Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Am Soc Nephrol ; 33(6): 1120-1136, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35292439

RESUMO

BACKGROUND: Glomerular endothelial cell (GEnC) fenestrations are recognized as an essential component of the glomerular filtration barrier, yet little is known about how they are regulated and their role in disease. METHODS: We comprehensively characterized GEnC fenestral and functional renal filtration changes including measurement of glomerular Kf and GFR in diabetic mice (BTBR ob-/ob- ). We also examined and compared human samples. We evaluated Eps homology domain protein-3 (EHD3) and its association with GEnC fenestrations in diabetes in disease samples and further explored its role as a potential regulator of fenestrations in an in vitro model of fenestration formation using b.End5 cells. RESULTS: Loss of GEnC fenestration density was associated with decreased filtration function in diabetic nephropathy. We identified increased diaphragmed fenestrations in diabetes, which are posited to increase resistance to filtration and further contribute to decreased GFR. We identified decreased glomerular EHD3 expression in diabetes, which was significantly correlated with decreased fenestration density. Reduced fenestrations in EHD3 knockdown b.End5 cells in vitro further suggested a mechanistic role for EHD3 in fenestration formation. CONCLUSIONS: This study demonstrates the critical role of GEnC fenestrations in renal filtration function and suggests EHD3 may be a key regulator, loss of which may contribute to declining glomerular filtration function through aberrant GEnC fenestration regulation. This points to EHD3 as a novel therapeutic target to restore filtration function in disease.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Fenômenos Fisiológicos do Sistema Urinário , Animais , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Células Endoteliais/metabolismo , Glomérulos Renais/metabolismo , Camundongos
2.
Circ Res ; 126(6): 767-783, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32078435

RESUMO

RATIONALE: Central nervous system has low vascular permeability by organizing tight junction (TJ) and limiting endothelial transcytosis. While TJ has long been considered to be responsible for vascular barrier in central nervous system, suppressed transcytosis in endothelial cells is now emerging as a complementary mechanism. Whether transcytosis regulation is independent of TJ and its dysregulation dominantly causes diseases associated with edema remain elusive. Dll4 signaling is important for various vascular contexts, but its role in the maintenance of vascular barrier in central nervous system remains unknown. OBJECTIVE: To find a TJ-independent regulatory mechanism selective for transcytosis and identify its dysregulation as a cause of pathological leakage. METHODS AND RESULTS: We studied transcytosis in the adult mouse retina with low vascular permeability and employed a hypertension-induced retinal edema model for its pathological implication. Both antibody-based and genetic inactivation of Dll4 or Notch1 induce hyperpermeability by increasing transcytosis without junctional destabilization in arterial endothelial cells, leading to nonhemorrhagic leakage predominantly in the superficial retinal layer. Endothelial Sox17 deletion represses Dll4 in retinal arteries, phenocopying Dll4 blocking-driven vascular leakage. Ang II (angiotensin II)-induced hypertension represses arterial Sox17 and Dll4, followed by transcytosis-driven retinal edema, which is rescued by a gain of Notch activity. Transcriptomic profiling of retinal endothelial cells suggests that Dll4 blocking activates SREBP1 (sterol regulatory element-binding protein 1)-mediated lipogenic transcription and enriches gene sets favorable for caveolae formation. Profiling also predicts the activation of VEGF (vascular endothelial growth factor) signaling by Dll4 blockade. Inhibition of SREBP1 or VEGF-VEGFR2 (VEGF receptor 2) signaling attenuates both Dll4 blockade-driven and hypertension-induced retinal leakage. CONCLUSIONS: In the retina, Sox17-Dll4-SREBP1 signaling axis controls transcytosis independently of TJ in superficial arteries among heterogeneous regulations for the whole vessels. Uncontrolled transcytosis via dysregulated Dll4 underlies pathological leakage in hypertensive retina and could be a therapeutic target for treating hypertension-associated retinal edema.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Barreira Hematorretiniana/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Retinopatia Hipertensiva/metabolismo , Transcitose , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Artérias/metabolismo , Proteínas de Ligação ao Cálcio/genética , Cavéolas/metabolismo , Células Endoteliais/metabolismo , Proteínas HMGB/metabolismo , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Receptor Notch1/genética , Receptor Notch1/metabolismo , Fatores de Transcrição SOXF/metabolismo , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Junções Íntimas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
3.
Dev Biol ; 464(2): 137-144, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32565279

RESUMO

Tissue macrophages, which are ubiquitously present innate immune cells, play versatile roles in development and organogenesis. During development, macrophages prune transient or unnecessary synapses in neuronal development, and prune blood vessels in vascular development, facilitating appropriate tissue remodeling. In the present study, we identified that macrophages contributed to the development of pupillary morphology. Csf1op/op mutant mice, in which ocular macrophages are nearly absent, exhibited abnormal pupillary edges, with abnormal protrusions of excess iris tissue into the pupillary space. Macrophages located near the pupillary edge engulfed pigmented debris, which likely consisted of unnecessary iris protrusions that emerge during smoothening of the pupillary edge. Indeed, pupillary edge macrophages phenotypically possessed some features of M2 macrophages, consistent with robust tissue engulfment and remodeling activities. Interestingly, protruding irises in Csf1op/op mice were only detected in gaps between regressing blood vessels. Taken together, our findings uncovered a new role for ocular macrophages, demonstrating that this cell population is important for iris pruning during development.


