701
|
RNA activation of haploinsufficient Foxg1 gene in murine neocortex. Sci Rep 2016; 6:39311. [PMID: 27995975 PMCID: PMC5172352 DOI: 10.1038/srep39311] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 11/22/2016] [Indexed: 11/18/2022] Open
Abstract
More than one hundred distinct gene hemizygosities are specifically linked to epilepsy, mental retardation, autism, schizophrenia and neuro-degeneration. Radical repair of these gene deficits via genome engineering is hardly feasible. The same applies to therapeutic stimulation of the spared allele by artificial transactivators. Small activating RNAs (saRNAs) offer an alternative, appealing approach. As a proof-of-principle, here we tested this approach on the Rett syndrome-linked, haploinsufficient, Foxg1 brain patterning gene. We selected a set of artificial small activating RNAs (saRNAs) upregulating it in neocortical precursors and their derivatives. Expression of these effectors achieved a robust biological outcome. saRNA-driven activation (RNAa) was limited to neural cells which normally express Foxg1 and did not hide endogenous gene tuning. saRNAs recognized target chromatin through a ncRNA stemming from it. Gene upregulation required Ago1 and was associated to RNApolII enrichment throughout the Foxg1 locus. Finally, saRNA delivery to murine neonatal brain replicated Foxg1-RNAa in vivo.
Collapse
|
702
|
Dashkoff J, Lerner EP, Truong N, Klickstein JA, Fan Z, Mu D, Maguire CA, Hyman BT, Hudry E. Tailored transgene expression to specific cell types in the central nervous system after peripheral injection with AAV9. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16081. [PMID: 27933308 PMCID: PMC5142512 DOI: 10.1038/mtm.2016.81] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/22/2016] [Accepted: 10/24/2016] [Indexed: 01/06/2023]
Abstract
The capacity of certain adeno-associated virus (AAV) vectors to cross the blood–brain barrier after intravenous delivery offers a unique opportunity for noninvasive brain delivery. However, without a well-tailored system, the use of a peripheral route injection may lead to undesirable transgene expression in nontarget cells or organs. To refine this approach, the present study characterizes the transduction profiles of new self-complementary AAV9 (scAAV9) expressing the green fluorescent protein (GFP) either under an astrocyte (glial fibrillary acidic (GFA) protein) or neuronal (Synapsin (Syn)) promoter, after intravenous injection of adult mice (2 × 1013 vg/kg). ScAAV9-GFA-GFP and scAAV9-Syn-GFP robustly transduce astrocytes (11%) and neurons (17%), respectively, without aberrant expression leakage. Interestingly, while the percentages of GFP-positive astrocytes with scAAV9-GFA-GFP are similar to the performances observed with scAAV9-CBA-GFP (broadly active promoter), significant higher percentages of neurons express GFP with scAAV9-Syn-GFP. GFP-positive excitatory as well as inhibitory neurons are observed, as well as motor neurons in the spinal cord. Additionally, both activated (GFAP-positive) and resting astrocytes (GFAP-negative) express the reporter gene after scAAV9-GFA-GFP injection. These data thoroughly characterize the gene expression specificity of AAVs fitted with neuronal and astrocyte-selective promoters after intravenous delivery, which will prove useful for central nervous system (CNS) gene therapy approaches in which peripheral expression of transgene is a concern.
Collapse
Affiliation(s)
- Jonathan Dashkoff
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, USA; MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Eli P Lerner
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School , Charlestown, Massachusetts, USA
| | - Nhi Truong
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School , Charlestown, Massachusetts, USA
| | - Jacob A Klickstein
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School , Charlestown, Massachusetts, USA
| | - Zhanyun Fan
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School , Charlestown, Massachusetts, USA
| | - Dakai Mu
- Department of Neurology, The Massachusetts General Hospital, and NeuroDiscovery Center, Harvard Medical School , Boston, Massachusetts, USA
| | - Casey A Maguire
- Department of Neurology, The Massachusetts General Hospital, and NeuroDiscovery Center, Harvard Medical School , Boston, Massachusetts, USA
| | - Bradley T Hyman
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School , Charlestown, Massachusetts, USA
| | - Eloise Hudry
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School , Charlestown, Massachusetts, USA
| |
Collapse
|
703
|
Abstract
Microscopy continues to expand our knowledge of brain structure and function. Sarah Webb looks at some of the latest tools and techniques leading the charge.
Collapse
|
704
|
Systemic application of AAV vectors targeting GFAP-expressing astrocytes in Z -Q175-KI Huntington's disease mice. Mol Cell Neurosci 2016; 77:76-86. [DOI: 10.1016/j.mcn.2016.10.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 09/12/2016] [Accepted: 10/24/2016] [Indexed: 01/03/2023] Open
|
705
|
Fu H, McCarty DM. Crossing the blood–brain-barrier with viral vectors. Curr Opin Virol 2016; 21:87-92. [DOI: 10.1016/j.coviro.2016.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 08/08/2016] [Accepted: 08/09/2016] [Indexed: 02/05/2023]
|
706
|
Gustincich S, Zucchelli S, Mallamaci A. The Yin and Yang of nucleic acid-based therapy in the brain. Prog Neurobiol 2016; 155:194-211. [PMID: 27887908 DOI: 10.1016/j.pneurobio.2016.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 11/16/2016] [Accepted: 11/20/2016] [Indexed: 02/06/2023]
Abstract
The post-genomic era has unveiled the existence of a large repertory of non-coding RNAs and repetitive elements that play a fundamental role in cellular homeostasis and dysfunction. These may represent unprecedented opportunities to modify gene expression at the right time in the correct space in vivo, providing an almost unlimited reservoir of new potential pharmacological agents. Hijacking their mode of actions, the druggable genome can be extended to regulatory RNAs and DNA elements in a scalable fashion. Here, we discuss the state-of-the-art of nucleic acid-based drugs to treat neurodegenerative diseases. Beneficial effects can be obtained by inhibiting (Yin) and increasing (Yang) gene expression, depending on the disease and the drug target. Together with the description of the current use of inhibitory RNAs (small inhibitory RNAs and antisense oligonucleotides) in animal models and clinical trials, we discuss the molecular basis and applications of new classes of activatory RNAs at transcriptional (RNAa) and translational (SINEUP) levels.
Collapse
Affiliation(s)
- Stefano Gustincich
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT), Genova, Italy; Area of Neuroscience, SISSA, Trieste, Italy.
| | - Silvia Zucchelli
- Area of Neuroscience, SISSA, Trieste, Italy; Department of Health Sciences, Universita' del Piemonte Orientale, Novara, Italy
| | | |
Collapse
|
707
|
Dissen GA, Adachi K, Lomniczi A, Chatkupt T, Davidson BL, Nakai H, Ojeda SR. Engineering a gene silencing viral construct that targets the cat hypothalamus to induce permanent sterility: An update. Reprod Domest Anim 2016; 52 Suppl 2:354-358. [PMID: 27859771 DOI: 10.1111/rda.12834] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The intent of this contribution is to provide an update of the progress we have made towards developing a method/treatment to permanently sterilize cats. Our approach employs two complementary methodologies: RNA interference (RNAi) to silence genes involved in the central control of reproduction and a virus-based gene therapy system intended to deliver RNAi selectively to the hypothalamus (where these genes are expressed) via the systemic administration of modified viruses. We selected the hypothalamus because it contains neurons expressing Kiss1 and Tac3, two genes essential for reproduction and fertility. We chose the non-pathogenic adeno-associated virus (AAV) as a vector whose tropism could be modified to target the hypothalamus. The issues that must be overcome to utilize this vector as a delivery vehicle to induce sterility include modification of the wild-type AAV to target the hypothalamic region of the brain with a simultaneous reduction in targeting of peripheral tissues and non-hypothalamic brain regions, identification of RNAi targets that will effectively reduce the expression of Kiss1 and Tac3 without off-target effects, and determination if neutralizing antibodies to the AAV serotype of choice are present in cats. Successful resolution of these issues will pave the way for the development of a powerful tool to induce the permanent sterility in cats.
Collapse
Affiliation(s)
- G A Dissen
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - K Adachi
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - A Lomniczi
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - T Chatkupt
- Department of Comparative Medicine, Oregon Health and Science University, Portland, OR, USA
| | - B L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - H Nakai
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA.,Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - S R Ojeda
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| |
Collapse
|
708
|
Chou YY, Krupp A, Kaynor C, Gaudin R, Ma M, Cahir-McFarland E, Kirchhausen T. Inhibition of JCPyV infection mediated by targeted viral genome editing using CRISPR/Cas9. Sci Rep 2016; 6:36921. [PMID: 27841295 PMCID: PMC5107994 DOI: 10.1038/srep36921] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 10/21/2016] [Indexed: 12/16/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a debilitating disease resulting from infection of oligodendrocytes by the JC polyomavirus (JCPyV). Currently, there is no anti-viral therapeutic available against JCPyV infection. The clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) system (CRISPR/Cas9) is a genome editing tool capable of introducing sequence specific breaks in double stranded DNA. Here we show that the CRISPR/Cas9 system can restrict the JCPyV life cycle in cultured cells. We utilized CRISPR/Cas9 to target the noncoding control region and the late gene open reading frame of the JCPyV genome. We found significant inhibition of virus replication and viral protein expression in cells recipient of Cas9 together with JCPyV-specific single-guide RNA delivered prior to or after JCPyV infection.
Collapse
Affiliation(s)
- Yi-ying Chou
- Department of Cell Biology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Annabel Krupp
- Department of Neuroimmunology, Biogen, Cambridge, MA 02142, USA
| | - Campbell Kaynor
- Department of Neuroimmunology, Biogen, Cambridge, MA 02142, USA
| | - Raphaël Gaudin
- Department of Cell Biology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Minghe Ma
- Department of Cell Biology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | - Tom Kirchhausen
- Department of Cell Biology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
709
|
Jackson KL, Dayton RD, Deverman BE, Klein RL. Better Targeting, Better Efficiency for Wide-Scale Neuronal Transduction with the Synapsin Promoter and AAV-PHP.B. Front Mol Neurosci 2016; 9:116. [PMID: 27867348 PMCID: PMC5095393 DOI: 10.3389/fnmol.2016.00116] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/19/2016] [Indexed: 11/13/2022] Open
Abstract
Widespread genetic modification of cells in the central nervous system (CNS) with a viral vector has become possible and increasingly more efficient. We previously applied an AAV9 vector with the cytomegalovirus/chicken beta-actin (CBA) hybrid promoter and achieved wide-scale CNS transduction in neonatal and adult rats. However, this method transduces a variety of tissues in addition to the CNS. Thus we studied intravenous AAV9 gene transfer with a synapsin promoter to better target the neurons. We noted in systematic comparisons that the synapsin promoter drives lower level expression than does the CBA promoter. The engineered adeno-associated virus (AAV)-PHP.B serotype was compared with AAV9, and AAV-PHP.B did enhance the efficiency of expression. Combining the synapsin promoter with AAV-PHP.B could therefore be advantageous in terms of combining two refinements of targeting and efficiency. Wide-scale expression was used to model a disease with widespread pathology. Vectors encoding the amyotrophic lateral sclerosis (ALS)-related protein transactive response DNA-binding protein, 43 kDa (TDP-43) with the synapsin promoter and AAV-PHP.B were used for efficient CNS-targeted TDP-43 expression. Intracerebroventricular injections were also explored to limit TDP-43 expression to the CNS. The neuron-selective promoter and the AAV-PHP.B enhanced gene transfer and ALS disease modeling in adult rats.
Collapse
Affiliation(s)
- Kasey L Jackson
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center Shreveport, LA, USA
| | - Robert D Dayton
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center Shreveport, LA, USA
| | - Benjamin E Deverman
- Division of Biology and Biological Engineering, California Institute of Technology Pasadena, CA, USA
| | - Ronald L Klein
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center Shreveport, LA, USA
| |
Collapse
|
710
|
Jackson KL, Dhaibar HA, Dayton RD, Cananzi SG, Mayhan WG, Glasscock E, Klein RL. Severe respiratory changes at end stage in a FUS-induced disease state in adult rats. BMC Neurosci 2016; 17:69. [PMID: 27793099 PMCID: PMC5086065 DOI: 10.1186/s12868-016-0304-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/19/2016] [Indexed: 12/12/2022] Open
Abstract
Background Fused in sarcoma (FUS) is an RNA-binding protein associated with the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration. ALS manifests in patients as a progressive paralysis which leads to respiratory dysfunction and failure, the primary cause of death in ALS. We expressed human FUS in rats to determine if FUS would induce ALS relevant respiratory changes to serve as an early stage disease indicator. The FUS expression was initiated in adult rats by way of an intravenously administered adeno-associated virus vector serotype 9 (AAV9) providing an adult onset model. Results The rats developed progressive motor impairments observed as early as 2–3 weeks post gene transfer. Respiratory abnormalities manifested 4–7 weeks post gene transfer including increased respiratory frequency and decreased tidal volume. Rats with breathing abnormalities also had arterial blood acidosis. Similar detailed plethysmographic changes were found in adult rats injected with AAV9 TDP-43. FUS gene transfer to adult rats yielded a consistent pre-clinical model with relevant motor paralysis in the early to middle stages and respiratory dysfunction at the end stage. Both FUS and TDP-43 yielded a similar consistent disease state. Conclusions This modeling method yields disease relevant motor and respiratory changes in adult rats. The reproducibility of the data supports the use of this method to study other disease related genes and their combinations as well as a platform for disease modifying interventional strategies.
Collapse
Affiliation(s)
- Kasey L Jackson
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, LA, 71130, USA.
| | - Hemangini A Dhaibar
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, LA, 71130, USA
| | - Robert D Dayton
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, LA, 71130, USA
| | - Sergio G Cananzi
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, LA, 71130, USA
| | - William G Mayhan
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, LA, 71130, USA
| | - Edward Glasscock
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, LA, 71130, USA
| | - Ronald L Klein
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, LA, 71130, USA
| |
Collapse
|
711
|
Sjulson L, Cassataro D, DasGupta S, Miesenböck G. Cell-Specific Targeting of Genetically Encoded Tools for Neuroscience. Annu Rev Genet 2016; 50:571-594. [PMID: 27732792 DOI: 10.1146/annurev-genet-120215-035011] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genetically encoded tools for visualizing and manipulating neurons in vivo have led to significant advances in neuroscience, in large part because of the ability to target expression to specific cell populations of interest. Current methods enable targeting based on marker gene expression, development, anatomical projection pattern, synaptic connectivity, and recent activity as well as combinations of these factors. Here, we review these methods, focusing on issues of practical implementation as well as areas for future improvement.
Collapse
Affiliation(s)
- Lucas Sjulson
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016; .,Department of Neuroscience and Physiology, Smilow Neuroscience Program, and New York University Neuroscience Institute, New York, NY 10016
| | - Daniela Cassataro
- Department of Neuroscience and Physiology, Smilow Neuroscience Program, and New York University Neuroscience Institute, New York, NY 10016
| | - Shamik DasGupta
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, OX1 3SR, United Kingdom; .,Present address: Tata Institute of Fundamental Research, Mumbai, 400005, India
| | - Gero Miesenböck
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, OX1 3SR, United Kingdom;
| |
Collapse
|
712
|
Buckinx R, Timmermans JP. Targeting the gastrointestinal tract with viral vectors: state of the art and possible applications in research and therapy. Histochem Cell Biol 2016; 146:709-720. [PMID: 27665281 DOI: 10.1007/s00418-016-1496-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2016] [Indexed: 12/11/2022]
Abstract
While there is a large body of preclinical data on the use of viral vectors in gene transfer, relatively little is known about viral gene transfer in the gastrointestinal tract. Viral vector technology is especially underused in the field of neurogastroenterology when compared to brain research. This review provides an overview of the studies employing viral vectors-in particular retroviruses, adenoviruses and adeno-associated viruses-to transduce different cell types in the intestine. Early work mainly focused on mucosal transduction, but had limited success due to the harsh luminal conditions in the gastrointestinal tract and the high turnover rate of enterocytes. More recently, several studies have successfully employed viral gene transfer to target the enteric nervous system and its progenitors. Although several hurdles still need to be overcome, in particular on how to augment transduction efficiency and specific cell targeting, viral vector technology holds strong potential not only as a valid research tool in fundamental gastroenterological research but also as a therapeutic agent in translational (bio)medical research.
Collapse
Affiliation(s)
- Roeland Buckinx
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
713
|
Yamazaki Y, Painter MM, Bu G, Kanekiyo T. Apolipoprotein E as a Therapeutic Target in Alzheimer's Disease: A Review of Basic Research and Clinical Evidence. CNS Drugs 2016; 30:773-89. [PMID: 27328687 PMCID: PMC5526196 DOI: 10.1007/s40263-016-0361-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that causes progressive cognitive decline. The majority of AD cases are sporadic and late-onset (>65 years old) making it the leading cause of dementia in the elderly. While both genetic and environmental factors contribute to the development of late-onset AD (LOAD), APOE polymorphism is a major genetic risk determinant for LOAD. In humans, the APOE gene has three major allelic variants: ε2, ε3, and ε4, of which APOE ε4 is the strongest genetic risk factor for LOAD, whereas APOE ε2 is protective. Mounting evidence suggests that APOE ε4 contributes to AD pathogenesis through multiple pathways including facilitated amyloid-β deposition, increased tangle formation, synaptic dysfunction, exacerbated neuroinflammation, and cerebrovascular defects. Since APOE modulates multiple biological processes through its corresponding protein apolipoprotein E (apoE), APOE gene and apoE properties have been a promising target for therapy and drug development against AD. In this review, we summarize the current evidence regarding how the APOE ε4 allele contributes to the pathogenesis of AD and how relevant therapeutic approaches can be developed to target apoE-mediated pathways in AD.
Collapse
Affiliation(s)
- Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Meghan M Painter
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
714
|
van Zundert B, Brown RH. Silencing strategies for therapy of SOD1-mediated ALS. Neurosci Lett 2016; 636:32-39. [PMID: 27507699 DOI: 10.1016/j.neulet.2016.07.059] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset, lethal, paralytic disorder caused by the degeneration of motor neurons. Our understanding of this disease has been greatly facilitated by studies of familial ALS caused by mutations in the gene encoding superoxide dismutase 1 (SOD1). Evidence indicates that misfolded wild-type SOD1 may also be pathogenic in sporadic ALS. Mutant SOD1 is neurotoxic through multiple mechanisms. Because the pathogenicity of mutant SOD1 is proportional to the dose of the toxic protein, a rational approach to treating SOD1-related ALS is to reduce levels of the toxic SOD1 species. An advantage of this strategy is that it potentially obviates intervening in multiple, downstream pathological cascades. In recent years, several strategies to silence gene expression have been developed. The most clinically promising are predicated on approaches that enhance degradation of RNA, such as anti-sense oligonucleotides (ASO) and RNA interference (RNAi); the latter include small inhibitory RNA (siRNA), short hairpin RNA (shRNA) and microRNA (miR). Agents such as shRNA and either native or synthetic miR are capable of permeating the central nervous system (CNS) and efficiently silencing genes in the brain and spinal cord. Here we review recent progress in silencing SOD1, focusing on studies using artificial shRNA or miRNA in combination with potent viral vector delivery systems to mediate SOD1 silencing within the CNS in transgenic SOD1G93A mice and non-human primates.
Collapse
Affiliation(s)
- Brigitte van Zundert
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Avenida República 217, Santiago, Chile.
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01655, USA.
| |
Collapse
|
715
|
Stoica L, Sena-Esteves M. Adeno Associated Viral Vector Delivered RNAi for Gene Therapy of SOD1 Amyotrophic Lateral Sclerosis. Front Mol Neurosci 2016; 9:56. [PMID: 27531973 PMCID: PMC4969298 DOI: 10.3389/fnmol.2016.00056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/04/2016] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease caused by progressive loss of upper and lower motor neurons. Mutations in superoxide dismutase 1 (SOD1) are a leading cause of ALS, responsible for up to 20% of familial cases. Although the exact mechanism by which mutant SOD1 causes disease remains unknown, multiple studies have shown that reduction of the mutant species leads to delayed disease onset and extension of lifespan of animal models. This makes SOD1 an ideal target for gene therapy coupling adeno associated virus vector (AAV) gene delivery with RNAi molecules. In this review we summarize the studies done thus far attempting to decrease SOD1 gene expression, using AAV vectors as delivery tools, and RNAi as therapeutic molecules. Current hurdles to be overcome, such as the need for widespread gene delivery through the entire central nervous system (CNS), are discussed. Continued efforts to improve current AAV delivery methods and capsids will accelerate the application of these therapeutics to the clinic.
Collapse
Affiliation(s)
- Lorelei Stoica
- Gene Therapy Center, University of Massachusetts Medical SchoolWorcester, MA, USA; Department of Neurology, University of Massachusetts Medical SchoolWorcester, MA, USA
| | - Miguel Sena-Esteves
- Gene Therapy Center, University of Massachusetts Medical SchoolWorcester, MA, USA; Department of Neurology, University of Massachusetts Medical SchoolWorcester, MA, USA
| |
Collapse
|
716
|
Choudhury SR, Fitzpatrick Z, Harris AF, Maitland SA, Ferreira JS, Zhang Y, Ma S, Sharma RB, Gray-Edwards HL, Johnson JA, Johnson AK, Alonso LC, Punzo C, Wagner KR, Maguire CA, Kotin RM, Martin DR, Sena-Esteves M. In Vivo Selection Yields AAV-B1 Capsid for Central Nervous System and Muscle Gene Therapy. Mol Ther 2016; 24:1247-57. [PMID: 27117222 PMCID: PMC5088762 DOI: 10.1038/mt.2016.84] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/12/2016] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated viral (AAV) vectors have shown promise as a platform for gene therapy of neurological disorders. Achieving global gene delivery to the central nervous system (CNS) is key for development of effective therapies for many of these diseases. Here we report the isolation of a novel CNS tropic AAV capsid, AAV-B1, after a single round of in vivo selection from an AAV capsid library. Systemic injection of AAV-B1 vector in adult mice and cat resulted in widespread gene transfer throughout the CNS with transduction of multiple neuronal subpopulations. In addition, AAV-B1 transduces muscle, β-cells, pulmonary alveoli, and retinal vasculature at high efficiency. This vector is more efficient than AAV9 for gene delivery to mouse brain, spinal cord, muscle, pancreas, and lung. Together with reduced sensitivity to neutralization by antibodies in pooled human sera, the broad transduction profile of AAV-B1 represents an important improvement over AAV9 for CNS gene therapy.
Collapse
Affiliation(s)
- Sourav R Choudhury
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Zachary Fitzpatrick
- Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Anne F Harris
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Stacy A Maitland
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jennifer S Ferreira
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Yuanfan Zhang
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute and Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Shan Ma
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Ophthalmology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Rohit B Sharma
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Heather L Gray-Edwards
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Jacob A Johnson
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Aime K Johnson
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Laura C Alonso
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Claudio Punzo
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Ophthalmology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kathryn R Wagner
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute and Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Casey A Maguire
- Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert M Kotin
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Voyager Therapeutics, Cambridge, Massachusetts, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Miguel Sena-Esteves
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
717
|
Jackson KL, Dayton RD, Klein RL. Gene vector ‘magic bullet’: targeted expression in the central nervous system after peripheral delivery using the synapsin promoter. Expert Opin Ther Targets 2016; 20:1153-4. [DOI: 10.1080/14728222.2016.1212016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Kasey L. Jackson
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Robert D. Dayton
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Ronald L. Klein
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
718
|
Treweek JB, Gradinaru V. Extracting structural and functional features of widely distributed biological circuits with single cell resolution via tissue clearing and delivery vectors. Curr Opin Biotechnol 2016; 40:193-207. [PMID: 27393829 DOI: 10.1016/j.copbio.2016.03.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 03/10/2016] [Accepted: 03/15/2016] [Indexed: 12/13/2022]
Abstract
The scientific community has learned a great deal from imaging small and naturally transparent organisms such as nematodes and zebrafish. The consequences of genetic mutations on their organ development and survival can be visualized easily and with high-throughput at the organism-wide scale. In contrast, three-dimensional information is less accessible in mammalian subjects because the heterogeneity of light-scattering tissue elements renders their organs opaque. Likewise, genetically labeling desired circuits across mammalian bodies is prohibitively slow and costly via the transgenic route. Emerging breakthroughs in viral vector engineering, genome editing tools, and tissue clearing can render larger opaque organisms genetically tractable and transparent for whole-organ cell phenotyping, tract tracing and imaging at depth.
Collapse
Affiliation(s)
- Jennifer Brooke Treweek
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
719
|
Hocquemiller M, Giersch L, Audrain M, Parker S, Cartier N. Adeno-Associated Virus-Based Gene Therapy for CNS Diseases. Hum Gene Ther 2016; 27:478-96. [PMID: 27267688 PMCID: PMC4960479 DOI: 10.1089/hum.2016.087] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 06/07/2016] [Indexed: 12/11/2022] Open
Abstract
Gene therapy is at the cusp of a revolution for treating a large spectrum of CNS disorders by providing a durable therapeutic protein via a single administration. Adeno-associated virus (AAV)-mediated gene transfer is of particular interest as a therapeutic tool because of its safety profile and efficiency in transducing a wide range of cell types. The purpose of this review is to describe the most notable advancements in preclinical and clinical research on AAV-based CNS gene therapy and to discuss prospects for future development based on a new generation of vectors and delivery.
Collapse
Affiliation(s)
| | | | - Mickael Audrain
- Université Paris Descartes, Paris, France
- INSERM UMR1169, Université Paris-Sud,Université Paris-Saclay, Orsay, France
- CEA, DSV, IBM, MIRCen, Fontenay-aux-Roses, France
| | | | - Nathalie Cartier
- INSERM UMR1169, Université Paris-Sud,Université Paris-Saclay, Orsay, France
- CEA, DSV, IBM, MIRCen, Fontenay-aux-Roses, France
| |
Collapse
|
720
|
Lee H, Oh WC, Seong J, Kim J. Advanced Fluorescence Protein-Based Synapse-Detectors. Front Synaptic Neurosci 2016; 8:16. [PMID: 27445785 PMCID: PMC4927625 DOI: 10.3389/fnsyn.2016.00016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/13/2016] [Indexed: 11/13/2022] Open
Abstract
The complex information-processing capabilities of the central nervous system emerge from intricate patterns of synaptic input-output relationships among various neuronal circuit components. Understanding these capabilities thus requires a precise description of the individual synapses that comprise neural networks. Recent advances in fluorescent protein engineering, along with developments in light-favoring tissue clearing and optical imaging techniques, have rendered light microscopy (LM) a potent candidate for large-scale analyses of synapses, their properties, and their connectivity. Optically imaging newly engineered fluorescent proteins (FPs) tagged to synaptic proteins or microstructures enables the efficient, fine-resolution illumination of synaptic anatomy and function in large neural circuits. Here we review the latest progress in fluorescent protein-based molecular tools for imaging individual synapses and synaptic connectivity. We also identify associated technologies in gene delivery, tissue processing, and computational image analysis that will play a crucial role in bridging the gap between synapse- and system-level neuroscience.
Collapse
Affiliation(s)
- Hojin Lee
- Center for Functional Connectomics, Korea Institute of Science and TechnologySeoul, South Korea; Neuroscience Program, Korea University of Science and TechnologyDaejeon, South Korea
| | - Won Chan Oh
- Center for Functional Connectomics, Korea Institute of Science and Technology Seoul, South Korea
| | - Jihye Seong
- Neuroscience Program, Korea University of Science and TechnologyDaejeon, South Korea; Center for Diagnosis Treatment Care of Dementia, Korea Institute of Science and TechnologySeoul, South Korea
| | - Jinhyun Kim
- Center for Functional Connectomics, Korea Institute of Science and TechnologySeoul, South Korea; Neuroscience Program, Korea University of Science and TechnologyDaejeon, South Korea
| |
Collapse
|
721
|
Körbelin J, Dogbevia G, Michelfelder S, Ridder DA, Hunger A, Wenzel J, Seismann H, Lampe M, Bannach J, Pasparakis M, Kleinschmidt JA, Schwaninger M, Trepel M. A brain microvasculature endothelial cell-specific viral vector with the potential to treat neurovascular and neurological diseases. EMBO Mol Med 2016; 8:609-25. [PMID: 27137490 PMCID: PMC4888852 DOI: 10.15252/emmm.201506078] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Gene therapy critically relies on vectors that combine high transduction efficiency with a high degree of target specificity and that can be administered through a safe intravenous route. The lack of suitable vectors, especially for gene therapy of brain disorders, represents a major obstacle. Therefore, we applied an in vivo screening system of random ligand libraries displayed on adeno‐associated viral capsids to select brain‐targeted vectors for the treatment of neurovascular diseases. We identified a capsid variant showing an unprecedented degree of specificity and long‐lasting transduction efficiency for brain microvasculature endothelial cells as the primary target of selection. A therapeutic vector based on this selected viral capsid was used to markedly attenuate the severe cerebrovascular pathology of mice with incontinentia pigmenti after a single intravenous injection. Furthermore, the versatility of this selection system will make it possible to select ligands for additional in vivo targets without requiring previous identification of potential target‐specific receptors.
Collapse
Affiliation(s)
- Jakob Körbelin
- Hubertus Wald Cancer Center, Department of Oncology and Hematology University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Godwin Dogbevia
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Stefan Michelfelder
- Hubertus Wald Cancer Center, Department of Oncology and Hematology University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dirk A Ridder
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Agnes Hunger
- Hubertus Wald Cancer Center, Department of Oncology and Hematology University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Wenzel
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Henning Seismann
- Hubertus Wald Cancer Center, Department of Oncology and Hematology University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Lampe
- Hubertus Wald Cancer Center, Department of Oncology and Hematology University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jacqueline Bannach
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Manolis Pasparakis
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Centre for Molecular Medicine (CMMC), Institute for Genetics University of Cologne, Cologne, Germany
| | | | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Martin Trepel
- Hubertus Wald Cancer Center, Department of Oncology and Hematology University Medical Center Hamburg-Eppendorf, Hamburg, Germany Department of Hematology and Oncology, Augsburg Medical Center, Augsburg, Germany
| |
Collapse
|
722
|
Pulmonary Targeting of Adeno-associated Viral Vectors by Next-generation Sequencing-guided Screening of Random Capsid Displayed Peptide Libraries. Mol Ther 2016; 24:1050-1061. [PMID: 27018516 DOI: 10.1038/mt.2016.62] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/16/2016] [Indexed: 12/31/2022] Open
Abstract
Vectors mediating strong, durable, and tissue-specific transgene expression are mandatory for safe and effective gene therapy. In settings requiring systemic vector administration, the availability of suited vectors is extremely limited. Here, we present a strategy to select vectors with true specificity for a target tissue from random peptide libraries displayed on adeno-associated virus (AAV) by screening the library under circulation conditions in a murine model. Guiding the in vivo screening by next-generation sequencing, we were able to monitor the selection kinetics and to determine the right time point to discontinue the screening process. The establishment of different rating scores enabled us to identify the most specifically enriched AAV capsid candidates. As proof of concept, a capsid variant was selected that specifically and very efficiently delivers genes to the endothelium of the pulmonary vasculature after intravenous administration. This technical approach of selecting target-specific vectors in vivo is applicable to any given tissue of interest and therefore has broad implications in translational research and medicine.
Collapse
|