Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
PLoS Pathog ; 11(12): e1005281, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26625259

RESUMO

Adeno-associated viruses are members of the genus dependoviruses of the parvoviridae family. AAV vectors are considered promising vectors for gene therapy and genetic vaccination as they can be easily produced, are highly stable and non-pathogenic. Nevertheless, transduction of cells in vitro and in vivo by AAV in the absence of a helper virus is comparatively inefficient requiring high multiplicity of infection. Several bottlenecks for AAV transduction have previously been described, including release from endosomes, nuclear transport and conversion of the single stranded DNA into a double stranded molecule. We hypothesized that the bottlenecks in AAV transduction are, in part, due to the presence of host cell restriction factors acting directly or indirectly on the AAV-mediated gene transduction. In order to identify such factors we performed a whole genome siRNA screen which identified a number of putative genes interfering with AAV gene transduction. A number of factors, yielding the highest scores, were identified as members of the SUMOylation pathway. We identified Ubc9, the E2 conjugating enzyme as well as Sae1 and Sae2, enzymes responsible for activating E1, as factors involved in restricting AAV. The restriction effect, mediated by these factors, was validated and reproduced independently. Our data indicate that SUMOylation targets entry of AAV capsids and not downstream processes of uncoating, including DNA single strand conversion or DNA damage signaling. We suggest that transiently targeting SUMOylation will enhance application of AAV in vitro and in vivo.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Sumoilação/genética , Transdução Genética , Sequência de Bases , Western Blotting , Linhagem Celular , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Dados de Sequência Molecular , RNA Interferente Pequeno/genética , Transfecção
2.
Mol Ther ; 24(6): 1050-1061, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27018516

RESUMO

Vectors mediating strong, durable, and tissue-specific transgene expression are mandatory for safe and effective gene therapy. In settings requiring systemic vector administration, the availability of suited vectors is extremely limited. Here, we present a strategy to select vectors with true specificity for a target tissue from random peptide libraries displayed on adeno-associated virus (AAV) by screening the library under circulation conditions in a murine model. Guiding the in vivo screening by next-generation sequencing, we were able to monitor the selection kinetics and to determine the right time point to discontinue the screening process. The establishment of different rating scores enabled us to identify the most specifically enriched AAV capsid candidates. As proof of concept, a capsid variant was selected that specifically and very efficiently delivers genes to the endothelium of the pulmonary vasculature after intravenous administration. This technical approach of selecting target-specific vectors in vivo is applicable to any given tissue of interest and therefore has broad implications in translational research and medicine.


Assuntos
Capsídeo/metabolismo , Dependovirus/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Pulmão/metabolismo , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Dependovirus/metabolismo , Terapia Genética , Vetores Genéticos/administração & dosagem , Camundongos , Especificidade de Órgãos , Biblioteca de Peptídeos , Transdução Genética
3.
PLoS Pathog ; 9(10): e1003671, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204256

RESUMO

Disassembly of the nuclear lamina is essential in mitosis and apoptosis requiring multiple coordinated enzymatic activities in nucleus and cytoplasm. Activation and coordination of the different activities is poorly understood and moreover complicated as some factors translocate between cytoplasm and nucleus in preparatory phases. Here we used the ability of parvoviruses to induce nuclear membrane breakdown to understand the triggers of key mitotic enzymes. Nuclear envelope disintegration was shown upon infection, microinjection but also upon their application to permeabilized cells. The latter technique also showed that nuclear envelope disintegration was independent upon soluble cytoplasmic factors. Using time-lapse microscopy, we observed that nuclear disassembly exhibited mitosis-like kinetics and occurred suddenly, implying a catastrophic event irrespective of cell- or type of parvovirus used. Analyzing the order of the processes allowed us to propose a model starting with direct binding of parvoviruses to distinct proteins of the nuclear pore causing structural rearrangement of the parvoviruses. The resulting exposure of domains comprising amphipathic helices was required for nuclear envelope disintegration, which comprised disruption of inner and outer nuclear membrane as shown by electron microscopy. Consistent with Ca⁺⁺ efflux from the lumen between inner and outer nuclear membrane we found that Ca⁺⁺ was essential for nuclear disassembly by activating PKC. PKC activation then triggered activation of cdk-2, which became further activated by caspase-3. Collectively our study shows a unique interaction of a virus with the nuclear envelope, provides evidence that a nuclear pool of executing enzymes is sufficient for nuclear disassembly in quiescent cells, and demonstrates that nuclear disassembly can be uncoupled from initial phases of mitosis.