Assuntos
Macrófagos/metabolismo , Pupila , Animais , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/citologia , Camundongos , Camundongos Mutantes
4.
Diabetologia ; 61(11): 2422-2432, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30094465

RESUMO

AIMS/HYPOTHESIS: Diabetic retinopathy is increasing in prevalence worldwide and is fast becoming a global epidemic and a leading cause of visual loss. Current therapies are limited, and the development of effective treatments for diabetic retinopathy requires a greater in-depth knowledge of disease progression and suitable modelling of diabetic retinopathy in animals. The aim of this study was to assess the early pathological changes in retinal morphology and neuronal, inflammatory and vascular features consistent with diabetic retinopathy in the ob/ob mouse model of type 2 diabetes, to investigate whether features similar to those in human diabetic retinopathy were present. METHODS: Male and female wild-type (+/+), heterozygous (+/-) and homozygous (-/-) BTBR ob/ob mice were examined at 6, 10, 15 and 20 weeks of age. Animals were weighed and blood glucose was measured. TUNEL and brain-specific homeobox/POU domain protein 3A (BRN3A) markers were used to examine retinal ganglion cells. We used immunostaining (collagen IV and platelet endothelial cell adhesion molecule [PECAM]/CD31) to reveal retinal vessel degeneration. Spectral domain optical coherence tomography was used to reveal changes in the thickness and structure of the retinal layer. Vitreous fluorophotometry was used to investigate vascular permeability. A-waves, b-waves and oscillatory potentials were measured under photopic and scotopic conditions. Concanavalin A leucostasis and immunostaining with glial fibrillary acidic protein (GFAP) and ionised calcium-binding adapter molecule 1 (IBA-1) identified differences in inflammatory status. Paraffin sections and transmission electron microscopy were used to reveal changes in the thickness and structure of the retinal layer. RESULTS: Following the development of obesity and hyperglycaemia in 2-week-old and 3-week-old ob-/ob- mice, respectively (p < 0.001), early functional deficits (p < 0.001) and thinning of the inner retina (p < 0.001) were identified. Glial activation, leucostasis (p < 0.05) and a shift in microglia/macrophage phenotype were observed before microvascular degeneration (p < 0.05) and elevated vascular permeability occurred (p < 0.05). CONCLUSIONS/INTERPRETATION: The present characterisation of the development of diabetic retinopathy in the ob/ob mouse represents a platform that will enable the development of new therapies, particularly for the early stages of disease.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Retina/metabolismo , Retina/patologia , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/genética , Modelos Animais de Doenças , Feminino , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Obesidade/metabolismo , Obesidade/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia
5.
Am J Pathol ; 185(9): 2534-49, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26188133

RESUMO

Choroidal neovascularization (CNV) is a defining feature of wet age-related macular degeneration. We examined the functional role of CCR3 in the development of CNV in mice and primates. CCR3 was associated with spontaneous CNV lesions in the newly described JR5558 mice, whereas CCR3 ligands localized to CNV-associated macrophages and the retinal pigment epithelium/choroid complex. Intravitreal injection of neutralizing antibodies against vascular endothelial growth factor receptor 2, CCR3, CC chemokine ligand 11/eotaxin-1, and CC chemokine ligand 24/eotaxin-2 all reduced CNV area and lesion number in these mice. Systemic administration of the CCR3 antagonists GW766994X and GW782415X reduced spontaneous CNV in JR5558 mice and laser-induced CNV in mouse and primate models in a dose-dependent fashion. Combination treatment with antivascular endothelial growth factor receptor 2 antibody and GW766994X yielded additive reductions in CNV area and hyperpermeability in mice. Interestingly, topical GW766994X and intravitreal anti-CCR3 antibody yielded strong systemic effects, reducing CNV in the untreated, contralateral eye. Contrarily, ocular administration of GW782415X in primates failed to substantially elevate plasma drug levels or to reduce the development of grade IV CNV lesions. These findings suggest that CCR3 signaling may be an attractive therapeutic target for CNV, utilizing a pathway that is at least partly distinct from that of vascular endothelial growth factor receptor. The findings also demonstrate that systemic exposure to CCR3 antagonists may be crucial for CNV-targeted activity.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Permeabilidade Capilar/efeitos dos fármacos , Neovascularização de Coroide/tratamento farmacológico , Receptores CCR3/antagonistas & inibidores , Degeneração Macular Exsudativa/tratamento farmacológico , Animais , Permeabilidade Capilar/imunologia , Corioide/patologia , Neovascularização de Coroide/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Fator A de Crescimento do Endotélio Vascular/metabolismo , Degeneração Macular Exsudativa/patologia
6.
Proc Natl Acad Sci U S A ; 109(9): E553-62, 2012 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-22323600

RESUMO

The generation of diverse neuronal types and subtypes from multipotent progenitors during development is crucial for assembling functional neural circuits in the adult central nervous system. During mouse retinogenesis, early retinal progenitors give rise to several cell types, including ganglion, amacrine, horizontal, cone, and rod cells. It is unknown at present how each of these fates is selected from the multiple neuronal fates available to the early progenitor. By using a combination of bioinformatic, genetic, and biochemical approaches, we investigated the mechanism by which Foxn4 selects the amacrine and horizontal cell fates from multipotential retinal progenitors. These studies indicate that Foxn4 has an intrinsic activity to suppress the alternative photoreceptor cell fates of early retinal progenitors by selectively activating Dll4-Notch signaling. Gene expression and conditional ablation analyses reveal that Dll4 is directly activated by Foxn4 via phylogenetically conserved enhancers and that Dll4 can partly mediate the Foxn4 function by serving as a major Notch ligand to expand the progenitor pool and limit photoreceptor production. Our data together define a Foxn4-mediated molecular and signaling pathway that underlies the suppression of alternative cell fates of early retinal progenitors.


Assuntos
Proteínas do Olho/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Células Fotorreceptoras de Vertebrados/citologia , Receptores Notch/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Células Amácrinas/citologia , Animais , Proteínas de Ligação ao Cálcio , Diferenciação Celular/genética , Linhagem da Célula , Sequência Conservada , Elementos Facilitadores Genéticos , Proteínas do Olho/genética , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/fisiologia , Retina/embriologia , Retina/crescimento & desenvolvimento , Células Fotorreceptoras Retinianas Bastonetes/citologia , Transdução de Sinais/fisiologia
7.
Am J Pathol ; 182(4): 1379-90, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23416159

RESUMO

Vascular endothelial growth factor A (VEGF-A) is a validated therapeutic target in several angiogenic- and vascular permeability-related pathological conditions, including certain cancers and potentially blinding diseases, such as age-related macular degeneration and diabetic retinopathy. We and others have shown that VEGF-A also plays an important role in neuronal development and neuroprotection, including in the neural retina. Antagonism of VEGF-A function might therefore present a risk to neuronal survival as a significant adverse effect. Herein, we demonstrate that VEGF-A acts directly on retinal ganglion cells (RGCs) to promote survival. VEGF receptor-2 signaling via the phosphoinositide-3-kinase/Akt pathway was required for the survival response in isolated RGCs. These results were confirmed in animal models of staurosporine-induced RGC death and experimental hypertensive glaucoma. Importantly, we observed that VEGF-A blockade significantly exacerbated neuronal cell death in the hypertensive glaucoma model. Our findings highlight the need to better define the risks associated with use of VEGF-A antagonists in the ocular setting.


Assuntos
Glaucoma/tratamento farmacológico , Glaucoma/patologia , Fármacos Neuroprotetores/uso terapêutico , Retina/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Citoproteção/efeitos dos fármacos , Modelos Animais de Doenças , Glaucoma/enzimologia , Neuropilinas/metabolismo , Fármacos Neuroprotetores/farmacologia , Testes de Neutralização , Hipertensão Ocular/tratamento farmacológico , Hipertensão Ocular/enzimologia , Hipertensão Ocular/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Retina/efeitos dos fármacos , Retina/enzimologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/enzimologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Transdução de Sinais/efeitos dos fármacos , Testes de Toxicidade Aguda , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
J Exp Med ; 198(3): 483-9, 2003 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-12900522

