Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Exp Eye Res ; 175: 20-31, 2018 10.
Article in English | MEDLINE | ID: mdl-29864441

ABSTRACT

There is currently no efficient method available for the delivery of full length functional proteins into the cytoplasm of retinal cells in vivo. Historically, the most successful approach for the treatment of retinal diseases has been intravitreal injection of antibodies or recombinant proteins, but this approach is not yet utilized for the delivery of proteins that require intracellular access for a therapeutic effect. Here we describe a platform for the delivery of functional proteins into ganglion cells, photoreceptors and retinal pigment epithelium via intravitreal injection. A nucleolin binding aptamer, AS1411, was biotinylated and complexed with traptavidin and utilized as a platform for the delivery of GFP or X-linked inhibitor of apoptosis (XIAP) proteins by intravitreal injection in BALB/c mice. Retinal sections were analyzed for uptake of proteins in the retina. Apoptosis was induced by intravitreal injection of N-methyl-D-aspartate (NMDA). Retinas were harvested for analysis of TUNEL and caspase 3/7 activity. Intravitreal injection of AS1411-directed GFP or XIAP complexes enabled delivery of these proteins into ganglion cells, photoreceptors and retinal pigment epithelium in vivo. AS1411-XIAP complexes conferred significant protection to cells in the outer and inner nuclear layers following NMDA induced apoptosis. A concomitant decrease in activity of Caspase 3/7 was observed in eyes injected with the AS1411-XIAP complex. In conclusion, AS1411 can be used as a platform for the delivery of therapeutic proteins into retinal cells. This approach can potentially be utilized to introduce a large variety of therapeutically relevant proteins that are previously well characterized to maintain the structural integrity and function of retina, thus, preventing vision loss due to ocular trauma or inherited retinal degeneration.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors/administration & dosage , Drug Delivery Systems , Oligodeoxyribonucleotides/administration & dosage , Retina/drug effects , Retinal Degeneration/prevention & control , X-Linked Inhibitor of Apoptosis Protein/administration & dosage , Animals , Aptamers, Nucleotide/administration & dosage , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Electrophoresis, Polyacrylamide Gel , Excitatory Amino Acid Agonists/toxicity , G-Quadruplexes , Green Fluorescent Proteins/administration & dosage , In Situ Nick-End Labeling , Intravitreal Injections , Mice , Mice, Inbred BALB C , Microscopy, Confocal , N-Methylaspartate/toxicity , Retinal Degeneration/pathology
2.
Mol Ther ; 26(6): 1568-1580, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29678656

ABSTRACT

Uveitis is an inflammatory disorder of the eye responsible for approximately 10%-15% of blindness in the US. In this study, we examined the role of the complement membrane attack complex (MAC) and the NLRP3 inflammasome in the pathogenesis of experimental autoimmune uveitis (EAU) in normal and C9-/- mice that are incapable of assembling the MAC. We discovered that the MAC and the NLRP3 inflammasome and associated production of IL-1ß are elevated in EAU mice and that MAC may be involved in regulation of Th1 and Th17 cell differentiation. In contrast, MAC and the NLRP3 inflammasome were not elevated in C9-/- mice. However, EAU-associated pathophysiology including retinal structure and function were not rescued in C9-/- mice. Unexpectedly, AAV-mediated delivery of sCD59, an inhibitor of C9 incorporation into the MAC, successfully attenuated activation of the NLRP3 inflammasome and EAU pathology as well as MAC. Our studies provide an improved understanding of the role of the MAC and the NLRP3 inflammasome in EAU as well as suggest a novel approach for the treatment of uveitis.


Subject(s)
CD59 Antigens/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Uveitis/metabolism , Animals , CD59 Antigens/genetics , Complement Activation/genetics , Complement Activation/physiology , Inflammasomes/genetics , Interleukin-1beta/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Uveitis/genetics
3.
Mol Ther Nucleic Acids ; 8: 77-89, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28918058

ABSTRACT

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.
Gastroenterology ; 153(2): 566-578.e5, 2017 08.
Article in English | MEDLINE | ID: mdl-28456632

ABSTRACT

BACKGROUND & AIMS: Tumor necrosis factor (TNF) is an inflammatory cytokine expressed by human fetal liver cells (HFLCs) after infection with cell culture-derived hepatitis C virus (HCV). TNF has been reported to increase entry of HCV pseudoparticles into hepatoma cells and inhibit signaling by interferon alpha (IFNα), but have no effect on HCV-RNA replication. We investigated the effects of TNF on HCV infection of and spread among Huh-7 hepatoma cells and primary HFLCs. METHODS: Human hepatoma (Huh-7 and Huh-7.5) and primary HFLCs were incubated with TNF and/or recombinant IFNA2A, IFNB, IFNL1, and IFNL2 before or during HCV infection. We used 2 fully infectious HCV chimeric viruses of genotype 2A in these studies: J6/JFH (clone 2) and Jc1(p7-nsGluc2A) (Jc1G), which encodes a secreted luciferase reporter. We measured HCV replication, entry, spread, production, and release in hepatoma cells and HFLCs. RESULTS: TNF inhibited completion of the HCV infectious cycle in hepatoma cells and HFLCs in a dose-dependent and time-dependent manner. This inhibition required TNF binding to its receptor. Inhibition was independent of IFNα, IFNß, IFNL1, IFNL2, or Janus kinase signaling via signal transducer and activator of transcription. TNF reduced production of infectious viral particles by Huh-7 and HFLC, and thereby reduced the number of infected cells and focus size. TNF had little effect on HCV replicons and increased entry of HCV pseudoparticles. When cells were incubated with TNF before infection, the subsequent antiviral effects of IFNs were increased. CONCLUSIONS: In a cell culture system, we found TNF to have antiviral effects independently of, as well as in combination with, IFNs. TNF inhibits HCV infection despite increased HCV envelope glycoprotein-mediated infection of liver cells. These findings contradict those from other studies, which have reported that TNF blocks signal transduction in response to IFNs. The destructive inflammatory effects of TNF must be considered along with its antiviral effects.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Interferons/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Genotype , Hepacivirus/genetics , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Janus Kinases/metabolism , Liver/cytology , Liver Neoplasms/virology , Receptors, Tumor Necrosis Factor/metabolism , Replicon/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Virus Replication/drug effects
5.
Exp Eye Res ; 145: 380-392, 2016 04.
Article in English | MEDLINE | ID: mdl-26923800

ABSTRACT

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.


Subject(s)
Aptamers, Nucleotide/administration & dosage , Drug Delivery Systems/methods , Oligonucleotides/administration & dosage , Photoreceptor Cells, Vertebrate/metabolism , Recombinant Proteins/administration & dosage , Retinal Pigment Epithelium/metabolism , Analysis of Variance , Animals , Humans , Immunohistochemistry , Intravitreal Injections , MCF-7 Cells , Mice , Mice, Inbred BALB C , Oligonucleotides/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Recombinant Proteins/metabolism , Nucleolin
6.
Exp Eye Res ; 140: 171-178, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26368850

ABSTRACT

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.


Subject(s)
Aptamers, Nucleotide/administration & dosage , Choroidal Neovascularization/prevention & control , Disease Models, Animal , Oligodeoxyribonucleotides/administration & dosage , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Wet Macular Degeneration/prevention & control , Administration, Topical , Animals , Antigens, Differentiation/metabolism , Cell Movement/drug effects , Choroidal Neovascularization/metabolism , Endothelium, Vascular/drug effects , Glycosphingolipids/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Intravitreal Injections , Macrophages/physiology , Mice , Mice, Inbred C57BL , Microvessels/drug effects , Neovascularization, Pathologic/prevention & control , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Wet Macular Degeneration/metabolism , Nucleolin
7.
J Gene Med ; 17(10-12): 229-43, 2015.
Article in English | MEDLINE | ID: mdl-26369397

ABSTRACT

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.


Subject(s)
Complement C3/metabolism , Complement Factor H/therapeutic use , Adenoviridae , Animals , Complement C3/adverse effects , Complement Factor H/administration & dosage , Genetic Therapy , Genetic Vectors , Humans , Macular Degeneration/therapy , Mice , Retina/drug effects , Retina/pathology
8.
Eur J Immunol ; 45(7): 2072-83, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25929249

ABSTRACT

Absence of MHC class I expression is an important mechanism by which NK cells recognize a variety of target cells, yet the pathways underlying "missing-self" recognition, including the involvement of activating receptors, remain poorly understood. Using ethyl-N-nitrosourea mutagenesis in mice, we identified a germline mutant, designated Ace, with a marked defect in NK cell mediated recognition and elimination of "missing-self" targets. The causative mutation was linked to chromosome 11 and identified as a missense mutation (Thr428Ile) in the SH2 domain of Slp-76-a critical adapter molecule downstream of ITAM-containing surface receptors. The Slp-76 Ace mutation behaved as a hypomorphic allele-while no major defects were observed in conventional T-cell development/function, a marked defect in NK cell mediated elimination of ß2-microglobulin (ß2M) deficient target cells was observed. Further studies revealed Slp-76 to control NK-cell receptor expression and maturation; however, activation of Slp-76(ace/ace) NK cells through ITAM-containing NK-cell receptors or allogeneic/tumor target cells appeared largely unaffected. Imagestream analysis of the NK-ß2M(-/-) target cell synapse revealed a specific defect in actin recruitment to the conjugate synapse in Slp-76(ace/ace) NK cells. Overall these studies establish Slp-76 as a critical determinant of NK-cell development and NK cell mediated elimination of missing-self target cells in mice.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Killer Cells, Natural/immunology , Phosphoproteins/immunology , Self Tolerance/immunology , Animals , Flow Cytometry , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
9.
J Gene Med ; 17(6-7): 101-15, 2015.
Article in English | MEDLINE | ID: mdl-25917932

ABSTRACT

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.


Subject(s)
CD55 Antigens/genetics , CD59 Antigens/genetics , Complement Activation/genetics , Complement Inactivating Agents , Membrane Cofactor Protein/genetics , Transgenes , Animals , CD55 Antigens/chemistry , CD59 Antigens/chemistry , Complement Inactivating Agents/administration & dosage , Complement Inactivating Agents/chemistry , Dependovirus , Genetic Vectors , Humans , Membrane Cofactor Protein/chemistry , Mice , Protein Structure, Tertiary/genetics
10.
Front Immunol ; 5: 550, 2014.
Article in English | MEDLINE | ID: mdl-25426115

ABSTRACT

Hepatitis C virus (HCV) remains a global problem, despite advances in treatment. The low cost and high benefit of vaccines have made them the backbone of modern public health strategies, and the fight against HCV will not be won without an effective vaccine. Achievement of this goal will benefit from a robust understanding of virus-host interactions and protective immunity in HCV infection. In this review, we summarize recent findings on HCV-specific antibody responses associated with chronic and spontaneously resolving human infection. In addition, we discuss specific epitopes within HCV's envelope glycoproteins that are targeted by neutralizing antibodies. Understanding what prompts or prevents a successful immune response leading to viral clearance or persistence is essential to designing a successful vaccine.

11.
J Leukoc Biol ; 96(4): 535-48, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25015956

ABSTRACT

Despite the development of potent antiviral drugs, HCV remains a global health problem; global eradication is a long way off. In this review, we discuss the immune response to HCV infection and particularly, the interplay between viral strategies that delay the onset of antiviral responses and host strategies that limit or even eradicate infected cells but also contribute to pathogenesis. Although HCV can disable some cellular virus-sensing machinery, IFN-stimulated antiviral genes are induced in the infected liver. Whereas epitope evolution contributes to escape from T cell-mediated immunity, chronic high antigen load may also blunt the T cell response by activating exhaustion or tolerance mechanisms. The evasive maneuvers of HCV limit sterilizing humoral immunity through rapid evolution of decoy epitopes, epitope masking, stimulation of interfering antibodies, lipid shielding, and cell-to-cell spread. Whereas the majority of HCV infections progress to chronic hepatitis with persistent viremia, at least 20% of patients spontaneously clear the infection. Most of these are protected from reinfection, suggesting that protective immunity to HCV exists and that a prophylactic vaccine may be an achievable goal. It is therefore important that we understand the correlates of protective immunity and mechanisms of viral persistence.


