301
|
Bongiovanni D, Ziegler T, D’Almeida S, Zhang T, Ng JKM, Dietzel S, Hinkel R, Kupatt C. Thymosin β4 attenuates microcirculatory and hemodynamic destabilization in sepsis. Expert Opin Biol Ther 2015; 15 Suppl 1:S203-10. [DOI: 10.1517/14712598.2015.1006193] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
302
|
Rossi M, Cui Z, Nakajima KI, Hu J, Zhu L, Wess J. Virus-Mediated Expression of DREADDs for In Vivo Metabolic Studies. Methods Mol Biol 2015; 1335:205-221. [PMID: 26260603 PMCID: PMC9400095 DOI: 10.1007/978-1-4939-2914-6_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
During the past few years, CNO-sensitive designer G protein-coupled receptors (GPCRs) known as DREADDs (designer receptors exclusively activated by designer drugs) have emerged as powerful new tools for the study of GPCR physiology. In this chapter, we present protocols employing adeno-associated viruses (AAVs) to express a Gq-coupled DREADD (Dq) in two metabolically important cell types, AgRP neurons of the hypothalamus and hepatocytes of the liver. We also provide examples dealing with the metabolic analysis of the Dq mutant mice after administration of CNO in vivo. The approaches described in this chapter can be applied to other members of the DREADD family and, of course, different cell types. It is likely that the use of DREADD technology will identify physiologically important signaling pathways that can be targeted for therapeutic purposes.
Collapse
Affiliation(s)
- Mario Rossi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIH-NIDDK, Bethesda, MD, 20892, USA
| | | | | | | | | | | |
Collapse
|
303
|
Lentz TB, Samulski RJ. Insight into the mechanism of inhibition of adeno-associated virus by the Mre11/Rad50/Nbs1 complex. J Virol 2015; 89:181-94. [PMID: 25320294 PMCID: PMC4301101 DOI: 10.1128/jvi.01990-14] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 10/03/2014] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Adeno-associated virus (AAV) is a dependent virus of the family Parvoviridae. The gene expression and replication of AAV and derived recombinant AAV (rAAV) vectors are severely limited (>10-fold) by the cellular DNA damage-sensing complex made up of Mre11, Rad50, and Nbs1 (MRN). The AAV genome does not encode the means to circumvent this block to productive infection but relies on coinfecting helper virus to do so. Using adenovirus helper proteins E1B55k and E4orf6, which enhance the transduction of AAV via degradation of MRN, we investigated the mechanism through which this DNA damage complex inhibits gene expression from rAAV. We tested the substrate specificity of inhibition and the contribution of different functions of the MRN complex. Our results demonstrate that both single- and double-stranded rAAV vectors are inhibited by MRN, which is in contrast to the predominant model that inhibition is the result of a block to second-strand synthesis. Exploring the contribution of known functions of MRN, we found that inhibition of rAAV does not require downstream DNA damage response factors, including signaling kinases ATM and ATR. The nuclease domain of Mre11 appears to play only a minor role in inhibition, while the DNA binding domain makes a greater contribution. Additionally, mutation of the inverted terminal repeat of the rAAV genome, which has been proposed to be the signal for interaction with MRN, is tolerated by the mechanism of inhibition. These results articulate a model of inhibition of gene expression in which physical interaction is more important than enzymatic activity and several key downstream damage repair factors are dispensable. IMPORTANCE Many viruses modulate the host DNA damage response (DDR) in order to create a cellular environment permissive for infection. The MRN complex is a primary sensor of damage in the cell but also responds to invading viral genomes, often posing a block to infection. AAV is greatly inhibited by MRN and dependent on coinfecting helper virus, such as adenovirus, to remove this factor. Currently, the mechanism through which MRN inhibits AAV and other viruses is poorly understood. Our results reform the predominant model that inhibition of rAAV by MRN is due to limiting second-strand DNA synthesis. Instead, a novel mechanism of inhibition of gene expression independent of a block in rAAV DNA synthesis or downstream damage factors is indicated. These findings have clear implications for understanding this restriction to transduction of AAV and rAAV vectors, which have high therapeutic relevance and likely translate to other viruses that must navigate the DDR.
Collapse
Affiliation(s)
- Thomas B Lentz
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
304
|
Adeno-associated virus inverted terminal repeats stimulate gene editing. Gene Ther 2014; 22:190-5. [PMID: 25503695 DOI: 10.1038/gt.2014.109] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 10/09/2014] [Accepted: 11/03/2014] [Indexed: 12/13/2022]
Abstract
Advancements in genome editing have relied on technologies to specifically damage DNA which, in turn, stimulates DNA repair including homologous recombination (HR). As off-target concerns complicate the therapeutic translation of site-specific DNA endonucleases, an alternative strategy to stimulate gene editing based on fragile DNA was investigated. To do this, an episomal gene-editing reporter was generated by a disruptive insertion of the adeno-associated virus (AAV) inverted terminal repeat (ITR) into the egfp gene. Compared with a non-structured DNA control sequence, the ITR induced DNA damage as evidenced by increased gamma-H2AX and Mre11 foci formation. As local DNA damage stimulates HR, ITR-mediated gene editing was investigated using DNA oligonucleotides as repair substrates. The AAV ITR stimulated gene editing >1000-fold in a replication-independent manner and was not biased by the polarity of the repair oligonucleotide. Analysis of additional human DNA sequences demonstrated stimulation of gene editing to varying degrees. In particular, inverted yet not direct, Alu repeats induced gene editing, suggesting a role for DNA structure in the repair event. Collectively, the results demonstrate that inverted DNA repeats stimulate gene editing via double-strand break repair in an episomal context and allude to efficient gene editing of the human chromosome using fragile DNA sequences.
Collapse
|
305
|
Restoration of visual function by expression of a light-gated mammalian ion channel in retinal ganglion cells or ON-bipolar cells. Proc Natl Acad Sci U S A 2014; 111:E5574-83. [PMID: 25489083 DOI: 10.1073/pnas.1414162111] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Most inherited forms of blindness are caused by mutations that lead to photoreceptor cell death but spare second- and third-order retinal neurons. Expression of the light-gated excitatory mammalian ion channel light-gated ionotropic glutamate receptor (LiGluR) in retinal ganglion cells (RGCs) of the retina degeneration (rd1) mouse model of blindness was previously shown to restore some visual functions when stimulated by UV light. Here, we report restored retinal function in visible light in rodent and canine models of blindness through the use of a second-generation photoswitch for LiGluR, maleimide-azobenzene-glutamate 0 with peak efficiency at 460 nm (MAG0(460)). In the blind rd1 mouse, multielectrode array recordings of retinal explants revealed robust and uniform light-evoked firing when LiGluR-MAG0(460) was targeted to RGCs and robust but diverse activity patterns in RGCs when LiGluR-MAG0(460) was targeted to ON-bipolar cells (ON-BCs). LiGluR-MAG0(460) in either RGCs or ON-BCs of the rd1 mouse reinstated innate light-avoidance behavior and enabled mice to distinguish between different temporal patterns of light in an associative learning task. In the rod-cone dystrophy dog model of blindness, LiGluR-MAG0(460) in RGCs restored robust light responses to retinal explants and intravitreal delivery of LiGluR and MAG0(460) was well tolerated in vivo. The results in both large and small animal models of photoreceptor degeneration provide a path to clinical translation.
Collapse
|
306
|
Xiao X, Guo P, Prasadan K, Shiota C, Peirish L, Fischbach S, Song Z, Gaffar I, Wiersch J, El-Gohary Y, Husain SZ, Gittes GK. Pancreatic cell tracing, lineage tagging and targeted genetic manipulations in multiple cell types using pancreatic ductal infusion of adeno-associated viral vectors and/or cell-tagging dyes. Nat Protoc 2014; 9:2719-2724. [PMID: 25356582 PMCID: PMC4734891 DOI: 10.1038/nprot.2014.183] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Genetic manipulations, with or without lineage tracing for specific pancreatic cell types, are very powerful tools for studying diabetes, pancreatitis and pancreatic cancer. Nevertheless, the use of Cre/loxP systems to conditionally activate or inactivate the expression of genes in a cell type- and/or temporal-specific manner is not applicable to cell tracing and/or gene manipulations in more than one lineage at a time. Here we report a technique that allows efficient delivery of dyes for cell tagging into the mouse pancreas through the duct system, and that also delivers viruses carrying transgenes or siRNA under a specific promoter. When this technique is applied in genetically modified mice, it enables the investigator to perform either double lineage tracing or cell lineage tracing combined with gene manipulation in a second lineage. The technique requires <40 min.
Collapse
Affiliation(s)
- Xiangwei Xiao
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ping Guo
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Krishna Prasadan
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chiyo Shiota
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lauren Peirish
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shane Fischbach
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Zewen Song
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Iljana Gaffar
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - John Wiersch
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yousef El-Gohary
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sohail Z Husain
- Division of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - George K Gittes
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
307
|
Byrne LC, Dalkara D, Luna G, Fisher SK, Clérin E, Sahel JA, Léveillard T, Flannery JG. Viral-mediated RdCVF and RdCVFL expression protects cone and rod photoreceptors in retinal degeneration. J Clin Invest 2014; 125:105-16. [PMID: 25415434 DOI: 10.1172/jci65654] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 10/23/2014] [Indexed: 12/21/2022] Open
Abstract
Alternative splicing of nucleoredoxin-like 1 (Nxnl1) results in 2 isoforms of the rod-derived cone viability factor. The truncated form (RdCVF) is a thioredoxin-like protein secreted by rods that promotes cone survival, while the full-length isoform (RdCVFL), which contains a thioredoxin fold, is involved in oxidative signaling and protection against hyperoxia. Here, we evaluated the effects of these different isoforms in 2 murine models of rod-cone dystrophy. We used adeno-associated virus (AAV) to express these isoforms in mice and found that both systemic and intravitreal injection of engineered AAV vectors resulted in RdCVF and RdCVFL expression in the eye. Systemic delivery of AAV92YF vectors in neonates resulted in earlier onset of RdCVF and RdCVFL expression compared with that observed with intraocular injection using the same vectors at P14. We also evaluated the efficacy of intravitreal injection using a recently developed photoreceptor-transducing AAV variant (7m8) at P14. Systemic administration of AAV92YF-RdCVF improved cone function and delayed cone loss, while AAV92YF-RdCVFL increased rhodopsin mRNA and reduced oxidative stress by-products. Intravitreal 7m8-RdCVF slowed the rate of cone cell death and increased the amplitude of the photopic electroretinogram. Together, these results indicate different functions for Nxnl1 isoforms in the retina and suggest that RdCVF gene therapy has potential for treating retinal degenerative disease.
Collapse
|
308
|
Weinberg MS, Nicolson S, Bhatt AP, McLendon M, Li C, Samulski RJ. Recombinant adeno-associated virus utilizes cell-specific infectious entry mechanisms. J Virol 2014; 88:12472-84. [PMID: 25142580 PMCID: PMC4248914 DOI: 10.1128/jvi.01971-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 08/07/2014] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Understanding the entry and trafficking mechanism(s) of recombinant adeno-associated virus (rAAV) into host cells can lead to evolution in capsid and vector design and delivery methods, resulting in enhanced transduction and therapeutic gene expression. Variability of findings regarding the early entry pathway of rAAV supports the possibility that rAAV, like other viruses, can utilize more than one infectious entry pathway. We tested whether inhibition of macropinocytosis impacted rAAV transduction of HeLa cells compared to hepatocellular carcinoma cell lines. We found that macropinocytosis inhibitor cytochalasin D blocked rAAV transduction of HeLa cells (>2-fold) but enhanced (10-fold) transduction in HepG2 and Huh7 lines. Similar results were obtained with another macropinocytosis inhibitor, 5-(N-ethyl-N-isopropyl) amiloride (EIPA). The augmented transduction was due to neither viral binding nor promoter activity, affected multiple rAAV serotypes (rAAV2, rAAV2-R585E, and rAAV8), and influenced single-stranded and self-complementary virions to comparable extents. Follow-up studies using CDC42 inhibitor ML141 and p21-activated kinase 1 (PAK1) siRNA knockdown also resulted in enhanced HepG2 transduction. Microscopy revealed that macropinocytosis inhibition correlated with expedited nuclear entry of the rAAV virions into HepG2 cells. Enhancement of hepatocellular rAAV transduction extended to the mouse liver in vivo (4-fold enhancement) but inversely blocked heart tissue transduction (13-fold). This evidence of host cell-specific rAAV entry pathways confers a potent means for controlling and enhancing vector delivery and could help unify the divergent accounts of rAAV cellular entry mechanisms. IMPORTANCE There is a recognized need for improved rAAV vector targeting strategies that result in delivery of fewer total particles, averting untoward toxicity and/or an immune response against the vector. A critical step in rAAV transduction is entry and early trafficking through the host cellular machinery, the mechanisms of which are under continued study. However, should the early entry and trafficking mechanisms of rAAV differ across virus serotype or be dependent on host cell environment, this could expand our ability to target particular cells and tissue for selective transduction. Thus, the observation that inhibiting macropinocytosis leads to cell-specific enhancement or inhibition of rAAV transduction that extends to the organismic level exposes a new means of modulating vector targeting.
Collapse
Affiliation(s)
- Marc S Weinberg
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Sarah Nicolson
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Aadra P Bhatt
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Michael McLendon
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
309
|
Szikra T, Trenholm S, Drinnenberg A, Jüttner J, Raics Z, Farrow K, Biel M, Awatramani G, Clark DA, Sahel JA, da Silveira RA, Roska B. Rods in daylight act as relay cells for cone-driven horizontal cell-mediated surround inhibition. Nat Neurosci 2014; 17:1728-35. [PMID: 25344628 DOI: 10.1038/nn.3852] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 09/29/2014] [Indexed: 12/18/2022]
Abstract
Vertebrate vision relies on two types of photoreceptors, rods and cones, which signal increments in light intensity with graded hyperpolarizations. Rods operate in the lower range of light intensities while cones operate at brighter intensities. The receptive fields of both photoreceptors exhibit antagonistic center-surround organization. Here we show that at bright light levels, mouse rods act as relay cells for cone-driven horizontal cell-mediated surround inhibition. In response to large, bright stimuli that activate their surrounds, rods depolarize. Rod depolarization increases with stimulus size, and its action spectrum matches that of cones. Rod responses at high light levels are abolished in mice with nonfunctional cones and when horizontal cells are reversibly inactivated. Rod depolarization is conveyed to the inner retina via postsynaptic circuit elements, namely the rod bipolar cells. Our results show that the retinal circuitry repurposes rods, when they are not directly sensing light, to relay cone-driven surround inhibition.
Collapse
Affiliation(s)
- Tamas Szikra
- Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Stuart Trenholm
- 1] Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland. [2] Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| | - Antonia Drinnenberg
- 1] Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland. [2] University of Basel, Basel, Switzerland
| | - Josephine Jüttner
- Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Zoltan Raics
- Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Karl Farrow
- Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Martin Biel
- Department of Pharmacy-Center for Drug Research, Center for Integrated Protein Science Munich, Ludwig-Maximilians University, Munich, Germany
| | - Gautam Awatramani
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| | - Damon A Clark
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - José-Alain Sahel
- 1] Université Pierre et Marie Curie-Sorbonne Universités, Institut de la Vision, Paris, France. [2] Institut national de la santé et de la recherche médicale, Institut de la Vision, Paris, France. [3] Centre national de la recherche scientifique, Institut de la Vision, Paris, France. [4] Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Département Hospitalo-Universitaire ViewMaintain, Paris, France. [5] Fondation Ophtalmologique Adolphe de Rothschild, Paris, France
| | - Rava Azeredo da Silveira
- 1] Department of Physics, École Normale Supérieure, Paris, France. [2] Laboratoire de Physique Statistique, Centre National de la Recherche Scientifique, Université Pierre et Marie Curie, Université Denis Diderot, Paris, France
| | - Botond Roska
- Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| |
Collapse
|
310
|
Yagishita S, Hayashi-Takagi A, Ellis-Davies GCR, Urakubo H, Ishii S, Kasai H. A critical time window for dopamine actions on the structural plasticity of dendritic spines. Science 2014; 345:1616-20. [PMID: 25258080 PMCID: PMC4225776 DOI: 10.1126/science.1255514] [Citation(s) in RCA: 401] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Animal behaviors are reinforced by subsequent rewards following within a narrow time window. Such reward signals are primarily coded by dopamine, which modulates the synaptic connections of medium spiny neurons in the striatum. The mechanisms of the narrow timing detection, however, remain unknown. Here, we optically stimulated dopaminergic and glutamatergic inputs separately and found that dopamine promoted spine enlargement only during a narrow time window (0.3 to 2 seconds) after the glutamatergic inputs. The temporal contingency was detected by rapid regulation of adenosine 3',5'-cyclic monophosphate in thin distal dendrites, in which protein-kinase A was activated only within the time window because of a high phosphodiesterase activity. Thus, we describe a molecular basis of reinforcement plasticity at the level of single dendritic spines.
Collapse
Affiliation(s)
- Sho Yagishita
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan. Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Akiko Hayashi-Takagi
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan. Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan. Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | | | - Hidetoshi Urakubo
- Integrated Systems Biology Laboratory, Department of Systems Science, Graduate School of Informatics, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shin Ishii
- Integrated Systems Biology Laboratory, Department of Systems Science, Graduate School of Informatics, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Haruo Kasai
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan. Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
311
|
Murlidharan G, Samulski RJ, Asokan A. Biology of adeno-associated viral vectors in the central nervous system. Front Mol Neurosci 2014; 7:76. [PMID: 25285067 PMCID: PMC4168676 DOI: 10.3389/fnmol.2014.00076] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 09/04/2014] [Indexed: 01/11/2023] Open
Abstract
Gene therapy is a promising approach for treating a spectrum of neurological and neurodegenerative disorders by delivering corrective genes to the central nervous system (CNS). In particular, adeno-associated viruses (AAVs) have emerged as promising tools for clinical gene transfer in a broad range of genetic disorders with neurological manifestations. In the current review, we have attempted to bridge our understanding of the biology of different AAV strains with their transduction profiles, cellular tropisms, and transport mechanisms within the CNS. Continued efforts to dissect AAV-host interactions within the brain are likely to aid in the development of improved vectors for CNS-directed gene transfer applications in the clinic.
Collapse
Affiliation(s)
- Giridhar Murlidharan
- Curriculum in Genetics and Molecular Biology, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA ; Gene Therapy Center, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| | - Richard J Samulski
- Gene Therapy Center, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA ; Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill NC, USA
| | - Aravind Asokan
- Gene Therapy Center, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA ; Department of Genetics and Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| |
Collapse
|
312
|
Hu L, Lan W, Guo H, Chai GD, Huang K, Zhang L, Huang Y, Chen XF, Zhang L, Song NN, Chen L, Lang B, Wang Y, Wang QX, Zhang JB, McCaig C, Xu L, Ding YQ. A mouse line for inducible and reversible silencing of specific neurons. Mol Brain 2014; 7:68. [PMID: 25231486 PMCID: PMC4177062 DOI: 10.1186/s13041-014-0068-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 08/31/2014] [Indexed: 11/13/2022] Open
Abstract
Background Genetic methods for inducibly and reversibly inhibiting neuronal activity of specific neurons are critical for exploring the functions of neuronal circuits. The engineered human glycine receptor, called ivermectin (IVM)-gated silencing receptor (IVMR), has been shown to possess this ability in vitro. Results Here we generated a mouse line, in which the IVMR coding sequence was inserted into the ROSA26 locus downstream of a loxP-flanked STOP cassette. Specific Cre-mediated IVMR expression was revealed by mis-expression of Cre in the striatum and by crossing with several Cre lines. Behavioral alteration was observed in Rosa26-IVMR mice with unilateral striatal Cre expression after systemic administration of IVM, and it could be re-initiated when IVM was applied again. A dramatic reduction in neuron firing was recorded in IVM-treated free moving Rosa26-IVMR;Emx1-Cre mice, and neuronal excitability was reduced within minutes as shown by recording in brain slice. Conclusion This Rosa26-IVMR mouse line provides a powerful tool for exploring selective circuit functions in freely behaving mice. Electronic supplementary material The online version of this article (doi:10.1186/s13041-014-0068-8) contains supplementary material, which is available to authorized users.
Collapse
|
313
|
Feng L, Kwon O, Lee B, Oh WC, Kim J. Using mammalian GFP reconstitution across synaptic partners (mGRASP) to map synaptic connectivity in the mouse brain. Nat Protoc 2014; 9:2425-37. [PMID: 25232938 DOI: 10.1038/nprot.2014.166] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Many types of questions in neuroscience require the detection and mapping of synapses in the complex mammalian brain. A tool, mammalian GFP reconstitution across synaptic partners (mGRASP), offers a relatively easy, quick and economical approach to this technically challenging task. Here we describe in step-by-step detail the protocols for virus production, gene delivery, brain specimen preparation, fluorescence imaging and image analysis, calibrated substantially and specifically to make mGRASP-assisted circuit mapping (mGRASPing) practical in the mouse brain. The protocol includes troubleshooting suggestions and solutions to common problems. The mGRASP method is suitable for mapping mammalian synaptic connectivity at multiple scales: microscale for synapse-by-synapse or neuron-by-neuron analysis, and mesoscale for revealing local and long-range circuits. The entire protocol takes 5-6 weeks, including time for incubation and virus expression.
Collapse
Affiliation(s)
- Linqing Feng
- 1] Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Korea. [2]
| | - Osung Kwon
- 1] Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Korea. [2] Neuroscience program, University of Science and Technology, Daejeon, Korea. [3]
| | - Bokyoung Lee
- 1] Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Korea. [2] Neuroscience program, University of Science and Technology, Daejeon, Korea
| | - Won Chan Oh
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Jinhyun Kim
- 1] Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Korea. [2] Neuroscience program, University of Science and Technology, Daejeon, Korea
| |
Collapse
|
314
|
Byrne LC, Lin YJ, Lee T, Schaffer DV, Flannery JG. The expression pattern of systemically injected AAV9 in the developing mouse retina is determined by age. Mol Ther 2014; 23:290-6. [PMID: 25224467 DOI: 10.1038/mt.2014.181] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/09/2014] [Indexed: 02/03/2023] Open
Abstract
Systemic delivery of AAV9 offers the potential for widespread and efficient gene delivery to the retina, and may thus be a useful approach for treatment of disease where intraocular injections are not possible, for syndromes affecting multiple organs, or where early intervention is required. The expression resulting from intravenous injection of AAV9 is more efficient in neonates than adults, and here we characterize the effect of age on retinal transduction of AAV9 in the mouse retina. We find that the pattern of expression in neonatal mice is correlated to the development of the retinal vasculature, and that the area of the retinal transduction as well as the cell types infected vary depending on the age at injection. Furthermore, we demonstrate that sequential injections of AAV9 vectors carrying two different transgenes infect adjacent areas of the retina, providing a larger area of coverage. Lastly, we show that the retina's endogenous spatiotemporal expression pattern of Mfsd2a, a protein associated with the maturation of a functional blood-brain barrier, coincides with suppression of retinal transduction by intravenously-delivered AAV9, suggesting that AAV9 crosses the blood-retina barrier through transcytosis.
