Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(15)2021 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-34361120

RESUMO

A major limiting factor for systemically delivered gene therapies is the lack of novel tissue specific AAV (Adeno-associated virus) derived vectors. Bispecific antibodies can be used to redirect AAVs to specific target receptors. Here, we demonstrate that the insertion of a short linear epitope "2E3" derived from human proprotein-convertase subtilisin/kexin type 9 (PCSK9) into different surface loops of the VP capsid proteins can be used for AAV de-targeting from its natural receptor(s), combined with a bispecific antibody-mediated retargeting. We chose to target a set of distinct disease relevant membrane proteins-fibroblast activation protein (FAP), which is upregulated on activated fibroblasts within the tumor stroma and in fibrotic tissues, as well as programmed death-ligand 1 (PD-L1), which is strongly upregulated in many cancers. Upon incubation with a bispecific antibody recognizing the 2E3 epitope and FAP or PD-L1, the bispecific antibody/rAAV complex was able to selectively transduce receptor positive cells. In summary, we developed a novel, rationally designed vector retargeting platform that can target AAVs to a new set of cellular receptors in a modular fashion. This versatile platform may serve as a valuable tool to investigate the role of disease relevant cell types and basis for novel gene therapy approaches.


Assuntos
Anticorpos Biespecíficos/imunologia , Proteínas do Capsídeo/imunologia , Capsídeo/imunologia , Dependovirus/genética , Endopeptidases/imunologia , Epitopos/imunologia , Vetores Genéticos/administração & dosagem , Proteínas de Membrana/imunologia , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Endopeptidases/genética , Endopeptidases/metabolismo , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/imunologia , Pró-Proteína Convertase 9/metabolismo , Transdução Genética
2.
J Am Soc Nephrol ; 29(4): 1141-1153, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29335241

RESUMO

The complement system is essential for host defense, but uncontrolled complement system activation leads to severe, mostly renal pathologies, such as atypical hemolytic uremic syndrome or C3 glomerulopathy. Here, we investigated a novel combinational approach to modulate complement activation by targeting C3 and the terminal pathway simultaneously. The synthetic fusion protein MFHR1 links the regulatory domains of complement factor H (FH) with the C5 convertase/C5b-9 inhibitory fragment of the FH-related protein 1. In vitro, MFHR1 showed cofactor and decay acceleration activity and inhibited C5 convertase activation and C5b-9 assembly, which prevented C3b deposition and reduced C3a/C5a and C5b-9 generation. Furthermore, this fusion protein showed the ability to escape deregulation by FH-related proteins and form multimeric complexes with increased inhibitory activity. In addition to substantially inhibiting alternative and classic pathway activation, MFHR1 blocked hemolysis mediated by serum from a patient with aHUS expressing truncated FH. In FH-/- mice, MFHR1 administration augmented serum C3 levels, reduced abnormal glomerular C3 deposition, and ameliorated C3 glomerulopathy. Taking the unique design of MFHR1 into account, we suggest that the combination of proximal and terminal cascade inhibition together with the ability to form multimeric complexes explain the strong inhibitory capacity of MFHR1, which offers a novel basis for complement therapeutics.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/sangue , Proteínas Sanguíneas/deficiência , Proteínas Inativadoras do Complemento C3b/genética , Inativadores do Complemento/farmacologia , Terapia de Alvo Molecular , Proteínas Recombinantes de Fusão/farmacologia , Animais , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/imunologia , Complemento C3/metabolismo , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Complemento C3b/antagonistas & inibidores , Proteínas Inativadoras do Complemento C3b/deficiência , Complemento C5/metabolismo , Fator H do Complemento/genética , Inativadores do Complemento/isolamento & purificação , Inativadores do Complemento/uso terapêutico , Complexo de Ataque à Membrana do Sistema Complemento/biossíntese , Via Alternativa do Complemento , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Glomérulos Renais/química , Glomérulos Renais/patologia , Camundongos , Camundongos Knockout , Domínios Proteicos , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/uso terapêutico
3.
J Am Soc Nephrol ; 28(5): 1462-1474, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27932477

RESUMO

Genetic defects in complement regulatory proteins can lead to severe renal diseases, including atypical hemolytic uremic syndrome and C3 glomerulopathies, and age-related macular degeneration. The majority of the mutations found in patients with these diseases affect the glycoprotein complement factor H, the main regulator of the alternative pathway of complement activation. Therapeutic options are limited, and novel treatments, specifically those targeting alternative pathway activation, are highly desirable. Substitution with biologically active factor H could potentially treat a variety of diseases that involve increased alternative pathway activation, but no therapeutic factor H is commercially available. We recently reported the expression of full-length recombinant factor H in moss (Physcomitrella patens). Here, we present the production of an improved moss-derived recombinant human factor H devoid of potentially immunogenic plant-specific sugar residues on protein N-glycans, yielding approximately 1 mg purified moss-derived human factor H per liter of initial P. patens culture after a multistep purification process. This glycosylation-optimized factor H showed full in vitro complement regulatory activity similar to that of plasma-derived factor H and efficiently blocked LPS-induced alternative pathway activation and hemolysis induced by sera from patients with atypical hemolytic uremic syndrome. Furthermore, injection of moss-derived factor H reduced C3 deposition and increased serum C3 levels in a murine model of C3 glomerulopathy. Thus, we consider moss-produced recombinant human factor H a promising pharmaceutical product for therapeutic intervention in patients suffering from complement dysregulation.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Bryopsida , Proteínas do Sistema Complemento , Doenças do Sistema Imunitário/tratamento farmacológico , Animais , Bryopsida/metabolismo , Fator H do Complemento/biossíntese , Fator H do Complemento/metabolismo , Fator H do Complemento/uso terapêutico , Glicosilação , Humanos , Camundongos
4.
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
5.
Pediatr Nephrol ; 30(11): 1951-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25986912

RESUMO

BACKGROUND: C3 glomerulopathies (C3G) are characterized by uncontrolled activation of the alternative pathway of complement. In most patients these diseases progress towards end-stage renal disease, and the risk of recurrence after renal transplantation is high. In the majority of patients, only antibodies against the C3 convertase, termed C3Nef, can be found as a potential pathogenic factor. Although a large variety of therapeutic approaches have been used, no generally accepted therapy exists. METHODS: In four consecutive patients with C3G in whom all known complement factor mutations were excluded and only C3Nef could be identified as a potential cause of disease, a multimodal therapeutic regimen with plasma therapy, corticosteroids and mycophenolate mofetil was used. RESULTS: The multimodal regimen achieved normalization of renal function in all four patients, with complete remission in two patients and a distinct reduction of proteinuria in the other two patients. The single patient with C3 glomerulonephritis (C3GN) and marked terminal complement complex elevation only showed partial remission; further improvement was achieved following the addition of eculizumab to the therapeutic regimen. Repeatedly measured C3Nef levels did not correlate with disease course or therapeutic response in any of the patients. CONCLUSIONS: As this multimodal therapeutic approach was effective in all four treated patients with suspected autoimmune etiology of C3G, it offers a treatment option for severely affected patients with this rare disease until more specific regimens are available.


Assuntos
Fator Nefrítico do Complemento 3/metabolismo , Glomerulonefrite Membranoproliferativa/sangue , Glomerulonefrite Membranoproliferativa/terapia , Imunossupressores/uso terapêutico , Troca Plasmática , Adolescente , Corticosteroides/uso terapêutico , Criança , Terapia Combinada , Feminino , Humanos , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapêutico
6.
Mol Ther Methods Clin Dev ; 25: 360-369, 2022 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-35573045

RESUMO

Adeno-associated virus (AAV) vector applications are often limited by capsid-directed humoral immune responses, mainly through neutralizing antibodies (NAbs), which are present throughout the human population due to natural AAV infections. Currently, antibody levels are often quantified via ELISA-based protocols or by cellular NAb assays and less frequently by in vivo NAb assays in mice. These methods need optimization for each serotype and are often not applicable to AAV variants with poor in vitro transduction. To tackle these limitations, we have established Meso Scale Discovery (MSD)-based assays for the quantification of binding antibodies (BAbs) and NAbs against the three most commonly used AAV serotypes, AAV2, AAV8, and AAV9. Both assays detect anti-AAV-IgG1-3 with high sensitivity and consistency as shown in a screen of sera from 40 healthy human donors. Subsequently, BAb and NAb titers were determined for identification of seronegative animals in a non-human primate (NHP) cohort. Moreover, the MSD-based BAb assay protocol was extended to a panel of 14 different AAV serotypes. In summary, our platform allows a rapid and quantitative assessment of the immunological properties of any natural or engineered AAV variant irrespective of transduction efficiency and enables high-throughput screens.

7.
Stem Cell Reports ; 16(9): 2242-2256, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34525384

RESUMO

Gene therapies using adeno-associated viruses (AAVs) are among the most promising strategies to treat or even cure hereditary and acquired retinal diseases. However, the development of new efficient AAV vectors is slow and costly, largely because of the lack of suitable non-clinical models. By faithfully recreating structure and function of human tissues, human induced pluripotent stem cell (iPSC)-derived retinal organoids could become an essential part of the test cascade addressing translational aspects. Organ-on-chip (OoC) technology further provides the capability to recapitulate microphysiological tissue environments as well as a precise control over structural and temporal parameters. By employing our recently developed retina on chip that merges organoid and OoC technology, we analyzed the efficacy, kinetics, and cell tropism of seven first- and second-generation AAV vectors. The presented data demonstrate the potential of iPSC-based OoC models as the next generation of screening platforms for future gene therapeutic studies.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Células-Tronco Pluripotentes Induzidas/citologia , Dispositivos Lab-On-A-Chip , Organoides/metabolismo , Retina/metabolismo , Transdução Genética , Biomarcadores , Técnicas de Cultura de Células , Técnicas de Cultura de Células em Três Dimensões , Diferenciação Celular , Imunofluorescência , Expressão Gênica , Genes Reporter , Terapia Genética , Humanos , Organoides/citologia , Retina/citologia , Transgenes
8.
Cancer Res ; 67(8): 3518-23, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17440057

RESUMO

Interleukin-2 (IL-2) and its receptor (IL-2R) play a major role in cellular immunity. The monoclonal antibodies basiliximab and daclizumab directed against the IL-2R subunit CD25 are widely used to prevent graft or host rejection after allogeneic tissue transplantation. Although these antibodies have been used for this purpose for many years, their common epitope within the CD25 protein is unknown. We screened a random phage display library to isolate peptides specifically binding to basiliximab. A striking amino acid sequence motif was enriched. This motif is homologous to the peptide ERIYHFV comprising amino acid positions 116 to 122 within the extracellular domain of CD25, suggesting that this is the basiliximab epitope. Basiliximab and daclizumab binding of selected phage was specific, as no binding was observed to isotype antibody controls. Phage binding could be inhibited by the cognate peptide. In cells expressing mutant CD25, binding of basiliximab was abolished when two or more amino acids of the suspected epitope were changed. In contrast, basiliximab binding remained unaffected in cells expressing CD25 versions with mutations outside this epitope. We therefore conclude that the (116)ERIYHFV(122) string within CD25 is the epitope recognized by basiliximab and daclizumab. This epitope overlaps with the interaction site of CD25 and IL-2, thus revealing the structural basis for the inhibition of IL-2R binding by this class of immunosuppressive antibodies.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Epitopos/análise , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Imunossupressores/imunologia , Imunossupressores/farmacologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados , Basiliximab , Sítios de Ligação , Linhagem Celular , Daclizumabe , Humanos , Imunoglobulina G/química , Imunossupressores/química , Interleucina-2/química , Interleucina-2/imunologia , Interleucina-2/metabolismo , Subunidade alfa de Receptor de Interleucina-2/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Biblioteca de Peptídeos , Peptídeos/química , Peptídeos/imunologia , Conformação Proteica , Receptores de Interleucina-2/química , Receptores de Interleucina-2/imunologia , Receptores de Interleucina-2/metabolismo , Proteínas Recombinantes de Fusão/química
9.
Front Plant Sci ; 10: 260, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30949184

RESUMO

The human complement system is an important part of the immune system responsible for lysis and elimination of invading microorganisms and apoptotic body cells. Improper activation of the system due to deficiency, mutations, or autoantibodies of complement regulators, mainly factor H (FH) and FH-related proteins (FHRs), causes severe kidney and eye diseases. However, there is no recombinant FH therapeutic available on the market. The first successful recombinant production of FH was accomplished with the moss bioreactor, Physcomitrella patens. Recently, a synthetic regulator, MFHR1, was designed to generate a multitarget complement inhibitor that combines the activities of FH and the FH-related protein 1 (FHR1). The potential of MFHR1 was demonstrated in a proof-of-concept study with transiently transfected insect cells. Here, we present the stable production of recombinant glyco-engineered MFHR1 in the moss bioreactor. The key features of this system are precise genome engineering via homologous recombination, Good Manufacturing Practice-compliant production in photobioreactors, high batch-to-batch reproducibility, and product stability. Several potential biopharmaceuticals are being produced in this system. In some cases, these are even biobetters, i.e., the recombinant proteins produced in moss have a superior quality compared to their counterparts from mammalian systems as for example moss-made aGal, which successfully passed phase I clinical trials. Via mass spectrometry-based analysis of moss-produced MFHR1, we now prove the correct synthesis and modification of this glycoprotein with predominantly complex-type N-glycan attachment. Moss-produced MFHR1 exhibits cofactor and decay acceleration activities comparable to FH, and its mechanism of action on multiple levels within the alternative pathway of complement activation led to a strong inhibitory activity on the whole alternative pathway, which was higher than with the physiological regulator FH.

10.
Expert Opin Drug Deliv ; 5(6): 653-63, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18532921

RESUMO

BACKGROUND: Acute myeloid leukemia was among the first malignancies to be cured by drug therapy alone, but overall survival rates remain unsatisfactory and have changed little over the past 20 years. Conventional chemotherapeutic regimens, which almost invariably include cytarabine and anthracyclines, are untargeted, and more specific therapies are needed. OBJECTIVE: We have chosen acute myeloid leukemia as a disease prototype to review established and novel targeted approaches in leukemia treatment. METHODS: Our selection of the reviewed literature focused on drug delivery aspects. CONCLUSION: While the toxicity profile of chemotherapeutics has been improved by liposomal formulations and antibody conjugation for leukemia-directed uptake, their efficacy has probably not changed significantly. Drugs with an alternative mode of action, including kinase inhibitors, hold great promise. Further improvements may result from the characterization of novel acute myeloid leukemia (AML) cell surface receptors and of leukemic stem cells, as well as from the design of leukemia-targeted gene therapy vectors.


Assuntos
Antineoplásicos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Sistemas de Liberação de Medicamentos , Terapia Genética , Humanos , Integrina alfa4beta1/antagonistas & inibidores , Leucemia Mieloide Aguda/tratamento farmacológico , Lipossomos , Células-Tronco Neoplásicas/efeitos dos fármacos , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico
11.
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
12.
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
13.
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
14.
PLoS One ; 6(8): e23101, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21850255

RESUMO

Efficiency and specificity of viral vectors are vital issues in gene therapy. Insertion of peptide ligands into the adeno-associated viral (AAV) capsid at receptor binding sites can re-target AAV2-derived vectors to alternative cell types. Also, the use of serotypes AAV8 and -9 is more efficient than AAV2 for gene transfer to certain tissues in vivo. Consequently, re-targeting of these serotypes by ligand insertion could be a promising approach but has not been explored so far. Here, we generated AAV8 and -9 vectors displaying peptides in the threefold spike capsid domain. These peptides had been selected from peptide libraries displayed on capsids of AAV serotype 2 to optimize systemic gene delivery to murine lung tissue and to breast cancer tissue in PymT transgenic mice (PymT). Such peptide insertions at position 590 of the AAV8 capsid and position 589 of the AAV9 capsid changed the transduction properties of both serotypes. However, both peptides inserted in AAV8 did not result in the same changes of tissue tropism as they did in AAV2. While the AAV2 peptides selected on murine lung tissue did not alter tropism of serotypes 8 and -9, insertion of the AAV2-derived peptide selected on breast cancer tissue augmented tumor gene delivery in both serotypes. Further, this peptide mediated a strong but unspecific in vivo gene transfer for AAV8 and abrogated transduction of various control tissues for AAV9. Our findings indicate that peptide insertion into defined sites of AAV8 and -9 capsids can change and improve their efficiency and specificity compared to their wild type variants and to AAV2, making these insertion sites attractive for the generation of novel targeted vectors in these serotypes.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Peptídeos/metabolismo , Transdução Genética/métodos , Animais , Neoplasias da Mama/metabolismo , Capsídeo/química , Capsídeo/metabolismo , Linhagem Celular , Feminino , Humanos , Técnicas In Vitro , Pulmão/metabolismo , Camundongos , Camundongos Transgênicos , Peptídeos/genética , Sorotipagem
15.
Toxins (Basel) ; 2(12): 2754-68, 2010 12.
Artigo em Inglês | MEDLINE | ID: mdl-22069574

RESUMO

Vectors based on adeno-associated virus (AAV) are promising tools for gene therapy. The production of strongly toxic vectors, for example for cancer-directed gene transfer, is often unfeasible due to uncontrolled expression of toxic genes in vector-producing cells. Using an approach based on transcriptional repression, we have created novel AAV vectors carrying the genes coding for diphtheria toxin A (DTA) and the pro-apoptotic PUMA protein. The DTA vector had a significant toxic effect on a panel of tumor cell lines, and abrogation of protein synthesis could be shown. The PUMA vector had a toxic effect on HeLa and RPMI 8226 cells, and sensitized transduced cells to doxorubicin. To permit targeted gene transfer, we incorporated the DTA gene into a genetically modified AAV-2 capsid previously developed by our group that mediates enhanced transduction of murine breast cancer cells in vitro. This vector had a stronger cytotoxic effect on breast cancer cells than DTA vectors with wildtype AAV capsid or vectors with a random capsid modification. The vector production and application system presented here allows for easy exchange of promotors, transgenes and capsid specificity for certain target cells. It will therefore be of great possible value in a broad range of applications in cytotoxic gene therapy and significantly broadens the spectrum of available tools for AAV-based gene therapy.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Dependovirus/genética , Toxina Diftérica/genética , Vetores Genéticos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Células HEK293 , Células HeLa , Humanos , Camundongos , Plasmídeos , Células Tumorais Cultivadas
16.
Adv Genet ; 67: 29-60, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19914449

RESUMO

Efficient and specific delivery of genes to the cell type of interest is a crucial issue in gene therapy. Adeno-associated virus (AAV) has gained particular interest as gene vector recently and is therefore the focus of this chapter. Its low frequency of random integration into the genome and the moderate immune response make AAV an attractive platform for vector design. Like in most other vector systems, the tropism of AAV vectors limits their utility for certain tissues especially upon systemic application. This may in part be circumvented by using AAV serotypes with an in vivo gene transduction pattern most closely fitting the needs of the application. Also, the tropism of AAV capsids may be changed by combining parts of the natural serotype diversity. In addition, peptides mediating binding to the cell type of interest can be identified by random phage display library screening and subsequently be introduced into an AAV capsid region critical for receptor binding. Such peptide insertions can abrogate the natural tropism of AAV capsids and result in detargeting from the liver in vivo. In a novel approach, cell type-directed vector capsids can be selected from random peptide libraries displayed on viral capsids or serotype-shuffling libraries in vitro and in vivo for optimized transduction of the cell type or tissue of interest.


Assuntos
Dependovirus/genética , Vetores Genéticos , Capsídeo/química , Genoma Viral , Modelos Moleculares , Biblioteca de Peptídeos , Reação em Cadeia da Polimerase , Receptores Virais/metabolismo , Transgenes
17.
PLoS One ; 4(4): e5122, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19357785

RESUMO

Targeting viral vectors to certain tissues in vivo has been a major challenge in gene therapy. Cell type-directed vector capsids can be selected from random peptide libraries displayed on viral capsids in vitro but so far this system could not easily be translated to in vivo applications. Using a novel, PCR-based amplification protocol for peptide libraries displayed on adeno-associated virus (AAV), we selected vectors for optimized transduction of primary tumor cells in vitro. However, these vectors were not suitable for transduction of the same target cells under in vivo conditions. We therefore performed selections of AAV peptide libraries in vivo in living animals after intravenous administration using tumor and lung tissue as prototype targets. Analysis of peptide sequences of AAV clones after several rounds of selection yielded distinct sequence motifs for both tissues. The selected clones indeed conferred gene expression in the target tissue while gene expression was undetectable in animals injected with control vectors. However, all of the vectors selected for tumor transduction also transduced heart tissue and the vectors selected for lung transduction also transduced a number of other tissues, particularly and invariably the heart. This suggests that modification of the heparin binding motif by target-binding peptide insertion is necessary but not sufficient to achieve tissue-specific transgene expression. While the approach presented here does not yield vectors whose expression is confined to one target tissue, it is a useful tool for in vivo tissue transduction when expression in tissues other than the primary target is uncritical.


Assuntos
Dependovirus , Vetores Genéticos , Biblioteca de Peptídeos , Tropismo , Sequência de Aminoácidos , Animais , Neoplasias da Mama , Capsídeo/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Feminino , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase/métodos , Transdução Genética , Tropismo/genética , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA