental/metabolismo , Retina/metabolismo , Neovascularización Retiniana/metabolismo , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
2.
Clin Ophthalmol ; 16: 3895-3904, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36444207

RESUMEN

Purpose: To compare postoperative anterior chamber inflammation, pain, and patient preference following small incision lenticule extraction (SMILE) in eyes treated with a dexamethasone 0.4 mg intracanalicular insert (DEX) or topical prednisolone acetate (PRED). Patients and Methods: In this prospective, randomized, fellow eye-controlled trial, 20 patients underwent same-day, bilateral SMILE. One randomly-selected eye of each patient received DEX placed immediately postoperatively, and the fellow eye received topical PRED tapered over 2 weeks. Postoperative evaluations were performed on day 1, week 1, month 1, and month 3. Primary outcomes included postoperative pain, incidence of anterior chamber cell and flare, and patient preference of steroid therapy. Results: No eyes in either group had any clinically evident cell or flare at any postoperative time point. Mean pain scores (0-10 by subjective report) and incidence of any pain were statistically similar at all postoperative visits. Uncorrected distance visual acuity improved in all eyes, 91% of which achieved 20/25 or better. No eyes lost any lines of corrected distance visual acuity. Three eyes developed a steroid-related rise in intraocular pressure, all of which resolved with 2 of the 3 eyes requiring topical therapy. At 1 week, 1 month, and 3 months, 70%, 65%, and 53% of patients preferred DEX over PRED therapy, respectively. Conclusion: The DEX insert was preferred by more patients and controlled postoperative inflammation and pain comparably to topical PRED in eyes undergoing SMILE. There were no statistically significant differences in visual outcomes between the two groups.

3.
JCI Insight ; 7(13)2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35653189

RESUMEN

Most patients with neovascular age-related macular degeneration (nvAMD), the leading cause of severe vision loss in elderly US citizens, respond inadequately to current therapies targeting a single angiogenic mediator, vascular endothelial growth factor (VEGF). Here, we report that aqueous fluid levels of a second vasoactive mediator, angiopoietin-like 4 (ANGPTL4), can help predict the response of patients with nvAMD to anti-VEGF therapies. ANGPTL4 expression was higher in patients who required monthly treatment with anti-VEGF therapies compared with patients who could be effectively treated with less-frequent injections. We further demonstrate that ANGPTL4 acts synergistically with VEGF to promote the growth and leakage of choroidal neovascular (CNV) lesions in mice. Targeting ANGPTL4 expression was as effective as targeting VEGF expression for treating CNV in mice, while simultaneously targeting both was more effective than targeting either factor alone. To help translate these findings to patients, we used a soluble receptor that binds to both VEGF and ANGPTL4 and effectively inhibited the development of CNV lesions in mice. Our findings provide an assay that can help predict the response of patients with nvAMD to anti-VEGF monotherapy and suggest that therapies targeting both ANGPTL4 and VEGF will be a more effective approach for the treatment of this blinding disease.


Asunto(s)
Neovascularización Coroidal , Degeneración Macular , Proteína 4 Similar a la Angiopoyetina , Animales , Humor Acuoso/metabolismo , Neovascularización Coroidal/tratamiento farmacológico , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Ratones , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
J Clin Invest ; 132(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34874918

RESUMEN

BackgroundTo reduce the treatment burden for patients with neovascular age-related macular degeneration (nvAMD), emerging therapies targeting vascular endothelial growth factor (VEGF) are being designed to extend the interval between treatments, thereby minimizing the number of intraocular injections. However, which patients will benefit from longer-acting agents is not clear.MethodsEyes with nvAMD (n = 122) underwent 3 consecutive monthly injections with currently available anti-VEGF therapies, followed by a treat-and-extend protocol. Patients who remained quiescent 12 weeks from their prior treatment entered a treatment pause and were switched to pro re nata (PRN) treatment (based on vision, clinical exam, and/or imaging studies). Proteomic analysis was performed on aqueous fluid to identify proteins that correlate with patients' response to treatment.ResultsAt the end of 1 year, 38 of 122 eyes (31%) entered a treatment pause (≥30 weeks). Conversely, 21 of 122 eyes (17%) failed extension and required monthly treatment at the end of year 1. Proteomic analysis of aqueous fluid identified proteins that correlated with patients' response to treatment, including proteins previously implicated in AMD pathogenesis. Interestingly, apolipoprotein-B100 (ApoB100), a principal component of drusen implicated in the progression of nonneovascular AMD, was increased in treated patients who required less frequent injections. ApoB100 expression was higher in AMD eyes compared with controls but was lower in eyes that develop choroidal neovascularization (CNV), consistent with a protective role. Accordingly, mice overexpressing ApoB100 were partially protected from laser-induced CNV.FundingThis work was supported by the National Eye Institute, National Institutes of Health grants R01EY029750, R01EY025705, and R01 EY27961; the Research to Prevent Blindness, Inc.; the Alcon Research Institute; and Johns Hopkins University through the Robert Bond Welch and Branna and Irving Sisenwein professorships in ophthalmology.ConclusionAqueous biomarkers could help identify patients with nvAMD who may not require or benefit from long-term treatment with anti-VEGF therapy.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Apolipoproteína B-100/metabolismo , Neovascularización Coroidal , Proteínas del Ojo/metabolismo , Degeneración Macular , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Animales , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/metabolismo , Femenino , Humanos , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Masculino , Ratones , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
J Clin Invest ; 129(11): 4593-4608, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31545295

RESUMEN

The majority of patients with diabetic macular edema (DME), the most common cause of vision loss in working-age Americans, do not respond adequately to current therapies targeting VEGFA. Here, we show that expression of angiopoietin-like 4 (ANGPTL4), a HIF-1-regulated gene product, is increased in the eyes of diabetic mice and patients with DME. We observed that ANGPTL4 and VEGF act synergistically to destabilize the retinal vascular barrier. Interestingly, while ANGPTL4 modestly enhanced tyrosine phosphorylation of VEGF receptor 2, promotion of vascular permeability by ANGPTL4 was independent of this receptor. Instead, we found that ANGPTL4 binds directly to neuropilin 1 (NRP1) and NRP2 on endothelial cells (ECs), leading to rapid activation of the RhoA/ROCK signaling pathway and breakdown of EC-EC junctions. Treatment with a soluble fragment of NRP1 (sNRP1) prevented ANGPTL4 from binding to NRP1 and blocked ANGPTL4-induced activation of RhoA as well as EC permeability in vitro and retinal vascular leakage in diabetic animals in vivo. In addition, sNRP1 reduced the stimulation of EC permeability by aqueous fluid from patients with DME. Collectively, these data identify the ANGPTL4/NRP/RhoA pathway as a therapeutic target for the treatment of DME.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/metabolismo , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Edema Macular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Diabetes Mellitus Experimental/patología , Retinopatía Diabética/patología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Edema Macular/patología , Ratones , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
6.
PLoS One ; 12(8): e0183320, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28832635

RESUMEN

The recent success of therapies directly targeting the angiogenic mediator, vascular endothelial growth factor (VEGF), for the treatment of proliferative diabetic retinopathy has encouraged clinicians to extend the use of anti-VEGF therapies for the treatment of another ischemic retinal vascular disease, proliferative sickle cell retinopathy (PSR), the most common cause of irreversible blindness in patients with sickle cell disease. However, results from case reports evaluating anti-VEGF therapies for PSR have been mixed. This highlights the need to identify alternative therapeutic targets for the treatment of retinal neovascularization in sickle cell patients. In this regard, angiopoietin-like 4 (ANGPTL4) is a novel angiogenic factor regulated by the transcription factor, hypoxia-inducible factor 1, the master regulator of angiogenic mediators (including VEGF) in ischemic retinal disease. In an effort to identify alternative targets for the treatment of sickle cell retinopathy, we have explored the expression of ANGPTL4 in the eyes of patients with PSR. To this end, we examined expression and localization of ANGPTL4 by immunohistochemistry in autopsy eyes from patients with known PSR (n = 5 patients). Complementary studies were performed using enzyme-linked immunosorbent assays in aqueous (n = 8; 7 patients, 2 samples from one eye of same patient) and vitreous (n = 3 patients) samples from a second group of patients with active PSR. We detected expression of ANGPTL4 in neovascular tissue and in the ischemic inner retina in PSR, but not control, eyes. We further observed elevated expression of ANGPTL4 in the aqueous and vitreous of PSR patients compared to controls. These results suggest that ANGPTL4 could contribute to the development of retinal neovascularization in sickle cell patients and could therefore be a therapeutic target for the treatment of PSR.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Angiopoyetinas/metabolismo , Ceguera/metabolismo , Neovascularización Patológica/metabolismo , Proteína 4 Similar a la Angiopoyetina , Ceguera/etiología , Humanos , Neovascularización Patológica/etiología , Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Invest Ophthalmol Vis Sci ; 57(15): 6739-6746, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27951596

RESUMEN

Purpose: Retinal vascular occlusions in sickle cell anemia patients cause tissue ischemia and the release of angiogenic mediators that promote the development of retinal neovascularization, initiating proliferative sickle retinopathy (PSR). Laser photocoagulation (LPC) has emerged as the most common treatment for PSR. Nonetheless, only two randomized controlled clinical trials have evaluated the use of LPC for PSR, and both failed to definitively demonstrate efficacy of this approach. This may be due to a lack of knowledge regarding the appropriate location for placement of laser coagulations in PSR eyes. To help address this question, we examined the expression of hypoxia-inducible factor (HIF)-1α and vascular endothelial growth factor (VEGF) in PSR eyes. Methods: The expression pattern of HIF-1α and VEGF in PSR (n = 5) and control (n = 3) eyes was examined by immunohistochemistry in different retinal regions defined by the presence or absence of retinal vessels. Results: Hypoxia-inducible factor 1α and VEGF were expressed in the inner retina of 5/5 untreated PSR eyes adjacent to retinal neovascularization; expression of HIF-1α was not detected (and VEGF only lightly detected) in normal retinal and choroidal vasculature of 3/3 control eyes. Hypoxia-inducible factor 1α and VEGF were strongly expressed in retinal cells within avascular (nonperfused) retina, anterior to the boundary between perfused and nonperfused retina, as well as in posterior ischemic retina in the presence or absence of neovascular sea fans. Conclusions: If the goal of LPC in PSR is to quench the expression of HIF-1-driven angiogenic mediators, our results support broad application of peripheral laser for its treatment.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Coagulación con Láser , Retina/metabolismo , Neovascularización Retiniana/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Vitreorretinopatía Proliferativa/metabolismo , Anemia de Células Falciformes/diagnóstico , Cadáver , Coroides/irrigación sanguínea , Coroides/metabolismo , Coroides/patología , Humanos , Inmunohistoquímica , Retina/patología , Retina/cirugía , Neovascularización Retiniana/etiología , Neovascularización Retiniana/cirugía , Vasos Retinianos/diagnóstico por imagen , Vasos Retinianos/metabolismo , Vitreorretinopatía Proliferativa/etiología , Vitreorretinopatía Proliferativa/cirugía
8.
Proc Natl Acad Sci U S A ; 112(23): E3030-9, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26039997

RESUMEN

Diabetic eye disease is the most common cause of severe vision loss in the working-age population in the developed world, and proliferative diabetic retinopathy (PDR) is its most vision-threatening sequela. In PDR, retinal ischemia leads to the up-regulation of angiogenic factors that promote neovascularization. Therapies targeting vascular endothelial growth factor (VEGF) delay the development of neovascularization in some, but not all, diabetic patients, implicating additional factor(s) in PDR pathogenesis. Here we demonstrate that the angiogenic potential of aqueous fluid from PDR patients is independent of VEGF concentration, providing an opportunity to evaluate the contribution of other angiogenic factor(s) to PDR development. We identify angiopoietin-like 4 (ANGPTL4) as a potent angiogenic factor whose expression is up-regulated in hypoxic retinal Müller cells in vitro and the ischemic retina in vivo. Expression of ANGPTL4 was increased in the aqueous and vitreous of PDR patients, independent of VEGF levels, correlated with the presence of diabetic eye disease, and localized to areas of retinal neovascularization. Inhibition of ANGPTL4 expression reduced the angiogenic potential of hypoxic Müller cells; this effect was additive with inhibition of VEGF expression. An ANGPTL4 neutralizing antibody inhibited the angiogenic effect of aqueous fluid from PDR patients, including samples from patients with low VEGF levels or receiving anti-VEGF therapy. Collectively, our results suggest that targeting both ANGPTL4 and VEGF may be necessary for effective treatment or prevention of PDR and provide the foundation for studies evaluating aqueous ANGPTL4 as a biomarker to help guide individualized therapy for diabetic eye disease.


Asunto(s)
Angiopoyetinas/fisiología , Retinopatía Diabética/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Angiopoyetinas/metabolismo , Retinopatía Diabética/metabolismo , Ojo/irrigación sanguínea , Ojo/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Patológica , Factor A de Crecimiento Endotelial Vascular/sangre
9.
Diabetes ; 62(11): 3863-73, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23884892

RESUMEN

In proliferative diabetic retinopathy (PDR), retinal ischemia promotes neovascularization (NV), which can lead to profound vision loss in diabetic patients. Treatment for PDR, panretinal photocoagulation, is inherently destructive and has significant visual consequences. Therapies targeting vascular endothelial growth factor (VEGF) have transformed the treatment of diabetic eye disease but have proven inadequate for treating NV, prompting exploration for additional therapeutic options for PDR patients. In this regard, extracellular proteolysis is an early and sustained activity strictly required for NV. Extracellular proteolysis in NV is facilitated by the dysregulated activity of matrix metalloproteinases (MMPs). Here, we set out to better understand the regulation of MMPs by ischemia in PDR. We demonstrate that accumulation of hypoxia-inducible factor-1α in Müller cells induces the expression of VEGF, which, in turn, promotes increased MMP-2 expression and activity in neighboring endothelial cells (ECs). MMP-2 expression was detected in ECs in retinal NV tissue from PDR patients, whereas MMP-2 protein levels were elevated in the aqueous of PDR patients compared with controls. Our findings demonstrate a complex interplay among hypoxic Müller cells, secreted angiogenic factors, and neighboring ECs in the regulation of MMP-2 in retinal NV and identify MMP-2 as a target for the treatment of PDR.


Asunto(s)
Retinopatía Diabética/fisiopatología , Células Ependimogliales/metabolismo , Metaloproteinasa 2 de la Matriz/biosíntesis , Neovascularización Retiniana/etiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Digoxina/farmacología , Activación Enzimática , Células Ependimogliales/efectos de los fármacos , Femenino , Hipoxia/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL
10.
PLoS One ; 8(3): e58138, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23536784

RESUMEN

New insight into the biomechanics of cancer cell motility in 3D extracellular matrix (ECM) environments would significantly enhance our understanding of aggressive cancers and help identify new targets for intervention. While several methods for measuring the forces involved in cell-matrix interactions have been developed, previous to this study none have been able to measure forces in a fibrillar environment. We have developed a novel assay for simultaneously measuring cell mechanotransduction and motility in 3D fibrillar environments. The assay consists of a controlled-density fibrillar collagen gel atop a controlled-stiffness polyacrylamide (PAA) surface. Forces generated by living cells and their migration in the 3D collagen gel were measured with the 3D motion of tracer beads within the PAA layer. Here, this 3D fibril force assay is used to study the role of the invasion-associated protein kinase Src in mechanotransduction and motility. Src expression and activation are linked with proliferation, invasion, and metastasis, and have been shown to be required in 2D for invadopodia membranes to direct and mediate invasion. Breast cancer cell line MDA-MD-231 was stably transfected with GFP-tagged constitutively active Src or wild-type Src. In 3D fibrillar collagen matrices we found that, relative to wild-type Src, constitutively active Src: 1) increased the strength of cell-induced forces on the ECM, 2) did not significantly change migration speed, and 3) increased both the duration and the length, but not the number, of long membrane protrusions. Taken together, these results support the hypothesis that Src controls invasion by controlling the ability of the cell to form long lasting cellular protrusions to enable penetration through tissue barriers, in addition to its role in promoting invadopodia matrix-degrading activity.


Asunto(s)
Colágeno/metabolismo , Neoplasias/metabolismo , Familia-src Quinasas/metabolismo , Fenómenos Biomecánicos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular , Extensiones de la Superficie Celular/metabolismo , Activación Enzimática , Matriz Extracelular/metabolismo , Femenino , Adhesiones Focales/metabolismo , Expresión Génica , Humanos , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 14 de la Matriz/metabolismo , Neoplasias/genética , Transporte de Proteínas , Transfección , Familia-src Quinasas/genética
11.
Mol Syst Biol ; 9: 636, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23340842

RESUMEN

Escherichia coli were genetically modified to enable programmed motility, sensing, and actuation based on the density of features on nearby surfaces. Then, based on calculated feature density, these cells expressed marker proteins to indicate phenotypic response. Specifically, site-specific synthesis of bacterial quorum sensing autoinducer-2 (AI-2) is used to initiate and recruit motile cells. In our model system, we rewired E. coli's AI-2 signaling pathway to direct bacteria to a squamous cancer cell line of head and neck (SCCHN), where they initiate synthesis of a reporter (drug surrogate) based on a threshold density of epidermal growth factor receptor (EGFR). This represents a new type of controller for targeted drug delivery as actuation (synthesis and delivery) depends on a receptor density marking the diseased cell. The ability to survey local surfaces and initiate gene expression based on feature density represents a new area-based switch in synthetic biology that will find use beyond the proposed cancer model here.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Receptores ErbB/metabolismo , Escherichia coli/genética , Neoplasias de Cabeza y Cuello/genética , Homoserina/análogos & derivados , Lactonas/metabolismo , Línea Celular Tumoral , Receptores ErbB/genética , Escherichia coli/metabolismo , Regulación de la Expresión Génica , Ingeniería Genética/métodos , Neoplasias de Cabeza y Cuello/patología , Homoserina/genética , Homoserina/metabolismo , Humanos , Nanotecnología , Percepción de Quorum
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...