Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Gene Ther ; 18(1): 38-42, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20882054

ABSTRACT

Previously (Shan et al, 2005), we reported that adenoviral vector-mediated transfer of the human aquaporin-1 (hAQP1) cDNA to minipig parotid glands following irradiation (IR) transiently restored salivary flow to near normal levels. This study evaluated a serotype 2, adeno-associated viral (AAV2) vector for extended correction of IR (single dose; 20 Gy)-induced, parotid salivary hypofunction in minipigs. At 16 weeks following the IR parotid salivary flow decreased by 85-90%. AAV2hAQP1 administration at week 17 transduced only duct cells and resulted in a dose-dependent increase in salivary flow to approximately 35% of pre-IR levels (to approximately 1 ml per 10 min) after 8 weeks (peak response). Administration of a control AAV2 vector or saline was without effect. Little change was observed in clinical chemistry and hematology values after AAV2hAQP1 delivery. Vector-treated animals generated high anti-AAV2 neutralizing antibody titers by week 4 (approximately 1:1600) and significant elevations in salivary (approximately 15%), but not serum, granulocyte macrophage colony-stimulating factor levels. Following vector administration, salivary [Na(+)] was dramatically increased, from approximately 10 to approximately 55 mM (at 4 weeks) and finally to 39 mM (8 weeks). The findings demonstrate that localized delivery of AAV2hAQP1 to IR-damaged parotid glands leads to increased fluid secretion from surviving duct cells, and may be useful in providing extended relief of salivary hypofunction in previously irradiated patients.


Subject(s)
Aquaporin 1/genetics , Dependovirus/genetics , Parotid Gland/metabolism , Parotid Gland/radiation effects , Animals , Aquaporin 1/administration & dosage , DNA, Complementary/metabolism , Genetic Vectors/genetics , Humans , Parotid Gland/chemistry , Swine
2.
Oral Dis ; 16(3): 269-77, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20374510

ABSTRACT

OBJECTIVES: Salivary glands are useful target organs for local and systemic gene therapeutics. For such applications, the regulation of transgene expression is important. Previous studies by us in murine submandibular glands showed that a rapamycin transcriptional regulation system in a single serotype 2, adeno-associated viral (AAV2) vector was effective for this purpose. This study evaluated if such a vector was similarly useful in rhesus macaque parotid glands. METHODS: A recombinant AAV2 vector (AAV-TF-RhEpo-2.3w), encoding rhesus erythropoietin (RhEpo) and a rapamycin-inducible promoter, was constructed. The vector was administered to macaques at either of two doses [1.5 x 10(11) (low dose) or 1.5 x 10(12) (high dose) vector genomes] via cannulation of Stensen's duct. Animals were followed up for 12-14 weeks and treated at intervals with rapamycin (0.1 or 0.5 mg kg(-1)) to induce gene expression. Serum chemistry, hematology, and RhEpo levels were measured at interval. RESULTS: AAV-TF-RhEpo-2.3w administration led to low levels of rapamycin-inducible RhEpo expression in the serum of most macaques. In five animals, no significant changes were seen in serum chemistry and hematology values over the study. One macaque, however, developed pneumonia, became anemic and subsequently required euthanasia. After the onset of anemia, a single administration of rapamycin led to significant RhEpo production in this animal. CONCLUSION: Administration of AAV-TF-RhEpo-2.3w to macaque parotid glands was generally safe, but led only to low levels of serum RhEpo in healthy animals following rapamycin treatment.


Subject(s)
Gene Expression Regulation/drug effects , Genetic Vectors/administration & dosage , Parotid Gland/metabolism , Sirolimus/pharmacology , Transduction, Genetic , Adenoviridae/genetics , Animals , Dose-Response Relationship, Drug , Erythropoietin/blood , Erythropoietin/genetics , Erythropoietin/metabolism , Macaca mulatta , Male , Promoter Regions, Genetic , Recombinant Proteins , Transgenes
3.
Gene Ther ; 17(1): 50-60, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19759566

