Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 32(2): 101263, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38827250

ABSTRACT

Prenatal somatic cell gene therapy (PSCGT) could potentially treat severe, early-onset genetic disorders such as spinal muscular atrophy (SMA) or muscular dystrophy. Given the approval of adeno-associated virus serotype 9 (AAV9) vectors in infants with SMA by the U.S. Food and Drug Administration, we tested the safety and biodistribution of AAV9-GFP (clinical-grade and dose) in fetal lambs to understand safety and efficacy after umbilical vein or intracranial injection on embryonic day 75 (E75) . Umbilical vein injection led to widespread biodistribution of vector genomes in all examined lamb tissues and in maternal uteruses at harvest (E96 or E140; term = E150). There was robust GFP expression in brain, spinal cord, dorsal root ganglia (DRGs), without DRG toxicity and excellent transduction of diaphragm and quadriceps muscles. However, we found evidence of systemic toxicity (fetal growth restriction) and maternal exposure to the viral vector (transient elevation of total bilirubin and a trend toward elevation in anti-AAV9 antibodies). There were no antibodies against GFP in ewes or lambs. Analysis of fetal gonads demonstrated GFP expression in female (but not male) germ cells, with low levels of integration-specific reads, without integration in select proto-oncogenes. These results suggest potential therapeutic benefit of AAV9 PSCGT for neuromuscular disorders, but warrant caution for exposure of female germ cells.

2.
Toxicol Pathol ; 50(4): 415-431, 2022 06.
Article in English | MEDLINE | ID: mdl-35658751

ABSTRACT

Biodistribution of self-complementary adeno-associated virus-9 (scAAV9)-chicken ß-actin promoter-green fluorescent protein (GFP) was assessed in juvenile cynomolgus macaques infused intrathecally via lumbar puncture or the intracisterna magna (1.0×1013 or 3.0×1013 vg/animal), with necropsy 28 days later. Our results characterized central nervous system biodistribution compared with systemic organs/tissues by droplet digital polymerase chain reaction for DNA and in situ hybridization. Green fluorescent protein expression was characterized by Meso Scale Discovery electrochemiluminescence immunosorbent assay and immunohistochemistry (IHC). Biodistribution was widespread but variable, with vector DNA and GFP expression greatest in the spinal cord, dorsal root ganglia (DRG), and certain systemic tissues (e.g., liver), with low concentrations in many brain regions despite direct cerebrospinal fluid administration. Transduction and expression were observed primarily in perivascular astrocytes in the brain, with a paucity in neurons. Greater GFP expression was observed in hepatocytes, striated myocytes, cardiomyocytes, spinal cord lower motor neurons, and DRG sensory neurons by IHC. These results should be considered when evaluating scAAV9-based intrathecal delivery with the current expression cassette as a modality for neurologic diseases that require widespread brain neuronal expression. This capsid/expression cassette combination may be better suited for diseases that express a secreted protein and/or do not require widespread brain neuronal transduction.


Subject(s)
Dependovirus , Genetic Vectors , Animals , Dependovirus/genetics , Dependovirus/metabolism , Green Fluorescent Proteins/genetics , Macaca fascicularis/genetics , Sensory Receptor Cells , Tissue Distribution
3.
Hum Vaccin Immunother ; 16(9): 2114-2122, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32783701

ABSTRACT

Lyme disease is the most common vector-borne disease in North America. The etiological agent is the spirochete Borreliella burgdorferi, transmitted to mammalian hosts by the Ixodes tick. In recent years there has been an increase in the number of cases of Lyme disease. Currently, there is no vaccine on the market for human use. We describe the development of a novel synthetically engineered DNA vaccine, pLD1 targeting the outer-surface protein A (OspA) of Borreliella burgdorferi. Immunization of C3 H/HeN mice with pLD1 elicits robust humoral and cellular immune responses that confer complete protection against a live Borreliella burgdorferi bacterial challenge. We also assessed intradermal (ID) delivery of pLD1 in Hartley guinea pigs, demonstrating the induction of robust and durable humoral immunity that lasts at least 1 year. We provide evidence of the potency of pLD1 by showing that antibodies targeting the OspA epitopes which have been associated with protection are prominently raised in the immunized guinea pigs. The described study provides the basis for the advancement of pDL1 as a potential vaccine for Lyme disease control.


Subject(s)
Borrelia burgdorferi Group , Borrelia burgdorferi , Lyme Disease , Vaccines, DNA , Animals , Antibodies, Bacterial , Antigens, Surface , Bacterial Outer Membrane Proteins , Bacterial Vaccines , Borrelia burgdorferi/genetics , Guinea Pigs , Lyme Disease/prevention & control , Mice , North America
4.
J Immunol ; 205(3): 648-660, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32591390

ABSTRACT

mAbs are a possible adjunct to vaccination and drugs in treatment of influenza virus infection. However, questions remain whether small animal models accurately predict efficacy in humans. We have established the pig, a large natural host animal for influenza, with many physiological similarities to humans, as a robust model for testing mAbs. We show that a strongly neutralizing mAb (2-12C) against the hemagglutinin head administered prophylactically at 15 mg/kg reduced viral load and lung pathology after pandemic H1N1 influenza challenge. A lower dose of 1 mg/kg of 2-12C or a DNA plasmid-encoded version of 2-12C reduced pathology and viral load in the lungs but not viral shedding in nasal swabs. We propose that the pig influenza model will be useful for testing candidate mAbs and emerging delivery platforms prior to human trials.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Orthomyxoviridae Infections , Animals , Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/drug therapy , Swine
5.
Mol Ther ; 27(5): 974-985, 2019 05 08.
Article in English | MEDLINE | ID: mdl-30962164

ABSTRACT

Zika virus (ZIKV) infection is endemic to several world regions, and many others are at high risk for seasonal outbreaks. Synthetic DNA-encoded monoclonal antibody (DMAb) is an approach that enables in vivo delivery of highly potent mAbs to control infections. We engineered DMAb-ZK190, encoding the mAb ZK190 neutralizing antibody, which targets the ZIKV E protein DIII domain. In vivo-delivered DMAb-ZK190 achieved expression levels persisting >10 weeks in mice and >3 weeks in non-human primate (NHPs), which is protective against ZIKV infectious challenge. This study is the first demonstration of infectious disease control in NHPs following in vivo delivery of a nucleic acid-encoded antibody, supporting the importance of this new platform.


Subject(s)
Antibodies, Neutralizing/pharmacology , DNA/pharmacology , Viral Envelope Proteins/immunology , Zika Virus Infection/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , DNA/immunology , Humans , Mice , Primates , Viral Envelope Proteins/antagonists & inhibitors , Zika Virus/genetics , Zika Virus/immunology , Zika Virus/pathogenicity , Zika Virus Infection/immunology , Zika Virus Infection/therapy , Zika Virus Infection/virology
6.
Adv Genet ; 90: 103-131, 2015.
Article in English | MEDLINE | ID: mdl-26296934

ABSTRACT

MicroRNAs (miRNAs) are small regulatory RNAs that modulate the translation of mRNA. They have emerged over the past few years as indispensable entities in the transcriptional regulation of genes. Their discovery has added additional layers of complexity to regulatory networks that control cellular homeostasis. Also, their dysregulated pattern of expression is now well demonstrated in myriad diseases and pathogenic processes. In the current review, we highlight the role of miRNAs in Lesch-Nyhan disease (LND), a rare neurogenetic syndrome caused by mutations in the purine metabolic gene encoding the hypoxanthine-guanine phosphoribosyltransferase (HPRT) enzyme. We describe how experimental and biocomputational approaches have helped to unravel genetic and signaling pathways that provide mechanistic understanding of some of the molecular and cellular basis of this ill-defined neurogenetic disorder. Through miRNA-based target predictions, we have identified signaling pathways that may be of significance in guiding biological therapeutic discovery for this incurable neurological disorder. We also propose a model to explain how a gene such as HPRT, mostly known for its housekeeping metabolic functions, can have pleiotropic effects on disparate genes and signal transduction pathways. Our hypothetical model suggests that HPRT mRNA transcripts may be acting as competitive endogenous RNAs (ceRNAs) intertwined in multiregulatory cross talk between key neural transcripts and miRNAs. Overall, this approach of using miRNA-based genomic approaches to elucidate the molecular and cellular basis of LND and guide biological target identification might be applicable to other ill-defined rare inborn-error metabolic diseases.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/therapy , MicroRNAs/therapeutic use , Animals , Gene Expression Regulation , Humans , Lesch-Nyhan Syndrome/genetics , RNA, Messenger/genetics
7.
PLoS One ; 9(5): e96575, 2014.
Article in English | MEDLINE | ID: mdl-24804781

ABSTRACT

Lesch-Nyhan Syndrome (LNS) is a neurodevelopmental disorder caused by mutations in the gene encoding the purine metabolic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT). This syndrome is characterized by an array of severe neurological impairments that in part originate from striatal dysfunctions. However, the molecular and cellular mechanisms underlying these dysfunctions remain largely unidentified. In this report, we demonstrate that HPRT-deficiency causes dysregulated expression of key genes essential for striatal patterning, most notably the striatally-enriched transcription factor B-cell leukemia 11b (Bcl11b). The data also reveal that the down-regulated expression of Bcl11b in HPRT-deficient immortalized mouse striatal (STHdh) neural stem cells is accompanied by aberrant expression of some of its transcriptional partners and other striatally-enriched genes, including the gene encoding dopamine- and cAMP-regulated phosphoprotein 32, (DARPP-32). Furthermore, we demonstrate that components of the BDNF/TrkB signaling, a known activator of DARPP-32 striatal expression and effector of Bcl11b transcriptional activation are markedly increased in HPRT-deficient cells and in the striatum of HPRT knockout mouse. Consequently, the HPRT-deficient cells display superior protection against reactive oxygen species (ROS)-mediated cell death upon exposure to hydrogen peroxide. These findings suggest that the purine metabolic defect caused by HPRT-deficiency, while it may provide neuroprotection to striatal neurons, affects key genes and signaling pathways that may underlie the neuropathogenesis of LNS.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Corpus Striatum/pathology , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/genetics , Receptor, trkB/genetics , Signal Transduction/genetics , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Death/genetics , Cell Differentiation/genetics , Corpus Striatum/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Lesch-Nyhan Syndrome/metabolism , Lesch-Nyhan Syndrome/pathology , Mice , Mice, Knockout , Neurons/metabolism , Reactive Oxygen Species/metabolism , Receptor, trkB/metabolism
8.
Hum Mol Genet ; 22(22): 4502-15, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23804752

ABSTRACT

Lesch-Nyhan syndrome (LNS) is a neurodevelopmental disorder caused by mutations in the gene encoding the purine metabolic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT). A series of motor, cognitive and neurobehavioral anomalies characterize this disease phenotype, which is still poorly understood. The clinical manifestations of this syndrome are believed to be the consequences of deficiencies in neurodevelopmental pathways that lead to disordered brain function. We have used microRNA array and gene ontology analysis to evaluate the gene expression of differentiating HPRT-deficient human neuron-like cell lines. We set out to identify dysregulated genes implicated in purine-based cellular functions. Our approach was based on the premise that HPRT deficiency affects preeminently the expression and the function of purine-based molecular complexes, such as guanine nucleotide exchange factors (GEFs) and small GTPases. We found that several microRNAs from the miR-17 family cluster and genes encoding GEF are dysregulated in HPRT deficiency. Most notably, our data show that the expression of the exchange protein activated by cAMP (EPAC) is blunted in HPRT-deficient human neuron-like cell lines and fibroblast cells from LNS patients, and is altered in the cortex, striatum and midbrain of HPRT knockout mouse. We also show a marked impairment in the activation of small GTPase RAP1 in the HPRT-deficient cells, as well as differences in cytoskeleton dynamics that lead to increased motility for HPRT-deficient neuron-like cell lines relative to control. We propose that the alterations in EPAC/RAP1 signaling and cell migration in HPRT deficiency are crucial for neuro-developmental events that may contribute to the neurological dysfunctions in LNS.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Lesch-Nyhan Syndrome/genetics , MicroRNAs/genetics , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Line , Cell Movement/physiology , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Cytoskeleton/metabolism , Gene Ontology , Guanine Nucleotide Exchange Factors/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Male , Mesencephalon/metabolism , Mice , Mice, Knockout , MicroRNAs/metabolism , Multigene Family , Oligonucleotide Array Sequence Analysis , Signal Transduction , rap1 GTP-Binding Proteins/genetics
9.
PLoS One ; 8(5): e63333, 2013.
Article in English | MEDLINE | ID: mdl-23691025

ABSTRACT

Lesch-Nyhan Disease (LND) is the result of mutations in the X-linked gene encoding the purine metabolic enzyme, hypoxanthine guanine phosphoribosyl transferase (HPRT). LND gives rise to severe neurological anomalies including mental retardation, dystonia, chorea, pyramidal signs and a compulsive and aggressive behavior to self injure. The neurological phenotype in LND has been shown to reflect aberrant dopaminergic signaling in the basal ganglia, however there are little data correlating the defect in purine metabolism to the neural-related abnormalities. In the present studies, we find that HPRT-deficient neuronal cell lines have reduced CREB (cAMP response element-binding protein) expression and intracellular cyclic AMP (cAMP), which correlates with attenuated CREB-dependent transcriptional activity and a reduced phosphorylation of protein kinase A (PKA) substrates such as synapsin (p-syn I). Of interest, we found increased expression of phosphodiesterase 10A (PDE10A) in HPRT-deficient cell lines and that the PDE10 inhibitor papaverine and PDE10A siRNA restored cAMP/PKA signaling. Furthermore, reconstitution of HPRT expression in mutant cells partly increased cAMP signaling synapsin phosphorylation. In conclusion, our data show that HPRT-deficiency alters cAMP/PKA signaling pathway, which is in part due to the increased of PDE10A expression and activity. These findings suggest a mechanistic insight into the possible causes of LND and highlight PDE10A as a possible therapeutic target for this intractable neurological disease.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Gene Expression Regulation, Enzymologic , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome/drug therapy , Phosphoric Diester Hydrolases/metabolism , Signal Transduction , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Knockdown Techniques , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Lesch-Nyhan Syndrome/pathology , MicroRNAs/genetics , Molecular Targeted Therapy , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synapsins/genetics , Transcription, Genetic
10.
Hum Mol Genet ; 21(3): 609-22, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22042773

ABSTRACT

Mutations in the gene encoding the purine biosynthetic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT) cause the intractable neurodevelopmental Lesch-Nyhan disease (LND) associated with aberrant development of brain dopamine pathways. In the current study, we have identified an increased expression of the microRNA miR181a in HPRT-deficient human dopaminergic SH-SY5Y neuroblastoma cells. Among the genes potentially regulated by miR181a are several known to be required for neural development, including Engrailed1 (En1), Engrailed2 (En2), Lmx1a and Brn2. We demonstrate that these genes are down-regulated in HPRT-deficient SH-SY5Y cells and that over-expression of miR181a significantly reduces endogenous expression of these genes and inhibits translation of luciferase plasmids bearing the En1/2 or Lmx1a 3'UTR miRNA-binding elements. Conversely, inhibition of miR181a increases the expression of these genes and enhances translation of luciferase constructs bearing the En1/2 and Lmx1a 3'UTR miRNA-binding sequences. We also demonstrate that key neurodevelopmental genes (e.g. Nurr1, Pitx3, Wnt1 and Mash1) known to be functional partners of Lmx1a and Brn2 are also markedly down-regulated in SH-SY5Y cells over-expressing miR181a and in HPRT-deficient cells. Our findings in SH-SY5Y cells demonstrate that HPRT deficiency is accompanied by dysregulation of some of the important pathways that regulate the development of dopaminergic neurons and dopamine pathways and that this defect is associated with and possibly due at least partly to aberrant expression of miR181a. Because aberrant expression of miR181a is not as apparent in HPRT-deficient LND fibroblasts, the relevance of the SH-SY5Y neuroblastoma cells to human disease remains to be proven. Nevertheless, we propose that these pleiotropic neurodevelopment effects of miR181a may play a role in the pathogenesis of LND.