Collapse
Affiliation(s)
- Leah C Byrne
- 1] Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, USA [2] Department of Chemical Engineering and Department of Bioengineering, University of California, Berkeley, California, USA
| | - Yvonne J Lin
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, USA
| | - Trevor Lee
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, USA
| | - David V Schaffer
- 1] Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, USA [2] Department of Chemical Engineering and Department of Bioengineering, University of California, Berkeley, California, USA
| | - John G Flannery
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
315
|
Abstract
The motor function of the spinal cord requires the computation of the local neuronal circuits within the same segments as well as the long-range coordination of different spinal levels. Implicated players in this process are the propriospinal neurons (PPNs) that project their axons across different levels of the spinal cord. However, their cellular, molecular, and functional properties remain unknown. Here we use a recombinant rabies virus-based method to label a specific type of long-projecting premotor PPNs in the mouse upper spinal cord that are monosynaptically connected to the motor neurons in the lumbar spinal cord. With a whole spinal cord imaging method, we find that these neurons are distributed along the entire length of the upper spinal cord with more in the lower thoracic levels. Among them, a subset of thoracic PPNs receive substantial numbers of sensory inputs, suggesting a function in coordinating the activity of trunk and hindlimb muscles. Although many PPNs in the cervical and thoracic spinal cord receive the synaptic inputs from corticospinal tract or serotonergic axons, limited bouton numbers suggested that these supraspinal inputs might not be major regulators of the PPNs in intact animals. Molecularly, these PPNs appear to be distinct from other known premotor interneurons, but some are derived from Chx10+ lineages. This study provides an anatomical basis for further exploring different functions of PPNs.
Collapse
|
316
|
Wu Q, He S, Wei X, Shao B, Luo S, Guo F, Zhang H, Wang Y, Gong C, Yang L. Synergistic Antitumor Effect of Recombinant Adeno-Associated Virus-Mediated Pigment Epithelium-Derived Factor with Hyperthermia on Solid Tumor. Hum Gene Ther 2014; 25:811-23. [PMID: 25003563 DOI: 10.1089/hum.2013.150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Qinjie Wu
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Shasha He
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Xiawei Wei
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Bin Shao
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Shuntao Luo
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Fuchun Guo
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Hailong Zhang
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Yongsheng Wang
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Changyang Gong
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Li Yang
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| |
Collapse
|
317
|
Adamantidis AR, Zhang F, de Lecea L, Deisseroth K. Optogenetics: opsins and optical interfaces in neuroscience. Cold Spring Harb Protoc 2014; 2014:815-22. [PMID: 25086025 DOI: 10.1101/pdb.top083329] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Optogenetics is defined as the integration of optics and genetics to control well-defined events within specified cells of living tissue. In this introduction, we focus on the basic techniques necessary for employing microbial opsins as optogenetic tools in mammalian brains. We provide a guide for the fundamentals of optogenetic application-selecting an opsin, implementing expression of opsins based on the neuroscientific experimental requirements, and adapting the corresponding optical hardware for delivery of light into mammalian brains.
Collapse
|
318
|
Chuong AS, Miri ML, Busskamp V, Matthews GAC, Acker LC, Sørensen AT, Young A, Klapoetke NC, Henninger MA, Kodandaramaiah SB, Ogawa M, Ramanlal SB, Bandler RC, Allen BD, Forest CR, Chow BY, Han X, Lin Y, Tye KM, Roska B, Cardin JA, Boyden ES. Noninvasive optical inhibition with a red-shifted microbial rhodopsin. Nat Neurosci 2014; 17:1123-9. [PMID: 24997763 PMCID: PMC4184214 DOI: 10.1038/nn.3752] [Citation(s) in RCA: 383] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/01/2014] [Indexed: 12/11/2022]
Abstract
Optogenetic inhibition of the electrical activity of neurons enables the causal assessment of their contributions to brain functions. Red light penetrates deeper into tissue than other visible wavelengths. We present a red-shifted cruxhalorhodopsin, Jaws, derived from Haloarcula (Halobacterium) salinarum (strain Shark) and engineered to result in red light-induced photocurrents three times those of earlier silencers. Jaws exhibits robust inhibition of sensory-evoked neural activity in the cortex and results in strong light responses when used in retinas of retinitis pigmentosa model mice. We also demonstrate that Jaws can noninvasively mediate transcranial optical inhibition of neurons deep in the brains of awake mice. The noninvasive optogenetic inhibition opened up by Jaws enables a variety of important neuroscience experiments and offers a powerful general-use chloride pump for basic and applied neuroscience.
Collapse
Affiliation(s)
- Amy S Chuong
- 1] Media Lab, Department of Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2] McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [3] Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mitra L Miri
- 1] Department of Neurobiology, Yale School of Medicine, Yale University, New Haven, Connecticut, USA. [2]
| | - Volker Busskamp
- 1] Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland. [2] Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA. [3]
| | - Gillian A C Matthews
- 1] Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2]
| | - Leah C Acker
- 1] Media Lab, Department of Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2] McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [3] Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [4]
| | - Andreas T Sørensen
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Andrew Young
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Nathan C Klapoetke
- 1] Media Lab, Department of Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2] McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [3] Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mike A Henninger
- 1] Media Lab, Department of Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2] McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [3] Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Suhasa B Kodandaramaiah
- 1] Media Lab, Department of Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2] McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [3] Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [4] George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Masaaki Ogawa
- 1] Media Lab, Department of Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2] McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [3] Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Shreshtha B Ramanlal
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - Rachel C Bandler
- Media Lab, Department of Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Brian D Allen
- Media Lab, Department of Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Craig R Forest
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Brian Y Chow
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - Yingxi Lin
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Kay M Tye
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Botond Roska
- Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Jessica A Cardin
- 1] Department of Neurobiology, Yale School of Medicine, Yale University, New Haven, Connecticut, USA. [2] Kavli Institute for Neuroscience, Yale University, New Haven, Connecticut, USA
| | - Edward S Boyden
- 1] Media Lab, Department of Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2] McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [3] Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
319
|
Ishikawa K, Fish KM, Tilemann L, Rapti K, Aguero J, Santos-Gallego CG, Lee A, Karakikes I, Xie C, Akar FG, Shimada YJ, Gwathmey JK, Asokan A, McPhee S, Samulski J, Samulski RJ, Sigg DC, Weber T, Kranias EG, Hajjar RJ. Cardiac I-1c overexpression with reengineered AAV improves cardiac function in swine ischemic heart failure. Mol Ther 2014; 22:2038-2045. [PMID: 25023328 DOI: 10.1038/mt.2014.127] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 07/03/2014] [Indexed: 02/07/2023] Open
Abstract
Cardiac gene therapy has emerged as a promising option to treat advanced heart failure (HF). Advances in molecular biology and gene targeting approaches are offering further novel options for genetic manipulation of the cardiovascular system. The aim of this study was to improve cardiac function in chronic HF by overexpressing constitutively active inhibitor-1 (I-1c) using a novel cardiotropic vector generated by capsid reengineering of adeno-associated virus (BNP116). One month after a large anterior myocardial infarction, 20 Yorkshire pigs randomly received intracoronary injection of either high-dose BNP116.I-1c (1.0 × 10(13) vector genomes (vg), n = 7), low-dose BNP116.I-1c (3.0 × 10(12) vg, n = 7), or saline (n = 6). Compared to baseline, mean left ventricular ejection fraction increased by 5.7% in the high-dose group, and by 5.2% in the low-dose group, whereas it decreased by 7% in the saline group. Additionally, preload-recruitable stroke work obtained from pressure-volume analysis demonstrated significantly higher cardiac performance in the high-dose group. Likewise, other hemodynamic parameters, including stroke volume and contractility index indicated improved cardiac function after the I-1c gene transfer. Furthermore, BNP116 showed a favorable gene expression pattern for targeting the heart. In summary, I-1c overexpression using BNP116 improves cardiac function in a clinically relevant model of ischemic HF.
Collapse
Affiliation(s)
- Kiyotake Ishikawa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kenneth M Fish
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lisa Tilemann
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kleopatra Rapti
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jaume Aguero
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carlos G Santos-Gallego
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ahyoung Lee
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ioannis Karakikes
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chaoqin Xie
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fadi G Akar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yuichi J Shimada
- Department of Medicine, Beth Israel Medical Center, University Hospital and Manhattan Campus for the Albert Einstein College of Medicine, New York, New York, USA
| | | | - Aravind Asokan
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | | | | - Daniel C Sigg
- Department of Integrative Biology and Physiology, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Thomas Weber
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
320
|
Zacchigna S, Zentilin L, Giacca M. Adeno-associated virus vectors as therapeutic and investigational tools in the cardiovascular system. Circ Res 2014; 114:1827-46. [PMID: 24855205 DOI: 10.1161/circresaha.114.302331] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The use of vectors based on the small parvovirus adeno-associated virus has gained significant momentum during the past decade. Their high efficiency of transduction of postmitotic tissues in vivo, such as heart, brain, and retina, renders these vectors extremely attractive for several gene therapy applications affecting these organs. Besides functional correction of different monogenic diseases, the possibility to drive efficient and persistent transgene expression in the heart offers the possibility to develop innovative therapies for prevalent conditions, such as ischemic cardiomyopathy and heart failure. Therapeutic genes are not only restricted to protein-coding complementary DNAs but also include short hairpin RNAs and microRNA genes, thus broadening the spectrum of possible applications. In addition, several spontaneous or engineered variants in the virus capsid have recently improved vector efficiency and expanded their tropism. Apart from their therapeutic potential, adeno-associated virus vectors also represent outstanding investigational tools to explore the function of individual genes or gene combinations in vivo, thus providing information that is conceptually similar to that obtained from genetically modified animals. Finally, their single-stranded DNA genome can drive homology-directed gene repair at high efficiency. Here, we review the main molecular characteristics of adeno-associated virus vectors, with a particular view to their applications in the cardiovascular field.
Collapse
Affiliation(s)
- Serena Zacchigna
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy (S.Z., L.Z., M.G.); and Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy (S.Z., M.G.)
| | - Lorena Zentilin
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy (S.Z., L.Z., M.G.); and Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy (S.Z., M.G.)
| | - Mauro Giacca
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy (S.Z., L.Z., M.G.); and Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy (S.Z., M.G.).
| |
Collapse
|
321
|
Paydar A, Lee B, Gangadharan G, Lee S, Hwang EM, Shin HS. Extrasynaptic GABAA receptors in mediodorsal thalamic nucleus modulate fear extinction learning. Mol Brain 2014; 7:39. [PMID: 24886120 PMCID: PMC4066285 DOI: 10.1186/1756-6606-7-39] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 05/23/2014] [Indexed: 11/14/2022] Open
Abstract
Background The gamma-amino-butyric acid (GABA) system is a critical mediator of fear extinction process. GABA can induce “phasic” or “tonic” inhibition in neurons through synaptic or extrasynaptic GABAA receptors, respectively. However, role of the thalamic “tonic GABA inhibition” in cognition has not been explored. We addressed this issue in extinction of conditioned fear in mice. Results Here, we show that GABAA receptors in the mediodorsal thalamic nucleus (MD) modulate fear extinction. Microinjection of gabazine, a GABAA receptor antagonist, into the MD decreased freezing behavior in response to the conditioned stimulus and thus facilitated fear extinction. Interestingly, microinjection of THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol), a preferential agonist for the δ-subunit of extrasynaptic GABAA receptors, into the MD attenuated fear extinction. In the opposite direction, an MD-specific knock-out of the extrasynaptic GABAA receptors facilitated fear extinction. Conclusions Our results suggest that “tonic GABA inhibition” mediated by extrasynaptic GABAA receptors in MD neurons, suppresses fear extinction learning. These results raise a possibility that pharmacological control of tonic mode of GABAA receptor activation may be a target for treatment of anxiety disorders like post-traumatic stress disorder.
Collapse
Affiliation(s)
| | | | | | | | | | - Hee-Sup Shin
- Center for Cognition and Sociality, Institute for Basic Science (IBS), 70, Yusung-daero 1689-gil, Yusung-gu, Daejeon 305-811, Republic of Korea.
| |
Collapse
|
322
|
Lin Z, von Gise A, Zhou P, Gu F, Ma Q, Jiang J, Yau AL, Buck JN, Gouin KA, van Gorp PRR, Zhou B, Chen J, Seidman JG, Wang DZ, Pu WT. Cardiac-specific YAP activation improves cardiac function and survival in an experimental murine MI model. Circ Res 2014; 115:354-63. [PMID: 24833660 DOI: 10.1161/circresaha.115.303632] [Citation(s) in RCA: 329] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RATIONALE Yes-associated protein (YAP), the terminal effector of the Hippo signaling pathway, is crucial for regulating embryonic cardiomyocyte proliferation. OBJECTIVE We hypothesized that YAP activation after myocardial infarction (MI) would preserve cardiac function and improve survival. METHODS AND RESULTS We used a cardiac-specific, inducible expression system to activate YAP in adult mouse heart. Activation of YAP in adult heart promoted cardiomyocyte proliferation and did not deleteriously affect heart function. Furthermore, YAP activation after MI preserved heart function and reduced infarct size. Using adeno-associated virus subtype 9 (AAV9) as a delivery vector, we expressed human YAP (hYAP) in the adult murine myocardium immediately after MI. We found that AAV9:hYAP significantly improved cardiac function and mouse survival. AAV9:hYAP did not exert its salutary effects by reducing cardiomyocyte apoptosis. Rather, AAV9:hYAP stimulated adult cardiomyocyte proliferation. Gene expression profiling indicated that AAV9:hYAP stimulated expression of cell cycle genes and promoted a less mature cardiac gene expression signature. CONCLUSIONS Cardiac-specific YAP activation after MI mitigated myocardial injury, improved cardiac function, and enhanced survival. These findings suggest that therapeutic activation of YAP or its downstream targets, potentially through AAV-mediated gene therapy, may be a strategy to improve outcome after MI.
Collapse
Affiliation(s)
- Zhiqiang Lin
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Alexander von Gise
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Pingzhu Zhou
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Fei Gu
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Qing Ma
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Jianming Jiang
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Allan L Yau
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Jessica N Buck
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Katryna A Gouin
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Pim R R van Gorp
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Bin Zhou
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Jinghai Chen
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Jonathan G Seidman
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - Da-Zhi Wang
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.)
| | - William T Pu
- From the Departments of Cardiology, Boston Children's Hospital (Z.L., A.v.G., P.Z., F.G., Q.M., A.L.Y., J.N.B., K.A.G., P.R.R.v.G., B.Z., J.C., D.-Z.W., W.T.P.) and Genetics (J.J., J.G.S.), Harvard Medical School, Boston, MA; Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, Hannover, Germany (A.v.G.); Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (P.R.R.v.G.); Harvard Stem Cell Institute, Harvard University, Cambridge, MA (D.-Z.W., W.T.P.); and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (B.Z.).
| |
Collapse
|
323
|
Castle MJ, Gershenson ZT, Giles AR, Holzbaur ELF, Wolfe JH. Adeno-associated virus serotypes 1, 8, and 9 share conserved mechanisms for anterograde and retrograde axonal transport. Hum Gene Ther 2014; 25:705-20. [PMID: 24694006 DOI: 10.1089/hum.2013.189] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Adeno-associated virus (AAV) vectors often undergo long-distance axonal transport after brain injection. This leads to transduction of brain regions distal to the injection site, although the extent of axonal transport and distal transduction varies widely among AAV serotypes. The mechanisms driving this variability are poorly understood. This is a critical problem for applications that require focal gene expression within a specific brain region, and also impedes the utilization of vector transport for applications requiring widespread delivery of transgene to the brain. Here, we compared AAV serotypes 1 and 9, which frequently demonstrate distal transduction, with serotype 8, which rarely spreads beyond the injection site. To examine directional AAV transport in vitro, we used a microfluidic chamber to apply dye-labeled AAV to the axon termini or to the cell bodies of primary rat embryonic cortical neurons. All three serotypes were actively transported along axons, with transport characterized by high velocities and prolonged runs in both the anterograde and retrograde directions. Coinfection with pairs of serotypes indicated that AAV1, 8, and 9 share the same intracellular compartments for axonal transport. In vivo, both AAV8 and 9 demonstrated anterograde and retrograde transport within a nonreciprocal circuit after injection into adult mouse brain, with highly similar distributions of distal transduction. However, in mass-cultured neurons, we found that AAV1 was more frequently transported than AAV8 or 9, and that the frequency of AAV9 transport could be enhanced by increasing receptor availability. Thus, while these serotypes share conserved mechanisms for axonal transport both in vitro and in vivo, the frequency of transport can vary among serotypes, and axonal transport can be markedly increased by enhancing vector uptake. This suggests that variability in distal transduction in vivo likely results from differential uptake at the plasma membrane, rather than fundamental differences in transport mechanisms among AAV serotypes.
Collapse
Affiliation(s)
- Michael J Castle
- 1 Research Institute of the Children's Hospital of Philadelphia , Philadelphia, PA 19104
| | | | | | | | | |
Collapse
|
324
|
Luo Y, Lin L, Bolund L, Sørensen CB. Efficient construction of rAAV-based gene targeting vectors by Golden Gate cloning. Biotechniques 2014; 56:263-8. [PMID: 24806227 DOI: 10.2144/000114169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 02/14/2014] [Indexed: 11/23/2022] Open
Abstract
The recombinant adeno-associated virus (rAAV) has proven to be an efficient and attractive tool for targeted genome engineering. Here we present a novel method employing the Golden Gate cloning strategy for fast and efficient construction of rAAV-based gene knockout or single-nucleotide knockin vectors. Two vectors, pGolden-Neo and pGolden-Hyg, were generated as common assembling modules to confer antibiotic resistance to the targeting vector. To validate the method, we then generated two rAAV-based targeting vectors: pAAV-pTP53-KO and pAAV-hTau(P301L)-KI. Furthermore, we generated a pGolden-AAV plasmid that allows one-step generation of an rAAV-based targeting vector. Our new methodology for rAAV targeting vector assembly is efficient, accurate, time-saving, and cost-effective.
Collapse
Affiliation(s)
- Yonglun Luo
- Department of Biomedicine, Faculty of Heath, Aarhus University, Aarhus, Denmark
| | - Lin Lin
- Department of Biomedicine, Faculty of Heath, Aarhus University, Aarhus, Denmark
| | - Lars Bolund
- Department of Biomedicine, Faculty of Heath, Aarhus University, Aarhus, Denmark
| | - Charlotte Brandt Sørensen
- Department of Biomedicine, Faculty of Heath, Aarhus University, Aarhus, Denmark; Department of Clinical Medicine, Department of Cardiological Medicine B, Aarhus University, Aarhus, Denmark
| |
Collapse
|
325
|
Robbins KL, Glascock JJ, Osman EY, Miller MR, Lorson CL. Defining the therapeutic window in a severe animal model of spinal muscular atrophy. Hum Mol Genet 2014; 23:4559-68. [PMID: 24722206 DOI: 10.1093/hmg/ddu169] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by the loss of a single gene, Survival Motor Neuron-1 (SMN1). Administration of a self-complementary Adeno-Associated Virus vector expressing full-length SMN cDNA (scAAV-SMN) has proven an effective means to rescue the SMA phenotype in SMA mice, either by intravenous (IV) or intracerebroventricular (ICV) administration at very early time points. We have recently shown that ICV delivery of scAAV9-SMN is more effective than a similar dose of vector administered via an IV injection, thereby providing an important mechanism to examine a timeline for rescuing the disease and determining the therapeutic window in a severe model of SMA. In this report, we utilized a relatively severe mouse model of SMA, SMNΔ7. Animals were injected with scAAV9-SMN vector via ICV injection on a single day, from P2 through P8. At each delivery point from P2 through P8, scAAV9-SMN decreased disease severity. A near complete rescue was obtained following P2 injection while a P8 injection produced a ∼ 40% extension in survival. Analysis of the underlying neuromuscular junction (NMJ) pathology revealed that late-stage delivery of the vector failed to provide protection from NMJ defects despite robust SMN expression in the central nervous system. While our study demonstrates that a maximal benefit is obtained when treatment is delivered during pre-symptomatic stages, significant therapeutic benefit can still be achieved after the onset of disease symptoms.
Collapse
Affiliation(s)
- Kate L Robbins
- Department of Veterinary Pathobiology, Christopher S. Bond Life Sciences Center
| | - Jacqueline J Glascock
- Department of Veterinary Pathobiology, Christopher S. Bond Life Sciences Center, Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine and
| | - Erkan Y Osman
- Department of Veterinary Pathobiology, Christopher S. Bond Life Sciences Center, Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine and
| | - Madeline R Miller
- Genetics Area Program, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Christian L Lorson
- Department of Veterinary Pathobiology, Christopher S. Bond Life Sciences Center, Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine and
| |
Collapse
|
326
|
Byrne LC, Oztürk BE, Lee T, Fortuny C, Visel M, Dalkara D, Schaffer DV, Flannery JG. Retinoschisin gene therapy in photoreceptors, Müller glia or all retinal cells in the Rs1h-/- mouse. Gene Ther 2014; 21:585-92. [PMID: 24694538 PMCID: PMC4047144 DOI: 10.1038/gt.2014.31] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 01/28/2014] [Accepted: 02/25/2014] [Indexed: 01/21/2023]
Abstract
X-linked retinoschisis, a disease characterized by splitting of the retina, is caused by mutations in the retinoschisin gene, which encodes a secreted cell adhesion protein. Currently, there is no effective treatment for retinoschisis, though viral vector-mediated gene replacement therapies offer promise. We used intravitreal delivery of three different AAV vectors to target delivery of the RS1 gene to Müller glia, photoreceptors, or multiple cell types throughout the retina. Müller glia radially span the entire retina, are accessible from the vitreous, and remain intact throughout progression of the disease. However, photoreceptors, not glia, normally secrete retinoschisin. We compared the efficacy of rescue mediated by retinoschisin secretion from these specific subtypes of retinal cells in the Rs1h−/− mouse model of retinoschisis. Our results indicate that all three vectors deliver the RS1 gene, and that several cell types can secrete retinoschisin, leading to transport of the protein across the retina. The greatest long-term rescue was observed when photoreceptors produce retinoschisin. Similar rescue was observed with photoreceptor-specific or generalized expression, though photoreceptor secretion may contribute to rescue in the latter case. These results collectively point to the importance of cell targeting and appropriate vector choice in the success of retinal gene therapies.