ABSTRACT

Salivary glands are potentially useful target sites for multiple clinical applications of gene transfer. Previously, we have shown that serotype 2 adeno-associated viral (AAV2) vectors lead to stable gene transfer in the parotid glands of rhesus macaques. As AAV5 vectors result in considerably greater transgene expression in murine salivary glands than do AAV2 vectors, herein we have examined the use of AAV5 vectors in macaques at two different doses (n = 3 per group; 10(10) or 3 x 10(11) particles per gland). AAV5 vector delivery, as with AAV2 vectors, led to no untoward clinical, hematological or serum chemistry responses in macaques. The extent of AAV5-mediated expression of rhesus erythropoietin (RhEpo) was dose-dependent and similar to that seen with an AAV2 vector. However, unlike results with the AAV2 vector, AAV5 vector-mediated RhEpo expression was transient. Maximal expression peaked at day 56, was reduced by approximately 80% on day 84 and thereafter remained near background levels until day 182 (end of experiment). Quantitative PCR studies of high-dose vector biodistribution at this last time point showed much lower AAV5 copy numbers in the targeted parotid gland (approximately 1.7%) than found with the same AAV2 vector dose. Molecular analysis of the conformation of vector DNA indicated a markedly lower level of concatamerization for the AAV5 vector compared with that of a similar AAV2 vector. In addition, cellular immunological studies suggest that host response differences may occur with AAV2 and AAV5 vector delivery at this mucosal site. The aggregate data indicate that results with AAV5 vectors in murine salivary glands apparently do not extend to macaque glands.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Parotid Gland/metabolism , Animals , Genetic Therapy/methods , Macaca mulatta , Parotid Gland/virology , Transduction, Genetic , Transgenes
4.
Hum Gene Ther ; 18(11): 1109-18, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17939749

ABSTRACT

Salivary glands (SGs) have proven useful targets for clinical applications of gene therapeutics. In this toxicology and biodistribution study, which conforms to U.S. Food and Drug Administration Good Laboratory Practice regulations, four doses (10(7)-10(10) particles) of a serotype 2 adeno-associated viral (AAV2) vector encoding human erythropoietin were directly administered to the right submandibular gland of male and female BALB/c mice (n = 21 per gender dose group). Control-treated (saline administered; n = 66) and vector-treated (n = 168) animals did not differ in clinical appearance, morbidity and mortality rates, food and water consumption, weight gain ratios, and final weight. Clinical hematology values also were unaffected by AAV2 administration except for parameters influenced by the expression of the recombinant protein (e.g., hematocrit). Mice were killed on days 3, 30, 55, and 92. No major vector-related toxicity was uncovered after complete pathology and histopathology review. However, a significant gender-related difference in vector biodistribution was revealed by quantitative polymerase chain reaction. In male mice vector (group receiving 10(10) particles/animal) effectively transduced, and was primarily confined within, the SGs (i.e., approximately 800 times more copies in SGs than in liver; day 3) and long lived. In contrast, in female mice, SG transduction was less efficient (260-fold less than in males; day 3) and short lived, and vector was disseminated widely via both the bloodstream (SG:liver copy ratio, approximately 1) and saliva (30-fold greater than in males). The observed vector biodistribution is likely due to differences in AAV2 receptor targets and structural differences affecting SG integrity. Sexual dimorphism is a factor of major significance that could potentially affect gene therapy clinical applications in SGs.


Subject(s)
Dependovirus , Genetic Vectors/administration & dosage , Submandibular Gland/metabolism , Submandibular Gland/virology , Animals , Blood/virology , Body Weight , Dependovirus/genetics , Eating , Erythropoietin/blood , Erythropoietin/genetics , Female , Genetic Therapy/methods , Humans , Injections , Leukocyte Count , Male , Mice , Mice, Inbred BALB C , Saliva/virology , Sex Characteristics , Tissue Distribution/genetics
5.
Ann Rheum Dis ; 65(2): 195-200, 2006 Feb.
Article in English | MEDLINE | ID: mdl-15975969

ABSTRACT

BACKGROUND: Sjögren's syndrome (SS), an autoimmune exocrinopathy mainly affecting lachrymal and salivary glands, results in ocular and oral dryness (keratoconjunctivitis sicca and xerostomia). The aetiology and pathogenesis are largely unknown; currently, only palliative treatment is available. OBJECTIVE: To determine whether gene transfer of vasoactive intestinal peptide (VIP), based on its immunomodulatory properties, might be useful in the management of SS. METHODS: A recombinant serotype 2 adeno-associated virus encoding the human VIP transgene (rAAV2hVIP) was constructed and its efficacy tested in the female non-obese diabetic (NOD) mouse model for SS after retrograde instillation in submandibular glands (SMGs). 10(10) particles/gland of rAAV2hVIP or rAAV2LacZ (encoding beta-galactosidase; control vector) were administered at 8 weeks of age (before sialadenitis onset). Salivary flow rates were determined before vector delivery and at time of death (16 weeks). After death, saliva, serum, and SMGs were harvested. Salivary output, inflammatory infiltrates (focus scores), VIP protein expression, cytokine profile, and serum anti-VIP antibodies were analysed. RESULTS: rAAV2hVIP significantly improved the salivary flow, increased SMG and serum expression of VIP, and reduced SMG cytokines interleukin (IL) 2, IL10, IL12 (p70), and tumour necrosis factor alpha, and serum RANTES, compared with the control vector. No difference in focus scores or apoptotic rates was found; neutralising antibodies were not detected. CONCLUSIONS: Local delivery of rAAV2hVIP can have disease modifying and immunosuppressive effects in SMGs of the NOD mouse model of SS. The new strategy of employing VIP prophylactically may be useful for both understanding and managing the salivary component of SS.


Subject(s)
Genetic Therapy/methods , Sjogren's Syndrome/therapy , Transduction, Genetic/methods , Vasoactive Intestinal Peptide/genetics , Adenoviridae/genetics , Animals , Antibodies/blood , Apoptosis , Cytokines/analysis , Cytokines/blood , Epithelial Cells/pathology , Female , Gene Expression , Genetic Engineering , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Lac Operon , Mice , Mice, Inbred NOD , Models, Animal , Sjogren's Syndrome/immunology , Sjogren's Syndrome/pathology , Submandibular Gland/immunology , Submandibular Gland/pathology , Transgenes , Vasoactive Intestinal Peptide/analysis , Vasoactive Intestinal Peptide/blood
6.
Gene Ther ; 13(7): 594-601, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16341060

ABSTRACT

We previously demonstrated that recombinant adeno-associated virus vectors based on serotype 2 (rAAV2) can direct transgene expression in salivary gland cells in vitro and in vivo. However, it is not known how other rAAV serotypes perform when infused into salivary glands. The capsids of serotypes 4 and 5 are distinct from rAAV2 and from each other, suggesting that they may direct binding and entry into different cell types. In the present study, we investigated the tropisms, transduction efficiencies, and antibody response to AAV vectors based on AAV serotypes 2, 4, and 5. Administration of rAAV2beta-galactosidase (betagal), rAAV4betagal, or rAAV5betagal to murine submandibular salivary glands by retrograde ductal instillation resulted in efficient transduction of salivary epithelial cells, with AAV4 and AAV5 producing 2.3 and 7.3 times more betagal activity compared with AAV2. Improved transduction with AAV5 was confirmed by QPCR of DNA extracted from glands and immunohistochemical staining for transgene expression. Like AAV2, AAV5 primarily transduced striated and intercalated ductal cells. AAV4 transduction was evident in striated, intercalated, and excretory ductal cells, as well as in convoluted granular tubules. In keeping with the encapsulated nature of the salivary gland, the majority of persistent viral genomes were found in the gland and not in other tissues. Neutralizing antibodies (NABs) found in the serum of virus-infused animals were serotype specific and there was no crossreactivity between serotypes. No NABs were detected in saliva but sialic acid conjugates present in saliva could neutralize AAV4 at low dilutions. Together our data suggest that because of differences in receptor binding and transduction pathways, other serotypes may have improved utility as gene transfer vectors in the salivary gland and these differences could be exploited in gene therapy applications.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Submandibular Gland/metabolism , Transduction, Genetic/methods , Animals , Antibodies, Viral/blood , Dependovirus/immunology , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression , Genetic Vectors/genetics , Immunohistochemistry/methods , Male , Mice , Mice, Inbred BALB C , Parvoviridae Infections/immunology , Parvoviridae Infections/virology , Saliva/immunology , Serotyping , Submandibular Gland/virology , Transgenes , beta-Galactosidase/analysis , beta-Galactosidase/genetics
7.
Diabetes ; 50(3): 515-20, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11246870

ABSTRACT

The ability to transfer immunoregulatory, cytoprotective, or antiapoptotic genes into pancreatic islet cells may allow enhanced posttransplantation survival of islet allografts and inhibition of recurrent autoimmune destruction of these cells in type 1 diabetes. However, transient transgene expression and the tendency to induce host inflammatory responses have limited previous gene delivery studies using viral transfer vectors. We demonstrate here that recombinant adeno-associated virus (rAAV) serotype 2, a vector that can overcome these limitations, effectively transduces both human and murine pancreatic islet cells with reporter genes as well as potentially important immunoregulatory cytokine genes (interleukin-4, interleukin-10), although a very high multiplicity of infection (10,000 infectious units/islet equivalent) was required. This requirement was alleviated by switching to rAAV serotype 5, which efficiently transduced islets at a multiplicity of infection of 100. Although adenovirus (Ad) coinfection was required for efficient ex vivo expression at early time points, islets transduced without Ad expressed efficiently when they were transplanted under the renal capsule and allowed to survive in vivo. The rAAV-delivered transgenes did not interfere with islet cell insulin production and were expressed in both beta- and non-beta-cells. We believe rAAV will provide a useful tool to deliver therapeutic genes for modulating immune responses against islet cells and markedly enhance longterm graft survival.


Subject(s)
Dependovirus/genetics , Genetic Vectors , Islets of Langerhans/physiology , Recombination, Genetic , Transduction, Genetic , Adenoviridae/genetics , Animals , Cell Transplantation , Cells, Cultured , Dependovirus/immunology , Gene Transfer Techniques , Genes, Reporter , Humans , Insulin/metabolism , Interleukin-10/genetics , Interleukin-4/genetics , Islets of Langerhans/cytology , Kidney/physiology , Mice , Mice, Inbred C57BL , Serotyping
8.
J Virol ; 74(20): 9441-50, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11000213

ABSTRACT

Adeno-associated virus (AAV) type 2 Rep78 is a multifunctional protein required for AAV DNA replication, integration, and gene regulation. The biochemical activities of Rep78 have been described, but the effects of Rep proteins on the cell have not been characterized. We have analyzed Rep-mediated cytotoxicity. We demonstrated that Rep78 expression is sufficient to induce cell death and disruption of the cell cycle. Cell death was found to be mediated by apoptosis. Rep78 expression resulted in the activation of caspase-3, a terminal caspase directly involved in the execution of cell death. A peptidic inhibitor of caspase-3, Z-Asp-Glu-Val-Asp-fluoromethylketone (Z-DEVD-FMK), abrogated Rep78-induced apoptosis, indicating that Rep78-mediated apoptosis is caspase-3 dependent. Rep78 induced apoptosis in wild-type p53-containing human embryonal carcinoma NT-2 cells and in p53-null promyelocytic human HL-60 cells, indicating that at least one pathway of Rep78-induced apoptosis is p53 independent. Apoptosis was shown to occur during the G(1) and early S phases of the cell cycle. By analyzing the effects of Rep78 mutations on cell viability, the cause of cell death was attributed in part to two biochemical activities of Rep78, DNA binding and ATPase/helicase activity. The endonuclease activity of Rep78 did not contribute to apoptosis induction.


Subject(s)
Apoptosis , Caspases/physiology , DNA-Binding Proteins/physiology , Dependovirus/physiology , Tumor Suppressor Protein p53/physiology , Viral Proteins/physiology , Adenosine Triphosphatases/physiology , Caspase 3 , DNA Damage , DNA Replication , Enzyme Activation , G1 Phase , HL-60 Cells , Humans
9.
Intervirology ; 42(4): 213-20, 1999.
Article in English | MEDLINE | ID: mdl-10567839

ABSTRACT

Two previous reports indicated that recombinant adeno-associated virus (rAAV) vectors were dependent on helper adenovirus (Ad) for efficient conversion of single-stranded (ss) rAAV DNA to the double-stranded (ds) form. This finding is somewhat paradoxical, however, since during a latent infection wild-type (wt)-AAV is rapidly converted to a ds form in the absence of Ad. Our hypothesis was that the effect observed in the previous studies was due to kinetic factors, i.e. to a relative delay in conversion to ds-DNA rather than to an absolute requirement for Ad. To test this, Hela cells were infected with a rAAV-CMV-green fluorescent protein (GFP) vector either in the presence or absence of Ad. Within the first 2 days, Ad infection resulted in a 4-fold increase in AAV vector expression and an augmentation of conversion to a ds-AAV DNA. By 6 days, however, the total number of GFP-expressing cells in the Ad-free culture had exceeded the original number in the Ad co-infected cells, and the conversion to ds-DNA episomes was substantial and ongoing.


Subject(s)
DNA, Recombinant/metabolism , Dependovirus/physiology , Genetic Vectors/physiology , Adenoviridae/physiology , Cell Line, Transformed , Dependovirus/genetics , Genetic Vectors/genetics , HeLa Cells , Humans , Time Factors , Transfection , Transformation, Genetic , Tumor Cells, Cultured , Virus Integration , Virus Latency
10.
J Virol ; 73(5): 4293-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10196327

ABSTRACT

Adeno-associated virus (AAV) replication depends on two viral components for replication: the AAV nonstructural proteins (Rep) in trans, and inverted terminal repeat (ITR) sequences in cis. AAV type 5 (AAV5) is a distinct virus compared to the other cloned AAV serotypes. Whereas the Rep proteins and ITRs of other serotypes are interchangeable and can be used to produce recombinant viral particles of a different serotype, AAV5 Rep proteins cannot cross-complement in the packaging of a genome with an AAV2 ITR. In vitro replication assays indicated that the block occurs at the level of replication instead of at viral assembly. AAV2 and AAV5 Rep binding activities demonstrate similar affinities for either an AAV2 or AAV5 ITR; however, comparison of terminal resolution site (TRS) endonuclease activities showed a difference in specificity for the two DNA sequences. AAV2 Rep78 cleaved only a type 2 ITR DNA sequence, and AAV5 Rep78 cleaved only a type 5 probe efficiently. Mapping of the AAV5 ITR TRS identified a distinct cleavage site (AGTG TGGC) which is absent from the ITRs of other AAV serotypes. Comparison of the TRSs in the AAV2 ITR, the AAV5 ITR, and the AAV chromosome 19 integration locus identified some conserved nucleotides downstream of the cleavage site but little homology upstream.


Subject(s)
DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Dependovirus/metabolism , Terminal Repeat Sequences , Viral Proteins/metabolism , Base Sequence , Binding Sites , DNA Replication , DNA-Binding Proteins/genetics , Dependovirus/genetics , HeLa Cells , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serotyping , Substrate Specificity , Viral Proteins/genetics
11.
Gene Ther ; 3(9): 748-55, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8875221

ABSTRACT

Adeno-associated virus-2 (AAV) can integrate in a site-specific manner to human chromosome 19 and is currently in phase I clinical trials for cystic fibrosis (CF) at Johns Hopkins Hospital. The goal of this study was to determine the fate of recombinant AAV containing the CFTR cDNA (AAV-CFTR) in an immortalized pseudotetraploid CF bronchial epithelial cell line (IB3-1) established from a patient with CF. Fluorescence in situ hybridization (FISH) and Southern blotting of DNA from IB3-1 cells infected with wild-type (wt) or recombinant AAV-CFTR were performed. CFRH2, an IB3-1 cell line with an estimated 15-20 integrated copies of CFTR cDNA, was used to test FISH sensitivity. All metaphase spreads had integrated copies: a single site in 36 of 56 (64.3%) and two sites within the same metaphase spread in 20 of 56 (35.7%). 3-CF-8, an IB3-1 cell line with integration of a partial CFTR cDNA (3.9 kb) was also analyzed by FISH. Integration was observed in 56 of 157 (35.7%) metaphase spreads examined. IB3-1 cells infected with wild-type AAV showed integration in 51 of 86 (59%) metaphase spreads examined. Of 51 integrations, 48 (94%) were to chromosome 19. Examination of 67 metaphase chromosome spreads of IB3-1 cells infected with AAV-CFTR vector (Azero) identified four integrations (6%) to different chromosomes. No integration was to chromosome 19 which differs significantly (P < 0.0001) from wild-type AAV. We then analyzed the A35 cell line, a clone of Azero selected for stable CFTR expression. Genomic DNA from A35 cells did not show a single site of integration; however episomal AAV-CFTR sequences were abundant in the low molecular weight DNA fraction. Examination of 68 metaphase chromosome preparations identified eight distinct integrations, none to chromosome 19. These studies show that FISH is sensitive for the detection of a partial CFTR cDNA integration. Wild-type AAV integrates in a predominantly site-specific fashion. Recombinant AAV-CFTR integrates at low frequency in a nonspecific manner and persists in episomal form in this epithelial cell line.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Virus Integration , Bronchi/cytology , Cell Line, Transformed , Chromosomes, Human, Pair 19/genetics , Cystic Fibrosis , DNA, Recombinant/analysis , Epithelial Cells , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Plasmids/genetics
12.
Gene Ther ; 3(8): 658-68, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8854091

ABSTRACT

Gene therapy for cystic fibrosis (CF) would ideally be accomplished with a vector capable of long-term expression of the cystic fibrosis transmembrane conductance regulator (CFTR) in the absence of a host inflammatory response. Recombinant adeno-associated virus (AAV)-CFTR vectors possess these characteristics in rabbits. Because the utility of AAV vectors as gene transfer agents has only been recognized recently, AAV vector-mediated transduction has never been modeled in a primate host, which is an important step before its use in humans. In order to test the safety and biological activity of AAV-CFTR, single doses of AAV-CFTR vector were administered by fiberoptic bronchoscopy to the posterior basal segment of the right lower lobe (RLL) of the lungs of 10 rhesus macaques with four matched vehicle-treated controls. Animals were followed for 10, 21, 90 or 180 days following vector instillation. Vector DNA transfer occurred in bronchial epithelial cells in the RLL of each animal that received vector as assessed by in situ DNA PCR. Vector mRNA was detectable for 180 days after administration as detected by RT-PCR and by RNase protection assay. Safety of vector administration was determined by measurements of pulmonary mechanics, arterial blood gas analysis, chest radiographs, and bronchoalveolar lavage (BAL) fluid analysis including cell count and quantification of inflammatory cytokines. Gross and microscopic pathologic examination were also performed. There was no evidence of inflammation or other toxicity, although vector DNA was found in extrapulmonary organs of some animals. These results indicate that transduction of the primate airway epithelium with the AAV-CFTR mediates long-term CFTR cDNA transfer and is relatively safe.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/pharmacology , Lung/drug effects , Animals , Bronchoalveolar Lavage , Bronchoscopy , Cystic Fibrosis/therapy , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Female , Genetic Therapy/methods , Lung/diagnostic imaging , Macaca mulatta , Male , Polymerase Chain Reaction , RNA, Viral/biosynthesis , Radiography , Respiratory Function Tests
13.
J Virol ; 70(5): 3235-41, 1996 May.
Article in English | MEDLINE | ID: mdl-8627804

ABSTRACT

Gene therapy vectors based on human DNA viruses could be mobilized or rescued from individuals who are subsequently infected with the corresponding wild-type (wt) helper viruses. This phenomenon has been effectively modeled in vitro with both adenovirus (Ad) and adeno-associated virus (AAV) vectors but has not previously been studied in vivo. In the current study, we have developed an in vivo model to study the interactions of a recombinant AAV vector (AAV-CFTR) with wt AAV type 2 (AAV2) and a host range mutant Ad (Ad2HR405) for which monkey cells are permissive (D.E.Brough, S.A.Rice, S.Sell, and D.F.Klessig, J. Virol. 55:206-212, 1985). AAV-CFTR was administered to the respiratory epithelium of the nose or lung of rhesus macaques. Primary cells were harvested from the infusion site at time points up to 3 months after vector administration to confirm vector DNA persistence. Vector DNA was present in episomal form and could be rescued in vitro only by addition of wt AAV2 and Ad. In in vivo rescue studies, vector was administered before or after wt-AAV2 and Ad2HR405 infection, and the shedding of AAV-CFTR was examined. Ad2HR405 and wt-AAV2 infections were established in the nose with concomitant administration. wt-AAV2 replication occurred in the lung when virus was administered directly at a high titer to the lower respiratory tract. AAV-CFTR vector rescue was also observed in the latter setting. Although these studies were performed with small numbers of animals within each group, it appears that AAV-CFTR DNA persists in the primate respiratory tract and that this model may be useful for studies of recombinant AAV vector rescue.


Subject(s)
Dependovirus/physiology , Genetic Vectors , Adenoviridae/physiology , Animals , Base Sequence , Bronchi/cytology , Bronchi/virology , Bronchoalveolar Lavage Fluid/virology , Cell Line , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , DNA Primers , Dependovirus/genetics , Dependovirus/isolation & purification , Epithelial Cells , Epithelium/virology , Genetic Therapy/methods , HeLa Cells , Helper Viruses/physiology , Humans , In Situ Hybridization, Fluorescence , Macaca mulatta , Molecular Sequence Data , Mutagenesis , Polymerase Chain Reaction , Recombination, Genetic , Virus Shedding
15.
Gene Ther ; 2(1): 29-37, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7712331

ABSTRACT

Adeno-associated virus (AAV) vectors are potentially useful for gene therapy of a number of human diseases. However, the use of these vectors has been limited by the lack of stable vector-packaging cell lines. The difficulties in developing packaging cell lines relate to low levels of rep gene expression from the AAV-p5 promoter, and to the propensity of Rep proteins to suppress continued growth of immortalized cell lines. We describe here two new techniques which allow these problems to be circumvented. First, we have demonstrated that expression of rep from the human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter results in a 10-fold improvement of packaging efficiency. Second, we have overcome the inefficiency of vector plasmid transfection by generating cell populations containing rescuable AAV recombinant genomes. These improvements yielded a net increase of 50-fold in the packaging efficiency of the AAVp5neo and AAVp5lacZ recombinant vectors. The AAVp5lacZ vector packaged with this method was administered systemically to recently weaned C57BL mice, and mediated efficient expression of the beta-galactosidase reporter gene in cells of the airway epithelium and spleen. This indicates the in vivo activity of these vector stocks, and their potential utility for gene therapy.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Transfection/methods , Animals , Cell Line , DNA, Recombinant/genetics , DNA-Binding Proteins/genetics , Gene Expression , Genes, Reporter , HIV Long Terminal Repeat/genetics , Lac Operon , Lung/enzymology , Lung/virology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Spleen/enzymology , Spleen/virology , Viral Proteins/genetics
16.
Am J Respir Cell Mol Biol ; 11(5): 517-21, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7946381

ABSTRACT

Adeno-associated virus type 2 (AAV2)-based vectors are capable of stable expression in the airway epithelium and may be useful for gene therapy for human diseases, such as cystic fibrosis. Certain virus vectors, such as retroviruses, require active cell division for integration and expression, but this has not been formally evaluated in the case of AAV2. The cystic fibrosis bronchial epithelial cell line, IB3-1, which can be transduced by AAV2 vectors, was shown to undergo a decrease in DNA synthesis to undetectable levels when grown to confluence. Cultures in which < 0.1% of cells were dividing could still be efficiently transduced with AAV-lacZ or AAV-neo vectors, with a linear dose response, up to 91% with a multiplicity of 3,000 vector particles per cell. The fate of vector DNA in nondividing target cells was investigated by Southern blotting of both low molecular weight, nonintegrated DNA and high molecular weight, genomic DNA fractions. Detectable levels of vector DNA were only seen in the nonintegrated state. These results indicate that AAV2-based vectors, unlike retrovirus vectors, do not require active cell division or integration for expression to occur and thus possess a unique profile of biologic properties.


Subject(s)
DNA, Viral/biosynthesis , Dependovirus/genetics , Gene Expression Regulation, Viral/physiology , Genetic Vectors , Bronchi/cytology , Bronchi/metabolism , Bronchi/virology , Cell Division , Cell Line , Cystic Fibrosis/metabolism , Cystic Fibrosis/virology , DNA, Viral/genetics , DNA, Viral/metabolism , Dependovirus/physiology , Epithelial Cells , Epithelium/metabolism , Epithelium/virology , Humans , Transfection , Virus Integration
17.
Proc Natl Acad Sci U S A ; 90(22): 10613-7, 1993 Nov 15.
Article in English | MEDLINE | ID: mdl-7504271

ABSTRACT

Adeno-associated virus (AAV) vectors expressing the normal cystic fibrosis transmembrane conductance regulator (CFTR) cDNA complement the cystic fibrosis (CF) defect in vitro. Unlike other DNA virus vectors, AAV is a stably integrating virus, which could make possible long-term in vivo complementation of the CF defect in the airway epithelium. We report AAV-CFTR gene transfer and expression after infection of primary CF nasal polyp cells and after in vivo delivery of AAV-CFTR vector to one lobe of the rabbit lung through a fiberoptic bronchoscope. In the rabbit, vector DNA could be detected in the infected lobe up to 6 months after administration. A 26-amino acid polypeptide sequence unique to the recombinant AAV-CFTR protein was used to generate both oligonucleotide probes and a polyclonal antibody which allowed the unambiguous identification of vector RNA and CFTR protein expression. With these reagents, CFTR RNA and protein were detected in the airway epithelium of the infected lobe for up to 6 months after vector administration. AAV vectors do, therefore, efficiently promote in vivo gene transfer to the airway epithelium which is stable over several months. These findings indicate that AAV-CFTR vectors could potentially be very useful for gene therapy.


Subject(s)
Cystic Fibrosis/therapy , Membrane Proteins/genetics , Amino Acid Sequence , Animals , Cystic Fibrosis Transmembrane Conductance Regulator , Dependovirus/genetics , Gene Expression , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Humans , In Vitro Techniques , Lung/metabolism , Membrane Proteins/metabolism , Molecular Sequence Data , Nasal Polyps/microbiology , RNA, Messenger/genetics , Rabbits , Transfection
18.
J Biol Chem ; 268(5): 3781-90, 1993 Feb 15.
Article in English | MEDLINE | ID: mdl-7679117

ABSTRACT

Adeno-associated virus type 2 (AAV) vectors have been used for gene expression in respiratory epithelial cells and may be useful in gene therapy for diseases like cystic fibrosis (CF) which affect the airways. The AAV p5 promoter together with the AAV inverted terminal repeat (ITR) forms a 263-base pair cassette which mediated efficient expression in a CF bronchial epithelial cell line. We report here that the ITR itself can mediate gene expression. In stable transfection assays, AAV-CF vectors expressing the full-length cystic fibrosis transmembrane conductance regulator (CFTR) cDNA from either the p5 promoter or the ITR restored cAMP regulation of the chloride efflux characteristic of CFTR function. An AAV-ITR-CF vector deleted for the amino terminus of CFTR was also functional. This vector was packaged into AAV particles and used to transduce cells without selection. Transduced cells also exhibited cAMP-regulated Cl- efflux. The complemented cell lines showed increased levels of CFTR protein immunofluorescence, and the presence of intact AAV-CF vector sequence was confirmed by Southern blot analysis of rescued vector sequences. These studies provide novel insights into AAV gene expression, and this newly described promoter allows for the production of AAV vectors expressing CFTR in those differentiated cells affected in CF.


Subject(s)
Dependovirus/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Promoter Regions, Genetic , Base Sequence , Cell Line , Chlorides/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator , Fluorescent Antibody Technique , Gene Expression , Genetic Vectors , Humans , Kinetics , Membrane Proteins/analysis , Molecular Sequence Data , Oligodeoxyribonucleotides , Plasmids , Polymerase Chain Reaction/methods , Signal Transduction , Transfection
19.
Am J Respir Cell Mol Biol ; 7(3): 349-56, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1325813

ABSTRACT

Lung diseases such as cystic fibrosis (CF) might be treated by gene therapy using viral vectors delivered to the airway. One potential vector is the defective human parvovirus, adeno-associated virus (AAV). We examined the AAV p5 transcription promoter for gene expression in immortalized cell lines derived from the airway (IB3-1) or pancreas (CFPAC-1) of CF patients. AAV vectors expressing the prokaryotic genes cat (pAAVp5cat) or neo (pAAVp5neo) from the p5 promoter were evaluated after introduction into IB3-1 or CFPAC-1 cells by lipofection. In transient assays in both cell lines, the cat gene was expressed 5- to 10-fold more efficiently from the p5 promoter than from a simian virus 40 early gene promoter (pSVcat). IB3-1 cells were transformed stably to geneticin resistance by pAAVp5neo at a 5-fold higher efficiency than by an SVneo vector. The AAV inverted terminal repeat (ITR) region immediately upstream of the p5 promoter appears to have an enhancer effect and the promoter also contains a CREB site which confers a response to forskolin. In IB3-1 cells, expression of the cat gene from a p5 promoter was decreased about 5-fold by deletion of both the upstream ITR and the CREB site. The AAVp5neo vector was also packaged into AAV particles and used to infect IB3-1 cells as a transducing virus. Under these conditions, 60 to 70% of the cells could be stably transformed to geneticin resistance. Thus, AAV transducing vectors appear to be a highly efficient delivery system for stable integration and expression of genes in cultured airway epithelial cells.


Subject(s)
Bronchi/metabolism , Cystic Fibrosis/therapy , Dependovirus/genetics , Genetic Vectors , Base Sequence , Bronchi/cytology , Cell Line , Cystic Fibrosis/metabolism , DNA , Epithelial Cells , Epithelium/metabolism , Gene Expression , Genetic Therapy , Humans , Molecular Sequence Data , Transduction, Genetic , Transfection
20.
Nature ; 358(6387): 581-4, 1992 Aug 13.
Article in English | MEDLINE | ID: mdl-1380129

ABSTRACT

Cystic fibrosis (CF) is a lethal genetic disease resulting in a reduced Cl- permeability, increased mucous sulphation, increased Na+ absorption and defective acidification of lysosomal vesicles. The CF gene encodes a protein (the cystic fibrosis transmembrane conductance regulator, CFTR) that can function as a low-conductance Cl- channel with a linear current-voltage relationship whose regulation is defective in CF patients. Larger conductance, outwardly rectifying Cl- channels are also defective in CF and fail to activate when exposed either to cyclic AMP-dependent protein kinase A or to protein kinase C. The role of the outwardly rectifying Cl- channel in CF has been questioned. We report here that expression of recombinant CF genes using adeno-associated virus vectors in CF bronchial epithelial cells corrects defective Cl- secretion, that it induces the appearance of small, linear conductance Cl- channels, and restores protein kinase A activation of outwardly rectifying Cl- channels. These results re-establish an involvement of outwardly rectifying Cl- channels in CF and suggest that CFTR regulates more than one conductance pathway in airway tissues.


Subject(s)
Chlorides/metabolism , Cystic Fibrosis/physiopathology , Membrane Proteins/physiology , Protein Kinases/metabolism , Transfection , Adenosine Triphosphate/metabolism , Cell Line , Cell Membrane/physiology , Chloride Channels , Cystic Fibrosis Transmembrane Conductance Regulator , Electric Conductivity , Humans , Membrane Potentials , Membrane Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...