1
|
Kul E, Okoroafor U, Dougherty A, Palkovic L, Li H, Valiño-Ramos P, Aberman L, Young SM. Development of adenoviral vectors that transduce Purkinje cells and other cerebellar cell-types in the cerebellum of a humanized mouse model. Mol Ther Methods Clin Dev 2024; 32:101243. [PMID: 38605812 PMCID: PMC11007541 DOI: 10.1016/j.omtm.2024.101243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 03/22/2024] [Indexed: 04/13/2024]
Abstract
Viral vector gene therapy has immense promise for treating central nervous system (CNS) disorders. Although adeno-associated virus vectors (AAVs) have had success, their small packaging capacity limits their utility to treat the root cause of many CNS disorders. Adenoviral vectors (Ad) have tremendous potential for CNS gene therapy approaches. Currently, the most common vectors utilize the Group C Ad5 serotype capsid proteins, which rely on the Coxsackievirus-Adenovirus receptor (CAR) to infect cells. However, these Ad5 vectors are unable to transduce many neuronal cell types that are dysfunctional in many CNS disorders. The human CD46 (hCD46) receptor is widely expressed throughout the human CNS and is the primary attachment receptor for many Ad serotypes. Therefore, to overcome the current limitations of Ad vectors to treat CNS disorders, we created chimeric first generation Ad vectors that utilize the hCD46 receptor. Using a "humanized" hCD46 mouse model, we demonstrate these Ad vectors transduce cerebellar cell types, including Purkinje cells, that are refractory to Ad5 transduction. Since Ad vector transduction properties are dependent on their capsid proteins, these chimeric first generation Ad vectors open new avenues for high-capacity helper-dependent adenovirus (HdAd) gene therapy approaches for cerebellar disorders and multiple neurological disorders.
Collapse
Affiliation(s)
- Emre Kul
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Uchechi Okoroafor
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
- Cell Developmental Biology Graduate Program, University of Iowa, Iowa City, IA 52242, USA
| | - Amanda Dougherty
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Lauren Palkovic
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Hao Li
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Paula Valiño-Ramos
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Leah Aberman
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Samuel M. Young
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
- Cell Developmental Biology Graduate Program, University of Iowa, Iowa City, IA 52242, USA
- Department of Otolaryngology, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
2
|
Ingusci S, Hall BL, Goins WF, Cohen JB, Glorioso JC. Viral vectors for gene delivery to the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:59-81. [PMID: 39341663 DOI: 10.1016/b978-0-323-90120-8.00001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Brain diseases with a known or suspected genetic basis represent an important frontier for advanced therapeutics. The central nervous system (CNS) is an intricate network in which diverse cell types with multiple functions communicate via complex signaling pathways, making therapeutic intervention in brain-related diseases challenging. Nevertheless, as more information on the molecular genetics of brain-related diseases becomes available, genetic intervention using gene therapeutic strategies should become more feasible. There remain, however, several significant hurdles to overcome that relate to (i) the development of appropriate gene vectors and (ii) methods to achieve local or broad vector delivery. Clearly, gene delivery tools must be engineered for distribution to the correct cell type in a specific brain region and to accomplish therapeutic transgene expression at an appropriate level and duration. They also must avoid all toxicity, including the induction of inflammatory responses. Over the last 40 years, various types of viral vectors have been developed as tools to introduce therapeutic genes into the brain, primarily targeting neurons. This review describes the most prominent vector systems currently approaching clinical application for CNS disorders and highlights both remaining challenges as well as improvements in vector designs that achieve greater safety, defined tropism, and therapeutic gene expression.
Collapse
Affiliation(s)
- Selene Ingusci
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bonnie L Hall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - William F Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Justus B Cohen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
3
|
Wang J, Zhu M, Sun J, Feng L, Yang M, Sun B, Mao L. Gene therapy of adeno-associated virus (AAV) vectors in preclinical models of ischemic stroke. CNS Neurosci Ther 2023; 29:3725-3740. [PMID: 37551863 PMCID: PMC10651967 DOI: 10.1111/cns.14392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/15/2023] [Accepted: 07/27/2023] [Indexed: 08/09/2023] Open
Abstract
Stroke has been associated with devastating clinical outcomes, with current treatment strategies proving largely ineffective. Therefore, there is a need to explore alternative treatment options for addressing post-stroke functional deficits. Gene therapy utilizing adeno-associated viruses (AAVs) as a critical gene vector delivering genes to the central nervous system (CNS) gene delivery has emerged as a promising approach for treating various CNS diseases. This review aims to provide an overview of the biological characteristics of AAV vectors and the therapeutic advancements observed in preclinical models of ischemic stroke. The study further investigates the potential of manipulating AAV vectors in preclinical applications, emphasizing the challenges and prospects in the selection of viral vectors, drug delivery strategies, immune reactions, and clinical translation.