Collapse
Affiliation(s)
- L C Byrne
- 1] Department of Molecular and Cellular Biology, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA [2] Department of Chemical and Biomolecular Engineering, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA [3] Department of Bioengineering, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA
| | - B E Oztürk
- Department of Molecular and Cellular Biology, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA
| | - T Lee
- Department of Molecular and Cellular Biology, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA
| | - C Fortuny
- 1] Department of Molecular and Cellular Biology, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA [2] Department of Vision Science, The University of California, Berkeley, CA, USA
| | - M Visel
- Department of Molecular and Cellular Biology, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA
| | - D Dalkara
- 1] Department of Molecular and Cellular Biology, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA [2] Department of Chemical and Biomolecular Engineering, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA [3] Department of Bioengineering, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA
| | - D V Schaffer
- 1] Department of Chemical and Biomolecular Engineering, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA [2] Department of Bioengineering, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA
| | - J G Flannery
- 1] Department of Molecular and Cellular Biology, The Helen Wills Neuroscience Institute, The University of California, Berkeley, CA, USA [2] Department of Vision Science, The University of California, Berkeley, CA, USA
| |
Collapse
|
327
|
Gubrij IB, Martin SR, Pangle AK, Kurten R, Johnson LG. Attenuation of monocrotaline-induced pulmonary hypertension by luminal adeno-associated virus serotype 9 gene transfer of prostacyclin synthase. Hum Gene Ther 2014; 25:498-505. [PMID: 24512101 DOI: 10.1089/hum.2013.187] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Idiopathic pulmonary arterial hypertension (iPAH) is associated with high morbidity and mortality. We evaluated whether luminal delivery of the human prostacyclin synthase (hPGIS) cDNA with adeno-associated virus (AAV) vectors could attenuate PAH. AAV serotype 5 (AAV5) and AAV9 vectors containing the hPGIS cDNA under the control of a cytomegalovirus-enhanced chicken β-actin (CB) promoter or vehicle (saline) were instilled into lungs of rats. Two days later, rats were injected with monocrotaline (MCT, 60 mg/kg) or saline. Biochemical, hemodynamic, and morphologic assessments were performed when the rats developed symptoms (3-4 weeks) or at 6 weeks. Luminal (airway) administration of AAV5 and AAV9CBhPGIS vectors (MCT-AAV5 and MCT-AAV9 rats) significantly increased plasma levels of 6-keto-PGF1(α) as compared with MCT-controls, and closely resembled levels measured in rats not treated with MCT (saline-saline). Right ventricular (RV)/left ventricular (LV)+septum (S) ratios and RV systolic pressure (RVSP) were greater in MCT-control rats than in saline-saline rats, whereas the ratios and RVSP in MCT-AAV5CBhPGIS and MCT-AAV9CBhPGIS rats were similar to saline-saline rats. Thickening of the muscular media of small pulmonary arteries of MCT-control rats was detected in histological sections, whereas the thickness of the muscular media in MCT-AAV5CBhPGIS and MCT-AAV9CBhPGIS rats was similar to saline-saline controls. In experiments with different promoters, a trend toward increased levels of PGF1(α) expression was detected in lung homogenates, but not plasma, of MCT-treated rats transduced with an AAV9-hPGIS vector containing a CB promoter. This correlated with significant reductions in the RV/LV+S ratio and RVSP in MCT-AAV9CBhPGIS rats that resembled levels in saline-saline rats. No changes in levels of PGF1(α), RV/LV+S, or RVSP were detected in rats transduced with AAV9-hPGIS vectors containing a modified CB promoter (CB7) or a distal epithelial cell-specific promoter (CC10). Thus, AAV9CBhPGIS vectors prevented development of MCT-induced PAH and associated pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Igor B Gubrij
- 1 Division of Pulmonary and Critical Care, Department of Medicine, University of Arkansas for Medical Sciences , Little Rock, AR 72205
| | | | | | | | | |
Collapse
|
328
|
Lutz D, Loers G, Kleene R, Oezen I, Kataria H, Katagihallimath N, Braren I, Harauz G, Schachner M. Myelin basic protein cleaves cell adhesion molecule L1 and promotes neuritogenesis and cell survival. J Biol Chem 2014; 289:13503-18. [PMID: 24671420 DOI: 10.1074/jbc.m113.530238] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The cell adhesion molecule L1 is a Lewis(x)-carrying glycoprotein that plays important roles in the developing and adult nervous system. Here we show that myelin basic protein (MBP) binds to L1 in a Lewis(x)-dependent manner. Furthermore, we demonstrate that MBP is released by murine cerebellar neurons as a sumoylated dynamin-containing protein upon L1 stimulation and that this MBP cleaves L1 as a serine protease in the L1 extracellular domain at Arg(687) yielding a transmembrane fragment that promotes neurite outgrowth and neuronal survival in cell culture. L1-induced neurite outgrowth and neuronal survival are reduced in MBP-deficient cerebellar neurons and in wild-type cerebellar neurons in the presence of an MBP antibody or L1 peptide containing the MBP cleavage site. Genetic ablation of MBP in shiverer mice and mutagenesis of the proteolytically active site in MBP or of the MBP cleavage site within L1 as well as serine protease inhibitors and an L1 peptide containing the MBP cleavage site abolish generation of the L1 fragment. Our findings provide evidence for novel functions of MBP in the nervous system.
Collapse
Affiliation(s)
- David Lutz
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
329
|
Lagor WR, Johnston JC, Lock M, Vandenberghe LH, Rader DJ. Adeno-associated viruses as liver-directed gene delivery vehicles: focus on lipoprotein metabolism. Methods Mol Biol 2014; 1027:273-307. [PMID: 23912992 DOI: 10.1007/978-1-60327-369-5_13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Adeno-associated viral vectors have proven to be excellent gene delivery vehicles for somatic overexpression. These viral vectors can efficiently and selectively target the liver, which plays a central role in lipoprotein metabolism. Both liver-expressed as well as non-hepatic secreted proteins can be easily examined in different mouse models using this approach. The dosability of adeno-associated viral (AAV) vectors, as well as their potential for long-term expression, makes them an excellent choice for assessing gene function in vivo. This section will cover the use of AAV to study lipoprotein metabolism-including vector design, virus production and purification, and viral delivery, as well as monitoring of transgene expression and resulting phenotypic changes. Practical information is provided to assist the investigator in designing, interpreting, and troubleshooting experiments.
Collapse
Affiliation(s)
- William R Lagor
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
330
|
Chaanine AH, Nonnenmacher M, Kohlbrenner E, Jin D, Kovacic JC, Akar FG, Hajjar RJ, Weber T. Effect of bortezomib on the efficacy of AAV9.SERCA2a treatment to preserve cardiac function in a rat pressure-overload model of heart failure. Gene Ther 2014; 21:379-386. [PMID: 24572786 PMCID: PMC3976435 DOI: 10.1038/gt.2014.7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 12/24/2013] [Accepted: 01/09/2014] [Indexed: 01/14/2023]
Abstract
Adeno-associated virus (AAV)-based vectors are promising vehicles for therapeutic gene delivery, including for the treatment for heart failure. It has been demonstrated for each of the AAV serotypes 1 through 8 that inhibition of the proteasome results in increased transduction efficiencies. For AAV9, however, the effect of proteasome inhibitors on in vivo transduction has until now not been evaluated. Here we demonstrate, in a well-established rodent heart failure model, that concurrent treatment with the proteasome inhibitor bortezomib does not enhance the efficacy of AAV9.SERCA2a to improve cardiac function as examined by echocardiography and pressure volume analysis. Western blot analysis of SERCA2a protein and reverse transcription-PCR of SERCA2a mRNA demonstrated that bortezomib had no effect on either endogenous rat SERCA2a levels nor on expression levels of human SERCA2a delivered by AAV9.SERCA2a. Similarly, the number of AAV9 genomes in heart samples was unaffected by bortezomib treatment. Interestingly, whereas transduction of HeLa cells and neonatal rat cardiomyocytes by AAV9 was stimulated by bortezomib, transduction of adult rat cardiomyocytes was inhibited. These results indicate an organ/cell-type-specific effect of proteasome inhibition on AAV9 transduction. A future detailed analysis of the underlying molecular mechanisms promises to facilitate the development of improved AAV vectors.
Collapse
Affiliation(s)
- Antoine H Chaanine
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Mathieu Nonnenmacher
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Erik Kohlbrenner
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Dongzhu Jin
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jason C Kovacic
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Fadi G Akar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Thomas Weber
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
331
|
Ruff P, Koh KD, Keskin H, Pai RB, Storici F. Aptamer-guided gene targeting in yeast and human cells. Nucleic Acids Res 2014; 42:e61. [PMID: 24500205 PMCID: PMC3985672 DOI: 10.1093/nar/gku101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Gene targeting is a genetic technique to modify an endogenous DNA sequence in its genomic location via homologous recombination (HR) and is useful both for functional analysis and gene therapy applications. HR is inefficient in most organisms and cell types, including mammalian cells, often limiting the effectiveness of gene targeting. Therefore, increasing HR efficiency remains a major challenge to DNA editing. Here, we present a new concept for gene correction based on the development of DNA aptamers capable of binding to a site-specific DNA binding protein to facilitate the exchange of homologous genetic information between a donor molecule and the desired target locus (aptamer-guided gene targeting). We selected DNA aptamers to the I-SceI endonuclease. Bifunctional oligonucleotides containing an I-SceI aptamer sequence were designed as part of a longer single-stranded DNA molecule that contained a region with homology to repair an I-SceI generated double-strand break and correct a disrupted gene. The I-SceI aptamer-containing oligonucleotides stimulated gene targeting up to 32-fold in yeast Saccharomyces cerevisiae and up to 16-fold in human cells. This work provides a novel concept and research direction to increase gene targeting efficiency and lays the groundwork for future studies using aptamers for gene targeting.
Collapse
Affiliation(s)
- Patrick Ruff
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | | | | | | | | |
Collapse
|
332
|
Dong B, Duan X, Chow HY, Chen L, Lu H, Wu W, Hauck B, Wright F, Kapranov P, Xiao W. Proteomics analysis of co-purifying cellular proteins associated with rAAV vectors. PLoS One 2014; 9:e86453. [PMID: 24498275 PMCID: PMC3911921 DOI: 10.1371/journal.pone.0086453] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 12/11/2013] [Indexed: 12/05/2022] Open
Abstract
Recombinant adeno-associated vectors (rAAV) are commonly purified by either chromatography or equilibrium CsCl gradient. Nevertheless, even after purification various cellular proteins often associate with rAAV vector capsids. Such co-purifying cellular proteins may raise concern about safety of gene therapy. Here we report identification and characterization of the co-purifying cellular protein in the vector preparations by using a combination of two proteomics approaches, GeLC-MS (gel electrophoresis liquid chromatography-mass spectrometry) and 2DE (two-dimensional gel electrophoresis). Most prominent bands revealed by Coomassie Blue staining were mostly similar to the AAV capsid proteins. Posttranslational modifications of capsid proteins were detected by the proteomics analysis. A total of 13 cellular proteins were identified in the rAAV vectors purified by two rounds of cesium chloride gradient centrifugation, including 9 by the GeLC-MS analysis and 4 by the 2DE analysis. Selected cellular proteins were verified by western blot. Furthermore, the cellular proteins could be consistently found associated with different AAV serotypes and carrying different transgenes. Yet, the proteins were not integral components of the viral capsis since a stringent washing procedure by column purification could remove them. These co-purified proteins in AAV vector preparations may have a role in various stages of the AAV life cycle.
Collapse
Affiliation(s)
- Biao Dong
- Department of Microbiology and Immunology, Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Xunbao Duan
- Department of Microbiology and Immunology, Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Hoi Yee Chow
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Lingxia Chen
- Department of Microbiology and Immunology, Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Hui Lu
- Department of Microbiology and Immunology, Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Wenman Wu
- Department of Microbiology and Immunology, Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Bernd Hauck
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, and Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Fraser Wright
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, and Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Philipp Kapranov
- St. Laurent Institute, Cambridge, Massachusetts, United States of America
| | - Weidong Xiao
- Department of Microbiology and Immunology, Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
333
|
Huang ZJ, Taniguchi H, He M, Kuhlman S. Cre-dependent adeno-associated virus preparation and delivery for labeling neurons in the mouse brain. Cold Spring Harb Protoc 2014; 2014:190-194. [PMID: 24492777 DOI: 10.1101/pdb.prot080382] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Virus-mediated gene delivery is a powerful strategy for labeling and manipulating neurons in mammalian brains. A major drawback of this gene delivery method has been the lack of cell-type specificity. However, methods that combine Cre-knockin mice and Cre-activated adeno-associated virus (AAV) have now been developed to achieve high-level, stable, and cell-type-specific gene expression. Here, we describe a protocol for the design, production, and delivery of Cre-dependent AAVs. This method is simple and highly efficient, and allows chronic live imaging of defined classes of synapses in the mouse brain.
Collapse
|
334
|
Recombinant adeno-associated virus utilizes host cell nuclear import machinery to enter the nucleus. J Virol 2014; 88:4132-44. [PMID: 24478436 DOI: 10.1128/jvi.02660-13] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Recombinant adeno-associated viral (rAAV) vectors have garnered much promise in gene therapy applications. However, widespread clinical use has been limited by transduction efficiency. Previous studies suggested that the majority of rAAV accumulates in the perinuclear region of cells, presumably unable to traffic into the nucleus. rAAV nuclear translocation remains ill-defined; therefore, we performed microscopy, genetic, and biochemical analyses in vitro in order to understand this mechanism. Lectin blockade of the nuclear pore complex (NPC) resulted in inhibition of nuclear rAAV2. Visualization of fluorescently labeled particles revealed that rAAV2 localized to importin-β-dense regions of cells in late trafficking steps. Additionally, small interfering RNA (siRNA) knockdown of importin-β partially inhibited rAAV2 nuclear translocation and inhibited transduction by 50 to 70%. Furthermore, coimmunopreciptation (co-IP) analysis revealed that capsid proteins from rAAV2 could interact with importin-β and that this interaction was sensitive to the small GTPase Ran. More importantly, mutations to key basic regions in the rAAV2 capsid severely inhibited interactions with importin-β. We tested several other serotypes and found that the extent of importin-β interaction varied, suggesting that different serotypes may utilize alternative import proteins for nuclear translocation. Co-IP and siRNA analyses were used to investigate the role of other karyopherins, and the results suggested that rAAV2 may utilize multiple import proteins for nuclear entry. Taken together, our results suggest that rAAV2 interacts with importin-β alone or in complex with other karyopherins and enters the nucleus via the NPC. These results may lend insight into the design of novel AAV vectors that have an enhanced nuclear entry capability and transduction potential. IMPORTANCE Use of recombinant adeno-associated viral (rAAV) vectors for gene therapy applications is limited by relatively low transduction efficiency, in part due to cellular barriers that hinder successful subcellular trafficking to the nucleus, where uncoating and subsequent gene expression occur. Nuclear translocation of rAAV has been regarded as a limiting step for successful transduction but it remains ill-defined. We explored potential nuclear entry mechanisms for rAAV2 and found that rAAV2 can utilize the classical nuclear import pathway, involving the nuclear pore complex, the small GTPase Ran, and cellular karyopherins. These results could lend insight into the rational design of novel rAAV vectors that can more efficiently translocate to the nucleus, which may lead to more efficient transduction.
Collapse
|
335
|
Donegan JJ, Girotti M, Weinberg MS, Morilak DA. A novel role for brain interleukin-6: facilitation of cognitive flexibility in rat orbitofrontal cortex. J Neurosci 2014; 34:953-62. [PMID: 24431453 PMCID: PMC3891970 DOI: 10.1523/jneurosci.3968-13.2014] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/07/2013] [Accepted: 11/27/2013] [Indexed: 12/21/2022] Open
Abstract
Cytokines, small proteins released by the immune system to combat infection, are typically studied under inflammatory conditions. However, these molecules are also expressed in the brain in basal, nonpathological states, where they can regulate neuronal processes, such as learning and memory. However, little is known about how cytokine signaling in the brain may influence higher-order cognitive functions. Cognitive flexibility is one such executive process, mediated by the prefrontal cortex, which requires an adaptive modification of learned behaviors in response to environmental change. We explored the role of basal IL-6 signaling in the orbitofrontal cortex (OFC) in reversal learning, a form of cognitive flexibility that can be measured in the rat using the attentional set-shifting test. We found that inhibiting IL-6 or its downstream JAK/STAT signaling pathway in the OFC impaired reversal learning, suggesting that basal IL-6 and JAK/STAT signaling facilitate cognitive flexibility. Further, we demonstrated that elevating IL-6 in the OFC by adeno-associated virus-mediated gene delivery reversed a cognitive deficit induced by chronic stress, thus identifying IL-6 and the downstream JAK/STAT signaling pathway as potentially novel therapeutic targets for the treatment of stress-related psychiatric diseases associated with cognitive dysfunction.
Collapse
Affiliation(s)
- Jennifer J. Donegan
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, and
| | - Milena Girotti
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, and
| | - Marc S. Weinberg
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - David A. Morilak
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, and
| |
Collapse
|
336
|
Abstract
Traditionally, the ability to edit the mammalian genome was inhibited by the inherent low efficiency of homologous recombination (HR; approximately <1 in a million events) and the inability to deliver DNA efficiently to dividing and non-dividing cells/tissue. Despite these limitations, creative selections designed over 20 years ago, clearly demonstrated the powerful implications of gene knock-in and knockout technology for the genetic engineering of mice (Doetschman et al. Nat 330(6148): 576-578, 1987; Thomas and Capecchi. Cell 51(3): 503-512, 1987). The development and application of recombinant vectors based on adeno-associated virus (rAAV) have helped to overcome both of the initial limitations regarding DNA delivery and the frequency of HR. Considering DNA delivery, rAAV infects non-dividing and dividing cultured cells as well as most tissues in mouse and larger animal models (including humans). At the DNA editing level, rAAV genomes have been reported to increase the frequency of HR several orders of magnitude by serving as the repair substrate (Russell and Hirata. Nat Genet 18(4): 325-330, 1998). However, reports on the ability of rAAV genomes to stimulate HR, compared to plasmid DNA and oligonucleotides, are variable, and many labs have found it necessary to augment the frequency of rAAV-induced HR using site-specific endonucleases (Ellis et al. Gene Ther, 2012; Hirsch et al. Gene Ther 17(9): 1175-1180, 2010; Porteus et al. Mol Cell Biol 23(10): 3558-3565, 2003; Radecke et al. Mol Ther 14(6): 798-808, 2006). In this protocol, we describe a method to perform rAAV-mediated double-strand break (DSB) repair for precise genetic engineering in human cells.
Collapse
Affiliation(s)
- Matthew L Hirsch
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA
| | | |
Collapse
|
337
|
Short hairpin RNA against PTEN enhances regenerative growth of corticospinal tract axons after spinal cord injury. J Neurosci 2013; 33:15350-61. [PMID: 24068802 DOI: 10.1523/jneurosci.2510-13.2013] [Citation(s) in RCA: 232] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Developing approaches to promote the regeneration of descending supraspinal axons represents an ideal strategy for rebuilding neuronal circuits to improve functional recovery after spinal cord injury (SCI). Our previous studies demonstrated that genetic deletion of phosphatase and tensin homolog (PTEN) in mouse corticospinal neurons reactivates their regenerative capacity, resulting in significant regeneration of corticospinal tract (CST) axons after SCI. However, it is unknown whether nongenetic methods of suppressing PTEN have similar effects and how regenerating axons interact with the extrinsic environment. Herein, we show that suppressing PTEN expression with short-hairpin RNA (shRNA) promotes the regeneration of injured CST axons, and these axons form anatomical synapses in appropriate areas of the cord caudal to the lesion. Importantly, this model of increased CST regrowth enables the analysis of extrinsic regulators of CST regeneration in vivo. We find that regenerating axons avoid dense clusters of fibroblasts and macrophages in the lesion, suggesting that these cell types might be key inhibitors of axon regeneration. Furthermore, most regenerating axons cross the lesion in association with astrocytes, indicating that these cells might be important for providing a permissive bridge for axon regeneration. Lineage analysis reveals that these bridge-forming astrocytes are not derived from ependymal stem cells within the spinal cord, suggesting that they are more likely derived from a subset of mature astrocytes. Overall, this study reveals insights into the critical extrinsic and intrinsic regulators of axon regeneration and establishes shRNA as a viable means to manipulate these regulators and translate findings into other mammalian models.
Collapse
|
338
|
Yu Q, Wang Y, Chang Q, Wang J, Gong S, Li H, Lin X. Virally expressed connexin26 restores gap junction function in the cochlea of conditional Gjb2 knockout mice. Gene Ther 2013; 21:71-80. [PMID: 24225640 PMCID: PMC3881370 DOI: 10.1038/gt.2013.59] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/09/2013] [Accepted: 09/23/2013] [Indexed: 02/03/2023]
Abstract
Mutations in GJB2, which codes for the gap junction protein connexin26, are the most common causes of human nonsyndromic hereditary deafness. We inoculated modified adeno-associated viral vectors into the scala media of early postnatal conditional Gjb2 knockout mice to drive exogenous connexin26 expression. We found extensive virally-expressed connexin26 in cells lining the scala media, and intercellular gap junction network was re-established in the organ of Corti of mutant mouse cochlea. Widespread ectopic connexin26 expression neither formed ectopic gap junctions nor affected normal hearing thresholds in wild type mice, suggesting that autonomous cellular mechanisms regulate proper membrane trafficking of exogenously-expressed connexin26 and govern the functional manifestation of them. Functional recovery of gap-junction-mediated coupling among the supporting cells was observed. We found that both cell death in the organ of Corti and degeneration of spiral ganglion neurons in the cochlea of mutant mice were substantially reduced, although auditory brainstem responses did not show significant hearing improvement. This is the first report demonstrating that virally-mediated gene therapy restored extensive gap junction intercellular network among cochlear non-sensory cells in vivo. Such a treatment performed at early postnatal stages resulted in a partial rescue of disease phenotypes in the cochlea of the mutant mice.
Collapse
Affiliation(s)
- Q Yu
- 1] Department of Otolaryngology Head & Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China [2] Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - Y Wang
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Q Chang
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - J Wang
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - S Gong
- Department of Otolaryngology Head & Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - H Li
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - X Lin
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
339
|
Chahal PS, Schulze E, Tran R, Montes J, Kamen AA. Production of adeno-associated virus (AAV) serotypes by transient transfection of HEK293 cell suspension cultures for gene delivery. J Virol Methods 2013; 196:163-73. [PMID: 24239634 PMCID: PMC7113661 DOI: 10.1016/j.jviromet.2013.10.038] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 10/11/2013] [Accepted: 10/29/2013] [Indexed: 01/29/2023]
Abstract
Transient transfection of HEK293 suspension cells efficiently produce AAV vectors. Nine different AAV serotypes were produced with yields of 1E+13 Vg/L. AAV2 and AAV6 produced in 3-L bioreactors gave yields comparable to shake-flasks. The process is cGMP compatible using serum-free media and HEK293 master cell bank. Industrialization of the process is possible for manufacturing AAV serotypes.
Adeno-associated virus (AAV) is being used successfully in gene therapy. Different serotypes of AAV target specific organs and tissues with high efficiency. There exists an increasing demand to manufacture various AAV serotypes in large quantities for pre-clinical and clinical trials. A generic and scalable method has been described in this study to efficiently produce AAV serotypes (AAV1-9) by transfection of a fully characterized cGMP HEK293SF cell line grown in suspension and serum-free medium. First, the production parameters were evaluated using AAV2 as a model serotype. Second, all nine AAV serotypes were produced successfully with yields of 1013 Vg/L cell culture. Subsequently, AAV2 and AAV6 serotypes were produced in 3-L controlled bioreactors where productions yielded up to 1013 Vg/L similar to the yields obtained in shake-flasks. For example, for AAV2 1013 Vg/L cell culture (6.8 × 1011 IVP/L) were measured between 48 and 64 h post transfection (hpt). During this period, the average cell specific AAV2 yields of 6800 Vg per cell and 460 IVP per cell were obtained with a Vg to IVP ratio of less than 20. Successful operations in bioreactors demonstrated the potential for scale-up and industrialization of this generic process for manufacturing AAV serotypes efficiently.
Collapse
Affiliation(s)
- Parminder Singh Chahal
- Vaccine Program, Human Health Therapeutics Portfolio, National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, Canada H4P2R2
| | - Erica Schulze
- Vaccine Program, Human Health Therapeutics Portfolio, National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, Canada H4P2R2
| | - Rosa Tran
- Vaccine Program, Human Health Therapeutics Portfolio, National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, Canada H4P2R2
| | - Johnny Montes
- Vaccine Program, Human Health Therapeutics Portfolio, National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, Canada H4P2R2
| | - Amine A Kamen
- Vaccine Program, Human Health Therapeutics Portfolio, National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, Canada H4P2R2.
| |
Collapse
|
340
|
Promyelocytic leukemia protein is a cell-intrinsic factor inhibiting parvovirus DNA replication. J Virol 2013; 88:925-36. [PMID: 24198403 DOI: 10.1128/jvi.02922-13] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tripartite motif proteins are important viral restriction factors and affect processes ranging from uncoating to transcription to immune signaling. Specifically, the promyelocytic leukemia protein (TRIM19; also called PML) is a viral restriction factor inhibiting processes from uncoating to transcription to cell survival. Here we investigated PML's effect on adeno-associated virus (AAV), a parvovirus used for gene delivery. Although dependovirus (AAV) and autonomous parvovirus (minute virus of mice) replication centers can colocalize with PML, PML's functional effect on parvoviruses is unknown. Using PML knockout mice, we determined that PML knockout enhances recombinant AAV2 (rAAV2) transduction at a range of vector doses in both male and female mice. In fact, male and female PML knockout mice exhibited up to 56-fold and 28-fold increases in transduction, respectively. PML inhibited several rAAV serotypes, suggesting a conserved mechanism, and organ specificity correlated with PML expression. Mechanistically, PML inhibited rAAV second-strand DNA synthesis, precluding inhibition of self-complementary rAAV, and did not affect the prior steps in transduction. Furthermore, we confirmed the effect of human PML on rAAV transduction through small interfering RNA (siRNA)-mediated knockdown in HuH7 cells and determined that the highest level of inhibition was due to effects of PML isoform II (PMLII). Overexpression of PMLII resulted in inhibition of second-strand synthesis, vector production, and genome replication. Moreover, wild-type AAV2 production and infectivity were also inhibited by PMLII, demonstrating a PML interaction with wild-type AAV. These data have important implications for AAV-mediated gene therapy. Additionally, PMLII inhibition of AAV second-strand synthesis and replication, which are processes necessary for all parvoviruses, suggests implications for replication of other parvoviruses.
Collapse
|
341
|
de Winter F, Hoyng S, Tannemaat M, Eggers R, Mason M, Malessy M, Verhaagen J. Gene therapy approaches to enhance regeneration of the injured peripheral nerve. Eur J Pharmacol 2013; 719:145-152. [DOI: 10.1016/j.ejphar.2013.04.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 03/28/2013] [Accepted: 04/03/2013] [Indexed: 01/26/2023]
|
342
|
Johnson MC, Garland AL, Nicolson SC, Li C, Samulski RJ, Wang B, Tisch R. β-cell-specific IL-2 therapy increases islet Foxp3+Treg and suppresses type 1 diabetes in NOD mice. Diabetes 2013; 62:3775-84. [PMID: 23884888 PMCID: PMC3806588 DOI: 10.2337/db13-0669] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Interleukin-2 (IL-2) is a critical cytokine for the homeostasis and function of forkhead box p3-expressing regulatory T cells (Foxp3(+)Tregs). Dysregulation of the IL-2-IL-2 receptor axis is associated with aberrant Foxp3(+)Tregs and T cell-mediated autoimmune diseases such as type 1 diabetes. Treatment with recombinant IL-2 has been reported to enhance Foxp3(+)Tregs and suppress different models of autoimmunity. However, efficacy of IL-2 therapy is dependent on achieving sufficient levels of IL-2 to boost tissue-resident Foxp3(+)Tregs while avoiding the potential toxic effects of systemic IL-2. With this in mind, adeno-associated virus (AAV) vector gene delivery was used to localize IL-2 expression to the islets of NOD mice. Injection of a double-stranded AAV vector encoding IL-2 driven by a mouse insulin promoter (dsAAVmIP-IL2) increased Foxp3(+)Tregs in the islets but not the draining pancreatic lymph nodes. Islet Foxp3(+)Tregs in dsAAVmIP-IL2-treated NOD mice exhibited enhanced fitness marked by increased expression of Bcl-2, proliferation, and suppressor function. In contrast, ectopic IL-2 had no significant effect on conventional islet-infiltrating effector T cells. Notably, β-cell-specific IL-2 expression suppressed late preclinical type 1 diabetes in NOD mice. Collectively, these findings demonstrate that β-cell-specific IL-2 expands an islet-resident Foxp3(+)Tregs pool that effectively suppresses ongoing type 1 diabetes long term.
Collapse
Affiliation(s)
- Mark C. Johnson
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alaina L. Garland
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sarah C. Nicolson
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Bo Wang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Roland Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Corresponding author: Roland Tisch,
| |
Collapse
|
343
|
Wagner A, Röhrs V, Kedzierski R, Fechner H, Kurreck J. A novel method for the quantification of adeno-associated virus vectors for RNA interference applications using quantitative polymerase chain reaction and purified genomic adeno-associated virus DNA as a standard. Hum Gene Ther Methods 2013; 24:355-63. [PMID: 23987130 DOI: 10.1089/hgtb.2013.095] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors are promising tools in gene therapy, but accurate quantification of the vector dose remains a critical issue for their successful application. We therefore aimed at the precise determination of the titer of self-complementary AAV (scAAV) vectors to improve the reliability of RNA interference (RNAi)-mediated knockdown approaches. Vector titers were initially determined by quantitative polymerase chain reaction (qPCR) using four primer sets targeting different regions within the AAV vector genome (VG) and either coiled or linearized plasmid standards. Despite very low variability between replicates in each assay, these quantification experiments revealed up to 20-fold variation in vector titers. Therefore, we developed a novel approach for the reproducible determination of titers of scAAV vectors based on the use of purified genomic vector DNA as a standard (scAAVStd). Consistent results were obtained in qPCR assays using the four primer sets mentioned above. RNAi-mediated silencing of human cyclophilin B (hCycB) by short hairpin RNA-expressing scAAV vectors was investigated in HeLa cells using two independent vector preparations. We found that the required vector titers for efficient knockdown differed by a factor of 3.5 between both preparations. Hence, we also investigated the number of internalized scAAV vectors, termed transduction units (TUs). TUs were determined by qPCR applying the scAAVStd. Very similar values for 80% hCycB knockdown were obtained for the two AAV vector preparations. Thus, only the determination of TUs, rather than vector concentration, allows for reproducible results in functional analyses using AAV vectors.
Collapse
Affiliation(s)
- Anke Wagner
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin , Berlin 13355, Germany
| | | | | | | | | |
Collapse
|
344
|
Zhang W, Solanki M, Müther N, Ebel M, Wang J, Sun C, Izsvak Z, Ehrhardt A. Hybrid adeno-associated viral vectors utilizing transposase-mediated somatic integration for stable transgene expression in human cells. PLoS One 2013; 8:e76771. [PMID: 24116154 PMCID: PMC3792901 DOI: 10.1371/journal.pone.0076771] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/28/2013] [Indexed: 12/14/2022] Open
Abstract
Recombinant adeno-associated viral (AAV) vectors have been shown to be one of the most promising vectors for therapeutic gene delivery because they can induce efficient and long-term transduction in non-dividing cells with negligible side-effects. However, as AAV vectors mostly remain episomal, vector genomes and transgene expression are lost in dividing cells. Therefore, to stably transduce cells, we developed a novel AAV/transposase hybrid-vector. To facilitate SB-mediated transposition from the rAAV genome, we established a system in which one AAV vector contains the transposon with the gene of interest and the second vector delivers the hyperactive Sleeping Beauty (SB) transposase SB100X. Human cells were infected with the AAV-transposon vector and the transposase was provided in trans either by transient and stable plasmid transfection or by AAV vector transduction. We found that groups which received the hyperactive transposase SB100X showed significantly increased colony forming numbers indicating enhanced integration efficiencies. Furthermore, we found that transgene copy numbers in transduced cells were dose-dependent and that predominantly SB transposase-mediated transposition contributed to stabilization of the transgene. Based on a plasmid rescue strategy and a linear-amplification mediated PCR (LAM-PCR) protocol we analysed the SB100X-mediated integration profile after transposition from the AAV vector. A total of 1840 integration events were identified which revealed a close to random integration profile. In summary, we show for the first time that AAV vectors can serve as template for SB transposase mediated somatic integration. We developed the first prototype of this hybrid-vector system which with further improvements may be explored for treatment of diseases which originate from rapidly dividing cells.
Collapse
Affiliation(s)
- Wenli Zhang
- Max von Pettenkofer-Institute, Department of Virology, Ludwig-Maximilians-University Munich, Munich, Germany
- Institute of Virology and Microbiology, Center for Biomedical Education and Research, Department of Human Medicine, Faculty of Health, University Witten/Herdecke, Witten, Germany
| | - Manish Solanki
- Institute of Virology and Microbiology, Center for Biomedical Education and Research, Department of Human Medicine, Faculty of Health, University Witten/Herdecke, Witten, Germany
| | - Nadine Müther
- Max von Pettenkofer-Institute, Department of Virology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Melanie Ebel
- Institute of Virology and Microbiology, Center for Biomedical Education and Research, Department of Human Medicine, Faculty of Health, University Witten/Herdecke, Witten, Germany
| | - Jichang Wang
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Chuanbo Sun
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Anja Ehrhardt
- Max von Pettenkofer-Institute, Department of Virology, Ludwig-Maximilians-University Munich, Munich, Germany
- Institute of Virology and Microbiology, Center for Biomedical Education and Research, Department of Human Medicine, Faculty of Health, University Witten/Herdecke, Witten, Germany
- * E-mail:
| |
Collapse
|
345
|
Castle MJ, Perlson E, Holzbaur EL, Wolfe JH. Long-distance axonal transport of AAV9 is driven by dynein and kinesin-2 and is trafficked in a highly motile Rab7-positive compartment. Mol Ther 2013; 22:554-566. [PMID: 24100640 DOI: 10.1038/mt.2013.237] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/30/2013] [Indexed: 12/15/2022] Open
Abstract
Adeno-associated virus (AAV) vectors can move along axonal pathways after brain injection, resulting in transduction of distal brain regions. This can enhance the spread of therapeutic gene transfer and improve treatment of neurogenetic disorders that require global correction. To better understand the underlying cellular mechanisms that drive AAV trafficking in neurons, we investigated the axonal transport of dye-conjugated AAV9, utilizing microfluidic primary neuron cultures that isolate cell bodies from axon termini and permit independent analysis of retrograde and anterograde axonal transport. After entry, AAV was trafficked into nonmotile early and recycling endosomes, exocytic vesicles, and a retrograde-directed late endosome/lysosome compartment. Rab7-positive late endosomes/lysosomes that contained AAV were highly motile, exhibiting faster retrograde velocities and less pausing than Rab7-positive endosomes without virus. Inhibitor experiments indicated that the retrograde transport of AAV within these endosomes is driven by cytoplasmic dynein and requires Rab7 function, whereas anterograde transport of AAV is driven by kinesin-2 and exhibits unusually rapid velocities. Furthermore, increasing AAV9 uptake by neuraminidase treatment significantly enhanced virus transport in both directions. These findings provide novel insights into AAV trafficking within neurons, which should enhance progress toward the utilization of AAV for improved distribution of transgene delivery within the brain.
Collapse
Affiliation(s)
- Michael J Castle
- Research Institute of the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eran Perlson
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Erika Lf Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John H Wolfe
- Research Institute of the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
346
|
Byrne LC, Khalid F, Lee T, Zin EA, Greenberg KP, Visel M, Schaffer DV, Flannery JG. AAV-mediated, optogenetic ablation of Müller Glia leads to structural and functional changes in the mouse retina. PLoS One 2013; 8:e76075. [PMID: 24086689 PMCID: PMC3785414 DOI: 10.1371/journal.pone.0076075] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 08/20/2013] [Indexed: 11/18/2022] Open
Abstract
Müller glia, the primary glial cell in the retina, provide structural and metabolic support for neurons and are essential for retinal integrity. Müller cells are closely involved in many retinal degenerative diseases, including macular telangiectasia type 2, in which impairment of central vision may be linked to a primary defect in Müller glia. Here, we used an engineered, Müller-specific variant of AAV, called ShH10, to deliver a photo-inducibly toxic protein, KillerRed, to Müller cells in the mouse retina. We characterized the results of specific ablation of these cells on visual function and retinal structure. ShH10-KillerRed expression was obtained following intravitreal injection and eyes were then irradiated with green light to induce toxicity. Induction of KillerRed led to loss of Müller cells and a concomitant decrease of Müller cell markers glutamine synthetase and cellular retinaldehyde-binding protein, reduction of rhodopsin and cone opsin, and upregulation of glial fibrillary acidic protein. Loss of Müller cells also resulted in retinal disorganization, including thinning of the outer nuclear layer and the photoreceptor inner and outer segments. High resolution imaging of thin sections revealed displacement of photoreceptors from the ONL, formation of rosette-like structures and the presence of phagocytic cells. Furthermore, Müller cell ablation resulted in increased area and volume of retinal blood vessels, as well as the formation of tortuous blood vessels and vascular leakage. Electrophysiologic measures demonstrated reduced retinal function, evident in decreased photopic and scotopic electroretinogram amplitudes. These results show that loss of Müller cells can cause progressive retinal degenerative disease, and suggest that AAV delivery of an inducibly toxic protein in Müller cells may be useful to create large animal models of retinal dystrophies.
Collapse
Affiliation(s)
- Leah C. Byrne
- Department of Molecular and Cellular Biology and The Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Fakhra Khalid
- Department of Molecular and Cellular Biology and The Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Trevor Lee
- Department of Molecular and Cellular Biology and The Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Emilia A. Zin
- Department of Molecular and Cellular Biology and The Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | | | - Meike Visel
- Department of Molecular and Cellular Biology and The Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - David V. Schaffer
- Department of Chemical Engineering, Department of Bioengineering, and The Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - John G. Flannery
- Department of Molecular and Cellular Biology and The Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
347
|
Multiple roles for sialylated glycans in determining the cardiopulmonary tropism of adeno-associated virus 4. J Virol 2013; 87:13206-13. [PMID: 24067974 DOI: 10.1128/jvi.02109-13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Adeno-associated virus 4 (AAV4) is one of the most divergent serotypes among known AAV isolates. Mucins or O-linked sialoglycans have been identified as the primary attachment receptors for AAV4 in vitro. However, little is known about the role(s) played by sialic acid interactions in determining AAV4 tissue tropism in vivo. In the current study, we first characterized two loss-of-function mutants obtained by screening a randomly mutated AAV4 capsid library. Both mutants harbored several amino acid residue changes localized to the 3-fold icosahedral symmetry axes on the AAV4 capsid and displayed low transduction efficiency in vitro. This defect was attributed to decreased cell surface binding as well as uptake of mutant virions. These results were further corroborated by low transgene expression and recovery of mutant viral genomes in cardiac and lung tissue following intravenous administration in mice. Pharmacokinetic analysis revealed rapid clearance of AAV4 mutants from the blood circulation in conjunction with low hemagglutination potential ex vivo. These results were recapitulated with mice pretreated intravenously with sialidase, directly confirming the role of sialic acids in determining AAV4 tissue tropism. Taken together, our results support the notion that blood-borne AAV4 particles interact sequentially with O-linked sialoglycans expressed abundantly on erythrocytes followed by cardiopulmonary tissues and subsequently for viral cell entry.
Collapse
|
348
|
Mechanistic insights into the enhancement of adeno-associated virus transduction by proteasome inhibitors. J Virol 2013; 87:13035-41. [PMID: 24027330 DOI: 10.1128/jvi.01826-13] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Proteasome inhibitors (e.g., bortezomib, MG132) are known to enhance adeno-associated virus (AAV) transduction; however, whether this results from pleotropic proteasome inhibition or off-target serine and/or cysteine protease inhibition remains unresolved. Here, we examined recombinant AAV (rAAV) effects of a new proteasome inhibitor, carfilzomib, which specifically inhibits chymotrypsin-like proteasome activity and no other proteases. We determined that proteasome inhibitors act on rAAV through proteasome inhibition and not serine or cysteine protease inhibition, likely through positive changes late in transduction.
Collapse
|
349
|
Weinberg MS, Blake BL, McCown TJ. Opposing actions of hippocampus TNFα receptors on limbic seizure susceptibility. Exp Neurol 2013; 247:429-37. [PMID: 23333565 PMCID: PMC3636186 DOI: 10.1016/j.expneurol.2013.01.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 01/09/2013] [Indexed: 11/19/2022]
Abstract
Resected epileptic tissues exhibit elements of chronic neuroinflammation that include elevated TNFα and increased TNFα receptor activation, but the seizure related consequences of chronic TNFα expression remain unknown. Twenty four hours after acute limbic seizures the rat hippocampus exhibited a rapid upregulation of TNFR1, but a simultaneous downregulation of TNFR2. These limbic seizures also evoked significant increases in measures of neuroinflammation and caused significant neuronal cell death in both the hilus and CA3 of the hippocampus. In order to mimic a state of chronic TNFα exposure, adeno-associated viral vectors were packaged with a TNF receptor 1 (TNFR1) specific agonist, human TNFα, or a TNF receptor 1/2 agonist, rat TNFα. Subsequently, chronic hippocampal overexpression of either TNFR ligand caused microglial activation and blood-brain barrier compromise, a pattern similar to limbic seizure-induced neuroinflammation. However, no evidence was found for neuronal cell death or spontaneous seizure activity. Thus, chronic, in vivo TNFα expression and the subsequent neuroinflammation alone did not cause cell death or elicit seizure activity. In contrast, chronic hippocampal activation of TNFR1 alone significantly increased limbic seizure sensitivity in both amygdala kainic acid and electrical amygdala kindling models, while chronic activation of both TNFR1 and TNFR2 significantly attenuated the amygdala kindling rate. With regard to endogenous TNFα, chronic hippocampal expression of a TNFα decoy receptor significantly reduced seizure-induced cell death in the hippocampus, but did not alter seizure susceptibility. These findings suggest that blockade of endogenous TNFα could attenuate seizure related neuropathology, while selective activation of TNFR2 could exert beneficial therapeutic effects on in vivo seizure sensitivity.
Collapse
Affiliation(s)
- Marc S. Weinberg
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Bonita L. Blake
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Thomas J. McCown
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
350
|
Oversized AAV transductifon is mediated via a DNA-PKcs-independent, Rad51C-dependent repair pathway. Mol Ther 2013; 21:2205-16. [PMID: 23939025 DOI: 10.1038/mt.2013.184] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 07/27/2013] [Indexed: 12/25/2022] Open
Abstract
A drawback of gene therapy using adeno-associated virus (AAV) is the DNA packaging restriction of the viral capsid (<4.7 kb). Recent observations demonstrate oversized AAV genome transduction through an unknown mechanism. Herein, AAV production using an oversized reporter (6.2 kb) resulted in chloroform and DNase-resistant particles harboring distinct "fragment" AAV (fAAV) genomes (5.0, 2.4, and 1.6 kb). Fractionation experiments determined that only the larger "fragments" mediated transduction in vitro, and relatively efficient transduction was also demonstrated in the muscle, the eye, and the liver. In contrast with concatemerization-dependent large-gene delivery by split AAV, fAAV transduction is independent of the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) in vitro and in vivo while disproportionately reliant on the DNA strand-annealing protein Rad51C. Importantly, fAAV's unique dependence on DNA repair proteins, compared with intact AAV, strongly suggests that the majority of oversized AAV transduction is mediated by fragmented genomes. Although fAAV transduction is less efficient than intact AAV, it is enhanced fourfold in muscle and sevenfold in the retina compared with split AAV transduction. Furthermore, fAAV carrying codon-optimized therapeutic dysferlin cDNA in a 7.5 kb expression cassette restored dysferlin levels in a dystrophic model. Collectively, oversized AAV genome transduction requires unique DNA repair pathways and offers an alternative, more efficient strategy for large-gene therapy.
Collapse
|