RESUMO

Hypoxia-induced VEGF governs both physiological retinal vascular development and pathological retinal neovascularization. In the current paper, the mechanisms of physiological and pathological neovascularization are compared and contrasted. During pathological neovascularization, both the absolute and relative expression levels for VEGF164 increased to a greater degree than during physiological neovascularization. Furthermore, extensive leukocyte adhesion was observed at the leading edge of pathological, but not physiological, neovascularization. When a VEGF164-specific neutralizing aptamer was administered, it potently suppressed the leukocyte adhesion and pathological neovascularization, whereas it had little or no effect on physiological neovascularization. In parallel experiments, genetically altered VEGF164-deficient (VEGF120/188) mice exhibited no difference in physiological neovascularization when compared with wild-type (VEGF+/+) controls. In contrast, administration of a VEGFR-1/Fc fusion protein, which blocks all VEGF isoforms, led to significant suppression of both pathological and physiological neovascularization. In addition, the targeted inactivation of monocyte lineage cells with clodronate-liposomes led to the suppression of pathological neovascularization. Conversely, the blockade of T lymphocyte-mediated immune responses with an anti-CD2 antibody exacerbated pathological neovascularization. These data highlight important molecular and cellular differences between physiological and pathological retinal neovascularization. During pathological neovascularization, VEGF164 selectively induces inflammation and cellular immunity. These processes provide positive and negative angiogenic regulation, respectively. Together, new therapeutic approaches for selectively targeting pathological, but not physiological, retinal neovascularization are outlined.


Assuntos
Fatores de Crescimento Endotelial/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Isquemia/metabolismo , Linfocinas/metabolismo , Retina/metabolismo , Neovascularização Retiniana/metabolismo , Animais , Animais Recém-Nascidos , Adesão Celular/fisiologia , Modelos Animais de Doenças , Fatores de Crescimento Endotelial/genética , Fatores de Crescimento Endotelial/imunologia , Feminino , Inflamação , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Isquemia/patologia , Leucócitos/metabolismo , Linfocinas/genética , Linfocinas/imunologia , Masculino , Camundongos , Camundongos Knockout , Monócitos/citologia , Monócitos/fisiologia , Neovascularização Fisiológica , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Ratos , Receptores de Interleucina-2/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Retina/patologia , Neovascularização Retiniana/patologia , Vasos Retinianos , Linfócitos T/fisiologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
9.
Cells ; 9(6)2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32503129

RESUMO

Fenestrae are transcellular plasma membrane pores that mediate blood-tissue exchange in specialised vascular endothelia. The composition and biogenesis of the fenestra remain enigmatic. We isolated and characterised the protein composition of large patches of fenestrated plasma membrane, termed sieve plates. Loss-of-function experiments demonstrated that two components of the sieve plate, moesin and annexin II, were positive and negative regulators of fenestra formation, respectively. Biochemical analyses showed that moesin is involved in the formation of an actin-fodrin submembrane cytoskeleton that was essential for fenestra formation. The link between the fodrin cytoskeleton and the plasma membrane involved the fenestral pore protein PV-1 and Na,K-ATPase, which is a key regulator of signalling during fenestra formation both in vitro and in vivo. These findings provide a conceptual framework for fenestra biogenesis, linking the dynamic changes in plasma membrane remodelling to the formation of a submembrane cytoskeletal signalling complex.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Células Endoteliais/metabolismo , Proteínas dos Microfilamentos/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Anexina A2/metabolismo , Linhagem Celular , Membrana Celular/ultraestrutura , Citoesqueleto/ultraestrutura , Células Endoteliais/ultraestrutura , Masculino , Camundongos , Ouabaína/farmacologia , Ratos Sprague-Dawley
10.
Biochem Soc Trans ; 37(Pt 6): 1201-6, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19909247

RESUMO

The longer splice isoforms of VEGF (vascular endothelial growth factor)-A, including VEGF(164(165)), contain a highly basic HBD (heparin-binding domain). This domain allows these isoforms to interact with and localize to the HS (heparan sulfate)-rich extracellular matrix, and bind to the co-receptor Nrp-1 (neuropilin-1). Heparin-binding VEGF-A isoforms are critical for survival: mice engineered to express exclusively the non-heparin-binding VEGF(120) have diminished vascular branching during embryonic development and die from postnatal angiogenesis defects shortly after birth. Although it is thought that the HBD contributes to the diverse functions of VEGF-A in both physiological and pathological processes, little is known about the molecular features within this domain that enable these functions. In the present paper, we discuss the roles of the VEGF HBD in normal and disease conditions, with a particular focus on the VEGF(164(165)) isoform.


Assuntos
Heparina/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Relação Estrutura-Atividade , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/metabolismo , Processamento Alternativo , Animais , Oftalmopatias/metabolismo , Oftalmopatias/patologia , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Camundongos , Modelos Moleculares , Neoplasias/metabolismo , Neovascularização Patológica , Ligação Proteica , Conformação Proteica , Isoformas de Proteínas/genética , Estrutura Terciária de Proteína , Fator A de Crescimento do Endotélio Vascular/genética
11.
J Cell Biol ; 157(7): 1161-73, 2002 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-12070132

RESUMO

Characterization of mammalian NSF (G274E) and Drosophila NSF (comatose) mutants revealed an evolutionarily conserved NSF activity distinct from ATPase-dependent SNARE disassembly that was essential for Golgi membrane fusion. Analysis of mammalian NSF function during cell-free assembly of Golgi cisternae from mitotic Golgi fragments revealed that NSF disassembles Golgi SNAREs during mitotic Golgi fragmentation. A subsequent ATPase-independent NSF activity restricted to the reassembly phase is essential for membrane fusion. NSF/alpha-SNAP catalyze the binding of GATE-16 to GOS-28, a Golgi v-SNARE, in a manner that requires ATP but not ATP hydrolysis. GATE-16 is essential for NSF-driven Golgi reassembly and precludes GOS-28 from binding to its cognate t-SNARE, syntaxin-5. We suggest that this occurs at the inception of Golgi reassembly to protect the v-SNARE and regulate SNARE function.


Assuntos
Adenosina Trifosfatases/fisiologia , Proteínas de Transporte/fisiologia , Complexo de Golgi/fisiologia , Membranas Intracelulares/fisiologia , Fusão de Membrana/fisiologia , Proteínas de Membrana/metabolismo , Proteínas de Transporte Vesicular , Adenosina Trifosfatases/metabolismo , Animais , Transporte Biológico Ativo , Células CHO , Células Cultivadas , Cricetinae , Drosophila , Evolução Molecular , Complexo de Golgi/metabolismo , Proteínas de Membrana/genética , Mitose , Mutação , Proteínas Sensíveis a N-Etilmaleimida , Proteínas Recombinantes de Fusão/metabolismo , Proteínas SNARE
12.
Nat Rev Drug Discov ; 5(2): 123-32, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16518379

RESUMO

Aptamers are oligonucleotide ligands that are selected for high-affinity binding to molecular targets. Pegaptanib sodium (Macugen; Eyetech Pharmaceuticals/Pfizer) is an RNA aptamer directed against vascular endothelial growth factor (VEGF)-165, the VEGF isoform primarily responsible for pathological ocular neovascularization and vascular permeability. After nearly a decade of preclinical development to optimize and characterize its biological effects, pegaptanib was shown in clinical trials to be effective in treating choroidal neovascularization associated with age-related macular degeneration. Pegaptanib therefore has the notable distinction of being the first aptamer therapeutic approved for use in humans, paving the way for future aptamer applications.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Aptâmeros de Nucleotídeos/uso terapêutico , Oftalmopatias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese/farmacocinética , Inibidores da Angiogênese/farmacologia , Animais , Aptâmeros de Nucleotídeos/farmacocinética , Aptâmeros de Nucleotídeos/farmacologia , Ensaios Clínicos como Assunto , Retinopatia Diabética/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Olho/irrigação sanguínea , Humanos
13.
Invest Ophthalmol Vis Sci ; 49(2): 662-70, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18235012

RESUMO

PURPOSE: To investigate the effect of the combined treatment of photodynamic therapy and specific VEGF165 inhibition with pegaptanib sodium (Macugen; Eyetech Pharmaceuticals, Lexington, MA) on ocular neovascularization. METHODS: Photodynamic therapy's (PDT's) effects on the integrity of pegaptanib sodium were analyzed by HPLC, a VEGF165-binding assay, and a VEGF165-induced tissue factor gene expression assay. The effects of mono- or combined treatment on vessel growth and regression were determined in a murine corneal neovascularization model. The effects of combined treatment on vessel growth were also determined in a murine choroidal neovascularization model. RESULTS: PDT did not affect the chemical composition of pegaptanib sodium nor the efficacy of pegaptanib sodium in the inhibition of VEGF165 binding to Flt-1 and VEGF165-induced gene expression. In an animal model of effects on existing ocular neovascular lesions (corneal neovascularization), PDT monotherapy yielded an initial regression of these vessels, but there followed a rapid regrowth. In contrast, pegaptanib sodium monotherapy yielded little regression but potently abrogated further vessel growth. The combination of pegaptanib sodium and PDT resulted in the regression of the neovascular lesions, as observed with PDT alone, but also prevented significant vessel regrowth, leading to a significantly greater reduction in lesion size than did each monotherapy. In addition, there was a significantly greater effect of the combination of pegaptanib sodium and PDT on lesion size in choroidal neovascularization than with each monotherapy. Pretreatment with pegaptanib sodium appeared to decrease the efficacy of PDT-induced vessel regression in corneal neovascularization, and as such the enhanced efficacy over monotherapy when the agents were delivered simultaneously was not observed. CONCLUSIONS: Although the combined simultaneous treatment of ocular neovascularization with PDT and pegaptanib sodium may provide a more effective approach for the regression and overall treatment of CNV associated with AMD, the order of addition of these treatments may play a role in achieving optimal efficacy.


Assuntos
Aptâmeros de Nucleotídeos/uso terapêutico , Neovascularização de Coroide/tratamento farmacológico , Neovascularização da Córnea/tratamento farmacológico , Modelos Animais de Doenças , Fotoquimioterapia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Cromatografia Líquida de Alta Pressão , Córnea/irrigação sanguínea , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Quimioterapia Combinada , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/uso terapêutico , RNA Mensageiro/metabolismo , Tromboplastina/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Verteporfina
14.
J Exp Med ; 215(3): 963-983, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29444818

RESUMO

High-grade glioma (HGG) is highly angiogenic, but antiangiogenic therapy has transient clinical benefit in only a fraction of patients. Vascular regulators of these heterogeneous responses remain undetermined. We found up-regulation of Sox7 and down-regulation of Sox17 in tumor endothelial cells (tECs) in mouse HGG. Sox7 deletion suppressed VEGFR2 expression, vascular abnormality, hypoxia-driven invasion, regulatory T cell infiltration, and tumor growth. Conversely, Sox17 deletion exacerbated these phenotypes by up-regulating Sox7 in tECs. Anti-VEGFR2 antibody treatment delayed tumor growth by normalizing Sox17-deficient abnormal vessels with high Sox7 levels but promoted it by regressing Sox7-deficient vessels, recapitulating variable therapeutic responses to antiangiogenic therapy in HGG patients. Our findings establish that Sox7 promotes tumor growth via vessel abnormalization, and its level determines the therapeutic outcome of VEGFR2 inhibition in HGG. In 189 HGG patients, Sox7 expression was heterogeneous in tumor vessels, and high Sox7 levels correlated with poor survival, early recurrence, and impaired vascular function, emphasizing the clinical relevance of Sox7 in HGG.


Assuntos
Vasos Sanguíneos/anormalidades , Glioma/metabolismo , Glioma/patologia , Fatores de Transcrição SOXF/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Proteínas de Ligação ao Cálcio , Regulação para Baixo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Deleção de Genes , Glioma/irrigação sanguínea , Glioma/imunologia , Humanos , Imunidade , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Gradação de Tumores , Prognóstico , Regulação para Cima , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
15.
Invest Ophthalmol Vis Sci ; 48(4): 1767-72, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17389510

RESUMO

PURPOSE: Photodynamic therapy (PDT) and the administration of compounds acting against vascular endothelial growth factor (anti-VEGF) are approved for the treatment of choroidal neovascularization (CNV) secondary to age-related macular degeneration (AMD). Experimental evidence that the combined use of both treatment options may improve therapeutic outcome is presented. METHODS: Fertilized chick eggs were incubated until day 12 of embryo development (EDD12) and were treated by PDT using two different photosensitizing agents (liposomal formulation of BPD-MA; m-THPP encapsulated in polymeric nanoparticles) and were visualized using an epifluorescence microscope. Vascular occlusion of the treated zones of the chorioallantoic membrane (CAM) was assessed by fluorescence angiography 24 and 48 hours after treatment. Alternatively, PDT-treated areas were exposed to a soluble VEGF receptor antagonist (sFlt-1) 6 hours after treatment and were analyzed. RESULTS: Vascular occlusion in the PDT-treated areas was observed with both photosensitizers 24 hours after treatment. Reperfusion of preexisting blood vessels and first signs of revascularization were visible 48 hours after PDT. Topical administration of sFlt-1 to the treated areas augmented occlusion and limited subsequent angiogenesis in a dose-dependent manner. CONCLUSIONS: The combined use of PDT and of agents targeting angiogenic cytokines may synergistically improve therapeutic outcome after combined treatment in patients with CNV secondary to AMD.


Assuntos
Membrana Corioalantoide/efeitos dos fármacos , Degeneração Macular/tratamento farmacológico , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Modelos Animais de Doenças , Sinergismo Farmacológico , Angiofluoresceinografia , Humanos , Lipossomos , Microscopia de Fluorescência , Nanopartículas , Neovascularização Fisiológica/efeitos dos fármacos , Porfirinas/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Verteporfina
16.
Invest Ophthalmol Vis Sci ; 48(3): 1212-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17325165

RESUMO

PURPOSE: Retinal ganglion cell (RGC) loss occurs in response to increased intraocular pressure (IOP) and/or retinal ischemia in glaucoma and leads to impairment of vision. This study was undertaken to test the efficacy of erythropoietin (EPO) in providing neuroprotection to RGCs in vivo. METHODS: The neuroprotective effects of EPO were studied in the DBA/2J mouse model of glaucoma. Mice were intraperitoneally injected with control substances or various doses of EPO, starting at the age of 6 months and continuing for an additional 2, 4, or 6 months. RGCs were labeled retrogradely by a gold tracer. IOP was measured with a microelectric-mechanical system, and EPO receptor (EPOR) expression was detected by immunohistochemistry. Axonal death in the optic nerve was quantified by para-phenylenediamine staining, and a complete blood count system was used to measure the number of erythrocytes. RESULTS: In DBA/2J mice, the average number of viable RGCs significantly decreased from 4 months to 10 months, with an inverse correlation between the number of dead optic nerve axons and viable RGCs. Treatment with EPO at doses of 3000, 6000, and 12,000 U/kg body weight per week all prevented significant RGC loss, compared with untreated DBA/2J control animals. EPO effects were similar to those of memantine, a known neuroprotective agent. IOP, in contrast, was unchanged by both EPO and memantine. Finally, EPOR was expressed in the RGC layer in both DBA/2J and C57BL/6J mice. CONCLUSIONS: EPO promoted RGC survival in DBA/2J glaucomatous mice without affecting IOP. These results suggest that EPO may be a potential therapeutic neuroprotectant in glaucoma.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Eritropoetina/farmacologia , Glaucoma/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Doenças do Nervo Óptico/prevenção & controle , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Axônios/efeitos dos fármacos , Modelos Animais de Doenças , Técnica Indireta de Fluorescência para Anticorpo , Glaucoma/metabolismo , Glaucoma/patologia , Pressão Intraocular/efeitos dos fármacos , Memantina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Microscopia de Fluorescência , Nervo Óptico/efeitos dos fármacos , Doenças do Nervo Óptico/metabolismo , Doenças do Nervo Óptico/patologia , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia
17.
Mol Vis ; 12: 1243-9, 2006 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-17110907

RESUMO

PURPOSE: The wet form of age-related macular degeneration (AMD) occurs as a consequence of abnormal blood vessel growth from the choroid into the retina. Pathological angiogenesis during tumor growth and ocular disease has been associated with specific exposure of cryptic extracellular matrix epitopes. We investigated the presence of cryptic collagen IV epitopes in a murine model of choroidal neovascularization (CNV), and tested the effect on blood vessel growth of H8, a humanized antibody directed against a cryptic collagen type IV epitope. METHODS: To induce experimental CNV in adult C57BL/6 mice, Bruch's membrane was ruptured using a diode laser. Subsequently, mice were treated with daily intraperitoneal (i.p.) injections of either H8 (10 mg/kg or 30 mg/kg) or an isotype-matched antibody control. Two weeks postinjection, choroidal flat mounts were immunostained with the blood vessel marker platelet/endothelial cell adhesion molecule-1 (PECAM-1) and H8. CNV was visualized using fluorescence microscopy and the CNV lesion area measured using Open Lab software. RESULTS: Collagen type IV and the cryptic epitope were observed at the site of laser-induced lesions. Staining with H8 was first observed three days post injury, two days after MMP2 expression in CNV lesions, becoming most intense five days following laser injury and extending beyond the area of neovascularization. At 14 days post injury, H8 staining was reduced in intensity, colocalized with the area of CNV, and was nearly absent from the underlying choroidal vessels. In addition, mice treated with H8 had a significant dose-dependent decrease in the area of CNV as compared to isotype-matched antibody controls. CONCLUSIONS: Results suggest that exposure of cryptic collagen type IV epitopes is associated with the incidence of CNV and that the humanized antibody H8 may provide a new treatment for CNV.


Assuntos
Anticorpos/farmacologia , Neovascularização de Coroide/prevenção & controle , Colágeno Tipo IV/imunologia , Epitopos/imunologia , Lasers , Animais , Anticorpos/imunologia , Neovascularização de Coroide/etiologia , Neovascularização de Coroide/imunologia , Epitopos/metabolismo , Matriz Extracelular/imunologia , Olho/efeitos dos fármacos , Olho/metabolismo , Traumatismos Oculares/imunologia , Traumatismos Oculares/metabolismo , Temperatura Alta , Imuno-Histoquímica/métodos , Masculino , Metaloproteinase 2 da Matriz/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Desnaturação Proteica , Lesões por Radiação/imunologia , Lesões por Radiação/metabolismo , Coloração e Rotulagem , Fatores de Tempo
18.
Mol Vis ; 12: 532-80, 2006 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-16735995

RESUMO

The Third International Symposium on Retinopathy of Prematurity (ROP) was convened with the aim of cross fertilizing the horizons of basic and clinical scientists with an interest in the pathogenesis and management of infants with ROP. Ten speakers in the clinical sciences and ten speakers in the basic sciences were recruited on the basis of their research to provide state of the art talks. The meeting was held November 9, 2003 immediately prior to the American Academy of Ophthalmology meeting; scholarships were provided for outreach to developing countries and young investigators. This review contain the summaries of the 20 platform presentations prepared by the authors and the abstracts of presented posters. Each author was asked to encapsulate the current state of understanding, identify areas of controversy, and make recommendations for future research. The basic science presentations included insights into the development of the human retinal vasculature, animal models for ROP, growth factors that affect normal development and ROP, and promising new therapeutic approaches to treating ROP like VEGF targeting, inhibition of proteases, stem cells, ribozymes to silence genes, and gene therapy to deliver antiangiogenic agents. The clinical presentations included new insights into oxygen management, updates on the CRYO-ROP and ETROP studies, visual function in childhood following ROP, the neural retina in ROP, screening for ROP, management of stage 3 and 4 ROP, ROP in the third world, and the complications of ROP in adult life. The meeting resulted in a penetrating exchange between clinicians and basic scientists, which provided great insights for conference attendees. The effect of preterm delivery on the normal cross-talk of neuroretinal and retinal vascular development is a fertile ground for discovering new understanding of the processes involved both in normal development and in retinal neovascular disorders. The meeting also suggested promising potential therapeutic interventions on the horizon for ROP.


Assuntos
Retinopatia da Prematuridade/etiologia , Retinopatia da Prematuridade/terapia , Humanos , Recém-Nascido
19.
EMBO Mol Med ; 8(11): 1265-1288, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27742718

RESUMO

Anti-angiogenic therapies using biological molecules that neutralize vascular endothelial growth factor-A (VEGF-A) have revolutionized treatment of retinal vascular diseases including age-related macular degeneration (AMD). This study reports preclinical assessment of a strategy to enhance anti-VEGF-A monotherapy efficacy by targeting both VEGF-A and angiopoietin-2 (ANG-2), a factor strongly upregulated in vitreous fluids of patients with retinal vascular disease and exerting some of its activities in concert with VEGF-A. Simultaneous VEGF-A and ANG-2 inhibition was found to reduce vessel lesion number, permeability, retinal edema, and neuron loss more effectively than either agent alone in a spontaneous choroidal neovascularization (CNV) model. We describe the generation of a bispecific domain-exchanged (crossed) monoclonal antibody (CrossMAb; RG7716) capable of binding, neutralizing, and depleting VEGF-A and ANG-2. RG7716 showed greater efficacy than anti-VEGF-A alone in a non-human primate laser-induced CNV model after intravitreal delivery. Modification of RG7716's FcRn and FcγR binding sites disabled the antibodies' Fc-mediated effector functions. This resulted in increased systemic, but not ocular, clearance. These properties make RG7716 a potential next-generation therapy for neovascular indications of the eye.


Assuntos
Angiopoietina-2/antagonistas & inibidores , Anticorpos Monoclonais/administração & dosagem , Oftalmopatias/tratamento farmacológico , Fatores Imunológicos/administração & dosagem , Neovascularização Patológica/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/farmacologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Oftalmopatias/patologia , Fatores Imunológicos/isolamento & purificação , Fatores Imunológicos/farmacologia , Macaca fascicularis , Resultado do Tratamento
20.
Invest Ophthalmol Vis Sci ; 45(2): 368-74, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14744874

RESUMO

PURPOSE: Vascular endothelial growth factor (VEGF) induces angiogenesis and vascular permeability and is thought to be operative in several ocular vascular diseases. The VEGF isoforms are highly conserved among species; however, little is known about their differential biological functions in adult tissue. In the current study, the inflammatory potential of two prevalent VEGF isoform splice variants, VEGF(120(121)) and VEGF(164(165)), was studied in the transparent and avascular adult mouse cornea. METHODS: Controlled-release pellets containing equimolar amounts of VEGF(120) and VEGF(164) were implanted in corneas. The mechanisms underlying this differential response of VEGF isoforms were explored. The response of VEGF in cultured endothelial cells was determined by Western blot analysis. The response of VEGF isoforms in leukocytes was also investigated. RESULTS: VEGF(164) was found to be significantly more potent at inducing inflammation. In vivo blockade of VEGF receptor (VEGFR)-1 significantly suppressed VEGF(164)-induced corneal inflammation. In vitro, VEGF(165) more potently stimulated intracellular adhesion molecule (ICAM)-1 expression on endothelial cells, an effect that was mediated by VEGFR2. VEGF(164) was also more potent at inducing the chemotaxis of monocytes, an effect that was mediated by VEGFR1. In an immortalized human leukocyte cell line, VEGF(165) was found to induce tyrosine phosphorylation of VEGFR1 more efficiently. CONCLUSIONS: Taken together, these data identify VEGF(164(165)) as a proinflammatory isoform and identify multiple mechanisms underlying its proinflammatory biology.


Assuntos
Córnea/efeitos dos fármacos , Neovascularização da Córnea/induzido quimicamente , Endotélio Vascular/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Western Blotting , Técnicas de Cultura de Células , Quimiotaxia de Leucócito/efeitos dos fármacos , Córnea/patologia , Neovascularização da Córnea/patologia , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Células Jurkat/efeitos dos fármacos , Contagem de Leucócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Isoformas de Proteínas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA