1
|
Steininger T, Öttl V, Franken LE, Frank C, Ohland P, Lopez Ferreiro M, Klostermann S, Fritsch J, Hirschauer E, Sandmeir A, Hilgenfeld LD, Semmelmann F, Dürr MS, Konkel F, Pechmann G, Linder S, Haindl M, Yazicioglu MN, Ringler P, Lauer ME, Phichith D, Seeber S, Fakhiri J. Improved Recombinant Adeno-Associated Viral Vector Production via Molecular Evolution of the Viral Rep Protein. Int J Mol Sci 2025; 26:1319. [PMID: 39941089 PMCID: PMC11818820 DOI: 10.3390/ijms26031319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
In the dynamic field of gene therapy, recombinant adeno-associated viruses (rAAVs) have become leading viral vectors due to their safety, long-term expression, and wide-ranging cell and tissue tropism. With numerous FDA approvals and commercial products underscoring their potential, there is a critical need for efficient production processes to achieve high vector titers and quality. A major challenge in rAAV production is the efficient packaging of the genome into the viral capsid, with empty or partially filled capsids often representing over 90% of the produced material. To tackle this issue, we engineered the replication and packaging proteins of an AAV (Rep) to boost their functionality and improve vector titers. We subjected a complex Rep library derived from the AAV serotypes 1-13 to directed evolution in an AAV producer cell line. After each round of selection, single clones were analyzed, showing enrichment of specific hybrid Rep domains. Comparative analysis of these selected clones revealed considerable differences in their ability to package AAV2-based viral genomes, with hybrid Rep proteins achieving up to a 2.5-fold increase in packaging efficiency compared to their parental counterparts. These results suggest that optimizing rep gene variants through directed evolution is an effective strategy to enhance rAAV production efficiency.
Collapse
Affiliation(s)
- Thomas Steininger
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (T.S.); (V.Ö.); (C.F.); (J.F.); (E.H.); (F.K.); (S.L.); (S.S.)
- Faculty of Bioengineering, University of Applied Sciences Weihenstephan-Triesdorf, Am Hofgarten 4, 85354 Freising, Germany
- Faculty 06, Munich University of Applied Sciences, Lothstraße 34, 80335 Munich, Germany
| | - Veronika Öttl
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (T.S.); (V.Ö.); (C.F.); (J.F.); (E.H.); (F.K.); (S.L.); (S.S.)
- Faculty of Medicine, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Linda E. Franken
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland; (L.E.F.); (P.O.); (M.L.F.); (P.R.); (M.E.L.); (D.P.)
| | - Cornelius Frank
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (T.S.); (V.Ö.); (C.F.); (J.F.); (E.H.); (F.K.); (S.L.); (S.S.)
- Gene Therapy Technical Research & Development, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (M.-S.D.); (G.P.); (M.H.)
| | - Philip Ohland
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland; (L.E.F.); (P.O.); (M.L.F.); (P.R.); (M.E.L.); (D.P.)
- Gene Therapy Technical Research & Development, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (M.-S.D.); (G.P.); (M.H.)
| | - Miriam Lopez Ferreiro
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland; (L.E.F.); (P.O.); (M.L.F.); (P.R.); (M.E.L.); (D.P.)
| | - Stefan Klostermann
- Roche Pharma Research and Early Development, Data and Analytics, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany;
| | - Johannes Fritsch
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (T.S.); (V.Ö.); (C.F.); (J.F.); (E.H.); (F.K.); (S.L.); (S.S.)
| | - Evelyn Hirschauer
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (T.S.); (V.Ö.); (C.F.); (J.F.); (E.H.); (F.K.); (S.L.); (S.S.)
| | - Anna Sandmeir
- Technical Development Analytics, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (A.S.); (L.D.H.); (F.S.)
| | - Luisa D. Hilgenfeld
- Technical Development Analytics, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (A.S.); (L.D.H.); (F.S.)
| | - Florian Semmelmann
- Technical Development Analytics, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (A.S.); (L.D.H.); (F.S.)
| | - Marie-Sofie Dürr
- Gene Therapy Technical Research & Development, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (M.-S.D.); (G.P.); (M.H.)
| | - Fabian Konkel
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (T.S.); (V.Ö.); (C.F.); (J.F.); (E.H.); (F.K.); (S.L.); (S.S.)
| | - Gregor Pechmann
- Gene Therapy Technical Research & Development, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (M.-S.D.); (G.P.); (M.H.)
| | - Sabine Linder
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (T.S.); (V.Ö.); (C.F.); (J.F.); (E.H.); (F.K.); (S.L.); (S.S.)
| | - Markus Haindl
- Gene Therapy Technical Research & Development, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (M.-S.D.); (G.P.); (M.H.)
| | - Mustafa N. Yazicioglu
- Spark Therapeutics, Roche Holding AG, 3737 Market Street, Philadelphia, PA 19104, USA;
| | - Philippe Ringler
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland; (L.E.F.); (P.O.); (M.L.F.); (P.R.); (M.E.L.); (D.P.)
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Matthias E. Lauer
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland; (L.E.F.); (P.O.); (M.L.F.); (P.R.); (M.E.L.); (D.P.)
| | - Denis Phichith
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland; (L.E.F.); (P.O.); (M.L.F.); (P.R.); (M.E.L.); (D.P.)
| | - Stefan Seeber
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (T.S.); (V.Ö.); (C.F.); (J.F.); (E.H.); (F.K.); (S.L.); (S.S.)
| | - Julia Fakhiri
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany; (T.S.); (V.Ö.); (C.F.); (J.F.); (E.H.); (F.K.); (S.L.); (S.S.)
| |
Collapse
|
2
|
Sant’Anna TB, Araujo NM. Adeno-associated virus infection and its impact in human health: an overview. Virol J 2022; 19:173. [PMID: 36316711 PMCID: PMC9623951 DOI: 10.1186/s12985-022-01900-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 01/24/2023] Open
Abstract
Discovered as a contaminant of adenovirus stocks in the 1960s, adeno-associated virus (AAV) is a mono-stranded DNA virus that depends on helper factors to replicate. Even though AAV is endemic in the human population (35-80%), it is remarkable that many issues concerning the natural infection by this virus remain unanswered. In this study, we reflect on the main basic aspects of AAV biology and provide an overview of the studies exploring the impact of AAV infection on human health, focusing on three major research areas including, (i) cervical and (ii) liver cancer, and (iii) reproductive system disorders. Conflicting results have been obtained into the association of AAV infection with the occurrence of adverse reproductive outcomes, such as placental complications, spontaneous abortion, and fertility disorders, or with a protective role in HPV-related cervical carcinogenesis. Noteworthy, recent reports have identified AAV insertional mutagenesis as a novel risk factor for the development of hepatocellular carcinoma. This latest finding raises concern regarding the widespread usage of AAV vectors in liver-targeted gene therapy.
Collapse
Affiliation(s)
- Thaís B Sant’Anna
- grid.418068.30000 0001 0723 0931Laboratory of Molecular Virology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Natalia M Araujo
- grid.418068.30000 0001 0723 0931Laboratory of Molecular Virology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Meier AF, Fraefel C, Seyffert M. The Interplay between Adeno-Associated Virus and its Helper Viruses. Viruses 2020; 12:E662. [PMID: 32575422 PMCID: PMC7354565 DOI: 10.3390/v12060662] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 12/14/2022] Open
Abstract
The adeno-associated virus (AAV) is a small, nonpathogenic parvovirus, which depends on helper factors to replicate. Those helper factors can be provided by coinfecting helper viruses such as adenoviruses, herpesviruses, or papillomaviruses. We review the basic biology of AAV and its most-studied helper viruses, adenovirus type 5 (AdV5) and herpes simplex virus type 1 (HSV-1). We further outline the direct and indirect interactions of AAV with those and additional helper viruses.
Collapse
Affiliation(s)
| | | | - Michael Seyffert
- Institute of Virology, University of Zurich, CH-8057 Zurich, Switzerland; (A.F.M.); (C.F.)
| |
Collapse
|
4
|
Chen W, Tan L, Zhou Q, Li W, Li T, Zhang C, Wu J. AAVS1 site-specific integration of the CAR gene into human primary T cells using a linear closed-ended AAV-based DNA vector. J Gene Med 2020; 22:e3157. [PMID: 31901177 DOI: 10.1002/jgm.3157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/31/2019] [Accepted: 12/31/2019] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Use of chimeric antigen receptor (CAR) T cells has become a promising strategy in cancer immunotherapy. However, safety in clinical application is also one of the most controversial issues. METHODS In the present study, we investigated the application of a non-viral site-directed vector (CELiD [closed-ended linear duplex DNA]) dependent on adeno-associated virus (AAV) genomes for the purpose of safe CAR-T engineering. We co-electroporated CD19-CAR encoding "CELiD" vectors with plasmid pCMV-Rep into human T cells and ensured stably transfected CAR-T cells by G418 selection. The efficiency of AAVS1 site-specific integration was analyzed by a real-time polymerase chain reaction. RESULTS CAR-T cells engineered by CELiD vectors could be established within 20 days with up to 22.8% AAVS1 site-specific integration efficiency. CAR expression and cytokine secretion of CAR modified T cells were evaluated in vitro. Abundant effector cytokines were produced by the CAR-T cells engineered by CELiD vectors compared to control T cells and the killing efficiency of target cells was estimated to as high as 75% in vitro. CONCLUSIONS With the help of the AAV-derived CELiD vector, CAR genes were preferentially integrated into the AAVS1 site. This technology could be utilized in human T cell modification and remove the safety constraints of CAR-T therapy.
Collapse
Affiliation(s)
- Wei Chen
- Suzhou Red Cross Blood Center, Suzhou, China
| | - Liang Tan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | | | - WenSheng Li
- Aier School of Ophthalmology, Central South University, Changsha, China
| | - Taiming Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chun Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | | |
Collapse
|
5
|
Zhang F, Aguilera J, Beaudet JM, Xie Q, Lerch TF, Davulcu O, Colón W, Chapman MS, Linhardt RJ. Characterization of interactions between heparin/glycosaminoglycan and adeno-associated virus. Biochemistry 2013; 52:6275-85. [PMID: 23952613 PMCID: PMC3859860 DOI: 10.1021/bi4008676] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Adeno-associated virus (AAV) is a key candidate in the development of gene therapy. In this work, we used surface plasmon resonance spectroscopy to study the interaction between AAV and heparin and other glycosaminoglycans (GAGs). Surface plasmon resonance results revealed that heparin binds to AAV with an extremely high affinity. Solution competition studies showed that binding of AAV to heparin is chain length-dependent. AAV prefers to bind full chain heparin. All sulfo groups (especially N-sulfo and 6-O-sulfo groups) on heparin are important for the AAV-heparin interaction. Higher levels of sulfo group substitution in GAGs enhance their binding affinities. Atomic force microscopy was also performed to image AAV-2 in a complex with heparin.
Collapse
Affiliation(s)
- Fuming Zhang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Javier Aguilera
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Julie M. Beaudet
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Qing Xie
- Department of Biochemistry and Molecular Biology School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Thomas F. Lerch
- Department of Biochemistry and Molecular Biology School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Omar Davulcu
- Department of Biochemistry and Molecular Biology School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Wilfredo Colón
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Michael S. Chapman
- Department of Biochemistry and Molecular Biology School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Robert J. Linhardt
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Departments of Biology and Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
6
|
Berns KI. My life with adeno-associated virus: a long time spent studying a short genome. DNA Cell Biol 2013; 32:342-7. [PMID: 23781880 DOI: 10.1089/dna.2013.2120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
My 45 years of studying the molecular biology of adeno-associated virus are recounted. Additional activities as a mentor, department chair, and medical school administrator are described, as are my activities in the public sphere, which involved national issues related to science policy and medical education.
Collapse
Affiliation(s)
- Kenneth I Berns
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida 32608, USA.
| |
Collapse
|
7
|
Lisowski L, Lau A, Wang Z, Zhang Y, Zhang F, Grompe M, Kay MA. Ribosomal DNA integrating rAAV-rDNA vectors allow for stable transgene expression. Mol Ther 2012; 20:1912-23. [PMID: 22990671 DOI: 10.1038/mt.2012.164] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although recombinant adeno-associated virus (rAAV) vectors are proving to be efficacious in clinical trials, the episomal character of the delivered transgene restricts their effectiveness to use in quiescent tissues, and may not provide lifelong expression. In contrast, integrating vectors enhance the risk of insertional mutagenesis. In an attempt to overcome both of these limitations, we created new rAAV-rDNA vectors, with an expression cassette flanked by ribosomal DNA (rDNA) sequences capable of homologous recombination into genomic rDNA. We show that after in vivo delivery the rAAV-rDNA vectors integrated into the genomic rDNA locus 8-13 times more frequently than control vectors, providing an estimate that 23-39% of the integrations were specific to the rDNA locus. Moreover, a rAAV-rDNA vector containing a human factor IX (hFIX) expression cassette resulted in sustained therapeutic levels of serum hFIX even after repeated manipulations to induce liver regeneration. Because of the relative safety of integration in the rDNA locus, these vectors expand the usage of rAAV for therapeutics requiring long-term gene transfer into dividing cells.
Collapse
Affiliation(s)
- Leszek Lisowski
- Stanford University, Departments of Pediatrics and Genetics, Stanford, California 94305-5164, USA
| | | | | | | | | | | | | |
Collapse
|
8
|
Zacharias J, Romanova LG, Menk J, Philpott NJ. p53 inhibits adeno-associated viral vector integration. Hum Gene Ther 2011; 22:1445-51. [PMID: 21506732 DOI: 10.1089/hum.2011.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Adeno-associated viral (AAV) vectors preferentially integrate into the genome of cells that are defective in DNA repair, such as occurs with DNA-PKcs deficiency or poly(ADP-ribose) polymerase-1 down-regulation. As the tumor suppressor protein p53 regulates the transcription of many genes involved in DNA repair, we sought to determine whether functional p53 affects the efficiency of AAV integration. p53 is mutated in more than 50% of cancers, and site-specific integration of AAV into the AAVS1 site of human chromosome 19 has frequently been observed in transformed cancer cell lines, but rarely seen in primary cells or in vivo. We therefore hypothesized that p53-negative cells would be more permissive to AAV integration than p53-positive cells. The integration of a rep- and green fluorescent protein-encoding recombinant AAV vector was quantified in p53-expressing and p53-deficient HCT116 colon cancer cells. Our results show that there is a higher efficiency of AAV integration in p53-negative cells compared with p53-positive cells, indicating that p53 does indeed inhibit AAV integration. Further experiments suggest that this p53-mediated block to AAV integration is likely to be through binding of p53 to the AAV Rep protein and the consequent inhibition of Rep activity during AAV integration.
Collapse
Affiliation(s)
- Jeana Zacharias
- Division of Rheumatic and Autoimmune Diseases, Department of Medicine, Institute of Human Genetics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
9
|
|
10
|
Integration preferences of wildtype AAV-2 for consensus rep-binding sites at numerous loci in the human genome. PLoS Pathog 2010; 6:e1000985. [PMID: 20628575 PMCID: PMC2900306 DOI: 10.1371/journal.ppat.1000985] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Accepted: 06/03/2010] [Indexed: 01/21/2023] Open
Abstract
Adeno-associated virus type 2 (AAV) is known to establish latency by preferential integration in human chromosome 19q13.42. The AAV non-structural protein Rep appears to target a site called AAVS1 by simultaneously binding to Rep-binding sites (RBS) present on the AAV genome and within AAVS1. In the absence of Rep, as is the case with AAV vectors, chromosomal integration is rare and random. For a genome-wide survey of wildtype AAV integration a linker-selection-mediated (LSM)-PCR strategy was designed to retrieve AAV-chromosomal junctions. DNA sequence determination revealed wildtype AAV integration sites scattered over the entire human genome. The bioinformatic analysis of these integration sites compared to those of rep-deficient AAV vectors revealed a highly significant overrepresentation of integration events near to consensus RBS. Integration hotspots included AAVS1 with 10% of total events. Novel hotspots near consensus RBS were identified on chromosome 5p13.3 denoted AAVS2 and on chromsome 3p24.3 denoted AAVS3. AAVS2 displayed seven independent junctions clustered within only 14 bp of a consensus RBS which proved to bind Rep in vitro similar to the RBS in AAVS3. Expression of Rep in the presence of rep-deficient AAV vectors shifted targeting preferences from random integration back to the neighbourhood of consensus RBS at hotspots and numerous additional sites in the human genome. In summary, targeted AAV integration is not as specific for AAVS1 as previously assumed. Rather, Rep targets AAV to integrate into open chromatin regions in the reach of various, consensus RBS homologues in the human genome. This is the first unbiased genome-wide analysis of wildtype AAV integration combined with a thorough bioinformatic analysis of preferred genomic motifs and patterns in the neighbourhood of the integration sites identified. The preference of Rep-dependent AAV integration near multiple consensus Rep-binding sites was lost in the case of AAV vector integration in the absence of Rep expression. Our findings challenge the commonly accepted notion of site-specific AAV targeting to AAVS1 on chromosome 19q13.42. Although AAVS1 contains a canonical Rep-binding site, numerous additional sites including the newly identified hotspots AAVS2 on chromosome 5p13.3 and AAVS3 on chromosome 3p24.3 harbour functional Rep-binding sites suitable for AAV integration. AAV vectors are quickly moving forward in the clinic and Rep-dependent vector targeting strategies are being actively pursued. Detailed information of AAV wildtype versus recombinant AAV vector integration sites and preferences are needed to evaluate the safety profile of AAV vectors in gene therapy.
Collapse
|
11
|
Abstract
Cystic Fibrosis (CF) is an autosomal recessive disorder due to mutations in the CF transmembrane conductance regulator (CFTR) gene that lead to defective ion transport in the conducting pulmonary airways and exocrine glands. Through a process that is not fully understood, CFTR defects predispose affected patients to chronic endobronchial infections with organisms such as Pseudomonas aeruginosa and Staphylococcus aureus. Following the discovery of the CFTR gene in 1989, CF became one of the primary targets for gene therapy research. Early enthusiasm surrounded the new field of gene therapy during most of the 1990s and it led academics and clinicians on a big effort to apply gene therapy for cystic fibrosis. Clinical studies have been pursued using recombinant adenovirus, recombinant adeno-associated virus, cationic liposomes, and cationic polymer vectors. Although to this date no dramatic therapeutic benefits have been observed, a lot of information has been gained from the pre-clinical and clinical studies that were performed. This learning curve has led to the optimization of vector technology and an appreciation of immune and mechanical barriers that have to be overcome for successful delivery.
Collapse
|
12
|
Yamamoto N, Suzuki M, Kawano MA, Inoue T, Takahashi RU, Tsukamoto H, Enomoto T, Yamaguchi Y, Wada T, Handa H. Adeno-associated virus site-specific integration is regulated by TRP-185. J Virol 2006; 81:1990-2001. [PMID: 17151120 PMCID: PMC1797547 DOI: 10.1128/jvi.02014-06] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adeno-associated virus (AAV) integrates site specifically into the AAVS1 locus on human chromosome 19. Although recruitment of the AAV nonstructural protein Rep78/68 to the Rep binding site (RBS) on AAVS1 is thought to be an essential step, the mechanism of the site-specific integration, particularly, how the site of integration is determined, remains largely unknown. Here we describe the identification and characterization of a new cellular regulator of AAV site-specific integration. TAR RNA loop binding protein 185 (TRP-185), previously reported to associate with human immunodeficiency virus type 1 TAR RNA, binds to AAVS1 DNA. Our data suggest that TRP-185 suppresses AAV integration at the AAVS1 RBS and enhances AAV integration into a region downstream of the RBS. TRP-185 bound to Rep68 directly, changing the Rep68 DNA binding property and stimulating Rep68 helicase activity. We present a model in which TRP-185 changes the specificity of the AAV integration site from the RBS to a downstream region by acting as a molecular chaperone that promotes Rep68 complex formation competent for 3'-->5' DNA helicase activity.
Collapse
Affiliation(s)
- Noriaki Yamamoto
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
The driving interest in adeno-associated virus (AAV) has been its potential as a gene delivery vector. The early observation that AAV can establish a latent infection by integrating into the host chromosome has been central to this interest. However, chromosomal integration is a two-edged sword, imparting on one hand the ability to maintain the therapeutic gene in progeny cells, and on the other hand, the risk of mutations that are deleterious to the host. A clearer understanding of the mechanism and efficiency of AAV integration, in terms of contributing viral and host-cell factors and circumstances, will provide a context in which to evaluate these potential benefits and risks. Research to date suggests that AAV integration in any context is inefficient, and that the persistence of AAV gene delivery vectors in tissues is largely attributable to episomal genomes.
Collapse
Affiliation(s)
- Douglas M McCarty
- School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | | | |
Collapse
|
14
|
Song S, Lu Y, Choi YK, Han Y, Tang Q, Zhao G, Berns KI, Flotte TR. DNA-dependent PK inhibits adeno-associated virus DNA integration. Proc Natl Acad Sci U S A 2004; 101:2112-6. [PMID: 14766968 PMCID: PMC357060 DOI: 10.1073/pnas.0307833100] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent studies have shown that recombinant adeno-associated virus (rAAV) can persist in episomal form; however, factors affecting rAAV persistence are poorly understood. DNA-dependent PK (DNA-PK) is a DNA repair enzyme, which we previously found played an important role in determining the molecular fate of the rAAV genome in mouse skeletal muscle. In the present study, we tested the effect of DNA-PK on AAV serotype 2 integration in vitro and in vivo in mouse liver. In an in vitro integration system, addition of DNA-PK decreased AAV integration, whereas antibody against DNA-PKcs increased integration. In vivo, matched doses of a recombinant AAV serotype 2 vector were injected into the portal vein of either C57BL/6 (DNA-PKcs(+/+)) or severe combined immunodeficient (DNA-PKcs(-/-)) mice. After partial hepatectomy to stimulate hepatocyte proliferation, retention of vector genomes and of transgene expression was substantially higher in severe combined immunodeficient mice, indicating that in the absence of DNA-PKcs, a greater proportion of genomes integrated into the cellular genome. In summary, we have provided evidence that DNA-PK inhibits AAV integration both in vitro and in vivo.
Collapse
Affiliation(s)
- Sihong Song
- Department of Pharmaceutics, Powell Gene Therapy Center, Genetics Institute, University of Florida, Gainesville, FL 32610, USA.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Philpott NJ, Gomos J, Falck-Pedersen E. Transgene Expression After Rep-Mediated Site-Specific Integration into Chromosome 19. Hum Gene Ther 2004; 15:47-61. [PMID: 14965377 DOI: 10.1089/10430340460732454] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have used a plasmid-based transfection model of the adeno-associated virus (AAV) Rep-mediated site-specific integration (RMSSI) pathway to characterize the stability and expression of a site-specifically integrated transgene (either green fluorescent protein [GFP] or chloramphenicol acetyltransferase [CAT]). Three plasmids containing the AAV p5 integration efficiency element (p5IEE) have been used to study integration and transgene expression in HeLa cells: (1) pRepGFP(itr+) contains both AAV ITRs, rep, and p5IEE and can be used as either a plasmid or rAAV vehicle for integration; (2) pRepGFP(itr-) contains the AAV rep gene and the p5IEE; (3) pAd-p5CAT contains only the 138-bp p5IEE of AAV. The data presented demonstrate that in the absence of drug selection, all three constructs undergo site-specific integration (efficiencies of between 10 and 40% of transduced cell lines). At 6 weeks posttransfection most cell lines that underwent RMSSI also expressed the appropriate transgene product. By 18 weeks posttransfection cell lines that were established with rep in cis to the transgene showed a decline in transgene expression as well as a loss of transgene DNA. In many cell lines, there appears to be transgene-containing DNA that does not contribute to gene expression. Data support a model of gene expression and transgene instability through a Rep-mediated pathway. In contrast to rep-containing cell lines, clonal cell lines containing p5IEECAT (with Rep provided in trans) maintained both the integrated transgene and transgene expression throughout the entire experimental time course (18 weeks).
Collapse
Affiliation(s)
- Nicola J Philpott
- Department of Microbiology and Immunology, William Randolph Hearst Foundation Molecular Biology Graduate Program, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | |
Collapse
|
16
|
Huttner NA, Girod A, Schnittger S, Schoch C, Hallek M, Büning H. Analysis of site-specific transgene integration following cotransduction with recombinant adeno-associated virus and a rep encodingplasmid. J Gene Med 2003; 5:120-9. [PMID: 12539150 DOI: 10.1002/jgm.324] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Recombinant adeno-associated virus (rAAV) has many advantages for gene therapeutic applications in comparison with other vector systems. One of the most promising features is the ability of wild-type (wt) AAV to integrate site-specifically into human chromosome 19. However, this feature is lost in rAAV vectors due to the removal of the rep-coding sequences. METHODS HeLa cells were transfected with a rep expression plasmid, infected by rAAV and grown with or without selection pressure. Single cell clones were generated and genomic DNA was analyzed for site-specific integration by Southern blotting analysis and fluorescence in situ hybridization (FISH). RESULTS Transfection of HeLa cells with a rep expression plasmid followed by transduction with a rAAV vector resulted in site-specific integration of the transgene at AAVS1 on human chromosome 19 in 7 of 10 cell clones analyzed. In marked contrast, transduction of cells with rAAV alone did not result in any site-specific integration of the transgene. CONCLUSIONS The high frequency with which the site-specific integration took place in the presence of Rep protein is comparable with the results observed with wtAAV. These results offer opportunities for the development of specifically integrating rAAV vectors.
Collapse
Affiliation(s)
- Nadja A Huttner
- Gene Center of the Ludwig-Maximilians-University Munich, Feodor-Lynen-Str. 25, 81377 Munich, Germany
| | | | | | | | | | | |
Collapse
|
17
|
Amiss TJ, McCarty DM, Skulimowski A, Samulski RJ. Identification and characterization of an adeno-associated virus integration site in CV-1 cells from the African green monkey. J Virol 2003; 77:1904-15. [PMID: 12525625 PMCID: PMC140930 DOI: 10.1128/jvi.77.3.1904-1915.2003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2002] [Accepted: 11/04/2002] [Indexed: 01/09/2023] Open
Abstract
Adeno-associated virus (AAV) is a classification given to a group of nonpathogenic, single-stranded DNA viruses known to reside latently in primates. During latency in humans, AAV type 2 (AAV2) preferentially integrates at a site on chromosome 19q13.3ter by targeting a sequence composed of an AAV Rep binding element (RBE), a spacer, and a nicking site. Here, we report the DNA sequence of an African green monkey AAV integration site isolated from CV-1 cells. Overall, it has 98% homology to the analogous human site, including identical spacer and nicking sequences. However, the simian RBE is expanded, having five perfect directly repeated GAGC tetramers. We carried out a number of in vitro and in vivo assays to determine the effect of this expanded RBE sequence on the Rep-RBE interaction and AAV targeted integration. Using electromobility shift assays it was demonstrated that AAV4 Rep68 bound the expanded RBE with a sixfold-greater affinity than the human RBE. To determine the basis for the affinity increase, DNase I protection and methylation interference (MI) assays were performed. Comparison of footprints on both the human and simian RBEs revealed nearly identical protection; however, MI analysis suggested greater interaction with the guanine nucleotides of the expanded RBE, thus providing a biochemical basis for the increased binding activity. In vivo, integration targeted to the simian RBE was demonstrated by PCR analysis of latently infected Cos-7 cells. Interestingly, the frequency of site-specific integration was twofold greater in Cos-7 cells than in HeLa cells. Overall, these experiments establish that the simian RBE, identified in CV-1 cells, functions analogously to the human RBE and provide further evidence for a developing model that proposes individual roles for the RBE and the spacer and nicking site elements.
Collapse
Affiliation(s)
- Terry J Amiss
- Department of Pharmacology. Gene Therapy Center. Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
18
|
Philpott NJ, Gomos J, Berns KI, Falck-Pedersen E. A p5 integration efficiency element mediates Rep-dependent integration into AAVS1 at chromosome 19. Proc Natl Acad Sci U S A 2002; 99:12381-5. [PMID: 12221283 PMCID: PMC129453 DOI: 10.1073/pnas.182430299] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adeno-associated virus (AAV) undergoes site-specific integration into human chromosome 19 through a deletion-substitution mechanism at the well characterized AAVS1 site. We have shown previously that a cis element within the left end of the AAV genome enhances the efficiency of Rep-mediated site-specific integration into chromosome 19 when present in inverted terminal repeat-containing recombinant AAV (rAAV) plasmids. We now demonstrate that a 138-bp cis element, the p5 integration efficiency element (p5IEE), mediates efficient integration. The p5IEE is not only required for efficient site-specific integration, it is also sufficient. Integration mediated by the p5IEE occurs in the absence of the AAV inverted terminal-repeat elements. The data presented in this study demonstrate that the p5IEE is a multifunctional element, serving as the highly regulatable Rep promoter and the primary substrate for targeted integration.
Collapse
Affiliation(s)
- Nicola J Philpott
- Weill Medical College of Cornell University, Hearst Research Foundation Department of Microbiology and Immunology, Molecular Biology Graduate Program, New York, NY 10021, USA
| | | | | | | |
Collapse
|
19
|
Dong WJ, Wu XB, Liu DP, Li JL, Liu G, Zu ZX, Zhao N, Hou YD, Liang CC. Analysis of adeno-associated virus-mediated ex vivo transferred human beta-globin gene in bone marrow engrafted mice. J Biomed Sci 2002; 9:253-60. [PMID: 12065900 DOI: 10.1007/bf02256072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Adeno-associated virus (AAV)-2 was developed as a useful vector for human gene therapy. In this report, we analyzed the integration and expression of AAV-mediated ex vivo transferred human beta-globin gene in bone marrow (BM) reconstituted mice. Recombinant AAV (rAAV) containing human beta-globin gene was packaged by infecting individual G418-resistant BHK-21 cell clones integrated with the plasmid AV53HS432Deltabeta2.0Neo with recombinant herpes simplex virus, which can express rep and cap genes of wild-type AAV. The titer of rAAV was determined using slot blot hybridization with a result of 10(13) virus particles/ml (genome copy number). Low-density mononuclear cells were isolated from fetal livers of embryos from pregnant C57BL/6 mice at 14-16 days of gestation and were infected with rAAV. The transduced hematopoietic cells were then reinfused into lethally irradiated C57BL/6 recipient mice via tail vein injection. To analyze the provirus in short-term and long-term BM reconstituted mice, PCR/Southern blot and RT-PCR were performed to identify the integrity of the provirus and to detect the expression of human beta-globin gene, respectively. Genomic DNA was extracted from spleen nodules of BM reconstituted mice 12 days after transplantation. Human beta-globin gene was detected in 1 out of 6 nodules using PCR combined with Southern blot. Human beta-globin gene was also detected in the BM and thymus of mouse Y6161, in the thymus and spleen of mouse Y6162 and in the BM of mice Y6211 and Y6212. RT-PCR revealed low levels of expression of human beta-globin gene in the BM of mouse Y6211. Our results suggested that the efficiency of AAV-mediated human beta-globin gene integration into hematopoietic stem/progenitor cells was very low. It is necessary to perform further research on AAV biology before applying gene therapy that requires integration of a foreign gene into host chromosomes.
Collapse
Affiliation(s)
- Wen-Ji Dong
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Musatov SA, Dudus L, Parrish CM, Scully TA, Fisher KJ. Spontaneous mobilization of integrated recombinant adenoassociated virus in a cell culture model of virus latency. Virology 2002; 294:151-69. [PMID: 11886274 DOI: 10.1006/viro.2001.1267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A cell line containing integrated recombinant adenoassociated virus (AAV) was investigated for spontaneous mobilization of vector sequence. Detection of these rare events was facilitated by using a vector design that allowed the circular rescue product (cAAV) to be individually scored by bacterial transformation. Restriction and sequence analysis of captured clones revealed five highly ordered classes of cAAV, each of which contained a defined segment of the integrated vector locus. A common feature of all cAAV classes was the presence of a modified inverted terminal repeat that joined the ends of the liberated sequence. Assembly of extrachromosomal vector genomes was accompanied by deletions in the integration locus that could be mapped to one of the five cAAV classes, suggesting an excision-type mechanism. We propose that the spontaneous deletion and mobilization of vector sequence from the recombinant adenoassociated virus (rAAV) integration locus is mediated by a recombination event between the inverted terminal repeats that define the boundaries of the individual genome subunits.
Collapse
Affiliation(s)
- Sergei A Musatov
- Department of Pathology and Laboratory Medicine, Tulane University Medical Center, New Orleans, Louisiana, USA
| | | | | | | | | |
Collapse
|
21
|
Young SM, Samulski RJ. Adeno-associated virus (AAV) site-specific recombination does not require a Rep-dependent origin of replication within the AAV terminal repeat. Proc Natl Acad Sci U S A 2001; 98:13525-30. [PMID: 11707592 PMCID: PMC61074 DOI: 10.1073/pnas.241508998] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adeno-associated virus (AAV) is the only known eukaryotic virus capable of targeted integration in human cells. An AAV Rep binding element (RBE) and terminal resolution site (trs) identical to the viral terminal repeats required for AAV DNA replication are located on chromosome (ch) 19. Both ch-19 RBE and trs elements have been shown to be essential for viral targeting to this locus. To characterize the role of the AAV inverted terminal repeat (ITR) cis-acting sequences in targeted integration an AAV trs mutant incapable of supporting viral replication was tested. Wild-type and mutant substrates were assayed for targeted integration after cotransfection in the presence or absence of Rep. Our results demonstrated that, in the presence of Rep78, both ITR substrates targeted to ch-19 with similar frequency. Molecular characterization of the mutant ITR integrants confirmed the presence of the trs mutation in the majority of samples tested. Complementation analysis confirmed that the mutant targeted viral genomes were unable to rescue and replicate. In addition, Rep78 induced extensive rearrangement and amplification of ch-19 sequences independent of wild-type or mutant targeting substrate. These studies demonstrate that Rep-dependent nicking of the viral cis-acting trs sequence is not a prerequisite for site-specific recombination and suggests AAV targeting is mediated by Rep78/68-dependent replication from the ch-19 origin of replication (ori). These studies have significant impact toward the understanding of AAV site-specific recombination and the development of targeting vectors.
Collapse
Affiliation(s)
- S M Young
- Curriculum in Genetics and Molecular Biology, Gene Therapy Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
22
|
Corsini J, Cotmore SF, Tattersall P, Winocour E. The left-end and right-end origins of minute virus of mice DNA differ in their capacity to direct episomal amplification and integration in vivo. Virology 2001; 288:154-63. [PMID: 11543668 DOI: 10.1006/viro.2001.1076] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previously it was shown that a 53-nucleotide viral replication origin, derived from the left-end (3') telomere of minute virus of mice (MVM) DNA, directed integration of infecting MVM genomes into an Epstein-Barr virus (EBV)-based episome in cell culture. Integration depended upon the presence, in the episome, of a functional origin sequence which could be nicked by NS1, the viral initiator protein. Here we extend our studies to the genomic right-end (5') origin and report that three 131- to 135-nucleotide right-end origin sequences failed to target MVM episomal integration even though the same sequences were functional in NS1-driven DNA replication assays in vitro. Additionally, we observed amplification of episomal DNA in response to MVM infection in cell lines harboring episomes which directed integration, but not in cell lines containing episomes which did not direct integration, including those with inserts of the MVM right-end origin.
Collapse
Affiliation(s)
- J Corsini
- Math and Science Department, Chadron State College, Chadron, Nebraska 69337, USA
| | | | | | | |
Collapse
|
23
|
Lamartina S, Ciliberto G, Toniatti C. Selective cleavage of AAVS1 substrates by the adeno-associated virus type 2 rep68 protein is dependent on topological and sequence constraints. J Virol 2000; 74:8831-42. [PMID: 10982325 PMCID: PMC102077 DOI: 10.1128/jvi.74.19.8831-8842.2000] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The adeno-associated virus type 2 (AAV-2) Rep78 and Rep68 proteins are required for replication of the virus as well as its site-specific integration into a unique site, called AAVS1, of human chromosome 19. Rep78 and Rep68 initiate replication by binding to a Rep binding site (RBS) contained in the AAV-2 inverted terminal repeats (ITRs) and then specifically nicking at a nearby site called the terminal resolution site (trs). Similarly, Rep78 and Rep68 are postulated to trigger the integration process by binding and nicking RBS and trs homologues present in AAVS1. However, Rep78 and Rep68 cleave in vitro AAVS1 duplex-linear substrates much less efficiently than hairpinned ITRs. In this study, we show that the AAV-2 Rep68 endonuclease activity is affected by the topology of the substrates in that it efficiently cleaves in vitro in a site- and strand-specific manner the AAVS1 trs only if this sequence is in a supercoiled (SC) conformation. DNA sequence mutagenesis in the context of SC templates allowed us to elucidate for the first time the AAVS1 trs sequence and position requirements for Rep68-mediated cleavage. Interestingly, Rep68 did not cleave SC templates containing RBS from other sites of the human genome. These findings have intriguing implications for AAV-2 site-specific integration in vivo.
Collapse
Affiliation(s)
- S Lamartina
- Department of Gene Therapy, Istituto di Ricerche di Biologia Molecolare, 00040 Pomezia, Rome, Italy
| | | | | |
Collapse
|
24
|
Meneses P, Berns KI, Winocour E. DNA sequence motifs which direct adeno-associated virus site-specific integration in a model system. J Virol 2000; 74:6213-6. [PMID: 10846109 PMCID: PMC112124 DOI: 10.1128/jvi.74.13.6213-6216.2000] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2000] [Accepted: 03/25/2000] [Indexed: 11/20/2022] Open
Abstract
The DNA sequence motifs which direct adeno-associated virus type 2 site-specific integration are being investigated using a shuttle vector, propagated as a stable episome in cultured cell lines, as the target for integration. Previously, we reported that the minimum episomal targeting elements comprise a 16-bp binding motif (Rep binding site [RBS]) for a viral regulatory protein (Rep) separated by a short DNA spacer from a sequence (terminal resolution site [TRS]) that can serve as a substrate for Rep-mediated nicking activity (R. M. Linden, P. Ward, C. Giraud, E. Winocour, and K. I. Berns, Proc. Natl. Acad. Sci. USA 93:11288-11294, 1996; R. M. Linden, E. Winocour, and K. I. Berns, Proc. Natl. Acad. Sci. USA 93:7966-7972, 1996). We now report that episomal integration depends upon both the sequence and the position of the spacer DNA separating the RBS and TRS motifs. The spacer thus constitutes a third element required for site-specific episomal integration.
Collapse
Affiliation(s)
- P Meneses
- Department of Pathology, Harvard Medical School, Boston, MA 02215, USA
| | | | | |
Collapse
|
25
|
Young SM, McCarty DM, Degtyareva N, Samulski RJ. Roles of adeno-associated virus Rep protein and human chromosome 19 in site-specific recombination. J Virol 2000; 74:3953-66. [PMID: 10756007 PMCID: PMC111909 DOI: 10.1128/jvi.74.9.3953-3966.2000] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adeno-associated virus type 2 (AAV) is the only known eucaryotic virus capable of targeted integration in human cells. AAV integrates preferentially into human chromosome (ch) 19q13.3qter. The nonstructural proteins of AAV-2, Rep78 and Rep68, are essential for targeted integration. Rep78 and Rep68 are multifunctional proteins with diverse biochemical activities, including site-specific binding to AAV and ch-19 target sequences, helicase activity, and strand-specific, site-specific endonuclease activities. Both a Rep DNA binding element (RBE) and a nicking site essential for AAV replication present within the viral terminal repeats are also located on ch-19. Recently, identical RBE sequences have been identified at other locations in the human genome. This fact raises numerous questions concerning AAV targeted integration; specifically, how many RBE sequences are in the human genome? How does Rep discriminate between these and the ch-19 RBE sequence? Does Rep interact with all sites and, if so, how is targeted integration within a fixed time frame facilitated? To better characterize the role of Rep in targeted integration, we established a Rep-dependent filter DNA binding assay using a highly purified Rep-68 fusion protein. Electron microscopy (EM) analysis was also performed to determine the characteristics of the Rep-RBE interaction. Our results determined that the Rep affinity for ch-19 is not distinct compared to other RBEs in the human genome when utilizing naked DNA. In fact, a minimum-binding site (GAGYGAGC) efficiently associated with Rep, suggesting that as many as 2 x 10(5) sites may exist. In addition, such sites also exist frequently in nonprimate mammalian genomes, although AAV integrates site specifically into primate genomes. EM analysis demonstrated that only one Rep-DNA complex was formed on ch-19 target DNA. Surprisingly, identically sized complexes were observed on all substrates containing a RBE sequence, but never on DNA lacking an RBE. Rep-DNA complexes involved a multimeric protein structure that spanned ca. 60 bp. Immunoprecipitation of AAV latently infected cells determined that 1,000 to 4,000 copies of Rep78 and Rep68 protein are expressed per cell. Comparison of the Rep association constant with those of established DNA binding proteins indicates that sufficient molecules of Rep are present to interact with all potential RBE sites. Moreover, Rep expression in the absence of AAV cis-acting substrate resulted in Rep-dependent amplification and rearrangement of the target sequence in ch-19. This result suggests that this locus is a hot spot for Rep-dependent recombination. Finally, we engineered mice to carry a single 2.7-kb human ch-19 insertion containing the AAV ch-19 target locus. Using cells derived from these mice, we demonstrated that this sequence was sufficient for site-specific recombination after infection with transducing vectors expressing Rep. This result indicates that any host factors required for targeting are conserved between human and mouse. Furthermore, the human ch-19 cis sequences and chromatin structure required for site-specific recombination are contained within this fragment. Overall, these results indicate that the specificity of targeted recombination to human ch-19 is not dictated by differential Rep affinities for RBE sites. Instead, specificity is likely dictated by human ch-19 sequences that serve as a Rep protein-mediated origin of replication, thus facilitating viral targeting through Rep-Rep interactions and host enzymes, resulting in site-specific recombination. Control of specificity is clearly dictated by the ch-19 sequences, since transfer of these sequences into the mouse genome are sufficient to achieve Rep-dependent site-specific integration.
Collapse
Affiliation(s)
- S M Young
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|