Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
Nat Commun ; 12(1): 4219, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244505

ABSTRACT

Streptococcus pyogenes (Spy) Cas9 has potential as a component of gene therapeutics for incurable diseases. One of its limitations is its large size, which impedes its formulation and delivery in therapeutic applications. Smaller Cas9s are an alternative, but lack robust activity or specificity and frequently recognize longer PAMs. Here, we investigated four uncharacterized, smaller Cas9s and found three employing a "GG" dinucleotide PAM similar to SpyCas9. Protein engineering generated synthetic RNA-guided nucleases (sRGNs) with editing efficiencies and specificities exceeding even SpyCas9 in vitro and in human cell lines on disease-relevant targets. sRGN mRNA lipid nanoparticles displayed manufacturing advantages and high in vivo editing efficiency in the mouse liver. Finally, sRGNs, but not SpyCas9, could be packaged into all-in-one AAV particles with a gRNA and effected robust in vivo editing of non-human primate (NHP) retina photoreceptors. Human gene therapy efforts are expected to benefit from these improved alternatives to existing CRISPR nucleases.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Gene Editing/methods , Staphylococcus/enzymology , Animals , CRISPR-Associated Protein 9/isolation & purification , Cell Line, Tumor , Dependovirus , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HEK293 Cells , Humans , Macaca fascicularis , Male , Mice , Parvovirinae/genetics , Protein Engineering , Ribonucleases , Staphylococcus/genetics , Substrate Specificity , Usher Syndromes/genetics , Usher Syndromes/therapy , RNA, Guide, CRISPR-Cas Systems
3.
Sci Signal ; 12(568)2019 02 12.
Article in English | MEDLINE | ID: mdl-30755477

ABSTRACT

DNA-alkylating agents are commonly used to kill cancer cells, but the base excision repair (BER) pathway they trigger can also produce toxic intermediates that cause tissue damage, such as retinal degeneration (RD). Apoptosis, a process of programmed cell death, is assumed to be the main mechanism of this alkylation-induced photoreceptor (PR) cell death in RD. Here, we studied the involvement of necroptosis (another programmed cell death process) and inflammation in alkylation-induced RD. Male mice exposed to a methylating agent exhibited a reduced number of PR cell rows, active gliosis, and cytokine induction and macrophage infiltration in the retina. Dying PRs exhibited a necrotic morphology, increased 8-hydroxyguanosine abundance (an oxidative damage marker), and overexpression of the necroptosis-associated genes Rip1 and Rip3 The activity of PARP1, which mediates BER, cell death, and inflammation, was increased in PR cells and associated with the release of proinflammatory chemokine HMGB1 from PR nuclei. Mice lacking the anti-inflammatory cytokine IL-10 exhibited more severe RD, whereas deficiency of RIP3 (also known as RIPK3) conferred partial protection. Female mice were partially protected from alkylation-induced RD, showing reduced necroptosis and inflammation compared to males. PRs in mice lacking the BER-initiating DNA glycosylase AAG did not exhibit alkylation-induced necroptosis or inflammation. Our findings show that AAG-initiated BER at alkylated DNA bases induces sex-dependent RD primarily by triggering necroptosis and activating an inflammatory response that amplifies the original damage and, furthermore, reveal new potential targets to prevent this side effect of chemotherapy.


Subject(s)
DNA Glycosylases/metabolism , DNA Repair , Inflammation/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Retinal Degeneration/metabolism , Animals , Antineoplastic Agents, Alkylating/adverse effects , Apoptosis/drug effects , Apoptosis/genetics , Cell Death/drug effects , Cell Death/genetics , DNA Glycosylases/genetics , Female , Inflammation/genetics , Inflammation/pathology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Retinal Degeneration/chemically induced , Retinal Degeneration/genetics
4.
Oncotarget ; 8(40): 68707-68720, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978150

ABSTRACT

Alkylating agents are commonly used to treat cancer. Although base excision repair (BER) is a major pathway for repairing DNA alkylation damage, under certain conditions, the initiation of BER produces toxic repair intermediates that damage healthy tissues. The initiation of BER by the alkyladenine DNA glycosylase (AAG, a.k.a. MPG) can mediate alkylation-induced cytotoxicity in specific cells in the retina and cerebellum of male mice. Cytotoxicity in both wild-type and Aag-transgenic (AagTg) mice is abrogated in the absence of Poly(ADP-ribose) polymerase-1 (PARP1). Here, we tested whether PARP inhibitors can also prevent alkylation-induced retinal and cerebellar degeneration in male and female WT and AagTg mice. Importantly, we found that WT mice display sex-dependent alkylation-induced retinal damage (but not cerebellar damage), with WT males being more sensitive than females. Accordingly, estradiol treatment protects males against alkylation-induced retinal degeneration. In AagTg male and female mice, the alkylation-induced tissue damage in both the retina and cerebellum is exacerbated and the sex difference in the retina is abolished. PARP inhibitors, much like Parp1 gene deletion, protect against alkylation-induced AAG-dependent neuronal degeneration in WT and AagTg mice, regardless of the gender, but their efficacy in preventing alkylation-induced neuronal degeneration depends on PARP inhibitor characteristics and doses. The recent surge in the use of PARP inhibitors in combination with cancer chemotherapeutic alkylating agents might represent a powerful tool for obtaining increased therapeutic efficacy while avoiding the collateral effects of alkylating agents in healthy tissues.

5.
Cell Death Dis ; 8(7): e2947, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28726787

ABSTRACT

Regulated necrosis has emerged as a major cell death mechanism in response to different forms of physiological and pharmacological stress. The AlkB homolog 7 (ALKBH7) protein is required for regulated cellular necrosis in response to chemotherapeutic alkylating agents but its role within a whole organism is unknown. Here, we show that ALKBH7 modulates alkylation-induced cellular death through a tissue and sex-specific mechanism. At the whole-animal level, we find that ALKBH7 deficiency confers increased resistance to MMS-induced toxicity in male but not female mice. Moreover, ALKBH7-deficient mice exhibit protection against alkylation-mediated cytotoxicity in retinal photoreceptor and cerebellar granule cells, two cell types that undergo necrotic death through the initiation of the base excision repair pathway and hyperactivation of the PARP1/ARTD1 enzyme. Notably, the protection against alkylation-induced cerebellar degeneration is specific to ALKBH7-deficient male but not female mice. Our results uncover an in vivo role for ALKBH7 in mediating a sexually dimorphic tissue response to alkylation damage that could influence individual responses to chemotherapies based upon alkylating agents.


Subject(s)
AlkB Enzymes/metabolism , Alkylating Agents/adverse effects , Photoreceptor Cells, Vertebrate/metabolism , Sex Characteristics , Spinocerebellar Degenerations/chemically induced , AlkB Enzymes/genetics , Alkylating Agents/pharmacology , Animals , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Necrosis , Photoreceptor Cells, Vertebrate/pathology , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/metabolism , Spinocerebellar Degenerations/pathology
6.
Oncotarget ; 7(29): 44950-44965, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27391435

ABSTRACT

Nephrotoxicity is a common toxic side-effect of chemotherapeutic alkylating agents. Although the base excision repair (BER) pathway is essential in repairing DNA alkylation damage, under certain conditions the initiation of BER produces toxic repair intermediates that damage healthy tissues. We have shown that the alkyladenine DNA glycosylase, Aag (a.k.a. Mpg), an enzyme that initiates BER, mediates alkylation-induced whole-animal lethality and cytotoxicity in the pancreas, spleen, retina, and cerebellum, but not in the kidney. Cytotoxicity in both wild-type and Aag-transgenic mice (AagTg) was abrogated in the absence of Poly(ADP-ribose) polymerase-1 (Parp1). Here we report that Parp1-deficient mice expressing increased Aag (AagTg/Parp1-/-) develop sex-dependent kidney failure upon exposure to the alkylating agent, methyl methanesulfonate (MMS), and suffer increased whole-animal lethality compared to AagTg and wild-type mice. Macroscopic, histological, electron microscopic and immunohistochemical analyses revealed morphological kidney damage including dilated tubules, proteinaceous casts, vacuolation, collapse of the glomerular tuft, and deterioration of podocyte structure. Moreover, mice exhibited clinical signs of kidney disease indicating functional damage, including elevated blood nitrogen urea and creatinine, hypoproteinemia and proteinuria. Pharmacological Parp inhibition in AagTg mice also resulted in sensitivity to MMS-induced nephrotoxicity. These findings provide in vivo evidence that Parp1 modulates Aag-dependent MMS-induced nephrotoxicity in a sex-dependent manner and highlight the critical roles that Aag-initiated BER and Parp1 may play in determining the side-effects of chemotherapeutic alkylating agents.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , DNA Glycosylases/metabolism , Kidney/drug effects , Poly (ADP-Ribose) Polymerase-1/metabolism , Alkylation , Animals , DNA Damage/drug effects , DNA Repair/drug effects , Female , Male , Mice , Mice, Transgenic , Sex Characteristics
7.
Proc Natl Acad Sci U S A ; 111(45): E4878-86, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25349415

ABSTRACT

Inflammation is accompanied by the release of highly reactive oxygen and nitrogen species (RONS) that damage DNA, among other cellular molecules. Base excision repair (BER) is initiated by DNA glycosylases and is crucial in repairing RONS-induced DNA damage; the alkyladenine DNA glycosylase (Aag/Mpg) excises several DNA base lesions induced by the inflammation-associated RONS release that accompanies ischemia reperfusion (I/R). Using mouse I/R models we demonstrate that Aag(-/-) mice are significantly protected against, rather than sensitized to, I/R injury, and that such protection is observed across three different organs. Following I/R in liver, kidney, and brain, Aag(-/-) mice display decreased hepatocyte death, cerebral infarction, and renal injury relative to wild-type. We infer that in wild-type mice, Aag excises damaged DNA bases to generate potentially toxic abasic sites that in turn generate highly toxic DNA strand breaks that trigger poly(ADP-ribose) polymerase (Parp) hyperactivation, cellular bioenergetics failure, and necrosis; indeed, steady-state levels of abasic sites and nuclear PAR polymers were significantly more elevated in wild-type vs. Aag(-/-) liver after I/R. This increase in PAR polymers was accompanied by depletion of intracellular NAD and ATP levels plus the translocation and extracellular release of the high-mobility group box 1 (Hmgb1) nuclear protein, activating the sterile inflammatory response. We thus demonstrate the detrimental effects of Aag-initiated BER during I/R and sterile inflammation, and present a novel target for controlling I/R-induced injury.


Subject(s)
Brain/enzymology , DNA Glycosylases/metabolism , DNA Repair , Kidney/enzymology , Liver/enzymology , Reperfusion Injury/enzymology , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Brain/pathology , Brain Infarction/enzymology , Brain Infarction/genetics , Brain Infarction/pathology , Cell Death , DNA Damage , DNA Glycosylases/genetics , Enzyme Induction/genetics , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Hepatocytes/enzymology , Hepatocytes/pathology , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Kidney/pathology , Liver/pathology , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology
8.
PLoS One ; 6(7): e22166, 2011.
Article in English | MEDLINE | ID: mdl-21818300

ABSTRACT

BACKGROUND: Gene transfer using adeno-associated viral (AAV) vectors has been successfully applied in the retina for the treatment of inherited retinal dystrophies. Recently, microRNAs have been exploited to fine-tune transgene expression improving therapeutic outcomes. Here we evaluated the ability of retinal-expressed microRNAs to restrict AAV-mediated transgene expression to specific retinal cell types that represent the main targets of common inherited blinding conditions. METHODOLOGY/PRINCIPAL FINDINGS: To this end, we generated AAV2/5 vectors expressing EGFP and containing four tandem copies of miR-124 or miR-204 complementary sequences in the 3'UTR of the transgene expression cassette. These vectors were administered subretinally to adult C57BL/6 mice and Large White pigs. Our results demonstrate that miR-124 and miR-204 target sequences can efficiently restrict AAV2/5-mediated transgene expression to retinal pigment epithelium and photoreceptors, respectively, in mice and pigs. Interestingly, transgene restriction was observed at low vector doses relevant to therapy. CONCLUSIONS: We conclude that microRNA-mediated regulation of transgene expression can be applied in the retina to either restrict to a specific cell type the robust expression obtained using ubiquitous promoters or to provide an additional layer of gene expression regulation when using cell-specific promoters.


Subject(s)
Gene Expression Regulation , MicroRNAs/metabolism , Retina/metabolism , Transgenes/genetics , Animals , Base Sequence , Dependovirus/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Molecular Sequence Data , Organ Specificity/genetics , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/metabolism , Retina/cytology , Retinal Pigment Epithelium/metabolism , Sus scrofa , Transduction, Genetic
9.
Invest Ophthalmol Vis Sci ; 52(8): 5713-9, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21273543

ABSTRACT

PURPOSE: Mutations in the PDE6B gene cause recessive, severe retinitis pigmentosa (RP). PDE6B encodes the ß subunit of the rod-specific phosphodiesterase (ßPDE), which, when absent, results in toxic levels of intracellular Ca(2+) and photoreceptor cell death. Ca(2+) blockers, such as nilvadipine, as well as light restriction, slow photoreceptor degeneration in animal models of ßPDE deficiencies. The goal of the study was to evaluate the efficacy of AAV2/5- or AAV2/8-mediated gene replacement in combination with nilvadipine and/or with light restriction in the rd10 mouse bearing homozygous pde6b mutations. METHODS: AAV vectors encoding either ßPDE or EGFP were subretinally administered at postnatal day (P)2. Nilvadipine was administered from P7 to P28. For light restriction, pregnant rd10 mice were kept in a dark environment until their pups were 28 days old. All functional and histologic analyses were performed at P35. RESULTS: Significant morphologic photoreceptor protection was observed after subretinal administration of AAV vectors encoding EGFP. This protection further increased after administration of AAV2/8 or -2/5 encoding for ßPDE and was not associated with significant functional improvement. Photoreceptor protection was higher after AAV2/8- than after AAV2/5-mediated delivery and was not significantly augmented by additional drug therapy and/or light restriction. The protective effect was lost after P35. CONCLUSIONS: In conclusion, more efficient gene transfer tools than those used in this study, as well as a better understanding of the disease pathogenesis, should be explored to increase the effect of gene replacement and to design gene-based strategies that block the apoptotic pathways activated by ßPDE deficiency.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Genetic Therapy/methods , Nifedipine/analogs & derivatives , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Animals , COS Cells , Calcium Channel Blockers/pharmacology , Chlorocebus aethiops , Combined Modality Therapy , Darkness , Dependovirus/genetics , Disease Models, Animal , Female , Genetic Vectors/genetics , Homozygote , Intravitreal Injections , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nifedipine/pharmacology , Photoreceptor Cells, Vertebrate/pathology , Pregnancy , Retinitis Pigmentosa/drug therapy
10.
J Clin Invest ; 118(5): 1955-64, 2008 May.
Article in English | MEDLINE | ID: mdl-18414684

ABSTRACT

Vectors derived from adeno-associated virus (AAV) are promising for human gene therapy, including treatment for retinal blindness. One major limitation of AAVs as vectors is that AAV cargo capacity has been considered to be restricted to 4.7 kb. Here we demonstrate that vectors with an AAV5 capsid (i.e., rAAV2/5) incorporated up to 8.9 kb of genome more efficiently than 6 other serotypes tested, independent of the efficiency of the rAAV2/5 production process. Efficient packaging of the large murine Abca4 and human MYO7A and CEP290 genes, which are mutated in common blinding diseases, was obtained, suggesting that this packaging efficiency is independent of the specific sequence packaged. Expression of proteins of the appropriate size and function was observed following transduction with rAAV2/5 carrying large genes. Intraocular administration of rAAV2/5 encoding ABCA4 resulted in protein localization to rod outer segments and significant and stable morphological and functional improvement of the retina in Abca4(-/-) mice. This use of rAAV2/5 may be a promising therapeutic strategy for recessive Stargardt disease, the most common form of inherited macular degeneration. The possibility of packaging large genes in AAV greatly expands the therapeutic potential of this vector system.


Subject(s)
Dependovirus , Gene Transfer Techniques , Genetic Vectors , Retina , Serotyping , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Cycle Proteins , Cytoskeletal Proteins , Dependovirus/genetics , Dependovirus/metabolism , Dyneins/genetics , Dyneins/metabolism , Electroretinography , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Sequence Data , Myosin VIIa , Myosins/genetics , Myosins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Retina/cytology , Retina/metabolism
11.
Invest Ophthalmol Vis Sci ; 48(11): 5199-206, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17962474

ABSTRACT

PURPOSE: Delivery of glial cell-derived neurotrophic factor (GDNF), either as a recombinant protein or by retinal gene transfer results in photoreceptor (PR) neuroprotection in genetic models of retinitis pigmentosa (RP). The mechanism of GDNF action and its direct targets in the retina remain unknown. The goal of the present study was to test the neuroprotective effect of GDNF from light-induced damage, a commonly used stimulus of PR degeneration, and to determine whether protection occurs directly on PRs. METHODS: Adeno-associated viral vectors (AAV) were developed that expressed either GDNF or a constitutively (RetMen2A) or pharmacologically activated chimeric GDNF receptor (Fv2Ret). Fv2Ret homodimerization and activation are induced by the administration of the small dimerizer drug AP20187. AAV2/2 vectors and the cytomegalovirus (CMV) promoter were used to transduce GDNF in the retina, whereas RetMen2A and Fv2Ret were transduced by AAV2/5 vectors and their expression restricted to PRs by the rhodopsin promoter. In vivo GDNF levels were measured by ELISA, RetMen2A and Fv2Ret expression and activation in vitro and/or in vivo were assessed by Western blot and immunofluorescence analyses. ERG measurements and histologic analyses were performed to assess morphologic and functional rescue, respectively. RESULTS: GDNF gene transfer resulted in sustained protein expression in the eye. In addition, the results confirmed in vivo that PR-restricted activation of Ret signaling occurred after either AAV-mediated expression of RetMen2A or AP20187-dependent Fv2Ret activation. However, this or AAV-mediated GDNF retinal gene transfer did not result in functional or morphologic PR protection from light-induced damage. CONCLUSIONS: The results suggest that the apoptotic pathways responsible for light-induced PR degeneration are not inhibited by GDNF. However, GDNF signaling was shown to be regulated in time and levels in the retina by the AP20187/Fv2Ret system which is therefore available to be tested as gene-based therapeutic strategy in models of PR degeneration responsive to GDNF.


Subject(s)
Immunosuppressive Agents/pharmacology , Light , Photoreceptor Cells, Vertebrate/drug effects , Proto-Oncogene Proteins c-ret/metabolism , Radiation Injuries, Experimental/prevention & control , Retinal Degeneration/prevention & control , Tacrolimus/analogs & derivatives , Animals , Blotting, Western , Dependovirus/genetics , Electroretinography , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , Glial Cell Line-Derived Neurotrophic Factor/genetics , Male , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Multiple Endocrine Neoplasia Type 2a/genetics , Phosphorylation , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/radiation effects , Plasmids/genetics , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Tacrolimus/pharmacology , Transfection
12.
J Virol ; 81(20): 11372-80, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17699581

ABSTRACT

Severe inherited retinal diseases, such as retinitis pigmentosa and Leber congenital amaurosis, are caused by mutations in genes preferentially expressed in photoreceptors. While adeno-associated virus (AAV)-mediated gene transfer can correct retinal pigment epithelium (RPE) defects in animal models, approaches for the correction of photoreceptor-specific diseases are less efficient. We evaluated the ability of novel AAV serotypes (AAV2/7, AAV2/8, AAV2/9, AAV2rh.43, AAV2rh.64R1, and AAV2hu.29R) in combination with constitutive or photoreceptor-specific promoters to improve photoreceptor transduction, a limiting step in photoreceptor rescue. Based on a qualitative analysis, all AAV serotypes tested efficiently transduce the RPE as well as rod and cone photoreceptors after subretinal administration in mice. Interestingly, AAV2/9 efficiently transduces Müller cells. To compare photoreceptor transduction from different AAVs and promoters in both a qualitative and quantitative manner, we designed a strategy based on the use of a bicistronic construct expressing both enhanced green fluorescent protein and luciferase. We found that AAV2/8 and AAV2/7 mediate six- to eightfold higher levels of in vivo photoreceptor transduction than AAV2/5, considered so far the most efficient AAV serotype for photoreceptor targeting. In addition, following subretinal administration of AAV, the rhodopsin promoter allows significantly higher levels of photoreceptor expression than the other ubiquitous or photoreceptor-specific promoters tested. Finally, we show that AAV2/7, AAV2/8, and AAV2/9 outperform AAV2/5 following ex vivo transduction of retinal progenitor cells differentiated into photoreceptors. We conclude that AAV2/7 or AAV2/8 and the rhodopsin promoter provide the highest levels of photoreceptor transduction both in and ex vivo and that this may overcome the limitation to therapeutic success observed so far in models of inherited severe photoreceptor diseases.


Subject(s)
Dependovirus/genetics , Photoreceptor Cells/metabolism , Transduction, Genetic/methods , Animals , Genetic Vectors , Mice , Promoter Regions, Genetic , Retina/cytology , Rhodopsin/genetics , Serotyping , Transduction, Genetic/standards
13.
Mol Ther ; 14(5): 692-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16979943

ABSTRACT

Autosomal dominant retinitis pigmentosa caused by the frequent rhodopsin P23H mutation is characterized by progressive photoreceptor cell death eventually leading to blindness and for which no therapies are available. Considering the gain-of-function effect exerted by the P23H mutation, strategies aimed at silencing the expression of the mutated allele, like RNA interference, are desirable. We have designed small interfering RNAs (siRNA) to silence specifically the P23H rhodopsin allele expressed by a transgenic rat model of the disease. We have selected in vitro one siRNA and generated an adeno-associated viral (AAV) vector expressing the short hairpin RNA (shRNA) based on the selected siRNA. In vitro the shRNA significantly inhibits the expression of the P23H but not the wild-type rhodopsin allele. Subretinal administration of the AAV2/5 vector encoding the shRNA in P23H transgenic rats results in inhibition of rhodopsin P23H expression that is not able to prevent or block photoreceptor degeneration. Since rhodopsin is the most abundant rod photoreceptor protein, systems resulting in more robust shRNA expression in the retina may be required to achieve therapeutic efficacy in vivo.


Subject(s)
Gene Silencing , Mutation/genetics , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Rhodopsin/genetics , Alleles , Animals , Animals, Genetically Modified , Base Sequence , Dependovirus/genetics , Genetic Vectors/genetics , Mice , Models, Animal , Molecular Sequence Data , Proline/genetics , Proline/metabolism , RNA, Small Interfering/genetics , Rats , Retinal Degeneration/metabolism , Rhodopsin/metabolism
14.
Hum Gene Ther ; 17(5): 565-74, 2006 May.
Article in English | MEDLINE | ID: mdl-16716113

ABSTRACT

Duchenne muscular dystrophy (DMD) is a X-linked myopathy in which deletions and point mutations in the dystrophin gene abolish dystrophin expression. The defect can often be corrected at the posttranscriptional level by exon skipping. In an animal model of DMD, the mdx mouse, a point mutation in exon 23 of the dystrophin gene introduces a premature stop codon. Skipping of this exon reestablishes the open reading frame in the dystrophin mRNA. We have obtained persistent exon skipping in mdx mice by local muscle injection of AAV vectors expressing antisense sequences fused to either U1 or U7 small nuclear RNA (snRNA). In the transduced muscles, dystrophin expression, amelioration of muscle morphology, and significant force recovery were obtained. These data indicate that the expression of antisense snRNAs, combined with their efficient muscular delivery through AAV vectors, is a powerful strategy for the therapeutic treatment of DMD. Like U7 snRNA, spliceosomal U1 snRNA is also a suitable backbone for the expression of antisense molecules active in exon skipping.


Subject(s)
Dependovirus/genetics , Dystrophin/metabolism , Genetic Therapy , Muscular Dystrophy, Animal/therapy , RNA, Small Nuclear/genetics , Animals , Base Sequence , DNA, Recombinant , Genetic Vectors , Injections, Intramuscular , Mice , Mice, Inbred mdx/genetics , Molecular Sequence Data , Muscle Fibers, Skeletal/physiology , RNA, Antisense/therapeutic use , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use
15.
Proc Natl Acad Sci U S A ; 103(10): 3758-63, 2006 Mar 07.
Article in English | MEDLINE | ID: mdl-16501048

ABSTRACT

Duchenne muscular dystrophy is an X-linked muscle disease characterized by mutations in the dystrophin gene. Many of these can be corrected at the posttranscriptional level by skipping the mutated exon. We have obtained persistent exon skipping in mdx mice by tail vein injection with an adeno-associated viral (AAV) vector expressing antisense sequences as part of the stable cellular U1 small nuclear RNA. Systemic delivery of the AAV construct resulted in effective body-wide colonization, significant recovery of the functional properties in vivo, and lower creatine kinase serum levels, suggesting an overall decrease in muscle wasting. The transduced muscles rescued dystrophin expression and displayed a significant recovery of function toward the normal values at single muscle fiber level. This approach provides solid bases for a systemic use of AAV-mediated antisense-U1 small nuclear RNA expression for the therapeutic treatment of Duchenne muscular dystrophy.


Subject(s)
Genetic Therapy/methods , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Animals , Base Sequence , Dependovirus/genetics , Dystrophin/genetics , Exons , Genetic Vectors , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Molecular Sequence Data , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Oligodeoxyribonucleotides, Antisense/genetics , RNA, Small Nuclear/genetics
16.
Expert Opin Biol Ther ; 6(12): 1279-94, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17223737

ABSTRACT

Vectors based on the adeno-associated virus (rAAV) are able to transduce the retina of animal models, including non-human primates, for a long-term period, safely and at sustained levels. The ability of the various rAAV serotypes to transduce retinal target cells has been exploited to successfully transfer genes to photoreceptors, retinal pigment epithelium and the inner retina, which are affected in many inherited and non-inherited blinding diseases. rAAV-mediated, constitutive and regulated gene expression at therapeutic levels has been achieved in the retina of animal models, thus providing proof-of-principle of gene therapy efficacy and safety in models of dominant and recessive retinal disorders. In addition, gene transfer of molecules with either neurotrophic or antiangiogenic properties provides useful alternatives to the classic gene replacement for treatment of both mendelian and complex traits affecting the retina. Years of successful rAAV-mediated gene transfer to the retina have resulted in restoration of vision in dogs affected with congenital blindness. This has paved the way to the first attempts at treating inherited retinal diseases in humans with rAAV. Although the results of rAAV clinical trials for non-retinal diseases give a warning that the outcome of viral-mediated gene transfer in humans may be different from that predicted based on results in other species, the immune privilege of the retina combined with the versatility of rAAV serotypes may ultimately provide the first successful treatment of human inherited diseases using rAAV.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Retinal Diseases/genetics , Animals , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/therapeutic use , Humans , Retinal Diseases/therapy
17.
Mol Ther ; 10(5): 855-61, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509503

ABSTRACT

Molecules with neurotrophic activity are being evaluated for treatment of retinitis pigmentosa in animal models. In particular, great interest has been focused recently on erythropoietin (Epo). Evidence of its neurotrophic activity comes mainly from data demonstrating photoreceptor protection in a rodent light-damage model through systemic administration of a recombinant form of this hormone. Our goal was to test whether Epo retinal gene transfer can rescue or delay photoreceptor cell death. We delivered adeno-associated viral vectors encoding Epo intraocularly and, for comparison, intramuscularly to one light-induced and two genetic models of retinal degeneration. Intraocular Epo gene transfer resulted in sustained hormone expression in the eye, which was undetectable systemically. In contrast, Epo intramuscular gene transfer resulted in hormone secretion in the circulation, which was not detected in ocular fluids. The protein secreted from muscle and retina is of the same molecular weight as a commercial recombinant human Epo. Interestingly, following systemic but not intraocular Epo delivery, morphological photoreceptor protection was observed in the light-damage and rds/peripherin (Prph2) models of retinal degeneration. In the light-damage model, the morphological rescue was accompanied by a significant electrophysiological improvement of photoreceptor function. In contrast, no photoreceptor rescue was observed following Epo gene transfer in the rd10 model. This suggests that different apoptotic mechanisms, with varying sensitivities to Epo, occur in different retinal degeneration models. In conclusion, our data support Epo as a neuroprotective agent in some, but not all, retinal degenerations. Further, rescue is observed in specific models after systemic but not intraocular Epo gene transfer.


Subject(s)
Dependovirus/genetics , Erythropoietin/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Retinal Degeneration/therapy , Animals , Anterior Chamber/physiology , Cell Nucleus/chemistry , Electroretinography , Erythropoietin/analysis , Gene Transfer Techniques , Intermediate Filament Proteins/genetics , Light , Membrane Glycoproteins/genetics , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Peripherins , Photoreceptor Cells, Vertebrate/physiology , Rats , Rats, Inbred Lew , Retina/chemistry , Retina/metabolism , Retina/pathology , Retinal Degeneration/etiology , Retinal Degeneration/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...