Subject(s)
Dopaminergic Neurons/metabolism , Gene Expression Regulation , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome/genetics , MicroRNAs/metabolism , 3' Untranslated Regions , Cell Line, Tumor , Cells, Cultured , Down-Regulation , Fibroblasts/metabolism , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/antagonists & inhibitors , Hypoxanthine Phosphoribosyltransferase/genetics , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Lesch-Nyhan Syndrome/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , POU Domain Factors/genetics , POU Domain Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
PLoS One ; 6(1): e16572, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21305049

ABSTRACT

We have used microarray-based methods of global gene expression together with quantitative PCR and Western blot analysis to identify dysregulation of genes and aberrant cellular processes in human fibroblasts and in SH-SY5Y neuroblastoma cells made HPRT-deficient by transduction with a retrovirus stably expressing an shRNA targeted against HPRT. Analysis of the microarray expression data by Gene ontology (GO) and Gene Set Enrichment Analysis (GSEA) as well as significant pathway analysis by GeneSpring GX10 and Panther Classification System reveal that HPRT deficiency is accompanied by aberrations in a variety of pathways known to regulate neurogenesis or to be implicated in neurodegenerative disease, including the canonical Wnt/ß-catenin and the Alzheimer's disease/presenilin signaling pathways. Dysregulation of the Wnt/ß-catenin pathway is confirmed by Western blot demonstration of cytosolic sequestration of ß-catenin during in vitro differentiation of the SH-SY5Y cells toward the neuronal phenotype. We also demonstrate that two key transcription factor genes known to be regulated by Wnt signaling and to be vital for the generation and function of dopaminergic neurons; i.e., Lmx1a and Engrailed 1, are down-regulated in the HPRT knockdown SH-SY5Y cells. In addition to the Wnt signaling aberration, we found that expression of presenilin-1 shows severely aberrant expression in HPRT-deficient SH-SY5Y cells, reflected by marked deficiency of the 23 kDa C-terminal fragment of presenilin-1 in knockdown cells. Western blot analysis of primary fibroblast cultures from two LND patients also shows dysregulated presenilin-1 expression, including aberrant proteolytic processing of presenilin-1. These demonstrations of dysregulated Wnt signaling and presenilin-1 expression together with impaired expression of dopaminergic transcription factors reveal broad pleitropic neuro-regulatory defects played by HPRT expression and suggest new directions for investigating mechanisms of aberrant neurogenesis and neuropathology in LND and potential new targets for restoration of effective signaling in this neuro-developmental defect.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/deficiency , Neurogenesis/genetics , Presenilin-1/metabolism , Signal Transduction/genetics , Wnt Proteins/metabolism , Cell Line, Tumor , Dopamine , Gene Expression Profiling/methods , Humans , Lesch-Nyhan Syndrome , Transcription Factors
12.
Cold Spring Harb Protoc ; 2010(7): pdb.prot5420, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20647353

ABSTRACT

INTRODUCTION: A major obstacle to in vivo delivery of lentivirus or other retroviral vectors is their lability in the presence of serum. In vivo, these viral particles are rapidly destroyed by nonspecific complement-mediated degradation mechanisms. The eventual effective use of retroviral vectors for in vivo gene delivery would be greatly facilitated by the development of methods to protect the viral particles from such degradation. This protocol describes methods for the production of complement-stabilized lentiviral vectors either by pseudotyping the viral particles with a fusion envelope protein containing the complement-regulatory protein CD55 (decay accelerating factor, DAF) or by coassembly with the native DAF protein. An in vitro serum inactivation assay is also described.


Subject(s)
Complement Inactivator Proteins/pharmacology , Gene Transfer Techniques , Genetic Vectors/genetics , Lentivirus/classification , Lentivirus/genetics , Virus Inactivation , Cell Line , Filtration , Humans , Immunoassay , Plasmids/genetics , Serotyping , Transfection , Virus Inactivation/drug effects
13.
Mol Ther ; 18(1): 54-62, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19672249

ABSTRACT

Neuronal transcription factors play vital roles in the specification and development of neurons, including dopaminergic (DA) neurons. Mutations in the gene encoding the purine biosynthetic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT) cause the resulting intractable and largely untreatable neurological impairment of Lesch-Nyhan disease (LND). The disorder is associated with a defect in basal ganglia DA pathways. The mechanisms connecting the purine metabolic defect and the central nervous system (CNS) phenotype are poorly understood but have been presumed to reflect a developmental defect of DA neurons. We have examined the effect of HPRT deficiency on the differentiation of neurons in the well-established human (NT2) embryonic carcinoma neurogenesis model. We have used a retrovirus expressing a small hairpin RNA (shRNA) to knock down HPRT gene expression and have examined the expression of a number of transcription factors essential for neuronal differentiation and marker genes involved in DA biosynthetic pathway. HPRT-deficient NT2 cells demonstrate aberrant expression of several transcription factors and DA markers. Although differentiated HPRT-deficient neurons also demonstrate a striking deficit in neurite outgrowth during differentiation, resulting neurons demonstrate wild-type electrophysiological properties. These results represent direct experimental evidence for aberrant neurogenesis in HPRT deficiency and suggest developmental roles for other housekeeping genes in neurodevelopmental disease.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/metabolism , Neurogenesis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Electrophysiology , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Neurites/metabolism , Neurons/cytology , Neurons/metabolism
14.
J Virol Methods ; 148(1-2): 277-82, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18207578

ABSTRACT

The current study reports the production of baculoviral-virosomal vectors consisting of lipoplexes and of the viral glycoprotein (GP64) of baculovirus Autographa californica multiple nucleopolyhdrovirus (AcMNPV). This study demonstrates that such complexes have an increased transfection capability in a number of cells, including undifferentiated H9 human embryonic stem H9hES cells compared to lipoplexes alone. The GP64-mediated enhancement of gene transfer of lipoplexes is inhibited by the addition of anti-GP64 neutralizing antibody and by a modified GP64 protein, but is however less potent than vesicular stomatitis virus glycoprotein (VSV-G)-mediated enhancement of gene transfer of lipoplexes. This difference may be explained in part by the dissimilarity in the fusogenic properties of their respective viral glycoprotein.


Subject(s)
Liposomes/metabolism , Nucleopolyhedroviruses/genetics , Transfection/methods , Viral Envelope Proteins/biosynthesis , Virosomes/biosynthesis , Cell Line , Humans , Membrane Glycoproteins/metabolism , Viral Envelope Proteins/metabolism
15.
Hum Gene Ther ; 16(4): 489-501, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15871680

ABSTRACT

Chronic inflammation in tibialis anterior muscles of mdx mice was produced by a single injection of a recombinant adenovirus vector (AV) expressing an immunogenic beta-galactosidase (beta-gal). In regions of intense beta-gal staining, mononuclear infiltrates abounded, and muscle fibers showed strong extrasynaptic utrophin immunostaining, restoration of dystrophin-associated protein complex, and a marked reduction of the prevalence of centronucleation. Immunoblot analysis confirmed an increase of endogenous utrophin without an increase of the mRNA of the major muscle isoform utrA. Significantly better maximal tetanic force values were demonstrated in the inflammatory versus control mdx muscles. The resistance to lengthening contraction- induced damage was also significantly increased in the former. In muscles of mice lacking TNF-alpha gene, AV vector did not induce inflammation and extrajunctional utrophin increase did not occur. In the inflammatory mdx muscles, proteolytic activity of calcium-activated calpain was reduced, and in mdx myotubes in vitro, incubation with NO donors also reduced calpain-mediated utrophin proteolysis. Since utrophin was shown to be a natural substrate of calpain and known inhibitors of calpain in cultured mdx myotubes increased utrophin levels, the above results were consistent with the following conclusions: (1) extrasynaptic utrophin increase is mainly responsible for the antidystrophic effect; (2) extrasynaptic utrophin increase is a result of posttranscriptional mechanism(s) related to proinflammatory factors; and (3) reduction of endogenous muscle calpain activity by inflammatory cytokines has an important role in the stabilization and increase of the extrasynaptic utrophin.


Subject(s)
Myositis/metabolism , Sarcolemma/metabolism , Utrophin/metabolism , Adenoviridae/genetics , Animals , Animals, Newborn , Calcium/metabolism , Calpain/metabolism , Cells, Cultured , Chronic Disease , Cytokines/genetics , Cytokines/metabolism , Male , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myositis/etiology , Myositis/pathology , Nitric Oxide Donors/pharmacology , Protein Processing, Post-Translational , Synapses/metabolism , Utrophin/drug effects , Utrophin/genetics , beta-Galactosidase/adverse effects , beta-Galactosidase/genetics
16.
Mol Ther ; 11(4): 645-51, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15771967

ABSTRACT

The eventual development of efficient gene transfer vectors for in vivo gene delivery will require the development of a number of important new technologies such as stabilization of vectors against protective mechanisms that destroy or otherwise inactivate foreign infectious agents like gene transfer vectors. It is known that the baculovirus envelope protein GP64 of Autographa californica nucleopolyhedrovirus can efficiently pseudotype lentivirus vectors and that modified forms of the baculovirus envelope protein GP64 can also assemble efficiently into baculovirus particles to display functional foreign proteins on the baculovirus surface. In the present study we have combined these techniques to prepare HIV-based lentivirus vectors pseudotyped with GP64 envelope protein and coexpressing a fusion protein of GP64 with the complement-regulatory, decay accelerating factor (DAF, CD55). In addition, we have also prepared GP64-pseudotyped vectors in the presence of a DAF expression plasmid to allow the incorporation of DAF protein into viral particles. Our results demonstrate both the efficient expression and the high-titer production of GP64/GP64-DAF and GP64/DAF-pseudotyped particles and their stability against inactivation by human and nonhuman primate serum.


Subject(s)
CD55 Antigens/genetics , Complement Inactivator Proteins/genetics , Genetic Vectors/immunology , HIV-1/genetics , Viral Fusion Proteins/genetics , CD55 Antigens/metabolism , Complement Inactivator Proteins/metabolism , Complement System Proteins/immunology , Genetic Vectors/genetics , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Fusion Proteins/metabolism
17.
Am J Physiol Regul Integr Comp Physiol ; 287(4): R961-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15191902

ABSTRACT

Duchenne muscular dystrophy is characterized by myofiber necrosis, muscle replacement by connective tissue, and crippling weakness. Although the mdx mouse also lacks dystrophin, most muscles show little myofiber loss or functional impairment. An exception is the mdx diaphragm, which is phenotypically similar to the human disease. Here we tested the hypothesis that the mdx diaphragm has a defective regenerative response to necrotic injury, which could account for its severe phenotype. Massive necrosis was induced in mdx and wild-type (C57BL10) mouse diaphragms in vivo by topical application of notexin, which destroys mature myofibers while leaving myogenic precursor satellite cells intact. At 4 h after acute exposure to notexin, >90% of diaphragm myofibers in both wild-type and mdx mice demonstrated pathological sarcolemmal leakiness, and there was a complete loss of isometric force-generating capacity. Both groups of mice showed strong expression of embryonic myosin within the diaphragm at 5 days, which was largely extinguished by 20 days after injury. At 60 days postinjury, wild-type diaphragms exhibited a persistent loss ( approximately 25%) of isometric force-generating capacity, associated with a trend toward increased connective tissue infiltration. In contrast, mdx diaphragms achieved complete functional recovery of force generation to noninjured values, and there was no increase in muscle connective tissue over baseline. These data argue against any loss of intrinsic regenerative capacity within the mdx diaphragm, despite characteristic features of major dystrophic pathology being present. Our findings support the concept that significant latent regenerative capacity resides within dystrophic muscles, which could potentially be exploited for therapeutic purposes.


Subject(s)
Diaphragm/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Regeneration/physiology , Animals , Coloring Agents , Diaphragm/drug effects , Disease Models, Animal , Elapid Venoms/toxicity , Fibrosis , Immunohistochemistry , Isometric Contraction/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiopathology , Myosin Heavy Chains/metabolism
18.
Mol Ther ; 9(1): 76-84, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14741780

ABSTRACT

The production of potentially targetable VSV-G-pseudotyped retrovirus vectors has been hampered by inadequate understanding of the structure-function relationships of the VSV-G protein. In these studies we demonstrate assembly and production of MLV-based and HIV-1-based vector particles using VSV-G proteins modified by the insertion of a peptide ligand into a site corresponding to amino acid position 24 of the native VSV-G molecule. The inserted ligand represents the decapeptide encoding the collagen-binding domain of von Willebrand factor. We have used deconvolution microscopy to demonstrate that the modified VSV-G molecules sequester in perinuclear structures and are unavailable for assembly of infectious virus particles at the cell surface under standard tissue culture conditions at 37 degrees C. In contrast, at a lower permissive temperature of 30 degrees C, the modified VSV-G protein traffics appropriately to the cell surface and participates in useful titers. Furthermore, VSV-G-pseudotyped MLV-based and HIV-1-based vectors displaying the collagen-binding domain demonstrate a statistically significant increased attachment to a collagen matrix as indicated by an ELISA-like cell binding assay and by a focus transduction assay.


Subject(s)
Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Retroviridae/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Base Sequence , Cell Line , Collagen/metabolism , Gene Expression , Genetic Therapy , Genetic Vectors , HIV-1/genetics , HIV-1/physiology , Ligands , Molecular Sequence Data , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/physiology , Mutagenesis, Insertional , Phenotype , Protein Structure, Tertiary , Protein Transport , Temperature , Virus Assembly , von Willebrand Factor/chemistry
19.
Mol Ther ; 5(5 Pt 1): 538-46, 2002 May.
Article in English | MEDLINE | ID: mdl-11991744

ABSTRACT

Non-infectious, envelope protein-free, retrovirus-like particles (VLP) derived from either Moloney murine leukemia virus (MLV) or human HIV are able to bind efficiently to, but not infect, target cells. Upon subsequent addition to the bound particles of the G protein of vesicular stomatitis virus (VSV-G), an efficient surrogate retrovirus envelope protein, the VLP are efficiently taken up by the cells to produce infection. Cell attachment of the VLP is efficiently inhibited by soluble heparin and dextran sulfate and less efficiently abrogated by several other glycosaminoglycans (GAGs) including chondroitin sulfate A and chondroitin sulfate B (dermatan sulfate), as determined by deconvolution microscopic immunodetection of the viral gag protein and by quantitative binding studies of metabolically labeled (35)S-VLP. Enzymatic digestion of heparan sulfate (HS) from the cell surface with heparinase I also reduces VLP binding. Furthermore, VLP adsorption onto several CHO cell lines variably deficient in cell surface GAG is significantly but incompletely abrogated. De-sulfated heparins are less efficient than native heparin in inhibiting the Polybrene-mediated binding of VLP, whereas growth of human cells in the presence of sodium chlorate leads to significant reduction of Polybrene-mediated VLP binding. In addition, specific inhibition of VLP binding and infectivity of mature infectious VSV-G-pseudotyped virus is observed in the presence of heparin and HS under Polybrene-free conditions. We conclude from these studies that the presence of Polybrene, the degree of sulfation of cell surface GAG, and possibly the presence of charged cell surface macromolecules create an electrostatic environment that promotes optimum binding of VLP to cells. Additionally, our results demonstrate that, in the absence of Polybrene, initial attachments of non-infectious, envelope protein-free VLP and probably mature infectious virus particles are mediated by interactions of the virus particles with cell surface heparan sulfate, and possibly with other GAG molecules.


Subject(s)
Genetic Vectors/administration & dosage , HIV-1/physiology , Heparitin Sulfate/metabolism , Membrane Glycoproteins , Moloney murine leukemia virus/pathogenicity , Receptors, Virus/physiology , Retroviridae/genetics , Viral Envelope Proteins/metabolism , Animals , Cell Line , Cell-Free System , Culture Media , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/physiology , HeLa Cells , Humans , Moloney murine leukemia virus/classification , Moloney murine leukemia virus/genetics , RNA, Viral/physiology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...