Assuntos
Sinalização do Cálcio , Caspase 3/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Parvovirus H-1/metabolismo , Mitose , Membrana Nuclear/enzimologia , Infecções por Parvoviridae/enzimologia , Proteína Quinase C/metabolismo , Animais , Cálcio/metabolismo , Caspase 3/genética , Quinase 2 Dependente de Ciclina/genética , Parvovirus H-1/genética , Células HeLa , Humanos , Membrana Nuclear/genética , Membrana Nuclear/patologia , Membrana Nuclear/virologia , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/patologia , Proteína Quinase C/genética , Xenopus laevis
4.
J Virol ; 86(17): 9163-74, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22696661

RESUMO

Adeno-associated virus type 2 (AAV2) has gained much interest as a gene delivery vector. A hallmark of AAV2-mediated gene transfer is an intracellular conformational change of the virus capsid, leading to the exposure of infection-relevant protein domains. These protein domains, which are located on the N-terminal portion of the structural proteins VP1 and VP2, include a catalytic phospholipase A(2) domain and three clusters of basic amino acids. We have identified additional protein sequence motifs located on the VP1/2 N terminus that also proved to be obligatory for virus infectivity. These motifs include signals that are known to be involved in protein interaction, endosomal sorting and signal transduction in eukaryotic cells. Among different AAV serotypes they are highly conserved and mutation of critical amino acids of the respective motifs led to a severe infection-deficient phenotype. In particular, mutation of a YXXQ-sequence motif significantly reduced accumulation of virus capsids around the nucleus in comparison to wild-type AAV2. Interestingly, intracellular trafficking of AAV2 was shown to be independent of PLA(2) activity. Moreover, mutation of three PDZ-binding motifs, which are located consecutively at the very tip of the VP1 N terminus, revealed a nuclear transport-defective phenotype, suggesting a role in nuclear uptake of the virus through an as-yet-unknown mechanism.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Núcleo Celular/virologia , Dependovirus/metabolismo , Infecções por Parvoviridae/virologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteínas do Capsídeo/genética , Linhagem Celular , Núcleo Celular/metabolismo , Dependovirus/química , Dependovirus/genética , Humanos , Dados de Sequência Molecular , Infecções por Parvoviridae/metabolismo , Transporte Proteico , Alinhamento de Sequência
5.
J Virol ; 86(17): 9396-408, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22718833

RESUMO

Adeno-associated virus (AAV) has attracted considerable interest as a vector for gene therapy owing its lack of pathogenicity and the wealth of available serotypes with distinct tissue tropisms. One of the most promising isolates for vector development, based on its superior gene transfer efficiency to the liver in small animals compared to AAV type 2 (AAV2), is AAV8. Comparison of the in vivo gene transduction of rAAV2 and rAAV8 in mice showed that single amino acid exchanges in the 3-fold protrusions of AAV8 in the surface loops comprised of residues 581 to 584 and 589 to 592 to the corresponding amino acids of AAV2 and vice versa had a strong influence on transduction efficiency and tissue tropism. Surprisingly, not only did conversion of AAV8 to AAV2 cap sequences increase the transduction efficiency and change tissue tropism but so did the reciprocal conversion of AAV2 to AAV8. Insertion of new peptide motifs at position 590 in AAV8 also enabled retargeting of AAV8 capsids to specific tissues, suggesting that these sequences can interact with receptors on the cell surface. However, a neutralizing monoclonal antibody that binds to amino acids (588)QQNTA(592) of AAV8 does not prevent cell binding and virus uptake, indicating that this region is not necessary for receptor binding but rather that the antibody interferes with an essential step of postattachment processing in which the 3-fold protrusion is also involved. This study supports a multifunctional role of the 3-fold region of AAV capsids in the infection process.


Assuntos
Dependovirus/genética , Terapia Genética/instrumentação , Vetores Genéticos/genética , Transdução Genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Dependovirus/química , Dependovirus/fisiologia , Feminino , Vetores Genéticos/química , Vetores Genéticos/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Alinhamento de Sequência
6.
J Virol ; 86(23): 13038-48, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23015698

RESUMO

Adeno-associated virus (AAV) capsid assembly requires expression of the assembly-activating protein (AAP) together with capsid proteins VP1, VP2, and VP3. AAP is encoded by an alternative open reading frame of the cap gene. Sequence analysis and site-directed mutagenesis revealed that AAP contains two hydrophobic domains in the N-terminal part of the molecule that are essential for its assembly-promoting activity. Mutation of these sequences reduced the interaction of AAP with the capsid proteins. Deletions and a point mutation in the capsid protein C terminus also abolished capsid assembly and strongly reduced the interaction with AAP. Interpretation of these observations on a structural basis suggests an interaction of AAP with the VP C terminus, which forms the capsid protein interface at the 2-fold symmetry axis. This interpretation is supported by a decrease in the interaction of monoclonal antibody B1 with VP3 under nondenaturing conditions in the presence of AAP, indicative of steric hindrance of B1 binding to its C-terminal epitope by AAP. In addition, AAP forms high-molecular-weight oligomers and changes the conformation of nonassembled VP molecules as detected by conformation-sensitive monoclonal antibodies A20 and C37. Combined, these observations suggest a possible scaffolding activity of AAP in the AAV capsid assembly reaction.


Assuntos
Proteínas do Capsídeo/genética , Capsídeo/metabolismo , Dependovirus/metabolismo , Montagem de Vírus/genética , Anticorpos Monoclonais , Capsídeo/ultraestrutura , Proteínas do Capsídeo/metabolismo , Dependovirus/ultraestrutura , Immunoblotting , Imunoprecipitação , Microscopia Eletrônica , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Análise de Sequência de DNA
7.
J Virol ; 86(15): 7739-51, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22593150

RESUMO

Adeno-associated viruses (AAVs) are small single-stranded DNA viruses that can package and deliver nongenomic DNA for therapeutic gene delivery. AAV8, a liver-tropic vector, has shown great promise for the treatment of hemophilia A and B. However, as with other AAV vectors, host anti-capsid immune responses are a deterrent to therapeutic success. To characterize the antigenic structure of this vector, cryo-electron microscopy and image reconstruction (cryo-reconstruction) combined with molecular genetics, biochemistry, and in vivo approaches were used to define an antigenic epitope on the AAV8 capsid surface for a neutralizing monoclonal antibody, ADK8. Docking of the crystal structures of AAV8 and a generic Fab into the cryo-reconstruction for the AAV8-ADK8 complex identified a footprint on the prominent protrusions that flank the 3-fold axes of the icosahedrally symmetric capsid. Mutagenesis and cell-binding studies, along with in vitro and in vivo transduction assays, showed that the major ADK8 epitope is formed by an AAV variable region, VRVIII (amino acids 586 to 591 [AAV8 VP1 numbering]), which lies on the surface of the protrusions facing the 3-fold axis. This region plays a role in AAV2 and AAV8 cellular transduction. Coincidently, cell binding and trafficking assays indicate that ADK8 affects a postentry step required for successful virus trafficking to the nucleus, suggesting a probable mechanism of neutralization. This structure-directed strategy for characterizing the antigenic regions of AAVs can thus generate useful information to help re-engineer vectors that escape host neutralization and are hence more efficacious.


Assuntos
Anticorpos Antivirais/química , Antígenos Virais/química , Proteínas do Capsídeo/química , Dependovirus/química , Mapeamento de Epitopos , Fragmentos Fab das Imunoglobulinas/química , Transporte Ativo do Núcleo Celular , Animais , Anticorpos Antivirais/genética , Anticorpos Antivirais/imunologia , Antígenos Virais/genética , Antígenos Virais/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Núcleo Celular/genética , Núcleo Celular/imunologia , Núcleo Celular/virologia , Cristalografia por Raios X , Dependovirus/genética , Dependovirus/imunologia , Feminino , Técnicas de Transferência de Genes , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/imunologia , Camundongos , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
8.
Proc Natl Acad Sci U S A ; 107(22): 10220-5, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20479244

RESUMO

The volume available in icosahedral virus capsids limits the size of viral genomes. To overcome this limitation, viruses have evolved strategies to increase their coding capacity by using more than one ORF while keeping the genome length constant. The assembly of virus capsids requires the coordinated interaction of a large number of subunits to generate a highly ordered structure in which the viral genome can be enclosed. To understand this process, it is essential to know which viral and nonviral components are involved in the assembly reaction. Here, we show that the adeno-associated virus (AAV) encodes a protein required for capsid formation by means of a nested, alternative ORF of the cap gene. Translation is initiated at a nonconventional translation start site, resulting in the expression of a protein with a calculated molecular weight of 23 kDa. This protein, designated assembly-activating protein (AAP), is localized in the host cell nucleolus, where AAV capsid morphogenesis occurs. AAP targets newly synthesized capsid proteins to this organelle and in addition fulfils a function in the assembly reaction itself. Sequence analysis suggests that also all other species of the genus Dependovirus encode a homologous protein in their cap gene. The arrangement of different ORFs that encode capsid proteins and an assembly factor within the same mRNA facilitates a timely coordinated expression of the components involved in the assembly process.


Assuntos
Capsídeo/fisiologia , Nucléolo Celular/virologia , Dependovirus/fisiologia , Montagem de Vírus/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/fisiologia , Linhagem Celular , Primers do DNA/genética , DNA Viral/genética , Dependovirus/genética , Expressão Gênica , Genes Virais , Teste de Complementação Genética , Células HeLa , Humanos , Dados de Sequência Molecular , Fases de Leitura Aberta , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico
9.
J Gen Virol ; 93(Pt 10): 2131-2141, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22764318

RESUMO

Vectors based on adeno-associated virus serotype 2 (AAV2) belong to today's most promising and most frequently used viral vectors in human gene therapy. Like in many other vector systems, the broad but non-specific tropism limits their use for certain cell types or tissues. One approach to screen for transduction-improved vectors is the selection of random peptide libraries displayed directly on the AAV2 capsid. Although the AAV2 library system has been widely applied for the successful selection of improved gene therapy vectors, it remains unknown which steps of the transduction process are most affected and therefore critical for the selection of targeting peptides. Attachment to the cell surface is the first essential step of AAV-mediated gene transduction; however, our experiments challenge the conventional belief that enhanced gene transfer is equivalent to more efficient cell binding of recombinant AAV2 vectors. A comparison of the various steps of gene transfer by vectors carrying a wild-type AAV2 capsid or displaying two exemplary peptide ligands selected from AAV2 random libraries on different human tumour cell lines demonstrated strong alterations in cell binding, cellular uptake, as well as intracellular processing of these vectors. Combined, our results suggest that entry and post-entry events are decisive for the selection of the peptides NDVRSAN and GPQGKNS rather than their cell binding efficiency.


Assuntos
Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Linhagem Celular Tumoral , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/virologia , Células HEK293 , Humanos , Ligantes , Melanoma/genética , Melanoma/metabolismo , Melanoma/virologia , Biblioteca de Peptídeos , Transdução Genética , Replicação Viral/genética
10.
J Gen Virol ; 93(Pt 9): 1887-1898, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22694902

RESUMO

Adeno-associated virus (AAV) capsid assembly occurs in the nucleus. Newly synthesized capsid proteins VP1, VP2 and VP3 contain several basic regions (BRs), which may act as nuclear localization signals (NLSs). Mutation of BR2 and BR3, located at the VP1 and VP2 N termini, marginally reduced nuclear uptake of VP1 or VP2, but not of VP3, when expressed in the context of the whole AAV type 2 (AAV2) genome. Combined mutation of BR1, BR2 and BR3 resulted in capsids with slightly reduced amounts of VP1. Expression of isolated VP1/2 N termini revealed an influence of BR3 on nuclear transport, whilst BR1 or BR2 had no effect. However, deletion of an N-terminal fragment in front of the BR elements strongly reduced nuclear uptake of VP1/2 N termini. Mutation of BR4, present in all three capsid proteins, led to their retention in the cytoplasm and to the formation of speckles, resulting in a lack of capsid formation and a significant reduction in VP levels. In a VP fragment comprising BR2, BR3 and BR4, the BR4 element was not necessary for nuclear localization. Mutation of BR5 in the C-terminal part of the VPs resulted in a speckled protein distribution in the nucleus, strongly reduced capsid assembly, and low VP1 and VP2 levels. Taken together, these results showed that BR2 and BR3 have a weak influence on nuclear transport of VP1 and VP2, whilst combined mutation of BR1, BR2 and BR3 influences the stoichiometry of VPs in assembled capsids. BR4 and BR5 play a crucial role in capsid assembly but have no NLS activity.


Assuntos
Proteínas do Capsídeo/metabolismo , Núcleo Celular/virologia , Dependovirus/fisiologia , Infecções por Parvoviridae/virologia , Vírion/fisiologia , Montagem de Vírus , Transporte Ativo do Núcleo Celular , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Núcleo Celular/metabolismo , Dependovirus/química , Dependovirus/genética , Dados de Sequência Molecular , Sinais de Localização Nuclear , Infecções por Parvoviridae/metabolismo , Alinhamento de Sequência , Vírion/química , Vírion/genética
11.
Cytotherapy ; 12(1): 107-12, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19929455

RESUMO

BACKGROUND AND AIMS: Because of their pluripotency, human CD34(+) peripheral blood progenitor cells (PBPC) are targets of interest for the treatment of many acquired and inherited disorders using gene therapeutic approaches. Unfortunately, most current vector systems lack either sufficient transduction efficiency or an appropriate safety profile. Standard single-stranded recombinant adeno-associated virus 2 (AAV2)-based vectors offer an advantageous safety profile, yet lack the required efficiency in human PBPC. METHODS: A panel of pseudotyped AAV vectors (designated AAV2/x, containing the vector genome of serotype 2 and capsid of serotype x, AAV2/1-AAV2/6) was screened on primary human granulocyte-colony-stimulating factor (G-CSF)-mobilized CD34(+) PBPC to determine their gene transfer efficacy. Additionally, double-stranded self-complementary AAV (dsAAV) were used to determine possible second-strand synthesis limitations. RESULTS: AAV2/6 vectors proved to be the most efficient [12.8% (1.8-25.4%) transgene-expressing PBPC after a single transduction], being significantly more efficient (all P<0.005) than the other vectors [AAV2/2, 2.0% (0.2-7.3%); AAV2/1, 1.3% (0.1-2.9%); others, <; 1% transgene-expressing PBPC]. In addition, the relevance of the single-to-double-strand conversion block in transduction of human PBPC could be shown using pseudotyped dsAAV vectors: for dsAAV2/2 [9.3% (8.3-20.3%); P<0.001] and dsAAV2/6 [37.7% (23.6-61.0%); P<0.001) significantly more PBPC expressed the transgene compared with their single-stranded counterparts; for dsAAV2/1, no significant increase could be observed. CONCLUSIONS: We have shown that clinically relevant transduction efficiency levels using AAV-based vectors in human CD34(+) PBPC are feasible, thereby offering an efficient alternative vector system for gene transfer into this important target cell population.


Assuntos
Antígenos CD34/análise , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Células-Tronco/metabolismo , Adulto , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Células-Tronco/imunologia , Transdução Genética/métodos , Transgenes
12.
Exp Hematol ; 36(8): 957-64, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18495326

RESUMO

OBJECTIVE: Currently standard recombinant adeno-associated virus serotype 2(rAAV2)-based vectors lack the efficiency for gene transfer into primary human CD34(+) peripheral blood progenitor cells (PBPC). MATERIALS AND METHODS: An advancement in vector development now allows the generation of rAAV capsid mutants that offer higher target cell efficiency and specificity. To increase the gene transfer into hematopoietic progenitor cells, we applied this method for the first time on primary human CD34(+) PBPC cells. RESULTS: On a panel of leukemia cell lines (CML/AML), significantly higher gene transfer efficiency of the rAAV capsid mutants (up to 100% gene transfer) was observed compared to standard rAAV2 vectors. A higher transduction efficiency in the imatinib-resistant cell line LAMA84-R than in their sensitive counterpart LAMA84-S and a pronounced difference in susceptibility for the capsid mutants vs rAAV2 in LAMA84-S were particularly striking. On solid tumor cell lines, on the other hand, rAAV2 was more efficient than the capsid mutants, suggesting an increased specificity of our capsid mutants for hematopoietic progenitor cells. On primary human CD34(+) PBPC significantly higher (up to eightfold; 16% green fluorescent protein-positive) gene transfer could be obtained with the newly generated vectors compared to standard rAAV2 vectors. CONCLUSION: These novel vectors may enable efficient gene transfer using rAAV-based vectors into primary human blood progenitor cells for a future clinical application.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Biblioteca de Peptídeos , Capsídeo/metabolismo , Linhagem Celular , Evolução Molecular Direcionada , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Células-Tronco Hematopoéticas/virologia , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/virologia , Mutagênese , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Genet Vaccines Ther ; 6: 12, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18789140

RESUMO

BACKGROUND: For many promising target cells (e.g.: haematopoeitic progenitors), the susceptibility to standard adeno-associated viral (AAV) vectors is low. Advancements in vector development now allows the generation of target cell-selected AAV capsid mutants. METHODS: To determine its suitability, the method was applied on a chronic myelogenous leukaemia (CML) cell line (K562) to obtain a CML-targeted vector and the resulting vectors tested on leukaemia, non-leukaemia, primary human CML and CD34+ peripheral blood progenitor cells (PBPC); standard AAV2 and a random capsid mutant vector served as controls. RESULTS: Transduction of CML (BV173, EM3, K562 and Lama84) and AML (HL60 and KG1a) cell lines with the capsid mutants resulted in an up to 36-fold increase in CML transduction efficiency (K562: 2-fold, 60% +/- 2% green fluorescent protein (GFP)+ cells; BV173: 9-fold, 37% +/- 2% GFP+ cells; Lama84: 36-fold, 29% +/- 2% GFP+ cells) compared to controls. For AML (KG1a, HL60) and one CML cell line (EM3), no significant transduction (<1% GFP+ cells) was observed for any vector. Although the capsid mutant clone was established on a cell line, proof-of-principle experiments using primary human cells were performed. For CML (3.2-fold, mutant: 1.75% +/- 0.45% GFP+ cells, p = 0.03) and PBPC (3.5-fold, mutant: 4.21% +/- 3.40% GFP+ cells) a moderate increase in gene transfer of the capsid mutant compared to control vectors was observed. CONCLUSION: Using an AAV random peptide library on a CML cell line, we were able to generate a capsid mutant, which transduced CML cell lines and primary human haematopoietic progenitor cells with higher efficiency than standard recombinant AAV vectors.

14.
Exp Hematol ; 35(12): 1766-76, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17920758

RESUMO

OBJECTIVE: For acute myeloid leukemia (AML), gene therapy may be used to treat patients refractory to conventional chemotherapy. However, availability of vectors sufficiently and specifically transducing this cell type is very limited. METHOD: Here we report the selection of capsid-modified adeno-associated viral (AAV) vectors targeting Kasumi-1 AML cells by screening random AAV displayed peptide libraries. RESULTS: The peptide inserts of the enriched capsid mutants share a common sequence motif. The same motif was selected in an independent library screening on HL-60 AML cells. Recombinant targeted vectors displaying the selected peptides transduced the target leukemia cells they have been selected on up to 500-fold more efficiently compared to AAV vectors with control peptide inserts. One of the selected clones (NQVGSWS) also efficiently transduced all members of a panel of four other AML cell lines. Binding and blocking experiments showed that NQVGSWS binding to leukemia cells is independent of the wild-type AAV-2 receptor heparin sulfate proteoglycan. Transduction assays on a panel of hematopoietic and nonhematopoietic cell lines showed that the NQVGSWS capsid was able to overcome resistance to AAV transduction, especially in hematopoietic cancer cells, whereas normal peripheral blood mononuclear cells and CD34(+) hematopoietic progenitor cells were not transduced. CONCLUSIONS: Consequently, recombinant targeted NQVGSWS AAV vectors harboring a suicide gene conferred selective killing to Kasumi-1 cells, but not to control cells. This suggests that the AAV mutant selected here may be used as a tool to target therapeutic genes to AML cells.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos , Leucemia/tratamento farmacológico , Biblioteca de Peptídeos , Antivirais/uso terapêutico , Sequência de Bases , Linhagem Celular , Primers do DNA , Citometria de Fluxo , Ganciclovir/uso terapêutico , Humanos , Leucemia/genética , Reação em Cadeia da Polimerase , Timidina Quinase/genética , Transdução Genética
15.
J Virol Methods ; 140(1-2): 17-24, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17126418

RESUMO

Adeno-associated viruses (AAV) have been developed and evaluated as recombinant vectors for gene therapy. More recently, due to the advantages they offer for gene transfer, several AAV serotypes have gained increasing interest. However, monoclonal antibodies for the characterization and quantitation of vectors derived from different serotypes are at present not available. Serotype-specific monoclonal antibodies (mAbs) against the capsids of the serotypes 1/6, 4 and 5 are described. These antibodies, designated as ADK1a and b, ADK4 or ADK5a and b, reacted specifically with the indicated serotype capsids in cell lysates. ADK 1a and b cross-reacted with its highly related AAV6 serotype, but not with the other serotypes tested. The new antibodies recognized exclusively assembled capsids and neither free nor denatured capsid proteins as shown by fractionation experiments. In immunofluorescence experiments, the mAbs stained only distinct intranuclear foci in cells expressing the capsid protein. The development of capture ELISAs for quantitation of AAV1 and 6, AAV4 or AAV5 capsids illustrates that these novel monoclonal antibodies provide valuable tools for characterization of vector stocks.


Assuntos
Anticorpos Monoclonais/imunologia , Dependovirus/genética , Ensaio de Imunoadsorção Enzimática/métodos , Capsídeo/metabolismo , Linhagem Celular , Reações Cruzadas , Dependovirus/classificação , Imunofluorescência , Humanos , Plasmídeos , Sorotipagem
16.
Nat Biotechnol ; 21(9): 1040-6, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12897791

RESUMO

Characterizing the molecular diversity of the cell surface is critical for targeting gene therapy. Cell type-specific binding ligands can be used to target gene therapy vectors. However, targeting systems in which optimum eukaryotic vectors can be selected on the cells of interest are not available. Here, we introduce and validate a random adeno-associated virus (AAV) peptide library in which each virus particle displays a random peptide at the capsid surface. This library was generated in a three-step system that ensures encoding of displayed peptides by the packaged DNA. As proof-of-concept, we screened AAV-libraries on human coronary artery endothelial cells. We observed selection of particular peptide motifs. The selected peptides enhanced transduction in coronary endothelial cells but not in control nonendothelial cells. This vector targeting strategy has advantages over other combinatorial approaches such as phage display because selection occurs within the context of the capsid and may have a broad range of applications in biotechnology and medicine.


Assuntos
Dependovirus/genética , Dependovirus/metabolismo , Endotélio Vascular/metabolismo , Marcação de Genes/métodos , Terapia Genética/métodos , Vetores Genéticos/genética , Biblioteca de Peptídeos , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Capsídeo/metabolismo , Células Cultivadas , Vasos Coronários/metabolismo , Regulação Viral da Expressão Gênica , Técnicas de Transferência de Genes , Células HeLa , Humanos , Dados de Sequência Molecular , Análise de Sequência de Proteína , Transdução Genética/métodos , Proteínas Virais/genética
17.
Cardiovasc Res ; 70(1): 70-8, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16448634

RESUMO

OBJECTIVE: Vectors based on recombinant adeno-associated virus 2 (AAV-2) are a promising tool for cardiac gene transfer. However, potential therapeutic applications need to consider the predominant transduction of the liver once AAV-2 vectors enter the systemic circulation. We therefore aimed to increase efficiency and specificity of cardiac vector delivery by combining transcriptional and cell surface targeting. METHODS: For analysis of transcriptional targeting, recombinant AAV vectors were generated harboring a luciferase reporter gene under control of the cytomegalovirus (CMV) promoter or the 1.5-kb cardiac myosin light chain promoter fused to the CMV immediate-early enhancer (CMV(enh)/MLC1.5). Luciferase activities were determined in representative organs three weeks after intravenous injection of the vector into adult mice. Transductional targeting was studied using luciferase-reporter constructs crosspackaged into capsids of AAV serotypes 1 to 6 and modified AAV-2 capsids devoid of binding their primary receptor heparan sulfate proteoglycan. RESULTS: Intravenous injections of AAV-2 vectors harboring the CMV(enh)/MLC1.5 promoter enabled a specific and 50-fold higher reporter gene expression in left ventricular myocardium of adult mice compared to vectors containing the CMV promoter. Comparison of AAV-2 vector genomes crosspackaged into capsids of AAV-1 to -6 showed that AAV-1, -4, -5, and -6 capsids increased cardiac transduction efficiency by about 10-fold. However, transduction of other organs such as the liver was also increased after systemic administration. In contrast, AAV-2-based vectors with ablated binding to their primary receptor heparan sulfate proteoglycan enabled a significantly increased efficiency of cardiac gene transfer and reduced transduction of the liver. CONCLUSIONS: Combining transcriptional targeting by the CMV(enh)/MLC1.5 promoter and AAV vectors devoid of binding the AAV-2 primary receptor results in an efficient cardiac gene transfer with a significantly reduced hepatic transduction.


Assuntos
Dependovirus/genética , Marcação de Genes/métodos , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Miócitos Cardíacos/enzimologia , Animais , Capsídeo , Células Cultivadas , Citomegalovirus/genética , Feminino , Engenharia Genética , Vetores Genéticos/genética , Cardiopatias/terapia , Heparina/análogos & derivados , Heparina/genética , Injeções Intravenosas , Fígado/enzimologia , Luciferases/análise , Luciferases/genética , Camundongos , Camundongos Endogâmicos , Cadeias Leves de Miosina/genética , Regiões Promotoras Genéticas , Proteoglicanas/genética , Ratos , Ratos Sprague-Dawley , Transdução Genética/métodos
18.
EMBO Mol Med ; 8(6): 609-25, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27137490

RESUMO

Gene therapy critically relies on vectors that combine high transduction efficiency with a high degree of target specificity and that can be administered through a safe intravenous route. The lack of suitable vectors, especially for gene therapy of brain disorders, represents a major obstacle. Therefore, we applied an in vivo screening system of random ligand libraries displayed on adeno-associated viral capsids to select brain-targeted vectors for the treatment of neurovascular diseases. We identified a capsid variant showing an unprecedented degree of specificity and long-lasting transduction efficiency for brain microvasculature endothelial cells as the primary target of selection. A therapeutic vector based on this selected viral capsid was used to markedly attenuate the severe cerebrovascular pathology of mice with incontinentia pigmenti after a single intravenous injection. Furthermore, the versatility of this selection system will make it possible to select ligands for additional in vivo targets without requiring previous identification of potential target-specific receptors.


Assuntos
Encéfalo/patologia , Dependovirus/genética , Células Endoteliais/patologia , Terapia Genética/métodos , Vetores Genéticos , Incontinência Pigmentar/terapia , Microvasos/patologia , Animais , Modelos Animais de Doenças , Injeções Intravenosas , Camundongos , Transdução Genética , Resultado do Tratamento
19.
Hum Gene Ther ; 23(7): 733-41, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22401308

RESUMO

Cervical cancer is the second most common cancer in women worldwide. Persistent high-risk human papillomavirus (HPV) infection has been identified as the causative event for the development of this type of cancer. Recombinant adeno-associated viruses (rAAVs) are currently being developed and evaluated as vaccine vector. In previous work, we demonstrated that rAAVs administered intranasally in mice induced high titers and long-lasting neutralizing antibodies against HPV type 16 (HPV16). To extend this approach to a more human-related species, we immunized rhesus macaques (Macaca mulatta) with AAVs expressing an HPV16 L1 protein using rAAV5 and 9 vectors in an intranasal prophylactic setting. An rAAV5-L1 vector followed by a boost with rAAV9-L1 induced higher titers of L1-specific serum antibodies than a single rAAV5-L1 immunization. L1-specific antibodies elicited by AAV9 vector neutralized HPV16 pseudovirions and persisted for at least 7 months post immunization. Interestingly, nasal application of rAAV9 was immunogenic even in the presence of high AAV9 antibody titers, allowing reimmunization with the same serotype without prevention of the transgene expression. Two of six animals did not respond to AAV-mediated intranasal vaccination, although they were not tolerant, as both developed antibodies after intramuscular vaccination with HPV16 virus-like particles. These data clearly show the efficacy of an intranasal immunization using rAAV9-L1 vectors without the need of an adjuvant. We conclude from our results that rAAV9 vector is a promising candidate for a noninvasive nasal vaccination strategy.


Assuntos
Dependovirus/genética , Papillomavirus Humano 16/imunologia , Infecções por Papillomavirus/prevenção & controle , Vacinas contra Papillomavirus/genética , Vacinação , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Dependovirus/imunologia , Feminino , Vetores Genéticos , Células HEK293 , Células HeLa , Humanos , Imunidade Humoral , Macaca mulatta , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/imunologia , Infecções por Papillomavirus/imunologia , Infecções por Papillomavirus/virologia , Vacinas contra Papillomavirus/administração & dosagem
20.
Hum Gene Ther ; 23(5): 492-507, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22171602

RESUMO

Libraries based on the insertion of random peptide ligands into the capsid of adeno-associated virus type 2 (AAV2) have been widely used to improve the efficiency and selectivity of the AAV vector system. However, so far only libraries of 7-mer peptide ligands have been inserted at one well-characterized capsid position. Here, we expanded the combinatorial AAV2 display system to a panel of novel AAV libraries, displaying peptides of 5, 7, 12, 19, or 26 amino acids in length at capsid position 588 or displaying 7-mer peptides at position 453, the most prominently exposed region of the viral capsid. Library selections on two unrelated cell types-human coronary artery endothelial cells and rat cardiomyoblasts-revealed the isolation of cell type-characteristic peptides of different lengths mediating strongly improved target-cell transduction, except for the 26-mer peptide ligands. Characterization of vector selectivity by transduction of nontarget cells and comparative gene-transduction analysis using a panel of 44 human tumor cell lines revealed that insertion of different-length peptides allows targeting of distinct cellular receptors for cell entry with similar efficiency, but with different selectivity. The application of such novel AAV2 libraries broadens the spectrum of targetable receptors by capsid-modified AAV vectors and provides the opportunity to choose the best suited targeting ligand for a certain application from a number of different candidates.


Assuntos
Proteínas do Capsídeo/genética , Terapia Genética/métodos , Biblioteca de Peptídeos , Animais , Sítios de Ligação , Proteínas do Capsídeo/metabolismo , Linhagem Celular Tumoral , Dependovirus , Células Endoteliais , Vetores Genéticos , Humanos , Mutagênese Insercional , Mioblastos Cardíacos , Neoplasias/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica , Ratos , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA