Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Pediatr Ophthalmol Strabismus ; 59(4): 236-242, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34928772

RESUMO

PURPOSE: To review information pertaining to secondary glaucoma following infant lensectomy and provide evidence to support the mechanism responsible for this condition. METHODS: Reported risk factors and proposed mechanisms for infantile aphakic glaucoma are reviewed. Laboratory studies and clinical observations in affected patients with glaucoma are described. Evidence of postoperative anterior chamber fibrosis is reviewed and interpreted. RESULTS: Clinical evidence demonstrated the development of anterior chamber fibrosis following infant cataract surgery. Laboratory studies showed liberated lens epithelial cell transition to fibroblasts. CONCLUSIONS: The review and assessment of laboratory and clinical evidence support the proposal that infantile aphakic glaucoma is caused, in part, by postoperative anterior chamber fibroization related to lens cell dispersion, cytokine activation, and epithelial-mesenchymal transition with resultant filtration angle fibrosis and secondary loss of filtration function. [J Pediatr Ophthalmol Strabismus. 2022;59(4):236-242.].


Assuntos
Extração de Catarata , Catarata , Glaucoma , Hidroftalmia , Catarata/complicações , Extração de Catarata/efeitos adversos , Fibrose , Glaucoma/etiologia , Glaucoma/cirurgia , Humanos , Hidroftalmia/cirurgia , Lactente , Estudos Retrospectivos
2.
Exp Eye Res ; 184: 266-277, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31082363

RESUMO

Data from human dry and wet age-related macular degeneration (AMD) eyes support the hypothesis that constant 'tickover' of the alternative complement pathway results in chronic deposition of the complement membrane attack complex (MAC) on the choriocapillaris and the retinal pigment epithelium (RPE). Sub-lytic levels of MAC lead to cell signaling associated with tissue remodeling and the production of cytokines and inflammatory molecules. Lytic levels of MAC lead to cell death. CD59 is a naturally occurring inhibitor of the assembly of MAC. CD59 may thus be therapeutically efficacious against the pathophysiology of dry and wet AMD. The first gene therapy clinical trial for geographic atrophy - the advanced form of dry AMD has recently completed recruitment. This trial is studying the safety and tolerability of expressing CD59 from an adeno-associated virus (AAV) vector injected once into the vitreous. A second clinical trial assessing the efficacy of CD59 in wet AMD patients is also under way. Herein, the evidence for the role of MAC in the pathophysiology of dry as well as wet AMD and the scientific rationale underlying the use of AAV- delivered CD59 for the treatment of dry and wet AMD is discussed.


Assuntos
Antígenos CD59/uso terapêutico , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Terapia Genética , Atrofia Geográfica/fisiopatologia , Degeneração Macular Exsudativa/fisiopatologia , Animais , Antígenos CD59/genética , Ensaios Clínicos como Assunto , Dependovirus/genética , Vetores Genéticos , Atrofia Geográfica/terapia , Humanos , Injeções Intravítreas , Degeneração Macular Exsudativa/terapia
3.
Mol Ther Nucleic Acids ; 8: 77-89, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28918058

RESUMO

Non-viral gene delivery systems are being developed to address limitations of viral gene delivery. Many of these non-viral systems are modeled on the properties of viruses including cell surface binding, endocytosis, endosomal escape, and nuclear targeting. Most non-viral gene transfer systems exhibit little correlation between in vitro and in vivo efficiency, hampering a systematic approach to their development. Previously, we have described a 3.5 kDa peptide (peptide for ocular delivery [POD]) that targets cell surface sialic acid. When functionalized with polyethylene glycol (PEG) via a sulfhydryl group on the N-terminal cysteine of POD, PEG-POD could compact plasmid DNA, forming 120- to 180-nm homogeneous nanoparticles. PEG-POD enabled modest gene transfer and rescue of retinal degeneration in vivo. Systematic investigation of different stages of gene transfer by PEG-POD nanoparticles was hampered by their inability to deliver genes in vitro. Herein, we describe functionalization of POD with PEG using a reducible orthopyridyl disulfide bond. These reducible nanoparticles enabled gene transfer in vitro while retaining their in vivo gene transfer properties. These reducible PEG-POD nanoparticles were utilized to deliver human FLT1 to the retina in vivo, achieving a 50% reduction in choroidal neovascularization in a murine model of age-related macular degeneration.

4.
Exp Eye Res ; 145: 380-392, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26923800

RESUMO

There is currently no available method to efficiently deliver proteins across the plasma membrane of photoreceptor or retinal pigment epithelium (RPE) cells in vivo. Thus, current clinical application of recombinant proteins in ophthalmology is limited to the use of proteins that perform their biological function extracellularly. The ability to traverse biological membranes would enable the mobilization of a significantly larger number of proteins with previously well characterized properties. Nucleolin is abundantly present on the surface of rapidly dividing cells including cancer cells. Surprisingly, nucleolin is also present on the surface of photoreceptor cell bodies. Here we investigated whether nucleolin can be utilized as a gateway for the delivery of proteins into retinal cells following intravitreal injection. AS1411 is a G-quartet aptamer capable of targeting nucleolin. Subsequent to intravitreal injection, fluorescently labeled AS1411 localized to various retinal cell types including the photoreceptors and RPE. AS1411 linked to streptavidin (a ∼50 kDa protein) via a biotin bridge enabled the uptake of Streptavidin into photoreceptors and RPE. AS1411-Streptavidin conjugate applied topically to the cornea allowed for uptake of the conjugate into the nucleus and cytoplasm of corneal endothelial cells. Clinical relevance of AS1411 as a delivery vehicle was strongly indicated by demonstration of the presence of cell surface nucleolin on the photoreceptors, inner neurons and ganglion cells of human retina. These data support exploration of AS1411 as a means of delivering therapeutic proteins to diseased retina.


Assuntos
Aptâmeros de Nucleotídeos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Oligonucleotídeos/administração & dosagem , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas Recombinantes/administração & dosagem , Epitélio Pigmentado da Retina/metabolismo , Análise de Variância , Animais , Humanos , Imuno-Histoquímica , Injeções Intravítreas , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Oligonucleotídeos/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes/metabolismo , Nucleolina
5.
Exp Eye Res ; 140: 171-178, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26368850

RESUMO

Choroidal neovascularization (CNV) associated with the 'wet' form of age related macular degeneration (AMD) is one of the most common causes of central vision loss among the elderly. The 'wet' form of AMD is currently treated by intravitreal delivery of anti-VEGF agents. However, intravitreal injections are associated with complications and long-term inhibition of VEGF leads to macular atrophy. Thus, there is currently an unmet need for the development of therapies for CNV that target molecules other than VEGF. Here, we describe nucleolin as a novel target for the 'wet' form of AMD. Nucleolin was found on the surface of endothelial cells that migrate from the choroid into the subretinal space in the laser-induced model of 'wet' AMD. AS1411 is a previously described G-quartet oligonucleotide that has been shown to bind nucleolin. We found that AS1411 inhibited the formation of tubes by human umbilical vein endothelial cells (HUVECs) by approximately 27.4% in vitro. AS1411 co-localized with the site of laser induced CNV in vivo. Intravitreally injected AS1411 inhibited laser-induced CNV by 37.6% and attenuated infiltration of macrophages by 40.3%. Finally, topical application of AS1411 led to a 43.4% reduction in CNV. Our observations have potential implications for the development of therapies for CNV and specifically for the 'wet' form of AMD.


Assuntos
Aptâmeros de Nucleotídeos/administração & dosagem , Neovascularização de Coroide/prevenção & controle , Modelos Animais de Doenças , Oligodesoxirribonucleotídeos/administração & dosagem , Fosfoproteínas/antagonistas & inibidores , Proteínas de Ligação a RNA/antagonistas & inibidores , Degeneração Macular Exsudativa/prevenção & controle , Administração Tópica , Animais , Antígenos de Diferenciação/metabolismo , Movimento Celular/efeitos dos fármacos , Neovascularização de Coroide/metabolismo , Endotélio Vascular/efeitos dos fármacos , Glicoesfingolipídeos/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Injeções Intravítreas , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/efeitos dos fármacos , Neovascularização Patológica/prevenção & controle , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Degeneração Macular Exsudativa/metabolismo , Nucleolina
6.
J Gene Med ; 17(10-12): 229-43, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26369397

RESUMO

BACKGROUND: Age-related macular degeneration (AMD) is the most common cause of blindness in the elderly, with no therapy available for 90% of patients. Recent genetic evidence implicates activation of complement in the pathogenesis of AMD. We have recently discovered that adenovirus (Ad)-mediated expression of complement component C3 (AdCMVC3) in the murine retina recapitulates many of the pathological features found in human AMD. In the present study, utilizing a gene therapy approach, we examine whether Ad-mediated expression of complement Factor H (AdCAGfH) attenuates AdCMVC3-mediated retinal pathology. METHODS: AdCMVC3 was co-injected with either AdCAGfH or a negative control virus expressing green fluorescent protein (AdCMVGFP) into the subretinal space of adult mice. The resulting retinal pathology was analyzed by histology and immunocytochemistry and retinal function was quantified by electroretinography. RESULTS: Morphological and functional analyses indicated that AdCMVC3-mediated retinal pathology could be attenuated by AdCAGfH. Specifically, endothelial cell proliferation was reduced by 91% and atrophy of retinal pigment epithelium (RPE) could be attenuated by 69%. AdCAGfH injected eyes exhibited 90-150% greater A-wave and 120-180% greater B-wave amplitudes relative to control eyes. Immunocytochemical analysis of rhodopsin and RPE65 was consistent with the rescue of photoreceptors and RPE in AdCAGfH injected eyes. CONCLUSIONS: C3-induced pathology in murine retina can be attenuated by Ad-mediated expression of Factor H. Expression of Factor H is worthy of further study as a potential gene therapy for AMD.


Assuntos
Complemento C3/metabolismo , Fator H do Complemento/uso terapêutico , Adenoviridae , Animais , Complemento C3/efeitos adversos , Fator H do Complemento/administração & dosagem , Terapia Genética , Vetores Genéticos , Humanos , Degeneração Macular/terapia , Camundongos , Retina/efeitos dos fármacos , Retina/patologia
7.
J Gene Med ; 17(6-7): 101-15, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25917932

RESUMO

BACKGROUND: A variety of disorders are associated with the activation of complement. CD46, CD55 and CD59 are the major membrane associated regulators of complement on human cells. Previously, we have found that independent expression of CD55, CD46 or CD59 through gene transfer protects murine tissues against human complement mediated attack. In the present study, we investigated the potential of combining the complement regulatory properties of CD46, CD55 and CD59 into single gene products expressed from an adeno-associated virus (AAV) vector in a soluble non-membrane anchored form. METHODS: Minigenes encoding the complement regulatory domains from CD46, CD55 and CD59 (SACT) or CD55 and CD59 (DTAC) were cloned into an AAV vector. The specific regulatory activity of each component of SACT and DTAC was measured in vitro. The recombinant AAV vectors were injected into the peritoneum of mice and the efficacy of the transgene products for being able to protect murine liver vasculature against human complement, specifically the membrane attack complex (MAC), was measured. RESULTS: SACT and DTAC exhibited properties similar to CD46, CD55 and CD59 or CD55 and CD59, respectively, in vitro. AAV mediated delivery of SACT or DTAC protected murine liver vasculature from human MAC deposition by 63.2% and 56.7%, respectively. CONCLUSIONS: When delivered to mice in vivo via an AAV vector, SACT and DTAC are capable of limiting human complement mediated damage. SACT and DTAC merit further study as potential therapies for complement mediated disorders when delivered via a gene therapy approach.


Assuntos
Antígenos CD55/genética , Antígenos CD59/genética , Ativação do Complemento/genética , Inativadores do Complemento , Proteína Cofatora de Membrana/genética , Transgenes , Animais , Antígenos CD55/química , Antígenos CD59/química , Inativadores do Complemento/administração & dosagem , Inativadores do Complemento/química , Dependovirus , Vetores Genéticos , Humanos , Proteína Cofatora de Membrana/química , Camundongos , Estrutura Terciária de Proteína/genética
8.
PLoS One ; 8(10): e79661, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24167638

RESUMO

Diabetic retinopathy is the leading cause of visual dysfunction in working adults and is attributed to retinal vascular and neural cell damage. Recent studies have described elevated levels of membrane attack complex (MAC) and reduced levels of membrane associated complement regulators including CD55 and CD59 in the retina of diabetic retinopathy patients as well as in animal models of this disease. We have previously described the development of a soluble membrane-independent form of CD59 (sCD59) that when delivered via a gene therapy approach using an adeno-associated virus vector (AAV2/8-sCD59) to the eyes of mice, can block MAC deposition and choroidal neovascularization. Here, we examine AAV2/8-sCD59 mediated attenuation of MAC deposition and ensuing complement mediated damage to the retina of mice following streptozotocin (STZ) induced diabetes. We observed a 60% reduction in leakage of retinal blood vessels in diabetic eyes pre-injected with AAV2/8-sCD59 relative to negative control virus injected diabetic eyes. AAV2/8-sCD59 injected eyes also exhibited protection from non-perfusion of retinal blood vessels. In addition, a 200% reduction in retinal ganglion cell apoptosis and a 40% reduction in MAC deposition were documented in diabetic eyes pre-injected with AAV2/8-sCD59 relative to diabetic eyes pre-injected with the control virus. This is the first study characterizing a viral gene therapy intervention that targets MAC in a model of diabetic retinopathy. Use of AAV2/8-sCD59 warrants further exploration as a potential therapy for advanced stages of diabetic retinopathy.


Assuntos
Antígenos CD59/biossíntese , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Dependovirus , Diabetes Mellitus Experimental/terapia , Retinopatia Diabética/terapia , Transdução Genética , Animais , Antígenos CD59/genética , Complexo de Ataque à Membrana do Sistema Complemento/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/genética , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Humanos , Camundongos
9.
PLoS One ; 6(6): e21621, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21720565

RESUMO

Inappropriate activation of complement on the vascular endothelium of specific organs, or systemically, underlies the etiology of a number of diseases. These disorders include atypical hemolytic uremic syndrome, membranoproliferative glomerulonephritis, atherosclerosis, age-related macular degeneration, diabetic retinopathy, and transplant rejection. Inhibition of the terminal step of complement activation, i.e. formation of the membrane attack complex, using CD59 has the advantage of retaining the upstream processes of the complement cascade necessary for fighting pathogens and retaining complement's crucial role in tissue homeostasis. Previous studies have shown the necessity of membrane targeting of soluble CD59 in order for it to prove an effective inhibitor of complement deposition both in vitro and in vivo. In this study we have generated an in vivo model of human complement activation on murine liver vascular endothelium. This model should prove useful for the development of anti-complement therapies for complement-induced pathologies of vascular endothelium. Using this model, we have demonstrated the viability of a non membrane-targeted soluble CD59 to significantly inhibit complement deposition on the endothelium of murine liver vasculature when expressed in vivo from an adenovirus. This result, unanticipated based on prior studies, suggests that the use of non membrane-targeted sCD59 as an anti-complement therapy be re-visited.


Assuntos
Adenoviridae/genética , Antígenos CD59/imunologia , Ativação do Complemento/imunologia , Endotélio Vascular/imunologia , Fígado/irrigação sanguínea , Fígado/imunologia , Animais , Anticorpos/administração & dosagem , Anticorpos/imunologia , Aorta/imunologia , Vasos Sanguíneos/citologia , Vasos Sanguíneos/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Células Endoteliais/citologia , Células Endoteliais/imunologia , Endotélio Vascular/citologia , Humanos , Injeções Intraperitoneais , Fígado/citologia , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Ligação Proteica , Soro/imunologia , Solubilidade
10.
PLoS One ; 6(4): e19078, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21552568

RESUMO

Age related macular degeneration (AMD) is the most common cause of blindness amongst the elderly. Approximately 10% of AMD patients suffer from an advanced form of AMD characterized by choroidal neovascularization (CNV). Recent evidence implicates a significant role for complement in the pathogenesis of AMD. Activation of complement terminates in the incorporation of the membrane attack complex (MAC) in biological membranes and subsequent cell lysis. Elevated levels of MAC have been documented on choroidal blood vessels and retinal pigment epithelium (RPE) of AMD patients. CD59 is a naturally occurring membrane bound inhibitor of MAC formation. Previously we have shown that membrane bound human CD59 delivered to the RPE cells of mice via an adenovirus vector can protect those cells from human complement mediated lysis ex vivo. However, application of those observations to choroidal blood vessels are limited because protection from MAC- mediated lysis was restricted only to the cells originally transduced by the vector. Here we demonstrate that subretinal delivery of an adenovirus vector expressing a transgene for a soluble non-membrane binding form of human CD59 can attenuate the formation of laser-induced choroidal neovascularization and murine MAC formation in mice even when the region of vector delivery is distal to the site of laser induced CNV. Furthermore, this same recombinant transgene delivered to the intravitreal space of mice by an adeno-associated virus vector (AAV) can also attenuate laser-induced CNV. To our knowledge, this is the first demonstration of a non-membrane targeting CD59 having biological potency in any animal model of disease in vivo. We propose that the above approaches warrant further exploration as potential approaches for alleviating complement mediated damage to ocular tissues in AMD.


Assuntos
Antígenos CD59/química , Antígenos CD59/genética , Neovascularização de Coroide/genética , Neovascularização de Coroide/terapia , Terapia Genética/métodos , Degeneração Macular/fisiopatologia , Degeneração Macular/terapia , Adenoviridae/genética , Animais , Linhagem Celular , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Humanos , Lasers/efeitos adversos , Degeneração Macular/etiologia , Degeneração Macular/genética , Camundongos , Retina/metabolismo , Solubilidade
11.
Invest Ophthalmol Vis Sci ; 52(6): 3436-45, 2011 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-21357400

RESUMO

PURPOSE: Activation of complement has been implicated as one of the major causes of age-related macular degeneration (AMD). Evidence is accumulating for a role of complement in other retinal diseases, such as diabetic retinopathy and proliferative vitreoretinopathy. Because of the paucity of animal models that directly investigate the role of complement in retinal pathology, the authors sought to develop a model of increased complement expression and activation, specifically in the murine retina. METHODS: The authors constructed a recombinant adenovirus-expressing murine complement component 3 (C3, AdcmvC3). Adult mice were injected in the subretinal space with either AdcmvC3 or a control virus, AdcmvGFP. After 1 to 2 weeks of exogenous C3 expression, mice were analyzed by scotopic electroretinography and fluorescein angiography. Eyes were harvested for histologic, immunohistochemical, and quantitative RT-PCR analyses. RESULTS: Mice injected with C3-expressing adenovirus exhibited significantly increased vascular permeability, endothelial cell proliferation and migration, RPE atrophy, loss of photoreceptor outer segments, reactive gliosis, retinal detachment, and reduced retinal function relative to those injected with a control adenovirus. Deposition of the membrane attack complex was observed on endothelial cells and photoreceptor outer segments. CONCLUSIONS: Adenovirus-mediated delivery of C3 to murine RPE induces significant functional and anatomic changes that reproduce many of the features of AMD as well as those of other retinal diseases. This novel model may be useful in assessing the role of complement in retinal pathology and in developing anti-complement therapies for retinal diseases associated with complement activation.


Assuntos
Adenoviridae/genética , Complemento C3/genética , Regulação da Expressão Gênica/fisiologia , Degeneração Macular/genética , Retina/metabolismo , Descolamento Retiniano/genética , Animais , Atrofia , Permeabilidade Capilar , Movimento Celular , Proliferação de Células , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Eletrorretinografia , Endotélio Vascular/patologia , Angiofluoresceinografia , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Camundongos , Camundongos Endogâmicos C57BL , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Retina/patologia , Descolamento Retiniano/metabolismo , Descolamento Retiniano/patologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rodopsina/metabolismo
12.
J Gene Med ; 13(3): 158-70, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21344542

RESUMO

BACKGROUND: Cell-penetrating peptides (CPPs) can deliver molecules into cells by binding and penetrating the plasma membrane. However, the majority of CPPs get trapped in endosomes, resulting in degradation of the cargo molecule and inefficient delivery to the nucleus. The present study investigates the potential use of a nucleolin binding peptide (NBP) for the delivery of macromolecules including fluorophores, recombinant protein and DNA to the nuclei of ocular tissues in vivo. METHODS: Fluorescent dyes covalently linked to NBP or NBP-green fluorescent protein fusion protein were injected intravitreally or subretinally or topically applied to the cornea. Frozen sections were prepared for quantification of transduction. Delivery of plasmid DNA was studied using luciferase and LacZ DNA compacted with pegylated NBP. Levels of luciferase were quantified, and LacZ expression was localized in ocular tissues. RESULTS: We found that NBP-directed fluorophores exhibited retinal and corneal transduction. Subretinal injection transduced cell types throughout the retina, including photoreceptors, retinal pigment epithelium and neuronal cells. Intravitreal injection transduced neuronal cells in the retina, as well as cells in the cornea. Topically applied NBP lead to transduction of the superficial epithelial layer of the cornea. NBP localized to the nucleus upon exogenous application in vivo. Pegylated NBP nanoparticles significantly improved delivery and expression of transgenes over DNA alone without any measureable toxicity. CONCLUSIONS: The results obtained in the present study demonstrate that NBP can deliver small and large molecules into retinal and corneal cells and plasmid DNA into retinal cells and hence may be useful for the delivery of therapeutics to the eye.


Assuntos
Peptídeos Penetradores de Células , Córnea/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Proteína HMGN2/administração & dosagem , Peptídeos/metabolismo , Polietilenoglicóis/metabolismo , Retina/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Administração Tópica , Sequência de Aminoácidos , Animais , Núcleo Celular/metabolismo , Peptídeos Penetradores de Células/administração & dosagem , Peptídeos Penetradores de Células/metabolismo , DNA/administração & dosagem , DNA/genética , Eletrorretinografia , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/metabolismo , Proteínas de Fluorescência Verde , Proteína HMGN2/metabolismo , Humanos , Injeções Intravítreas , Luciferases/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Nanopartículas , Sinais de Localização Nuclear/metabolismo , Peptídeos/administração & dosagem , Peptídeos/genética , Fosfoproteínas/metabolismo , Plasmídeos/administração & dosagem , Plasmídeos/metabolismo , Polietilenoglicóis/administração & dosagem , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , beta-Galactosidase/genética , Nucleolina
13.
Invest Ophthalmol Vis Sci ; 51(12): 6776-83, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20688727

RESUMO

PURPOSE: Sequence variations in complement proteins are associated with age-related macular degeneration (AMD). The terminal pathway of complement results in the formation of the membrane attack complex (MAC) on the cell surface, resulting in their lysis. MAC has been documented on the retinal pigment epithelium (RPE), choroidal blood vessels, and drusen of AMD eyes. Here the investigators test the hypothesis that increasing the expression of decay accelerating factor (CD55) on RPE cells may result in reduced MAC-mediated damage. METHODS: The investigators constructed a recombinant adenovirus expressing human CD55 (AdCAGCD55). Mouse hepatocytes were infected with AdCAGCD55 or negative controls and subsequently incubated with normal human serum (NHS). Cell lysis and MAC formation were measured by FACS and immunocytochemistry, respectively. Adult mice were injected in the subretinal space with either AdCAGCD55 or controls; after 1 week of CD55 transgene expression, the eyecups were excised, challenged with NHS, and quantified for human MAC formation. RESULTS: Control-infected or uninfected mouse hepatocytes lyse at a rate of 93% and 94%, respectively. AdCAGCD55- infected mouse hepatocytes lyse at a rate of 29%. Lysis was confirmed to occur in the presence of MAC, which was reduced by 67% when cells were infected by AdCAGCD55. Mice injected in the subretinal space with AdCAGCD55 exhibited a 55.7% reduction in MAC formation on the RPE relative to controls. CONCLUSIONS: Adenovirus-mediated delivery of hCD55 to murine RPE confers protection against human complement. The investigators propose that the expression of hCD55 on RPE cells warrants investigation as a potential therapy for AMD.


Assuntos
Antígenos CD55/uso terapêutico , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas do Sistema Complemento/fisiologia , Terapia Genética , Degeneração Macular/terapia , Epitélio Pigmentado da Retina/metabolismo , Adenoviridae/genética , Animais , Western Blotting , Antígenos CD55/genética , Linhagem Celular , Citometria de Fluxo , Expressão Gênica/fisiologia , Vetores Genéticos , Hemólise/fisiologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Imuno-Histoquímica , Degeneração Macular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transgenes
14.
Mol Ther ; 18(11): 1917-26, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20700110

RESUMO

Peptide for ocular delivery (POD) is a novel cationic cell-penetrating peptide (CPP) which, when conjugated with polyethylene glycol (PEG-POD), can deliver plasmid DNA to the retinal pigment epithelium (RPE) of adult murine retina. PEG-POD nanoparticles containing an expression cassette for glial cell line-derived neurotrophic factor (PEG-POD~GDNF) were investigated for their ability to inhibit light-induced photoreceptor apoptosis. PEG-POD~GDNF, control nanoparticles, or buffer were injected into the subretinal space of adult murine retina and retinal degeneration induced by blue light. Animals injected with PEG-POD~GDNF showed a significant reduction (3.9-7.7 fold) in apoptosis relative to control-injected animals. The thickness of the outer nuclear layer (ONL) of the superior retina of PEG-POD~GDNF-injected eyes was significantly greater (23.6-39.3%) than control-injected retina 14 days post-light treatment. PEG-POD~GDNF-injected eyes showed a 27-39% greater functional response relative to controls, as measured by electroretinogram (ERG) 7 days post-light treatment. This is one of only two studies demonstrating histological and functional rescue of a mouse model of retinal degeneration following nonviral administration of a transgene into adult retina. Although rescue is short lived for clinical application, this study represents an important step in the development of nonviral gene therapy for retinal diseases.


Assuntos
Terapia Genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Nanopartículas , Lesões Experimentais por Radiação/terapia , Retina/efeitos da radiação , Degeneração Retiniana/terapia , Animais , Apoptose , Caspases/metabolismo , Sistemas de Liberação de Medicamentos , Eletrorretinografia , Luz/efeitos adversos , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/uso terapêutico , Polietilenoglicóis , Lesões Experimentais por Radiação/etiologia , Lesões Experimentais por Radiação/patologia , Ratos , Degeneração Retiniana/etiologia , Degeneração Retiniana/patologia
15.
J Gene Med ; 12(1): 86-96, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19937991

RESUMO

BACKGROUND: We have previously shown that a novel synthetic peptide for ocular delivery (POD) can efficiently compact DNA and deliver it to cells in vitro. This observation prompted us to develop use of POD as a nonviral vector in vivo. METHODS: POD peptide was modified using poly(ethylene) glycol (PEG-POD) and used to compact DNA into nanoparticles that were then analysed using electron microscopy, dynamic light scattering, and fluorescent labeling. Transfection efficiency and localization were determined 48 h post-injection into the subretinal space of the mouse eye using luciferase and LacZ, respectively. Efficiency of ocular transfection was compared to two other PEGylated peptides: PEG-TAT and PEG-CK30. RESULTS: PEG-POD can compact DNA and form discrete nanoparticles of approximately 136 nm that can penetrate and transduce the retinal pigment epithelium (RPE) in vivo. PEG-POD significantly increased expression of plasmid DNA by 215-fold, PEG-TAT by 56.52-fold, and PEG-CK30 by 24.73-fold relative to DNA injected alone. In all cases beta-galactosidase was observed primarily in the RPE layer after subretinal injection. Electrophysiological analyses of PEG-POD transduced retina indicates an absence of PEG-POD-mediated toxicity. PEG-POD can protect plasmid DNA from DNaseI digestion, resulting in significant transfection of the lung after intravenous injection in mice. CONCLUSIONS: PEG-POD was found to significantly increase gene delivery relative to both DNA alone and other pegylated peptides. These findings highlight the use of pegylated peptides, and specifically PEG-POD, as novel gene delivery vectors.


Assuntos
DNA/química , Técnicas de Transferência de Genes , Mitose , Nanopartículas/química , Peptídeos/química , Polietilenoglicóis/química , Epitélio Pigmentado da Retina/citologia , Animais , Linhagem Celular , DNA/ultraestrutura , Desoxirribonuclease I/metabolismo , Humanos , Injeções Intravenosas , Luciferases/metabolismo , Camundongos , Nanopartículas/toxicidade , Nanopartículas/ultraestrutura , Peptídeos/toxicidade , Polietilenoglicóis/toxicidade , Estrutura Quaternária de Proteína , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Transfecção
16.
Vision Res ; 50(7): 686-97, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-19733192

RESUMO

Recently we described a novel cell penetrating peptide, peptide for ocular delivery (POD) that could deliver small molecules including fluorescent dyes into retinal cells. The objective of the current study was to examine whether biologically relevant macromolecules such as proteins, genetically fused with POD could also be delivered into retinal tissues in vivo. We generated a POD-GFP fusion protein and examined its cell and tissue penetrating properties. We found that endogenously expressed POD-GFP fusion protein localized to the nucleus, suggesting that POD acts as a nuclear localization signal. Adenovirus (Ad) vectors expressing POD-GFP fusion protein were constructed and the recombinant protein was purified from Ad-infected human embryonic retinoblasts (HER). Exogenously supplied POD-GFP fusion protein rapidly transduced A549 and HER cells and colocalized in part with markers of late endosomes, from which it could escape. Following subretinal delivery, POD-GFP localized to the retinal pigment epithelium and the photoreceptor cell bodies. When injected into the vitreous, POD-GFP localized to the ganglion cells and the inner nuclear layer of the retina as well as the lens capsule. Topical application of POD-GFP to ocular surfaces resulted in uptake by the corneal epithelium. POD-GFP also transduced non-ocular tissues, including the epidermis of the skin following topical application.


Assuntos
Córnea/metabolismo , Peptídeos/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/metabolismo , Retina/metabolismo , Pele/metabolismo , Adenovírus Humanos/fisiologia , Administração Tópica , Transporte Biológico , Humanos , Peptídeos/metabolismo , Transfecção
17.
Invest Ophthalmol Vis Sci ; 51(4): 2219-28, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19892875

RESUMO

Purpose. Gene therapy for a number of retinal diseases necessitates efficient transduction of photoreceptor cells. Whereas adenovirus (Ad) serotype 5 (Ad5) does not transduce photoreceptors efficiently, previous studies have demonstrated improved photoreceptor transduction by Ad5 pseudotyped with Ad35 (Ad5/F35) or Ad37 (Ad5/F37) fiber or by the deletion of the RGD domain in the Ad5 penton base (Ad5DeltaRGD). However, each of these constructs contained a different transgene cassette, preventing the evaluation of the relative performance of these vectors, an important consideration before the use of these vectors in the clinic. The aim of this study was to evaluate these vectors in the retina and to attempt photoreceptor-specific transgene expression. Methods. Three Ad5-based vectors containing the same expression cassette were generated and injected into the subretinal space of adult mice. Eyes were analyzed for green fluorescence protein expression in flat-mounts, cross-sections, quantitative RT-PCR, and a modified stereological technique. A 257-bp fragment derived from the mouse opsin promoter was analyzed in the context of photoreceptor-specific transgene expression. Results. Each virus tested efficiently transduced the retinal pigment epithelium. The authors found no evidence that Ad5/F35 or Ad5/F37 transduced photoreceptors. Instead, they found that Ad5/F37 transduced Müller cells. Robust photoreceptor transduction by Ad5DeltaRGD was detected. Photoreceptor-specific transgene expression from the 257-bp mouse opsin promoter in the context of Ad5DeltaRGD vectors was found. Conclusions. Adenovirus vectors may be designed with tropism to distinct cell populations. Robust photoreceptor-specific transgene expression can be achieved in the context of Ad5DeltaRGD vectors.


Assuntos
Adenoviridae/genética , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Neurônios Retinianos/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Transfecção , Animais , Biomarcadores/metabolismo , Western Blotting , Linhagem Celular Tumoral , Proteínas do Olho/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Expressão Gênica , Proteína Cofatora de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Retina/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transgenes
18.
Vision Res ; 48(16): 1671-1680, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18565565

RESUMO

The majority of recent preclinical gene therapy studies targeting the retina have used adeno-associated virus (AAV) as the gene transfer vector. However, AAV has several limitations including the ability to generate innate inflammatory responses, the ability to cause insertional mutagenesis at a frequency of up to 56% in some tissues and a limited cloning capacity of 4.8Kb. Furthermore, AAV is known to generate limiting immune responses in humans despite the absence of similar immune responses in preclinical canine and murine studies. Three clinical trials to treat Leber's congenital amaurosis using AAV are under way. A clinical trial to treat Stargardt's using lentivirus vectors has also been recently announced. However, very limited evidence currently exists that lentivirus vectors can efficiently transduce photoreceptor cells. In contrast, very few preclinical ocular gene therapy studies have utilized adenovirus as the gene therapy vector. Nonetheless, the only two ocular gene therapy clinical trials performed to date have each used adenovirus as the vector and more significantly, in these published trials there has been no observed serious adverse event. These trials appear to be poised for Phase II/III status. Activation of cytotoxic T lymphocytes limits duration of transgene expression in the retina from first generation adenovirus vectors. However, an advanced class of adenovirus vectors referred to as Helper-dependent Adenovirus (Hd-Ad) have recently been shown to be capable of expressing transgenes in ocular tissues for more than one year. Hd-Ad vectors have many properties that potentially warrant their inclusion in the retinal gene therapy toolbox for the treatment of retinal degenerative diseases.


Assuntos
Terapia Genética/métodos , Degeneração Retiniana/terapia , Animais , Dependovirus/genética , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Camundongos , Mutagênese Insercional
19.
Invest Ophthalmol Vis Sci ; 49(9): 4126-36, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18487376

RESUMO

PURPOSE: Complement-mediated damage to the retinal pigment epithelium (RPE), Bruch membrane, and choroid has been associated with pathogenesis in age-related macular degeneration (AMD). The terminal step of complement activation involves lysis of cells by the insertion of the membrane attack complex (MAC) in the plasma membrane. The hypothesis that local overexpression of human CD59 (hCD59) delivered by an adenovirus (Ad) vector to primary murine RPE cells in vitro, RPE in vivo, or cornea ex vivo protects those cells from human MAC deposition and lysis was tested. METHODS: A humanized model of MAC deposition on murine cells and murine ocular tissues including RPE and cornea was developed to permit testing of human complement regulators in mice. A recombinant adenovirus-expressing hCD59 was generated, and this virus was injected into the subretinal space of adult mice. Subsequently, eyecups from these mice were exposed to human serum, and the levels of MAC deposition on the RPE were quantified. hCD59 was also expressed on murine cornea ex vivo and in murine hepatocytes, and primary RPE cells in vitro and levels of human MAC deposition and cell lysis were measured. RESULTS: Adenovirus-mediated delivery of hCD59 to the RPE, cornea, or cells in culture protects those cells from human MAC deposition and MAC-mediated damage and vesiculation. CONCLUSIONS: The humanized model of MAC deposition on murine ocular tissues allows testing of human complement regulators that may have potential in the treatment of AMD or other diseases associated with complement activation.


Assuntos
Antígenos CD59/uso terapêutico , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Degeneração Macular/tratamento farmacológico , Epitélio Pigmentado Ocular/fisiologia , Adenoviridae , Adulto , Envelhecimento , Animais , Antígenos CD59/administração & dosagem , Técnicas de Cultura de Células , Linhagem Celular , Embrião de Mamíferos , Citometria de Fluxo , Vetores Genéticos , Humanos , Degeneração Macular/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Epitélio Pigmentado Ocular/citologia
20.
Mol Ther ; 16(1): 107-14, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17923842

RESUMO

As in other organ systems, gene and drug delivery to ocular tissues such as the retina and cornea is hampered by inefficient penetration of therapeutic molecules across the plasma membrane. We describe the use of a novel peptide for ocular delivery (POD) with protein transduction properties, for delivery of small and large molecules across the plasma membrane. POD enters cells within 5 minutes in a temperature dependent manner. POD can compact and deliver plasmid DNA, achieving transgene expression in >50% of human embryonic retinoblasts. Delivery of small interfering RNA (siRNA) duplexes to cells using POD, allowed for silencing of transgene expression by >50%. POD could also be used to deliver quantum dots in vitro and in vivo. Upon ocular delivery, POD rapidly entered neural retina and localized to retinal pigment epithelium (RPE), photoreceptor, and ganglion cells. Additionally, POD was able to enter corneal epithelium, sclera, choroid, and the dura of the optic nerve via topical application. POD also functions as a bacteriostatic, a useful property for a carrier of molecules to post mitotic neural ocular tissues.


Assuntos
Permeabilidade da Membrana Celular , Córnea/metabolismo , Portadores de Fármacos/administração & dosagem , Sistemas de Liberação de Medicamentos , Peptídeos/administração & dosagem , Retina/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Permeabilidade da Membrana Celular/genética , Doenças da Córnea/tratamento farmacológico , Doenças da Córnea/metabolismo , Portadores de Fármacos/metabolismo , Humanos , Dados de Sequência Molecular , Peptídeos/genética , Peptídeos/metabolismo , Plasmídeos/administração & dosagem , Plasmídeos/metabolismo , RNA Interferente Pequeno/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA