1
|
del Rio D, Beucher B, Lavigne M, Wehbi A, Gonzalez Dopeso-Reyes I, Saggio I, Kremer EJ. CAV-2 Vector Development and Gene Transfer in the Central and Peripheral Nervous Systems. Front Mol Neurosci 2019; 12:71. [PMID: 30983967 PMCID: PMC6449469 DOI: 10.3389/fnmol.2019.00071] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/07/2019] [Indexed: 12/11/2022] Open
Abstract
The options available for genetic modification of cells of the central nervous system (CNS) have greatly increased in the last decade. The current panoply of viral and nonviral vectors provides multifunctional platforms to deliver expression cassettes to many structures and nuclei. These cassettes can replace defective genes, modify a given pathway perturbed by diseases, or express proteins that can be selectively activated by drugs or light to extinguish or excite neurons. This review focuses on the use of canine adenovirus type 2 (CAV-2) vectors for gene transfer to neurons in the brain, spinal cord, and peripheral nervous system. We discuss (1) recent advances in vector production, (2) why CAV-2 vectors preferentially transduce neurons, (3) the mechanism underlying their widespread distribution via retrograde axonal transport, (4) how CAV-2 vectors have been used to address structure/function, and (5) their therapeutic applications.
Collapse
Affiliation(s)
- Danila del Rio
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Bertrand Beucher
- PVM, BioCampus, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Marina Lavigne
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Amani Wehbi
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | | | - Isabella Saggio
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Institute of Structural Biology, School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| |
Collapse
|
2
|
Calcedo R, Chichester JA, Wilson JM. Assessment of Humoral, Innate, and T-Cell Immune Responses to Adeno-Associated Virus Vectors. Hum Gene Ther Methods 2019; 29:86-95. [PMID: 29668327 DOI: 10.1089/hgtb.2018.038] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Adeno-associated virus (AAV)-based gene therapy is being applied to treat a wide array of diseases. Preexisting host immune responses to AAV and immune responses elicited by AAV vector administration remain a problem that needs to be further studied. Here we present a series of protocols to assess immune responses before and after AAV vector administration that are applicable to multiple animal models and phase 1 clinical trials. More specifically, they may be use to evaluate (1) the humoral immune response, through levels of AAV-neutralizing and binding antibodies; (2) the innate immune response, through the acute induction of inflammatory cytokines; and (3) the T-cell immune response, through the activation of transgene- and vector-specific CD8+ and CD4+ T cells.
Collapse
Affiliation(s)
- Roberto Calcedo
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Jessica A Chichester
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Stav-Noraas TE, Edelmann RJ, Poulsen LLC, Sundnes O, Phung D, Küchler AM, Müller F, Kamen AA, Haraldsen G, Kaarbø M, Hol J. Endothelial IL-33 Expression Is Augmented by Adenoviral Activation of the DNA Damage Machinery. THE JOURNAL OF IMMUNOLOGY 2017; 198:3318-3325. [PMID: 28258201 DOI: 10.4049/jimmunol.1600054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/06/2017] [Indexed: 01/16/2023]
Abstract
IL-33, required for viral clearance by cytotoxic T cells, is generally expressed in vascular endothelial cells in healthy human tissues. We discovered that endothelial IL-33 expression was stimulated as a response to adenoviral transduction. This response was dependent on MRE11, a sensor of DNA damage that can also be activated by adenoviral DNA, and on IRF1, a transcriptional regulator of cellular responses to viral invasion and DNA damage. Accordingly, we observed that endothelial cells responded to adenoviral DNA by phosphorylation of ATM and CHK2 and that depletion or inhibition of MRE11, but not depletion of ATM, abrogated IL-33 stimulation. In conclusion, we show that adenoviral transduction stimulates IL-33 expression in endothelial cells in a manner that is dependent on the DNA-binding protein MRE11 and the antiviral factor IRF1 but not on downstream DNA damage response signaling.
Collapse
Affiliation(s)
- Tor Espen Stav-Noraas
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Reidunn J Edelmann
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Lars La Cour Poulsen
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Olav Sundnes
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Danh Phung
- Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Axel M Küchler
- Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Fredrik Müller
- Department of Microbiology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway; and
| | - Amine A Kamen
- Department of Bioengineering, McGill University, Montreal, Quebec H3A OC3, Canada
| | - Guttorm Haraldsen
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway; .,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Mari Kaarbø
- Department of Microbiology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway; and
| | - Johanna Hol
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| |
Collapse
|
4
|
Ahi YS, Mittal SK. Components of Adenovirus Genome Packaging. Front Microbiol 2016; 7:1503. [PMID: 27721809 PMCID: PMC5033970 DOI: 10.3389/fmicb.2016.01503] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022] Open
Abstract
Adenoviruses (AdVs) are icosahedral viruses with double-stranded DNA (dsDNA) genomes. Genome packaging in AdV is thought to be similar to that seen in dsDNA containing icosahedral bacteriophages and herpesviruses. Specific recognition of the AdV genome is mediated by a packaging domain located close to the left end of the viral genome and is mediated by the viral packaging machinery. Our understanding of the role of various components of the viral packaging machinery in AdV genome packaging has greatly advanced in recent years. Characterization of empty capsids assembled in the absence of one or more components involved in packaging, identification of the unique vertex, and demonstration of the role of IVa2, the putative packaging ATPase, in genome packaging have provided compelling evidence that AdVs follow a sequential assembly pathway. This review provides a detailed discussion on the functions of the various viral and cellular factors involved in AdV genome packaging. We conclude by briefly discussing the roles of the empty capsids, assembly intermediates, scaffolding proteins, portal vertex and DNA encapsidating enzymes in AdV assembly and packaging.
Collapse
Affiliation(s)
- Yadvinder S Ahi
- Department of Comparative Pathobiology, Purdue UniversityWest Lafayette, IN, USA; Purdue University Center for Cancer Research, Purdue UniversityWest Lafayette, IN, USA
| | - Suresh K Mittal
- Department of Comparative Pathobiology, Purdue UniversityWest Lafayette, IN, USA; Purdue University Center for Cancer Research, Purdue UniversityWest Lafayette, IN, USA; Purdue Institute for Immunology, Inflammation and Infectious Diseases, Purdue UniversityWest Lafayette, IN, USA
| |
Collapse
|
5
|
Zhou X, Ramke M, Chintakuntlawar AV, Lee JY, Rajaiya J, Chodosh J. Role of MyD88 in adenovirus keratitis. Immunol Cell Biol 2016; 95:108-116. [PMID: 27528076 PMCID: PMC5791738 DOI: 10.1038/icb.2016.73] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 07/25/2016] [Accepted: 08/09/2016] [Indexed: 12/29/2022]
Abstract
Pattern recognition receptors (PRRs) are critical to the early detection and innate immune responses to pathogens. In particular, the TLR system and its associated adaptor proteins play essential roles in early host responses to infection. Epidemic keratoconjunctivitis, caused by the human adenovirus, is a severe ocular surface infection associated with corneal inflammation (stromal keratitis). We previously showed that adenovirus capsid was a key molecular pattern in adenovirus keratitis, with viral DNA playing a lesser role. We have now investigated the role of the adaptor molecule MyD88 in a mouse model of adenovirus keratitis in which there is no viral replication. In MyD88−/− mice infected with human adenovirus type 37, clinical keratitis was markedly reduced, along with infiltration of CD45+ cells, and expression of inflammatory cytokines. Reduction of inflammatory cytokines was also observed in infected primary human corneal fibroblasts pretreated with a MyD88 inhibitory peptide. Keratitis similar to wild type mice was observed in TLR2, TLR9, and IL-1R knockout mice, but was reduced in TLR2/9 double knockout mice, consistent with synergy of TLR2 and TLR9 in the response to adenovirus infection. MyD88 co-immunoprecipitated with Src kinase in mice corneas and in human corneal fibroblasts infected with adenovirus, and MyD88 inhibitory peptide reduced Src phosphorylation, linking MyD88 activation to inflammatory gene expression through a signaling cascade previously shown to be directed by Src. Our findings reveal a critical role for the PRRs TLR2 and 9, and their adaptor protein MyD88, in corneal inflammation upon adenovirus infection.
Collapse
Affiliation(s)
- Xiaohong Zhou
- Howe Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Mirja Ramke
- Howe Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Ashish V Chintakuntlawar
- Howe Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Jeong Yoon Lee
- Howe Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Jaya Rajaiya
- Howe Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - James Chodosh
- Howe Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Piersanti S, Burla R, Licursi V, Brito C, La Torre M, Alves PM, Simao D, Mottini C, Salinas S, Negri R, Tagliafico E, Kremer EJ, Saggio I. Transcriptional Response of Human Neurospheres to Helper-Dependent CAV-2 Vectors Involves the Modulation of DNA Damage Response, Microtubule and Centromere Gene Groups. PLoS One 2015. [PMID: 26207738 PMCID: PMC4514711 DOI: 10.1371/journal.pone.0133607] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Brain gene transfer using viral vectors will likely become a therapeutic option for several disorders. Helper-dependent (HD) canine adenovirus type 2 vectors (CAV-2) are well suited for this goal. These vectors are poorly immunogenic, efficiently transduce neurons, are retrogradely transported to afferent structures in the brain and lead to long-term transgene expression. CAV-2 vectors are being exploited to unravel behavior, cognition, neural networks, axonal transport and therapy for orphan diseases. With the goal of better understanding and characterizing HD-CAV-2 for brain therapy, we analyzed the transcriptomic modulation induced by HD-CAV-2 in human differentiated neurospheres derived from midbrain progenitors. This 3D model system mimics several aspects of the dynamic nature of human brain. We found that differentiated neurospheres are readily transduced by HD-CAV-2 and that transduction generates two main transcriptional responses: a DNA damage response and alteration of centromeric and microtubule probes. Future investigations on the biochemistry of processes highlighted by probe modulations will help defining the implication of HD-CAV-2 and CAR receptor binding in enchaining these functional pathways. We suggest here that the modulation of DNA damage genes is related to viral DNA, while the alteration of centromeric and microtubule probes is possibly enchained by the interaction of the HD-CAV-2 fibre with CAR.
Collapse
Affiliation(s)
- Stefania Piersanti
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Romina Burla
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Valerio Licursi
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Rome, Italy
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780–901, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780–157, Oeiras, Portugal
| | - Mattia La Torre
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780–901, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780–157, Oeiras, Portugal
| | - Daniel Simao
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780–901, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780–157, Oeiras, Portugal
| | - Carla Mottini
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Sara Salinas
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Rodolfo Negri
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Rome, Italy
| | - Enrico Tagliafico
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Isabella Saggio
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Rome, Italy
- Institute of Molecular Biology and Pathology, CNR, Rome, Italy
- * E-mail:
| |
Collapse
|
7
|
The adenovirus genome contributes to the structural stability of the virion. Viruses 2014; 6:3563-83. [PMID: 25254384 PMCID: PMC4189039 DOI: 10.3390/v6093563] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 11/17/2022] Open
Abstract
Adenovirus (Ad) vectors are currently the most commonly used platform for therapeutic gene delivery in human gene therapy clinical trials. Although these vectors are effective, many researchers seek to further improve the safety and efficacy of Ad-based vectors through detailed characterization of basic Ad biology relevant to its function as a vector system. Most Ad vectors are deleted of key, or all, viral protein coding sequences, which functions to not only prevent virus replication but also increase the cloning capacity of the vector for foreign DNA. However, radical modifications to the genome size significantly decreases virion stability, suggesting that the virus genome plays a role in maintaining the physical stability of the Ad virion. Indeed, a similar relationship between genome size and virion stability has been noted for many viruses. This review discusses the impact of the genome size on Ad virion stability and emphasizes the need to consider this aspect of virus biology in Ad-based vector design.
Collapse
|
8
|
Hendrickx R, Stichling N, Koelen J, Kuryk L, Lipiec A, Greber UF. Innate immunity to adenovirus. Hum Gene Ther 2014; 25:265-84. [PMID: 24512150 DOI: 10.1089/hum.2014.001] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human adenoviruses are the most widely used vectors in gene medicine, with applications ranging from oncolytic therapies to vaccinations, but adenovirus vectors are not without side effects. In addition, natural adenoviruses pose severe risks for immunocompromised people, yet infections are usually mild and self-limiting in immunocompetent individuals. Here we describe how adenoviruses are recognized by the host innate defense system during entry and replication in immune and nonimmune cells. Innate defense protects the host and represents a major barrier to using adenoviruses as therapeutic interventions in humans. Innate response against adenoviruses involves intrinsic factors present at constant levels, and innate factors mounted by the host cell upon viral challenge. These factors exert antiviral effects by directly binding to viruses or viral components, or shield the virus, for example, soluble factors, such as blood clotting components, the complement system, preexisting immunoglobulins, or defensins. In addition, Toll-like receptors and lectins in the plasma membrane and endosomes are intrinsic factors against adenoviruses. Important innate factors restricting adenovirus in the cytosol are tripartite motif-containing proteins, nucleotide-binding oligomerization domain-like inflammatory receptors, and DNA sensors triggering interferon, such as DEAD (Asp-Glu-Ala-Asp) box polypeptide 41 and cyclic guanosine monophosphate-adenosine monophosphate synthase. Adenovirus tunes the function of antiviral autophagy, and counters innate defense by virtue of its early proteins E1A, E1B, E3, and E4 and two virus-associated noncoding RNAs VA-I and VA-II. We conclude by discussing strategies to engineer adenovirus vectors with attenuated innate responses and enhanced delivery features.
Collapse
Affiliation(s)
- Rodinde Hendrickx
- 1 Institute of Molecular Life Sciences, University of Zurich , CH-8057 Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
9
|
Piersanti S, Tagliafico E, Saggio I. DNA microarray to analyze adenovirus-host interactions. Methods Mol Biol 2013; 1089:89-104. [PMID: 24132480 DOI: 10.1007/978-1-62703-679-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Defining the molecular toxicity of viral vectors that are or will be in use for clinical trials is a prerequisite for their safe application in humans. DNA chips allow high-throughput evaluation of the profile of transduced cells and have contributed to underlining specific aspects of vector toxicity both in in vitro and in vivo assets. With gene chips we have been able to identify vector-specific properties, such as the cell cycle alteration induced by vector genomic DNA, along with the activation of specific innate immune pathways that can be ascribed to viral particles. We herein describe a detailed protocol for the use of gene chips to dissect the toxicogenomic signature of human and canine helper-dependent adenoviral vectors. We suggest specific procedures suited for the study of these viral vectors, but we also give indications that can be applied to different experimental contexts. In addition, we discuss the in silico elaboration of gene chip raw data which is a crucial step to extrapolate biological information from gene chip studies.
Collapse
Affiliation(s)
- Stefania Piersanti
- Dipartimento di Biologia e Biotecnologie, Sapienza, Università di Roma, Roma, Italy
| | | | | |
Collapse
|
10
|
Piacentino III V, Milano CA, Bolanos M, Schroder J, Messina E, Cockrell AS, Jones E, Krol A, Bursac N, Mao L, Devi GR, Samulski RJ, Bowles DE. X-linked inhibitor of apoptosis protein-mediated attenuation of apoptosis, using a novel cardiac-enhanced adeno-associated viral vector. Hum Gene Ther 2012; 23:635-46. [PMID: 22339372 PMCID: PMC3392616 DOI: 10.1089/hum.2011.186] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 02/09/2012] [Indexed: 12/31/2022] Open
Abstract
Successful amelioration of cardiac dysfunction and heart failure through gene therapy approaches will require a transgene effective at attenuating myocardial injury, and subsequent remodeling, using an efficient and safe delivery vehicle. Our laboratory has established a well-curated, high-quality repository of human myocardial tissues that we use as a discovery engine to identify putative therapeutic transgene targets, as well as to better understand the molecular basis of human heart failure. By using this rare resource we were able to examine age- and sex-matched left ventricular samples from (1) end-stage failing human hearts and (2) nonfailing human hearts and were able to identify the X-linked inhibitor of apoptosis protein (XIAP) as a novel target for treating cardiac dysfunction. We demonstrate that XIAP is diminished in failing human hearts, indicating that this potent inhibitor of apoptosis may be central in protecting the human heart from cellular injury culminating in heart failure. Efforts to ameliorate heart failure through delivery of XIAP compelled the design of a novel adeno-associated viral (AAV) vector, termed SASTG, that achieves highly efficient transduction in mouse heart and in cultured neonatal rat cardiomyocytes. Increased XIAP expression achieved with the SASTG vector inhibits caspase-3/7 activity in neonatal cardiomyocytes after induction of apoptosis through three common cardiac stresses: protein kinase C-γ inhibition, hypoxia, or β-adrenergic receptor agonist. These studies demonstrate the potential benefit of XIAP to correct heart failure after highly efficient delivery to the heart with the rationally designed SASTG AAV vector.
Collapse
Affiliation(s)
- Valentino Piacentino III
- Cardiothoracic Division, Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - Carmelo A. Milano
- Cardiothoracic Division, Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - Michael Bolanos
- Cardiothoracic Division, Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - Jacob Schroder
- Cardiothoracic Division, Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - Emily Messina
- Cardiothoracic Division, Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - Adam S. Cockrell
- Carolina Vaccine Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Edward Jones
- Cardiothoracic Division, Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - Ava Krol
- Department of Biomedical Engineering, Duke University, Durham, NC 27710
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC 27710
| | - Lan Mao
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710
| | - Gayathri R. Devi
- Department of Pathology, Duke University Medical Center, Durham, NC 27710
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Dawn E. Bowles
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
11
|
Baumgart J, Humbert L, Boulais É, Lachaine R, Lebrun JJ, Meunier M. Off-resonance plasmonic enhanced femtosecond laser optoporation and transfection of cancer cells. Biomaterials 2012; 33:2345-50. [DOI: 10.1016/j.biomaterials.2011.11.062] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 11/22/2011] [Indexed: 11/28/2022]
|
12
|
Montesinos MS, Chen Z, Young SM. pUNISHER: a high-level expression cassette for use with recombinant viral vectors for rapid and long term in vivo neuronal expression in the CNS. J Neurophysiol 2011; 106:3230-44. [PMID: 21957229 DOI: 10.1152/jn.00713.2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Fast onset and high-level neurospecific transgene expression in vivo is of importance for many areas in neuroscience, from basic to translational, and can significantly reduce the amount of vector load required to maintain transgene expression in vivo. In this study, we tested various cis elements to optimize transgene expression at transcriptional, posttranscriptional, and posttranslational levels and combined them together to create the high-level neuronal transgene expression cassette pUNISHER. Using a second-generation adenoviral vector system in combination with the pUNISHER cassette, we characterized its rate of onset of detectable expression and levels of expression compared with a neurospecific expression cassette driven by the 470-bp human synapsin promoter in vitro and in vivo. Our results demonstrate in primary neurons that the pUNISHER cassette, in a recombinant adenovirus type 5 background, led to a faster rate of onset of detectable transgene expression and higher level of transgene expression. More importantly, this cassette led to highly correlated neuronal expression in vivo and to stable transgene expression up to 30 days in the auditory brain stem with no toxicity on the characteristics of synaptic transmission and plasticity at the calyx of Held synapse. Thus the pUNISHER cassette is an ideal high-level neuronal expression cassette for use in vivo for neuroscience applications.
Collapse
Affiliation(s)
- Monica S Montesinos
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute, 5353 Parkside Drive MC19-RE, Jupiter, FL 33458, USA
| | | | | |
Collapse
|
13
|
Hirsch ML, Green L, Porteus MH, Samulski RJ. Self-complementary AAV mediates gene targeting and enhances endonuclease delivery for double-strand break repair. Gene Ther 2010; 17:1175-80. [PMID: 20463753 PMCID: PMC3152950 DOI: 10.1038/gt.2010.65] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2010] [Revised: 02/23/2010] [Accepted: 02/23/2010] [Indexed: 11/08/2022]
Abstract
Adeno-associated virus (AAV) mediates gene targeting in humans by providing exogenous DNA for allelic replacement through homologous recombination. In comparison to other methods of DNA delivery or alternative DNA substrates, AAV gene targeting is reported to be very efficient, perhaps due to its single-stranded DNA genome, the inverted terminal repeats (ITRs), and/or the consequence of induced cellular signals on infection or uncoating. These viral attributes were investigated in the presence and absence of an I-Sce endonuclease-induced double-strand break (DSB) within a chromosomal defective reporter in human embryonic kidney cells. Gene correction was evaluated using self-complementary (sc) AAV, which forms a duplexed DNA molecule and results in earlier and robust transgene expression compared with conventional single-strand (ss) AAV genomes. An scAAV repair substrate was modestly enhanced for reporter correction showing no dependency on ssAAV genomes for this process. The AAV ITR sequences were also investigated in a plasmid repair context. No correction was noted in the absence of a DSB, however, a modest inhibitory effect correlated with the increasing presence of ITR sequences. Similarly, signaling cascades stimulated upon recombinant AAV transduction had no effect on plasmid-mediated DSB repair. Noteworthy, was the 20-fold additional enhancement in reporter correction using scAAV vectors, over ss versions, to deliver both the repair substrate and the endonuclease. In this case, homologous recombination repaired the defective reporter in 4% of cells without any selection. This report provides novel insights regarding the recombination substrates used by AAV vectors in promoting homologous recombination and points to the initial steps in vector optimization that could facilitate their use in gene correction of genetic disorders.
Collapse
Affiliation(s)
- ML Hirsch
- UNC Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - L Green
- UNC Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - MH Porteus
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - RJ Samulski
- UNC Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
14
|
Tresilwised N, Pithayanukul P, Mykhaylyk O, Holm PS, Holzmüller R, Anton M, Thalhammer S, Adigüzel D, Döblinger M, Plank C. Boosting Oncolytic Adenovirus Potency with Magnetic Nanoparticles and Magnetic Force. Mol Pharm 2010; 7:1069-89. [DOI: 10.1021/mp100123t] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nittaya Tresilwised
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Pimolpan Pithayanukul
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Olga Mykhaylyk
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Per Sonne Holm
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Regina Holzmüller
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Martina Anton
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Stefan Thalhammer
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Denis Adigüzel
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Markus Döblinger
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Christian Plank
- Institute of Experimental Oncology and Therapy Research, Technische Universität München, Munich 81675, Germany, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand, Helmholtz Zentrum München, AG NanoAnalytics, Neuherberg 85764, Germany, and Department of Chemistry and Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| |
Collapse
|
15
|
Viral capsid is a pathogen-associated molecular pattern in adenovirus keratitis. PLoS Pathog 2010; 6:e1000841. [PMID: 20419141 PMCID: PMC2855317 DOI: 10.1371/journal.ppat.1000841] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Accepted: 03/03/2010] [Indexed: 11/25/2022] Open
Abstract
Human adenovirus (HAdV) infection of the human eye, in particular serotypes 8, 19 and 37, induces the formation of corneal subepithelial leukocytic infiltrates. Using a unique mouse model of adenovirus keratitis, we studied the role of various virus-associated molecular patterns in subsequent innate immune responses of resident corneal cells to HAdV-37 infection. We found that neither viral DNA, viral gene expression, or viral replication was necessary for the development of keratitis. In contrast, empty viral capsid induced keratitis and a chemokine profile similar to intact virus. Transfected viral DNA did not induce leukocyte infiltration despite CCL2 expression similar to levels in virus infected corneas. Mice without toll-like receptor 9 (Tlr9) signaling developed clinical keratitis upon HAdV-37 infection similar to wild type mice, although the absolute numbers of activated monocytes in the cornea were less in Tlr9−/− mice. Virus induced leukocytic infiltrates and chemokine expression in mouse cornea could be blocked by treatment with a peptide containing arginine glycine aspartic acid (RGD). These results demonstrate that adenovirus infection of the cornea induces chemokine expression and subsequent infiltration by leukocytes principally through RGD contact between viral capsid and the host cell, possibly through direct interaction between the viral capsid penton base and host cell integrins. Adenoviruses are nonenveloped DNA viruses that infect mucosal tissues, causing a wide array of diseases. Adenovirus infection of the cornea induces inflammation in the form of multifocal leukocytic infiltrates. Although studied extensively in tissue culture models, how adenoviruses induce inflammation in the living host is not well characterized in the cornea or elsewhere. Using a unique mouse model, we studied the role of viral components in the cornea, to determine which viral part(s) induce an innate immune response. We found that neither viral DNA or viral gene expression was necessary for the development of inflammation. In contrast, viral capsid, the protein coat of the virus, induced inflammation similar to intact virus. Mice lacking the toll-like receptor 9 (Tlr9) molecule, which acts as a pathogen DNA-sensing molecule within the cell, developed clinical inflammation upon adenovirus infection similar to wild type mice. Virus associated inflammation in the mouse cornea could be blocked by treatment with a peptide containing components of the adenoviral capsid. Adenovirus infection of the cornea induces inflammation principally through contact between the viral capsid and the host cell. Our study provides new insights into how the innate immune system in the eye responds to a clinically important viral pathogen.
Collapse
|
16
|
Liao IC, Chen S, Liu JB, Leong KW. Sustained viral gene delivery through core-shell fibers. J Control Release 2009; 139:48-55. [PMID: 19539680 DOI: 10.1016/j.jconrel.2009.06.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 06/08/2009] [Accepted: 06/10/2009] [Indexed: 12/18/2022]
Abstract
Although viral gene transfer is efficient in achieving transgene expression for tissue engineering, drawbacks of virus dissemination, toxicity and transient gene expression due to immune response have hindered its widespread application. Many tissue engineering studies thus opt to genetically engineer cells in vitro prior to their introduction in vivo. However, it would be attractive to obviate the need for in vitro manipulation by transducing the infiltrating progenitor cells in situ. This study introduces the fabrication of a virus-encapsulated electrospun fibrous scaffold to achieve sustained and localized transduction. Adenovirus encoding the gene for green fluorescent protein was efficiently encapsulated into the core of poly(epsilon-caprolactone) fibers through co-axial electrospinning and was subsequently released via a porogen-mediated process. HEK 293 cells seeded on the scaffolds expressed high level of transgene expression over a month, while cells inoculated by scaffold supernatant showed only transient expression for a week. RAW 264.7 cells cultured on the virus-encapsulated fibers produced a lower level of IL-1 beta, TNF-alpha and IFN-alpha, suggesting that the activation of macrophage cells by the viral vector was reduced when encapsulated in the core-shell PCL fibers. In demonstrating sustained and localized cell transduction, this study presents an attractive alternative mode of applying viral gene transfer for regenerative medicine.
Collapse
Affiliation(s)
- I-Chien Liao
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | | | | |
Collapse
|
17
|
Adenovirus induction of IRF3 occurs through a binary trigger targeting Jun N-terminal kinase and TBK1 kinase cascades and type I interferon autocrine signaling. J Virol 2009; 83:4081-91. [PMID: 19211767 DOI: 10.1128/jvi.02591-08] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Pathogen recognition is a critical function of immune sentinel cells. Naïve macrophages or dendritic cells (DCs) undergo pathogen-directed activation and maturation, and as mature antigen-presenting cells (APCs), they contribute essential functions to both innate and adaptive immunity. Using recombinant adenovirus (rAdV) as a model for murine APC activation by DNA viruses, we demonstrate a critical role for stress kinase activation in cell intrinsic and extrinsic antiviral signaling cascades. We propose two viral triggers, viral capsid and viral DNA, are required for APC activation. Endosomal escape and presentation of cytosolic rAdV DNA induces phosphorylation of TANK-binding kinase 1 (TBK1) at serine 172 but does not induce IkappaB kinase epsilon activity as determined by in vitro kinase assays. However, induction of TBK1 alone is not sufficient for interferon regulatory factor 3 (IRF3) phosphorylation. We show that capsid-dependent activation of Jun N-terminal kinase (JNK) stress kinase is a necessary step, licensing TBK1 phosphorylation of IRF3 at Ser 396. A second later phase of JNK activity is required to coordinate phosphorylation of JNK-dependent transcription factors (c-Jun/ATF2) with activated IRF3 in the induction of primary IRF3-responsive transcripts. Finally, we demonstrate that maximal JNK/TBK1/IRF3 stimulation by rAdV depends on an intact type I interferon (IFN) signaling cascade. By requiring multiple viral triggers and type I IFN autocrine regulation, APCs have an inherent fail-safe mechanism against inappropriate activation and maturation.
Collapse
|
18
|
Packaging of viral RNAs in virions of adenoviruses. Virol J 2009; 6:16. [PMID: 19196470 PMCID: PMC2647528 DOI: 10.1186/1743-422x-6-16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Accepted: 02/05/2009] [Indexed: 11/10/2022] Open
Abstract
Earlier, we detected viral RNAs packaged in the porcine adenovirus (PAdV) -3 virions. Using Southern blot analysis, we further demonstrated that the viral RNAs were predominantly packaged in CsCl purified mature capsids (containing viral genome) than empty/intermediate capsids. Some of the packaged viral RNAs appear to be polyadenylated. Real-time reverse transcription (RT)-PCR analysis indicated that the copy number of the tested viral mRNAs encoding E1Bsmall and fiber proteins was less than one per full capsid. Moreover, detection of viral RNA packaged in CsCl purified human adenovirus (HAdV) -5 virions indicates that the viral RNA packaging might be a common phenomenon in members of Adenoviridae family. Further quantitative analysis of viral protein, DNA, and RNA in CsCl purified mature and empty/intermediate capsids of recombinant HAdV-5 expressing enhanced green fluorescent protein indicated that the traceable viral RNA detected in empty/intermediate capsids seems associated with the presence of traceable viral genomic DNA. Taken together, our data suggest that the viral RNAs may be passively packaged in adenovirus virion during encapsidation of viral genomic DNA in cell nuclei. Thus, viral RNA packaging may be a characteristic feature of adenoviral genomic DNA encapsidation.
Collapse
|
19
|
Identification of cellular proteins that interact with the adeno-associated virus rep protein. J Virol 2008; 83:454-69. [PMID: 18971280 DOI: 10.1128/jvi.01939-08] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Adeno-associated virus (AAV) codes for four related nonstructural Rep proteins. AAV both replicates and assembles in the nucleus and requires coinfection with a helper virus, either adenovirus (Ad) or herpesvirus, for a productive infection. Like other more complex DNA viruses, it is believed that AAV interacts or modifies host cell proteins to carry out its infection cycle. To date, relatively little is known about the host proteins that interact with the viral Rep proteins, which are known to be directly involved in DNA replication, control of viral and cellular transcription, splicing, and protein translation. In this study, we used affinity-tagged Rep protein to purify cellular protein complexes that were associated with Rep in cells that had been infected with Ad and AAV. In all, we identified 188 cellular proteins from 16 functional categories, including 14 transcription factors, 6 translation factors, 15 potential splicing proteins, 5 proteins involved in protein degradation, and 13 proteins involved in DNA replication or repair. This dramatically increases the number of potential interactions over the current number of approximately 26. Twelve of the novel proteins found were further tested by coimmunoprecipitation or colocalization using confocal immunomicroscopy. Of these, 10 were confirmed as proteins that formed complexes with Rep, including proteins of the MCM complex (DNA replication), RCN1 (membrane transport), SMC2 (chromatin dynamics), EDD1 (ubiquitin ligase), IRS4 (signal transduction), and FUS (splicing). Computer analysis suggested that 45 and 28 of the 188 proteins could be placed in a pathway of interacting proteins involved in DNA replication and protein synthesis, respectively. Of the proteins involved in DNA replication, all of the previously identified proteins involved in AAV DNA replication were found, except Ad DBP. The only Ad protein found to interact with Rep was the E1b55K protein. In addition, we confirmed that Rep interacts with Ku70/80 helicase. In vitro DNA synthesis assays demonstrated that although Ku helicase activity could substitute for MCM to promote strand displacement synthesis, its presence was not essential. Our study suggests that the interaction of AAV with cellular proteins is much more complex than previously suspected and provides a resource for further studies of the AAV life cycle.
Collapse
|
20
|
McCaffrey AP, Fawcett P, Nakai H, McCaffrey RL, Ehrhardt A, Pham TTT, Pandey K, Xu H, Feuss S, Storm TA, Kay MA. The host response to adenovirus, helper-dependent adenovirus, and adeno-associated virus in mouse liver. Mol Ther 2008; 16:931-41. [PMID: 18388926 DOI: 10.1038/mt.2008.37] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Understanding host responses to viral gene therapy vectors is necessary for the development of safe and efficacious in vivo gene transfer agents. We describe the use of high-density spotted complementary DNA microarrays in monitoring the in vivo host transcriptional responses in mouse liver upon administration of either a "first-generation"adenoviral (Ad) vector, a helper-dependent "gutless" adenoviral (HD) vector, or an adeno-associated viral (AAV) vector containing human factor IX (hFIX) expression cassettes. Since HD and AAV do not contain any viral genes, they allow us to assess the host response to the viral capsid and packaged nonviral DNA in whole animals. Comparison of the host response to Ad and HD helps assess the importance of leaky adenoviral gene expression. While all three vectors induced characteristic temporally sequenced programs of gene expression, the gene expression programs induced by the Ad and HD adenovirus vectors were remarkably similar, including the induction of a prominent type I interferon (IFN)-dependent cluster within 6 hours of administration. In contrast, the AAV-based vector caused far fewer alterations of host-gene expression. Our results indicate that recognition of the Ad capsid or double-stranded DNA (of nonviral origin) in the vector elicits a robust type I IFN response that is, however, not elicited by AAV-derived vector transduction.
Collapse
Affiliation(s)
- Anton P McCaffrey
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa 52242, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Muruve DA, Pétrilli V, Zaiss AK, White LR, Clark SA, Ross PJ, Parks RJ, Tschopp J. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature 2008; 452:103-7. [PMID: 18288107 DOI: 10.1038/nature06664] [Citation(s) in RCA: 713] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2007] [Accepted: 01/04/2008] [Indexed: 02/08/2023]
Abstract
The innate immune system recognizes nucleic acids during infection and tissue damage. Whereas viral RNA is detected by endosomal toll-like receptors (TLR3, TLR7, TLR8) and cytoplasmic RIG-I and MDA5, endosomal TLR9 and cytoplasmic DAI bind DNA, resulting in the activation of nuclear factor-kappaB and interferon regulatory factor transcription factors. However, viruses also trigger pro-inflammatory responses, which remain poorly defined. Here we show that internalized adenoviral DNA induces maturation of pro-interleukin-1beta in macrophages, which is dependent on NALP3 and ASC, components of the innate cytosolic molecular complex termed the inflammasome. Correspondingly, NALP3- and ASC-deficient mice display reduced innate inflammatory responses to adenovirus particles. Inflammasome activation also occurs as a result of transfected cytosolic bacterial, viral and mammalian (host) DNA, but in this case sensing is dependent on ASC but not NALP3. The DNA-sensing pro-inflammatory pathway functions independently of TLRs and interferon regulatory factors. Thus, in addition to viral and bacterial components or danger signals in general, inflammasomes sense potentially dangerous cytoplasmic DNA, strengthening their central role in innate immunity.
Collapse
Affiliation(s)
- Daniel A Muruve
- Department of Medicine, University of Calgary, Alberta T2N 4N1, Canada
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Urosevic M, Fujii K, Calmels B, Laine E, Kobert N, Acres B, Dummer R. Type I IFN innate immune response to adenovirus-mediated IFN-gamma gene transfer contributes to the regression of cutaneous lymphomas. J Clin Invest 2007; 117:2834-46. [PMID: 17823660 PMCID: PMC1964512 DOI: 10.1172/jci32077] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Accepted: 06/13/2007] [Indexed: 12/30/2022] Open
Abstract
The fact that adenoviral vectors activate innate immunity and induce type I IFNs has not been fully appreciated in the context of cancer gene therapy. Type I IFNs influence different aspects of human immune response and are believed to be crucial for efficient tumor rejection. We performed transcriptional profiling to characterize the response of cutaneous lymphomas to intralesional adenovirus-mediated IFN-gamma (Ad-IFN-gamma) gene transfer. Gene expression profiles of skin lesions obtained from 19 cutaneous lymphoma patients before and after treatment with Ad-IFN-gamma revealed a distinct gene signature consisting of IFN-gamma- and numerous IFN-alpha-inducible genes (type II- and type I-inducible genes, respectively). The type I IFN response appears to have been induced by the vector itself, and its complexity, in terms of immune activation, was potentiated by the IFN-gamma gene insert. Intralesional IFN-gamma expression together with the induction of a combined type I/II IFN response to Ad-IFN-gamma gene transfer seem to underlie the objective (measurable) clinical response of the treated lesions. Biological effects of type I IFNs seem to enhance those set in motion by the transgene, in our case IFN-gamma. This combination may prove to be of therapeutic importance in cytokine gene transfer using Ads.
Collapse
Affiliation(s)
- Mirjana Urosevic
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
23
|
Huang BJ, Liu RY, Huang JL, Liang ZH, Gao GF, Wu JX, Huang W. Long-Term toxicity studies in Canine of E10A, an adenoviral vector for human endostatin gene. Hum Gene Ther 2007; 18:207-21. [PMID: 17346097 DOI: 10.1089/hum.2006.149] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
E10A, a recombinant adenovirus type 5 vector carrying the human endostatin gene, may be a promising gene therapy drug in the treatment of solid tumors by antiangiogenesis, but a preclinical safety evaluation of E10A has not yet been performed. With high and low doses equivalent to 30 and 7.5 times the human curative dose, respectively, intramuscular injections of E10A were given once daily, 6 days/week, for 3 months, followed by a 1-month recovery period. As of 4 months, all experimental animals appeared generally healthy: normal behavior and eating habits, no nausea, vomiting, or salivation, no abnormal changes in urination or defecation, and increased body weight with the time of experiment. Urinalysis, hemogram, blood biochemistry, electrocardiogram, macroscopic and microscopic studies of organs and tissues were done before treatment, at month 3 of treatment, and 1 month posttreatment. At all time points, no significant abnormal toxic effects were noted. Preliminary investigation of E10A immunotoxicity in dogs indicated that anti-adenoviral antibodies were generated, in a dose- and time-independent manner, after E10A injection. Our data demonstrated that, long term, high-dose intramuscular administration of recombinant human endostatin-carrying adenovirus (E10A) was not notably toxic and might be safe for clinical therapeutic use, although additional long-term toxicity studies by other administration routes are still necessary.
Collapse
Affiliation(s)
- Bi-Jun Huang
- State Key Laboratory of Oncology in Southern China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | | | | | | | | | | | | |
Collapse
|
24
|
Hensley SE, Amalfitano A. Toll-like Receptors Impact on Safety and Efficacy of Gene Transfer Vectors. Mol Ther 2007; 15:1417-22. [PMID: 17551505 DOI: 10.1038/sj.mt.6300217] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Innate immune responses are triggered when pattern-recognition receptors recognize specific conserved patterns on pathogens. The most extensively studied pattern-recognition receptors are toll-like receptors (TLRs), which are comprised of 11 different receptors, named TLR1-11. TLRs recognize motifs that are found on a wide range of pathogens, and activation of TLRs results in the production of large amounts of type I interferons and several proinflammatory cytokines. These cytokine responses are important in controlling pathogen replication and they also provide an initiation signal for the adaptive immune response. Although numerous manuscripts have reviewed the important role of TLRs in host defense against wild-type viruses, bacteria, and/or their subcomponents, none have focused on how TLRs recognize commonly utilized gene delivery vehicles such as adenovirus and adeno-associated virus (AAV) vectors. In this review, we discuss our understanding of how TLRs are activated by gene transfer vectors.
Collapse
Affiliation(s)
- Scott E Hensley
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | |
Collapse
|
25
|
Martina Y, Avitabile D, Piersanti S, Cherubini G, Saggio I. Different modulation of cellular transcription by adenovirus 5, DeltaE1/E3 adenovirus and helper-dependent vectors. Virus Res 2007; 130:71-84. [PMID: 17601622 DOI: 10.1016/j.virusres.2007.05.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 05/16/2007] [Accepted: 05/21/2007] [Indexed: 01/12/2023]
Abstract
One problem encountered in the use of adenoviral vectors for gene therapy is their toxicity. Although many studies have analyzed this question in vivo, few researches have investigated adenovirus vector effects at the cellular level using a large-scale approach. In particular, no such data are available for helper-dependent adenovirus vectors (HD), which are promising adenovirus vectors for clinical applications since they are devoid of all viral genes and can host large transgene cassettes. The present study used gene chips to examine (Affymetrix HG-U95Av2 interrogating 12,626 unique human transcripts) the effect on liver cells of HD vectors versus that of DeltaE1/E3 adenovirus vector and wild type Adenovirus (Ad5). The effects of the DeltaE1/E3 adenovirus and of HD vectors were comparable, and significantly milder than that of Ad5. Interestingly the expression signatures of DeltaE1/E3 adenovirus and HD vectors were non-overlapping both at the single gene and the pathway level, suggesting specific and different interactions between the host cell and the two gene therapy vectors.
Collapse
Affiliation(s)
- Yuri Martina
- Department of Genetics and Molecular Biology, University La Sapienza, and Fondazione Parco Biomedico S. Raffaele, Rome, Italy
| | | | | | | | | |
Collapse
|
26
|
Hartman ZC, Black EP, Amalfitano A. Adenoviral infection induces a multi-faceted innate cellular immune response that is mediated by the toll-like receptor pathway in A549 cells. Virology 2007; 358:357-72. [PMID: 17027060 DOI: 10.1016/j.virol.2006.08.041] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Revised: 07/18/2006] [Accepted: 08/23/2006] [Indexed: 01/15/2023]
Abstract
Adenovirus vectors are known to induce certain genes and impact innate response networks, but a broad understanding of this process and its mechanisms is currently lacking. For this reason, we chose to investigate and characterize Ad innate immunity using homogeneous, primary MEF cells replete with all the elements of the pathogen-sensing Toll-Like Receptor (TLR) pathway. By using an array-based approach to maximally define transcriptome changes induced upon Ad vector infection, we discovered that Ad infection induces a potent gene and transcription factor network response. This response is characterized by significant changes in the expression of genes involved in focal adhesion, tight junction, and RNA regulation, in addition to TLR pathway and other innate sensing genes. Further investigation using human A549 cells knocked down for various TLR pathway adaptors, revealed significant impacts on the Ad initiation of NF-kB and interferon responses, thus confirming TLR involvement in Ad-mediated immunity across diverse species.
Collapse
Affiliation(s)
- Zachary C Hartman
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
27
|
Cerullo V, Seiler MP, Mane V, Brunetti-Pierri N, Clarke C, Bertin TK, Rodgers JR, Lee B. Toll-like Receptor 9 Triggers an Innate Immune Response to Helper-dependent Adenoviral Vectors. Mol Ther 2007; 15:378-85. [PMID: 17235317 DOI: 10.1038/sj.mt.6300031] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A major obstacle to the clinical application of systemic adenoviral gene replacement therapy is the host innate immune response. Although recent studies have attempted to characterize the cellular basis for this response to systemically administered helper-dependent adenoviral vector (HD-Ad), the underlying molecular components of the innate immune repertoire required to recognize the viral vector have yet to be identified. Here, we show that primary macrophages can sense HD-Ad vectors via the Toll-like Receptor 9 (TLR9) and respond by increasing pro-inflammatory cytokine secretion. Moreover, TLR9 sensing is involved in the rapid innate immune response to HD-Ad in vivo. TLR9 deficiency attenuates the innate immune response to HD-Ad, whereas TLR9 blockade reduces the acute inflammatory response after intravenous injection of the vector. Moreover, HD-Ad upregulates TLR9 gene expression independent of TLR9 function, suggesting that additional innate signaling pathways work cooperatively with TLR9. The identification of the components of the innate immune response to adenovirus will facilitate the development of combinatorial therapy directed at increasing the maximal tolerated dose of systemically delivered adenoviral vectors.
Collapse
Affiliation(s)
- Vincenzo Cerullo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Odom GL, Gregorevic P, Chamberlain JS. Viral-mediated gene therapy for the muscular dystrophies: successes, limitations and recent advances. BIOCHIMICA ET BIOPHYSICA ACTA 2007; 1772:243-62. [PMID: 17064882 PMCID: PMC1894910 DOI: 10.1016/j.bbadis.2006.09.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Revised: 09/16/2006] [Accepted: 09/20/2006] [Indexed: 02/07/2023]
Abstract
Much progress has been made over the past decade elucidating the molecular basis for a variety of muscular dystrophies (MDs). Accordingly, there are examples of mouse models of MD whose disease progression has been halted in large part with the use of viral vector technology. Even so, we must acknowledge significant limitations of present vector systems that must be overcome prior to successful treatment of humans with such approaches. This review will present a variety of viral-mediated therapeutic strategies aimed at counteracting the muscle-wasting symptoms associated with muscular dystrophy. We include viral vector systems used for muscle gene transfer, with a particular emphasis on adeno-associated virus. Findings of several encouraging studies focusing on repair of the mutant dystrophin gene are also included. Lastly, we present a discussion of muscle compensatory therapeutics being considered that include pathways involved in the up-regulation of utrophin, promotion of cellular adhesion, enhancement of muscle mass, and antagonism of the inflammatory response. Considering the complexity of the muscular dystrophies, it appears likely that a multilayered approach tailored to a patient sub-group may be warranted in order to effectively contest the progression of this devastating disease.
Collapse
Affiliation(s)
- Guy L. Odom
- Department of Neurology Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Washington School of Medicine, 1959 NE Pacific Street, Seattle, WA, 98195-7720, USA
| | - Paul Gregorevic
- Department of Neurology Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Washington School of Medicine, 1959 NE Pacific Street, Seattle, WA, 98195-7720, USA
| | - Jeffrey S. Chamberlain
- Department of Neurology Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Washington School of Medicine, 1959 NE Pacific Street, Seattle, WA, 98195-7720, USA
| |
Collapse
|
29
|
Granberg F, Svensson C, Pettersson U, Zhao H. Adenovirus-induced alterations in host cell gene expression prior to the onset of viral gene expression. Virology 2006; 353:1-5. [PMID: 16860366 DOI: 10.1016/j.virol.2006.06.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2006] [Accepted: 06/19/2006] [Indexed: 12/17/2022]
Abstract
In this report, we have studied gene expression profiles in human primary lung fibroblasts (IMR-90) during the very early phase of an adenovirus infection. Eight out of twelve genes with known functions encoded transcription factors linked to two major cellular processes; inhibition of cell growth (ATF3, ATF4, KLF4, KLF6 and ELK3) and immune response (NR4A1 and CEBPB), indicating that the earliest consequences of an adenovirus infection are growth arrest and induction of an immune response. A time course analysis showed that the induction of these immediate-early response genes was transient and suppressed after the onset of the adenovirus early gene expression.
Collapse
Affiliation(s)
- Fredrik Granberg
- Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-751 85 Uppsala, Sweden
| | | | | | | |
Collapse
|
30
|
Wang J, Voutetakis A, Mineshiba F, Illei GG, Dang H, Yeh CK, Baum BJ. Effect of serotype 5 adenoviral and serotype 2 adeno- associated viral vector-mediated gene transfer to salivary glands on the composition of saliva. Hum Gene Ther 2006; 17:455-63. [PMID: 16610933 DOI: 10.1089/hum.2006.17.455] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Key to the development of a useful clinical therapy is the minimization of side effects. Routine safety testing, however, does not provide information about the physiological status of many potentially useful gene transfer target sites. In this study, we evaluated the longitudinal effects of intrasalivary duct delivery of recombinant serotype 5 adenoviral (rAd5; 10(9)-10(10) particles/gland in rats) and recombinant serotype 2 adeno-associated viral (rAAV2; 10(8)-10(9) particles/gland in mice) vectors on salivary composition. Both vectors led to modest, transient alterations in several salivary components that thereafter returned to normal. The changes suggested two initial specific consequences of rAd5 and rAAV2 vector administration: (1) a modest breach of the mucosal barrier in the targeted glands, indicated by elevations in salivary albumin, total protein, and Na+ levels, and (2) an innate host response, indicated by transient elevations in either salivary lactoferrin and IgA levels (rAd5) or mucin (rAAV2). These studies are consistent with the notion that administration of modest doses of rAd5 and rAAV2 vectors to salivary glands for a therapeutic purpose can be accomplished without severe or permanent injury to the target tissue, or compromise to its essential exocrine physiological function.
Collapse
Affiliation(s)
- Jianghua Wang
- Gene Therapy and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, DHHS, Bethesda, MD 20892-1190, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Morozov VN, Evanskey M, Tan YK, Shaffer D, Morozova TY, Bailey C. Ultrafiltration membrane for electrophoretic capturing of pathogens for AFM imaging. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2006; 22:1742-8. [PMID: 16460100 DOI: 10.1021/la051936u] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
This communication describes a simple and rapid technique for electrophoretically assisted capture of phages, viruses, and other pathogens on the surface of an ultrafiltration membrane that can be considered smooth at the nanoscale. The surface was prepared by coating commercial dialysis membrane with a micrometer-thick layer of cross-linked dextran or globular proteins. To ensure strong adherence of the coating, the surface of the dialysis membrane was activated in cold plasma. It was shown that the root-mean-square roughness of the coating was well below 1 nm when the polymer solution used for coating was allowed to slowly dry through a dialysis membrane left in direct contact with mica. Relatively small viral particles (e.g., fd phages 0.7 microm long and only 3.5 nm high in the dry state) are readily visible by AFM following electrophoretic capture from suspensions containing as few as 1 x 10(6) particles/mL onto membranes prepared as described.
Collapse
Affiliation(s)
- Victor N Morozov
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia 20110, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Glasgow JN, Everts M, Curiel DT. Transductional targeting of adenovirus vectors for gene therapy. Cancer Gene Ther 2006; 13:830-44. [PMID: 16439993 PMCID: PMC1781516 DOI: 10.1038/sj.cgt.7700928] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer gene therapy approaches will derive considerable benefit from adenovirus (Ad) vectors capable of self-directed localization to neoplastic disease or immunomodulatory targets in vivo. The ablation of native Ad tropism coupled with active targeting modalities has demonstrated that innate gene delivery efficiency may be retained while circumventing Ad dependence on its primary cellular receptor, the coxsackie and Ad receptor. Herein, we describe advances in Ad targeting that are predicated on a fundamental understanding of vector/cell interplay. Further, we propose strategies by which existing paradigms, such as nanotechnology, may be combined with Ad vectors to form advanced delivery vehicles with multiple functions.
Collapse
Affiliation(s)
- JN Glasgow
- Division of Human Gene Therapy, Departments of Medicine, Pathology and Surgery, Birmingham, AL, USA
| | - M Everts
- Division of Human Gene Therapy, Departments of Medicine, Pathology and Surgery, Birmingham, AL, USA
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - DT Curiel
- Division of Human Gene Therapy, Departments of Medicine, Pathology and Surgery, Birmingham, AL, USA
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
33
|
Altaras NE, Aunins JG, Evans RK, Kamen A, Konz JO, Wolf JJ. Production and formulation of adenovirus vectors. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2005; 99:193-260. [PMID: 16568893 DOI: 10.1007/10_008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Adenovirus vectors have attracted considerable interest over the past decade, with ongoing clinical development programs for applications ranging from replacement therapy for protein deficiencies to cancer therapeutics to prophylactic vaccines. Consequently, considerable product, process, analytical, and formulation development has been undertaken to support these programs. For example, "gutless" vectors have been developed in order to improve gene transfer capacity and durability of expression; new cell lines have been developed to minimize recombination events; production conditions have been optimized to improve volumetric productivities; analytical techniques and scaleable purification processes have advanced towards the goal of purified adenovirus becoming a "well-characterized biological"; and liquid formulations have been developed which maintain virus infectivity at 2-8 degrees C for over 18 months. These and other advances in the production of adenovirus vectors are discussed in detail in this review. In addition, the needs for the next decade are highlighted.
Collapse
Affiliation(s)
- Nedim E Altaras
- Fermentation and Cell Culture, Merck Research Laboratories, West Point, Pennsylvannia 19486-0004, USA
| | | | | | | | | | | |
Collapse
|
34
|
Ino A, Naito Y, Mizuguchi H, Handa N, Hayakawa T, Kobayashi I. A trial of somatic gene targeting in vivo with an adenovirus vector. GENETIC VACCINES AND THERAPY 2005; 3:8. [PMID: 16219108 PMCID: PMC1277836 DOI: 10.1186/1479-0556-3-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Accepted: 10/12/2005] [Indexed: 11/26/2022]
Abstract
Background Gene targeting in vivo provides a potentially powerful method for gene analysis and gene therapy. In order to sensitively detect and accurately measure designed sequence changes, we have used a transgenic mouse system, MutaMouse, which has been developed for detection of mutation in vivo. It carries bacteriophage lambda genome with lacZ+ gene, whose change to lacZ-negative allele is detected after in vitro packaging into bacteriophage particles. We have also demonstrated that gene transfer with a replication-defective adenovirus vector can achieve efficient and accurate gene targeting in vitro. Methods An 8 kb long DNA corresponding to the bacteriophage lambda transgene with one of two lacZ-negative single-base-pair-substitution mutant allele was inserted into a replication-defective adenovirus vector. This recombinant adenovirus was injected to the transgenic mice via tail-vein. Twenty-four hours later, genomic DNA was extracted from the liver tissue and the lambda::lacZ were recovered by in vitro packaging. The lacZ-negative phage was detected as a plaque former on agar with phenyl-beta-D-galactoside. Results The mutant frequency of the lacZ-negative recombinant adenovirus injected mice was at the same level with the control mouse (~1/10000). Our further restriction analysis did not detect any designed recombinant. Conclusion The frequency of gene targeting in the mouse liver by these recombinant adenoviruses was shown to be less than 1/20000 in our assay. However, these results will aid the development of a sensitive, reliable and PCR-independent assay for gene targeting in vivo mediated by virus vectors and other means.
Collapse
Affiliation(s)
- Asami Ino
- Department of Medical Genome Sciences, Graduate School of Frontier Science, University of Tokyo & Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Graduate Program in Biophysics and Biochemistry, Graduate School of Science the University of Tokyo
| | - Yasuhiro Naito
- Department of Medical Genome Sciences, Graduate School of Frontier Science, University of Tokyo & Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Department of Environmental Information, Keio University, 5322 Endo, Fujisawa, Kanagawa 252-8520, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Gene Transfer and Regulation, National Institute of Biomedical Innovation, Asagi 7-6-8, Saito, Ibaraki, Osaka 567-0085, Japan
| | - Naofumi Handa
- Department of Medical Genome Sciences, Graduate School of Frontier Science, University of Tokyo & Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Takao Hayakawa
- Pharmaceuticals and Medical Devices Agency, Shin-Kasumigaseki Bldg. 3-3-2, Kasumigaseki, Chiyoda-ku, Tokyo 100-0013, Japan
| | - Ichizo Kobayashi
- Department of Medical Genome Sciences, Graduate School of Frontier Science, University of Tokyo & Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Graduate Program in Biophysics and Biochemistry, Graduate School of Science the University of Tokyo
| |
Collapse
|
35
|
Scibetta AG, Copier J, Barrett A, Chaplin T, Taylor-Papadimitriou J. Gene expression changes induced by a recombinant E1-/E3- adenovirus type 5 vector in human mammary epithelial cells. Intervirology 2005; 48:350-61. [PMID: 16024939 DOI: 10.1159/000086062] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2004] [Accepted: 12/07/2004] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Adenoviral vectors are used in transferring exogenous genes to a variety of cells and tissue types both in vitro and in vivo. Gene expression changes induced by an E1/E3-defective adenovirus vector have been studied in human mammary epithelial cells by comparing the gene expression profile in infected and uninfected cells. METHODS The human mammary epithelial cell line HB2 was infected with an E1/E3-defective adenovirus type 5 vector. Total RNA was extracted from infected and uninfected cells 24 and 72 h after infection and subjected to microarray analysis using the Affymetrix U133A genomic chip system. Semiquantitative RT-PCR confirmed the regulation of genes observed by microarray analysis. RESULTS The microarray analysis showed 24 and 95 transcripts to be regulated 24 and 72 h after infection, respectively. A relatively high number of genes involved in innate and inflammatory host immune responses, including interleukin-8, interleukin-6, NF-kappaB(2), RELB and fos, were induced. As expected from an E1-defective virus, changes in the expression of genes involved in the G1-S transition and in the activation of cell proliferation were not detected. CONCLUSION Our study provides insight into the host transcriptional response following transduction of an adenoviral vector into mammary epithelial cells.
Collapse
Affiliation(s)
- Angelo G Scibetta
- Cancer Research UK, Breast Cancer Biology Group, Guys Hospital, London, UK.
| | | | | | | | | |
Collapse
|
36
|
Volk AL, Rivera AA, Page GP, Salazar-Gonzalez JF, Nettelbeck DM, Matthews QL, Curiel DT. Employment of microarray analysis to characterize biologic differences associated with tropism-modified adenoviral vectors: utilization of non-native cellular entry pathways. Cancer Gene Ther 2005; 12:162-74. [PMID: 15375380 DOI: 10.1038/sj.cgt.7700776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this study, we have applied high-density oligonucleotide microarray technology to characterize biologic changes associated with adenoviral vector-mediated target cell infection. We infected a human melanoma cell line, M21, with the tropism-modified vectors, Ad5lucRGD and Ad5/3luc1. In addition, we infected the M21 cell line with the Ad5luc1, a vector which primarily exploits the coxsackie and adenovirus receptor, as its primary native receptor. We found significant changes in gene expression of 5492 genes induced by Ad5luc1 infection, 2439 genes induced by Ad5/3luc1 infection, and 1251 genes induced by Ad5lucRGD infection, compared to uninfected cells. Among these changes in gene expression, 783 changes were common to Ad5/3luc1 and Ad5luc1 infections, 266 were common to Ad5lucRGD and Ad5luc1 infections, and 185 changes in gene expression were common to Ad5/3luc1 and Ad5lucRGD infections. Interestingly, 89 changes in gene expression were common to all the three groups, suggesting a commonly affected pathway. This analysis represents a unique application of microarray to study vector-related issues. Furthermore, these studies demonstrate the utility of microarray for characterizing the biologic sequelae of host-vector interaction.
Collapse
Affiliation(s)
- Andrea L Volk
- Division of Human Gene Therapy and The Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Li C, Samulski RJ. Serotype-specific replicating AAV helper constructs increase recombinant AAV type 2 vector production. Virology 2005; 335:10-21. [PMID: 15823602 DOI: 10.1016/j.virol.2005.02.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2004] [Revised: 12/16/2004] [Accepted: 02/07/2005] [Indexed: 11/26/2022]
Abstract
One of the major limitations of the use of adeno-associated virus (AAV) as a tool for gene therapy is the difficulty in providing sufficient quantities of the virus for pre-clinical and clinical trials. Here, we report a novel approach for amplifying AAV helper functions, which mimics the normal replication of wild type (wt) AAV resulting in a high yield of AAV vectors. Cotransfection of replicating but non-packaging AAV helper constructs in the presence of adenovirus (Ad) produces a high level of Rep and Cap proteins. Yield of AAV2/GFP vector obtained from this helper DNA replication system was approximately 20-fold higher than traditional methods. Molecular analysis suggested that virus yield was associated with capsid protein production. The transfection ratio was optimized using these novel helper constructs, resulting in an additional 2-fold increase in vector yield without presence of replication competent AAV (rcAAV). This strategy supports development of AAV packaging systems that retain normal virus replication capability without helper virus encapsidation.
Collapse
Affiliation(s)
- Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27510, USA
| | | |
Collapse
|
38
|
Piersanti S, Martina Y, Cherubini G, Avitabile D, Saggio I. Use of DNA microarrays to monitor host response to virus and virus-derived gene therapy vectors. ACTA ACUST UNITED AC 2005; 4:345-56. [PMID: 15651896 DOI: 10.2165/00129785-200404060-00002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Given the biological complexity of viral infections, the variability of the host response, and the safety concerns related to viral-mediated gene transfer, recent studies have made use of DNA mircoarrays to integrate multi-layered experimental approaches aimed at completely clarifying virus-host interactions. Particular attention has been given to those viruses that are implicated in clinical use and/or in life-threatening diseases. Examples of such use can be divided into three main categories, including: (i) the use of microarrays to study viral expression; (ii) the use of microarrays to analyze the host response to viral infection; and (iii) the use of microarrays to characterize the host response to viral vector-mediated transduction. Significant information on virus- and viral vector-host interactions can be obtained with the microarray approach, including the recognition of master pathways of virally-induced responses, the identification of new target genes for specific viruses, and indications on the molecular toxicity of specific gene transfer vectors currently used for gene therapy trials (in particular, adeno-associated viruses and adenovirus-derived vectors). We predict that the development of accessible repositories containing most of the DNA microarray data on viral infections will certainly help to elucidate the puzzling pictures of different viral infections. This will be crucially important for the correct handling of viral diseases and the intelligent amelioration of viral vectors for gene therapy.
Collapse
Affiliation(s)
- Stefania Piersanti
- Department of Genetics and Molecular Biology, University of Rome La Sapienza, Rome, Italy
| | | | | | | | | |
Collapse
|
39
|
Józkowicz A, Dulak J. Helper-dependent adenoviral vectors in experimental gene therapy. Acta Biochim Pol 2005; 52:589-99. [PMID: 16082408 PMCID: PMC1383728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2005] [Revised: 06/01/2005] [Accepted: 06/30/2005] [Indexed: 05/03/2023]
Abstract
In the majority of potential applications gene therapy will require an effective transfer of a transgene in vivo resulting in high-level and long-term transgene expression, all in the absence of significant toxicity or inflammatory responses. The most efficient vehicles for delivery of foreign genes to the target tissues are modified adenoviruses. Adenoviral vectors of the first generation, despite the high infection efficacy, have an essential drawback: they induce strong immune response, which leads to short term expression of the transgene, and limits their usefulness in clinical trials. In contrast, helper-dependent adenoviral vectors (HdAd) lacking all viral coding sequences display only minimal immunogenicity and negligible side-effects, allowing for long-term transgene expression. Thus, HdAd vehicles have become the carrier of choice for adenoviral vector-mediated experimental gene therapy, effectively used in animal models for delivery of transgenes into the liver, skeletal muscle, myocardium or brain. Strong and long-lasting expression of therapeutic genes has allowed for successful treatment of dyslipidemias, muscular dystrophy, obesity, hemophilia, and diabetes. Additionally, the large cloning capacity of HdAd, up to 37 kb, facilitates the use of physiologically regulated, endogenous promoters, instead of artificial viral promoter sequences. This enables also generation of the single vectors expressing multiple genes, which can be potentially useful for treatment of polygenic diseases. In this review we characterize the basic features of HdAd vectors and describe some of their experimental and potential clinical applications.
Collapse
Affiliation(s)
- Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biotechnology, Jagiellonian University, Kraków, Poland.
| | | |
Collapse
|
40
|
Abstract
Gene therapy is a clinical strategy that may potentially treat an array of genetic and nongenetic diseases, as well as a novel method for drug delivery and vaccination. To these ends, adenovirus vectors are a promising means to deliver specific genes of interest into the patient. A major limitation of the use of adenovirus vectors is the host immune response. Adenovirus vectors induce the innate arm of the immune system that results in inflammation of transduced tissues and efficient clearance of administered vectors. Unlike adaptive immunity, the innate response is mediated by the adenovirus particle and does not require viral transcription. In vivo, the innate immune response involves the induction of cytokines and activation of effector leukocytes that comprise the host response to these agents. A number of interactions with leukocytes and with epithelial and endothelial cells are essential in triggering the host response to adenovirus vectors. Signal transduction via MAP kinases and NF-kappaB-mediated gene transcription are triggered during early virus-cell interactions and are key events in the innate recognition of adenovirus vector transduction. This review aims to describe data examining cellular and molecular mechanisms involved in the adenovirus-mediated innate immune response.
Collapse
Affiliation(s)
- Daniel A Muruve
- Department of Medicine, University of Calgary, Calgary, AB, T2N 4N1 Canada.
| |
Collapse
|
41
|
Stilwell JL, Samulski RJ. Role of viral vectors and virion shells in cellular gene expression. Mol Ther 2004; 9:337-46. [PMID: 15006600 DOI: 10.1016/j.ymthe.2003.11.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2003] [Accepted: 11/14/2003] [Indexed: 11/24/2022] Open
Abstract
The role of the virion shell in viral pathogenesis is relatively unknown yet the use of viral vectors in human gene transfer experiments requires an understanding of these interactions. In this study, we used DNA microarrays to identify genes modulated during pathogenic adenovirus or nonpathogenic adeno-associated virus infections. Responses to wt viruses, recombinant vectors, or empty virion particles were compared. Adeno-associated virus shells induced nearly the full complement of changes elicited by the intact virus. The cellular genes elicited a nonpathogenic response, with antiproliferative genes being induced as a cluster. In contrast, adenovirus and adenovirus empty capsid infection yielded a broader response and subset, respectively, including induction of immune and stress-response genes associated with pathogenic effects. Our studies show that the impact of the viral capsid on cellular gene expression, and potential host toxicity, must be considered independent of the vector genome for safe gene transfer in the clinic.
Collapse
Affiliation(s)
- Jackie L Stilwell
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7352, USA
| | | |
Collapse
|