Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(7): e0236175, 2020.
Article in English | MEDLINE | ID: mdl-32697798

ABSTRACT

Adenoviruses cause upper respiratory infections, conjunctivitis, keratitis, and gastrointestinal illness. These can be fatal in immunocompromised individuals. Adenoviruses have also been engineered into viral vectors to deliver therapeutic genes or induce immunity as vaccine carriers. The success of ocular gene therapy is driven partly by the immunologic and biochemical influences of the intraocular environment. We have shown that versican and hyaluronan modulate adenoviral vector transgene expression through CD44 signaling. Herein we explored the role of these pathways on virus replication and viral protein expression of wild type adenovirus. We report that the addition of vitreous humor (which contains both versican and hyaluronan) increases viral hexon protein levels. Vitreous humor also increased wild type adenovirus DNA replication in vitro. Metalloproteinase and γ-secretase inhibitors, which inhibit CD44 proteolytic activation, blocked adenoviral replication in vitro. Similarly, protein kinase C and RhoA kinase inhibitors, both proteins associated with CD44 mediated pathways, also inhibited wild type adenoviral replication in vitro. Application of metalloproteinase and γ-secretase inhibitors to human conjunctival explants sharply decreased adenoviral vector gene expression. Our results demonstrate that pharmacologic delivery of these inhibitors is easily achievable. The inhibition of these enzymes should be explored as potential therapies of wild type adenoviral infections.


Subject(s)
Adenoviridae Infections/drug therapy , Adenoviridae/drug effects , Antiviral Agents/pharmacology , Genetic Vectors/drug effects , Virus Replication/drug effects , Adenoviridae/physiology , Adenoviridae Infections/virology , Administration, Ophthalmic , Amides/pharmacology , Amides/therapeutic use , Antiviral Agents/therapeutic use , Conjunctiva/metabolism , DNA, Viral/genetics , DNA, Viral/isolation & purification , Diamines/pharmacology , Diamines/therapeutic use , Dipeptides/pharmacology , Dipeptides/therapeutic use , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/physiology , HeLa Cells , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Maleimides/pharmacology , Maleimides/therapeutic use , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , Permeability , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Proteolysis/drug effects , Pyridines/pharmacology , Pyridines/therapeutic use , Signal Transduction/drug effects , Thiazoles/pharmacology , Thiazoles/therapeutic use , Versicans/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Vitreous Body/metabolism , rho-Associated Kinases/metabolism
2.
J Biol Chem ; 292(35): 14381-14390, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28684419

ABSTRACT

To examine the biochemical influences that may contribute to the success of gene therapy for ocular disorders, the role of versican, a vitreous component, in adenoviral-mediated transgene expression was examined. Versican is a large chondroitin sulfate-containing, hyaluronic acid-binding proteoglycan present in the extracellular matrix and in ocular vitreous body. Y79 retinoblastoma cells and CD44-negative SK-N-DZ neuroblastoma cells transduced with adenoviral vectors in the presence of versican respond with an activation of transgene expression. Proteolysis of versican generates a hyaluronan-binding G1 domain. The addition of recombinant versican G1 to SK-N-DZ cells results in a similar activation of transgene expression, and treatment with dasatinib, an inhibitor of Src family kinases, also mimics the effects of versican. Enhancement is accompanied by an increase in signal transducer and activator of transcription 5 (STAT5) phosphorylation and is abrogated by treatment with C188-9, a STAT3/5 inhibitor, or with ruxolitinib, a Janus kinase 1/2 (JAK1/2) inhibitor. These data implicate versican G1 in enhancing adenoviral vector transgene expression in a hyaluronic acid-CD44 independent manner that is down-regulated by inhibitors of the JAK/STAT pathway and enhanced by inhibitors of the Src kinase pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/therapy , Protein Kinase Inhibitors/pharmacology , Versicans/metabolism , Adenoviridae/growth & development , Adenoviridae/physiology , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , DNA, Recombinant/metabolism , DNA, Viral/metabolism , Genes, Reporter/drug effects , Genetic Vectors , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasms/metabolism , Neoplasms/virology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Versicans/chemistry , Versicans/genetics , Virus Replication/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
3.
PLoS One ; 11(6): e0157411, 2016.
Article in English | MEDLINE | ID: mdl-27299810

ABSTRACT

PURPOSE: To test the efficacy of systemic gene delivery of a mutant form of erythropoietin (EPO-R76E) that has attenuated erythropoietic activity, in a mouse model of autosomal dominant retinitis pigmentosa. METHODS: Ten-day old mice carrying one copy of human rhodopsin with the P23H mutation and both copies of wild-type mouse rhodopsin (hP23H RHO+/-,mRHO+/+) were injected into the quadriceps with recombinant adeno-associated virus (rAAV) carrying either enhanced green fluorescent protein (eGFP) or EpoR76E. Visual function (electroretinogram) and retina structure (optical coherence tomography, histology, and immunohistochemistry) were assessed at 7 and 12 months of age. RESULTS: The outer nuclear layer thickness decreased over time at a slower rate in rAAV.EpoR76E treated as compared to the rAAV.eGFP injected mice. There was a statistically significant preservation of the electroretinogram at 7, but not 12 months of age. CONCLUSIONS: Systemic EPO-R76E slows death of the photoreceptors and vision loss in hP23H RHO+/-,mRHO+/+ mice. Treatment with EPO-R76E may widen the therapeutic window for retinal degeneration patients by increasing the number of viable cells. Future studies might investigate if co-treatment with EPO-R76E and gene replacement therapy is more effective than gene replacement therapy alone.


Subject(s)
Erythropoietin/genetics , Gene Transfer Techniques , Genetic Therapy , Retinal Cone Photoreceptor Cells/pathology , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/therapy , Animals , Cell Death , Dependovirus/genetics , Disease Models, Animal , Erythropoietin/pharmacokinetics , Erythropoietin/therapeutic use , Humans , Mice , Opsins/analysis , Point Mutation , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/physiopathology , Vision, Ocular
4.
J Neuroinflammation ; 13: 39, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26876380

ABSTRACT

BACKGROUND: Glaucoma is a complex neurodegeneration and a leading cause of blindness worldwide. Current therapeutic strategies, which are all directed towards lowering the intraocular pressure (IOP), do not stop progression of the disease. We have demonstrated that recombinant adeno-associated virus (rAAV) gene delivery of a form of erythropoietin with attenuated erythropoietic activity (EpoR76E) can preserve retinal ganglion cells, their axons, and vision without decreasing IOP. The goal of this study was to determine if modulation of neuroinflammation or oxidative stress played a role in the neuroprotective activity of EPO.R76E. METHODS: Five-month-old DBA/2J mice were treated with either rAAV.EpoR76E or a control vector and collected at 8 months of age. Neuroprotection was assessed by quantification of axon transport and visual evoked potentials. Microglia number and morphology and cytokine and chemokine levels were quantified. Message levels of oxidative stress-related proteins were assessed. RESULTS: Axon transport and visual evoked potentials were preserved in rAAV.EpoR76E-treated mice. The number of microglia was decreased in retinas from 8-month-old rAAV.EpoR76E-treated mice, but proliferation was unaffected. The blood-retina barrier was also unaffected by treatment. Levels of some pro-inflammatory cytokines were decreased in retinas from rAAV.EpoR76E-treated mice including IL-1, IL-12, IL-13, IL-17, CCL4, and CCL5. TNFα messenger RNA (mRNA) was increased in retinas from 8-month-old mice compared to 3-month-old controls regardless of treatment. Expression of several antioxidant proteins was increased in retinas of rAAV.EpoR76E-treated 8-month-old mice. CONCLUSIONS: Treatment with rAAV.EpoR76E preserves vision in the DBA/2J model of glaucoma at least in part by decreasing infiltration of peripheral immune cells, modulating microglial reactivity, and decreasing oxidative stress.


Subject(s)
Cytokines/metabolism , Erythropoietin/therapeutic use , Genetic Therapy/methods , Glaucoma/complications , Glaucoma/therapy , Oxidative Stress/physiology , Animals , Calcium-Binding Proteins , Cholera Toxin/metabolism , Cytokines/genetics , Dependovirus/genetics , Disease Models, Animal , Erythropoietin/genetics , Evoked Potentials, Visual/physiology , Fluorescein Angiography , Gene Expression Regulation/physiology , Ki-67 Antigen/metabolism , Mice , Mice, Inbred DBA , Microfilament Proteins , Microglia/pathology , Photic Stimulation , Retina/pathology , Transduction, Genetic
5.
Mol Ther ; 24(2): 230-239, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26502777

ABSTRACT

Glaucoma, a common cause of blindness, is currently treated by intraocular pressure (IOP)-lowering interventions. However, this approach is insufficient to completely prevent vision loss. Here, we evaluate an IOP-independent gene therapy strategy using a modified erythropoietin, EPO-R76E, which has reduced erythropoietic function. We used two models of glaucoma, the murine microbead occlusion model and the DBA/2J mouse. Systemic recombinant adeno-associated virus-mediated gene delivery of EpoR76E (rAAV.EpoR76E) was performed concurrent with elevation of IOP. Axon structure and active anterograde transport were preserved in both models. Vision, as determined by the flash visual evoked potential, was preserved in the DBA/2J. These results show that systemic EpoR76E gene therapy protects retinal ganglion cells from glaucomatous degeneration in two different models. This suggests that EPO targets a component of the neurodegenerative pathway that is common to both models. The efficacy of rAAV.EpoR76E delivered at onset of IOP elevation supports clinical relevance of this treatment.


Subject(s)
Axons/pathology , Erythropoietin/genetics , Glaucoma/therapy , Mutation , Optic Nerve/pathology , Animals , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Genetic Vectors/administration & dosage , Glaucoma/genetics , Glaucoma/pathology , Humans , Intraocular Pressure , Mice
6.
Front Immunol ; 5: 523, 2014.
Article in English | MEDLINE | ID: mdl-25374571

ABSTRACT

Neuroinflammation is a normal and healthy response to neuronal damage. However, excessive or chronic neuroinflammation exacerbates neurodegeneration after trauma and in progressive diseases such as Alzheimer's, Parkinson's, age-related macular degeneration, and glaucoma. Therefore, molecules that modulate neuroinflammation are candidates as neuroprotective agents. Erythropoietin (EPO) is a known neuroprotective agent that indirectly attenuates neuroinflammation, in part, by inhibiting neuronal apoptosis. In this review, we provide evidence that EPO also modulates neuroinflammation upstream of apoptosis by acting directly on glia. Further, the signaling induced by EPO may differ depending on cell type and context possibly as a result of activation of different receptors. While significant progress has been made in our understanding of EPO signaling, this review also identifies areas for future study in terms of the role of EPO in modulating neuroinflammation.

7.
PLoS One ; 8(6): e63519, 2013.
Article in English | MEDLINE | ID: mdl-23826078

ABSTRACT

Verification that cell lines used for cancer research are derived from malignant cells in primary tumors is imperative to avoid invalidation of study results. Retinoblastoma is a childhood ocular tumor that develops from loss of functional retinoblastoma protein (pRb) as a result of genetic or epigenetic changes that affect both alleles of the RB1 gene. These patients contain unique identifiable genetic signatures specifically present in malignant cells. Primary cultures derived from retinoblastoma tumors can be established as non-adherent tumorspheres when grown in defined media or as attached monolayers when grown in serum-containing media. While the RB1 genotypes of tumorspheres match those of the primary tumor, adherent cultures have the germline RB1 genotype. Tumorspheres derived from pRb-negative tumors do not express pRb and express the neuroendocrine tumor markers synaptophysin and microtubule-associated protein 2 (MAP2). Adherent cells are synaptophysin-negative and express pRb, the epithelial cell marker cytokeratin that is expressed in the retinal pigmented epithelium and the vascular endothelial cell marker CD34. While tumorspheres are of malignant origin, our results cast doubt on the assumption that adherent tumor-derived cultures are always valid in vitro models of malignant cells and emphasize the need for validation of primary tumor cultures.


Subject(s)
Cell Adhesion , Eye Neoplasms/pathology , Retinoblastoma/pathology , Cell Differentiation , Eye Neoplasms/genetics , Genes, Retinoblastoma , Genotype , Humans , Mutation , Retinoblastoma/genetics , Tumor Cells, Cultured
8.
J Biol Chem ; 287(39): 32697-707, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22865879

ABSTRACT

The success of gene therapy in the ocular environment is partly due to the presence of hyaluronan in vitreous. Here we explore the mechanism of hyaluronan-mediated enhancement of adenoviral vector transgene expression. Introduction of hyaluronan receptor CD44 into CD44-negative cells followed by transduction in the presence of vitreous with an adenoviral vector containing an IL-12-coding transgene increases IL-12 secretion. We demonstrate that sequential CD44 proteolysis is responsible for hyaluronan-mediated enhancement. Metalloproteinase or γ-secretase inhibitors decrease adenoviral-mediated transgene expression. Deletion of these proteolytic sites in CD44 also inhibits transgene expression. Expression of CD44 with a mutation to prevent phosphorylation of serine 325 inhibits the response to vitreous. Expression of the CD44 intracellular domain enhances transgene expression in the absence of vitreous. CD44-mediated enhancement of gene expression was observed with vectors using different promoters and appears because of an increase in mRNA production, not because of an increase in vector transduction as determined by quantitative RT-PCR and quantitative PCR, respectively. These data fit a model where the interaction of hyaluronan in vitreous and CD44 modulates transgene expression by initiating CD44 proteolysis and release of the cytoplasmic domain, resulting in increased transgene transcription.


Subject(s)
Adenoviridae , Gene Expression , Genetic Vectors , Hyaluronan Receptors/metabolism , Proteolysis , Transgenes , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Humans , Hyaluronan Receptors/genetics , Hyaluronic Acid/genetics , Hyaluronic Acid/metabolism , Interleukin-12/biosynthesis , Interleukin-12/genetics , Mutation , Promoter Regions, Genetic , Protein Structure, Tertiary , Transcription, Genetic
9.
Invest Ophthalmol Vis Sci ; 53(7): 3454-62, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22562503

ABSTRACT

PURPOSE: Human retinoblastomas form during the proliferative phase of retina development and are caused by mutations that result in absent or functionally defective Rb protein. Similar tumors occur in mice only when multiple Rb gene family members are absent. We asked if retinal tumors can arise from an undifferentiated retinal cell. The tumor-initiating cells isolated from these tumors that formed in early embryonic murine retinas were characterized. METHODS: Transgenic mice were created using a Pax6 promoter to target expression of SV40 large T-antigen (T-Ag) in the undifferentiated murine embryonic retina. T-Ag, which sequesters all Rb family proteins and p53, is expressed in the retina and lens by murine embryonic day 10 (E10) and tumors are observed by E12.5. A cell line that is adherent in serum-containing media and forms neurospheres in supplemented serum-free media was developed from retinal tumors isolated on postnatal day 7. RESULTS: In all, 1.5% of attached cells form neurospheres when transferred to serum-free medium. All cultured cells express T-Ag, confirming that they derive from the original tumors; 0.5% of adherent cells express detectable levels of CD133. CD133+ FACS-sorted cells cultured in serum-free medium form 3-fold more neurospheres than do CD133- cells. Six of seven mice injected with CD133+ cells and one of seven mice injected with CD133- cells formed tumors during a 6-month period. Unlike primary adherent cells, primary and secondary tumors heterogeneously express markers of stem cells and differentiation similar to human retinoblastoma. CONCLUSIONS: CD133+ tumor-initiating cells can originate from proliferating undifferentiated precursor cells.


Subject(s)
Antigens, CD/metabolism , Antigens, Polyomavirus Transforming/genetics , Cell Transformation, Neoplastic/pathology , Glycoproteins/metabolism , Peptides/metabolism , Retina/embryology , Retinal Neoplasms/embryology , Retinoblastoma/embryology , AC133 Antigen , Animals , Disease Models, Animal , Eye Proteins/genetics , Flow Cytometry , Homeodomain Proteins/genetics , Humans , Immunophenotyping , Mice , Mice, Transgenic , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Retinoblastoma/metabolism , Retinoblastoma/pathology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
10.
J Vis Exp ; (54)2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21847079

ABSTRACT

Culturing retinoblastoma tumor cells in defined stem cell media gives rise to primary tumorspheres that can be grown and maintained for only a limited time. These cultured tumorspheres may exhibit markedly different cellular phenotypes when compared to the original tumors. Demonstration that cultured cells have the capability of forming new tumors is important to ensure that cultured cells model the biology of the original tumor. Here we present a protocol for propagating human retinoblastoma tumors in vivo using Rag2(-/-) immune deficient mice. Cultured human retinoblastoma tumorspheres of low passage or cells obtained from freshly harvested human retinoblastoma tumors injected directly into the vitreous cavity of murine eyes form tumors within 2-4 weeks. These tumors can be harvested and either further passaged into murine eyes in vivo or grown as tumorspheres in vitro. Propagation has been successfully carried out for at least three passages thus establishing a continuing source of human retinoblastoma tissue for further experimentation.


Subject(s)
Neoplasm Transplantation/methods , Retinal Neoplasms/immunology , Retinal Neoplasms/pathology , Retinoblastoma/immunology , Retinoblastoma/pathology , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Humans , Mice , Neoplasm Transplantation/immunology , Transplantation, Heterologous , Tumor Cells, Cultured
11.
Tissue Eng ; 13(2): 343-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17518568

ABSTRACT

Unlike established cell lines used in the biotechnology industry, primary cells used in tissue engineering require culture media to be supplemented with serum. The most common serum is fetal bovine serum (FBS); however, FBS is expensive, negatively affecting process economics. Less-costly alternative sera are commercially available, but their efficacy has not been documented. Therefore, bovine calf serum (BCS), bovine growth serum (BGS), and newborn calf serum (NCS) were compared with FBS. Porcine aortic valve interstitial cells (VICs) were cultured as 2-dimensional (2-D) monolayers or as 3-dimensional (3-D) collagen gels using medium supplemented with 10% FBS, BGS, BCS, or NCS. No significant difference was seen in cellular activity between VICs cultured in BCS and those cultured in FBS in 2-D cultures, whereas cells cultured in BGS and NCS had significantly lower specific growth rates coupled with markedly higher metabolic activity than cells cultured in FBS. No statistically significant differences were seen in cellular activity between any of the sera when cells were cultured in 3-D constructs. In conclusion, BCS is a suitable alternative to FBS for the 2-D and 3-D culture of VICs, which may be used to develop a tissue-engineered valve.


Subject(s)
Aortic Valve/cytology , Aortic Valve/physiology , Bioprosthesis , Heart Valve Prosthesis , Serum/metabolism , Tissue Engineering/methods , Animals , Cell Culture Techniques/methods , Cells, Cultured , Culture Media/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...