Subject(s)
Hepacivirus/immunology , Hepatitis C/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Gene Expression , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C/genetics , Hepatitis C/metabolism , Hepatitis C Antibodies/immunology , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/metabolism , Humans , Immunity, Cellular , Immunity, Innate , Interferons/genetics , Interferons/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
PLoS One ; 8(11): e82295, 2013.
Article in English | MEDLINE | ID: mdl-24278479

ABSTRACT

Retinitis pigmentosa (RP) is the most genetically heterogeneous disorder known to cause blindness, involving over 50 different genes. Previously, we have described nanoparticles (NPs) 150 nm in size, comprised of a 3.5 kD peptide (POD) complexed to PEG and DNA (PEGPOD DNA). These NPs expressing GDNF enabled rescue of photoreceptor degeneration in mice up to 11 days post injection. In the current study we examine use of scaffold/ matrix attachment regions (S/MARs), CpG depletion and titration of DNA content of PEGPOD DNA NPs to extend the duration of transgene expression. S/MARs and CpGs did not significantly influence the duration of transgene expression, but did influence its stability. These parameters enabled us to extend transgene expression from 48 hours to 10 weeks. At 77 days post injection, we observed a 76% rescue of the thickness of the retinal outer nuclear layer (ONL) and at 37 days post injection we observed 53% and 55% rescue of the A and B wave ERG amplitudes respectively and 60% rescue of the ONL. Our studies suggest that PEGPOD DNA NPs have potential as gene delivery vectors for the retina.


Subject(s)
Nanoparticles , Photoreceptor Cells, Vertebrate/metabolism , Polyethylene Glycols/chemistry , Transgenes , Animals , DNA/metabolism , Light , Mice, Inbred BALB C , Plasmids , Polymerase Chain Reaction
13.
Invest Ophthalmol Vis Sci ; 54(10): 7107-14, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24106121

ABSTRACT

PURPOSE: Immunocytochemical and genetic data implicate a significant role for the activation of complement in the pathology of AMD. Individuals homozygous for a Y402H polymorphism in Factor H have elevated levels of membrane attack complex (MAC) in their choroidal blood vessels and RPE relative to individuals homozygous for the wild-type allele. An R95X polymorphism in C9, a protein necessary for the final assembly of MAC, is partially protective against the formation of choroidal neovascularization (CNV) in AMD patients. Aurintricarboxylic Acid (ATA) is a small molecule inhibitor of MAC. Our hypothesis was that attenuation of the formation of MAC on ocular tissues by ATA may protect mice against laser-induced CNV. METHODS: The ability of ATA to inhibit human complement-mediated cell lysis, inhibit formation of human MAC, and inhibit formation of tubes by endothelial cells was examined in vitro. Subsequently, the Bruch's membrane of adult mice was damaged using an argon laser, followed by intravitreal injection of ATA. One week later, choroidal flat mounts from these mice were stained for the presence of MAC, endothelial cells, and macrophages. RESULTS: ATA protects cells from human complement-mediated lysis, attenuates assembly of the MAC, and inhibits tube formation by endothelial cells in vitro. ATA also attenuates CNV, MAC deposition, and macrophage infiltration in a murine model of exudative AMD. CONCLUSIONS: ATA warrants further study as a potential drug for the treatment of exudative and nonexudative AMD.


Subject(s)
Aurintricarboxylic Acid/pharmacology , Choroidal Neovascularization/prevention & control , Complement Membrane Attack Complex/antagonists & inhibitors , Macular Degeneration/complications , Adult , Animals , Apoptosis/drug effects , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Humans , Intravitreal Injections , Mice , Mice, Inbred C57BL
14.
PLoS One ; 8(10): e79661, 2013.
Article in English | MEDLINE | ID: mdl-24167638

ABSTRACT

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.


Subject(s)
CD59 Antigens/biosynthesis , Complement Membrane Attack Complex/metabolism , Dependovirus , Diabetes Mellitus, Experimental/therapy , Diabetic Retinopathy/therapy , Transduction, Genetic , Animals , CD59 Antigens/genetics , Complement Membrane Attack Complex/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Humans , Mice
15.
Oncoimmunology ; 1(6): 856-862, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-23162753

ABSTRACT

The immune system exerts a critical function as it recognizes and eliminates transformed or neoplastic cells, a process also referred to as immunosurveillance. NK cells play a particularly important role in that they are able to recognize tumor cells via "missing-self"-i.e., the absence of major histocompatibility complex Class I on target cells. Moreover, recent studies suggest that NK cells also participate in the onset and regulation of adaptive immune responses. The exact molecular pathways by which this occurs, however, remain poorly understood. To obtain further insight into the genes that are required for self-induced immune responses via NK cell-mediated cell death, our laboratory initiated a forward genetic approach using N-ethyl-N-nitrosourea (ENU) as a mutagen. Specifically, we tested the ability of NK cells from G3 ENU germline mice to recognize missing-self target cells and induce CD8+ T-cell responses following immunization with irradiated tumor cells. Here we present two ENU germline mutants, designated Ace and Chip, that are defective in the recognition of ß-2 microglobulin-deficient target cells, yet exhibit improved clearance of B16 melanoma cells in vivo. Coarse mapping and whole genome sequencing of the Chip mutation revealed a missense mutation causing a T'A amino acid substitution in the highly conserved third immuno-receptor tyrosine-based switch motif of CD244 (2B4). The forward genetic approach described here promises to reveal important insight into critical genes that are required for host responses involved in anticancer immunity.

16.
PLoS One ; 6(6): e21621, 2011.
Article in English | MEDLINE | ID: mdl-21720565

ABSTRACT

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.


Subject(s)
Adenoviridae/genetics , CD59 Antigens/immunology , Complement Activation/immunology , Endothelium, Vascular/immunology , Liver/blood supply , Liver/immunology , Animals , Antibodies/administration & dosage , Antibodies/immunology , Aorta/immunology , Blood Vessels/cytology , Blood Vessels/immunology , Complement Membrane Attack Complex/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelium, Vascular/cytology , Humans , Injections, Intraperitoneal , Liver/cytology , Mice , Mice, Inbred C57BL , Organ Specificity/immunology , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Protein Binding , Serum/immunology , Solubility
17.
PLoS One ; 6(4): e19078, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21552568

ABSTRACT

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.


Subject(s)
CD59 Antigens/chemistry , CD59 Antigens/genetics , Choroidal Neovascularization/genetics , Choroidal Neovascularization/therapy , Genetic Therapy/methods , Macular Degeneration/physiopathology , Macular Degeneration/therapy , Adenoviridae/genetics , Animals , Cell Line , Complement Membrane Attack Complex/metabolism , Dependovirus/genetics , Disease Models, Animal , Humans , Lasers/adverse effects , Macular Degeneration/etiology , Macular Degeneration/genetics , Mice , Retina/metabolism , Solubility
18.
Invest Ophthalmol Vis Sci ; 52(6): 3436-45, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21357400

ABSTRACT

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.


Subject(s)
Adenoviridae/genetics , Complement C3/genetics , Gene Expression Regulation/physiology , Macular Degeneration/genetics , Retina/metabolism , Retinal Detachment/genetics , Animals , Atrophy , Capillary Permeability , Cell Movement , Cell Proliferation , Complement Membrane Attack Complex/metabolism , Electroretinography , Endothelium, Vascular/pathology , Fluorescein Angiography , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mice , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retina/pathology , Retinal Detachment/metabolism , Retinal Detachment/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Reverse Transcriptase Polymerase Chain Reaction , Rhodopsin/metabolism
19.
J Gene Med ; 13(3): 158-70, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21344542

ABSTRACT

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.


Subject(s)
Cell-Penetrating Peptides , Cornea/metabolism , Drug Delivery Systems/methods , HMGN2 Protein/administration & dosage , Peptides/metabolism , Polyethylene Glycols/metabolism , Retina/metabolism , Active Transport, Cell Nucleus/physiology , Administration, Topical , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/metabolism , DNA/administration & dosage , DNA/genetics , Electroretinography , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/metabolism , Green Fluorescent Proteins , HMGN2 Protein/metabolism , Humans , Intravitreal Injections , Luciferases/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nanoparticles , Nuclear Localization Signals/metabolism , Peptides/administration & dosage , Peptides/genetics , Phosphoproteins/metabolism , Plasmids/administration & dosage , Plasmids/metabolism , Polyethylene Glycols/administration & dosage , RNA-Binding Proteins/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , beta-Galactosidase/genetics , Nucleolin
20.
Invest Ophthalmol Vis Sci ; 51(12): 6776-83, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20688727

ABSTRACT

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.


Subject(s)
CD55 Antigens/therapeutic use , Complement Membrane Attack Complex/metabolism , Complement System Proteins/physiology , Genetic Therapy , Macular Degeneration/therapy , Retinal Pigment Epithelium/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , CD55 Antigens/genetics , Cell Line , Flow Cytometry , Gene Expression/physiology , Genetic Vectors , Hemolysis/physiology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Immunohistochemistry , Macular Degeneration/metabolism , Mice , Mice, Inbred C57BL , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...