Collapse
Affiliation(s)
- Jing Wang
- Medical College of Qingdao UniversityQingdaoChina
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Mengna Zhu
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Jingyi Sun
- Department of Spinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Lina Feng
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Mingfeng Yang
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Baoliang Sun
- Medical College of Qingdao UniversityQingdaoChina
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Leilei Mao
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| |
Collapse
|
4
|
Matsunaga W, Gotoh A. Adenovirus as a Vector and Oncolytic Virus. Curr Issues Mol Biol 2023; 45:4826-4840. [PMID: 37367056 DOI: 10.3390/cimb45060307] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Adenoviral vectors, both oncolytic viruses and gene delivery vectors, are among the earliest approved and commercialised vectors for gene therapy. Adenoviruses have high cytotoxicity and immunogenicity. Therefore, lentiviruses or adeno-associated viruses as viral vectors and herpes simplex virus as an oncolytic virus have recently drawn attention. Thus, adenoviral vectors are often considered relatively obsolete. However, their high cargo limit and transduction efficiency are significant advantages over newer viral vectors. This review provides an overview of the new-generation adenoviral vectors. In addition, we describe the modification of the fiber knob region that enhances affinity of adenoviral vectors for cancer cells and the utilisation of cancer-cell-specific promoters to suppress expression of unwanted transgenes in non-malignant tissues.
Collapse
Affiliation(s)
- Wataru Matsunaga
- Joint-Use Research Facilities, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Japan
| | - Akinobu Gotoh
- Department of Education for Medical Research Base, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Japan
| |
Collapse
|
5
|
Mani S, Jindal D, Singh M. Gene Therapy, A Potential Therapeutic Tool for Neurological and Neuropsychiatric Disorders: Applications, Challenges and Future Perspective. Curr Gene Ther 2023; 23:20-40. [PMID: 35345999 DOI: 10.2174/1566523222666220328142427] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/18/2022] [Accepted: 02/02/2022] [Indexed: 02/08/2023]
Abstract
Neurological and neuropsychiatric disorders are the main risks for the health care system, exhibiting a huge socioeconomic load. The available range of pharmacotherapeutics mostly provides palliative consequences and fails to treat such conditions. The molecular etiology of various neurological and neuropsychiatric disorders is mostly associated with a change in genetic background, which can be inherited/triggered by other environmental factors. To address such conditions, gene therapy is considered a potential approach claiming a permanent cure of the disease primarily by deletion, silencing, or edition of faulty genes and by insertion of healthier genes. In gene therapy, vectors (viral/nonvial) play an important role in delivering the desired gene to a specific region of the brain. Targeted gene therapy has unraveled opportunities for the treatment of many neurological and neuropsychiatric disorders. For improved gene delivery, the current techniques mainly focus on designing a precise viral vector, plasmid transfection, nanotechnology, microRNA, and in vivo clustered regulatory interspaced short palindromic repeats (CRISPR)-based therapy. These latest techniques have great benefits in treating predominant neurological and neurodevelopmental disorders, including Parkinson's disease, Alzheimer's disease, and autism spectrum disorder, as well as rarer diseases. Nevertheless, all these delivery methods have their limitations, including immunogenic reactions, off-target effects, and a deficiency of effective biomarkers to appreciate the effectiveness of therapy. In this review, we present a summary of the current methods in targeted gene delivery, followed by the limitations and future direction of gene therapy for the cure of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Shalini Mani
- Department of Biotechnology, Centre for Emerging Diseases, Jaypee Institute of Information Technology, Noida, U.P., India
| | - Divya Jindal
- Department of Biotechnology, Centre for Emerging Diseases, Jaypee Institute of Information Technology, Noida, U.P., India
| | - Manisha Singh
- Department of Biotechnology, Centre for Emerging Diseases, Jaypee Institute of Information Technology, Noida, U.P., India
| |
Collapse
|
6
|
Eftekharpour E, Shcholok T. Cre-recombinase systems for induction of neuron-specific knockout models: a guide for biomedical researchers. Neural Regen Res 2023; 18:273-279. [PMID: 35900402 PMCID: PMC9396489 DOI: 10.4103/1673-5374.346541] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Gene deletion has been a valuable tool for unraveling the mysteries of molecular biology. Early approaches included gene trapping and gene targetting to disrupt or delete a gene randomly or at a specific location, respectively. Using these technologies in mouse embryos led to the generation of mouse knockout models and many scientific discoveries. The efficacy and specificity of these approaches have significantly increased with the advent of new technology such as clustered regularly interspaced short palindromic repeats for targetted gene deletion. However, several limitations including unwanted off-target gene deletion have hindered their widespread use in the field. Cre-recombinase technology has provided additional capacity for cell-specific gene deletion. In this review, we provide a summary of currently available literature on the application of this system for targetted deletion of neuronal genes. This article has been constructed to provide some background information for the new trainees on the mechanism and to provide necessary information for the design, and application of the Cre-recombinase system through reviewing the most frequent promoters that are currently available for genetic manipulation of neurons. We additionally will provide a summary of the latest technological developments that can be used for targeting neurons. This may also serve as a general guide for the selection of appropriate models for biomedical research.
Collapse
|
7
|
Tada T, Norton TD, Leibowitz R, Landau NR. Directly injected lentiviral vector-based T cell vaccine protects mice against acute and chronic viral infection. JCI Insight 2022; 7:161598. [PMID: 35972807 PMCID: PMC9675446 DOI: 10.1172/jci.insight.161598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/11/2022] [Indexed: 12/01/2022] Open
Abstract
Lentiviral vector–based dendritic cell vaccines induce protective T cell responses against viral infection and cancer in animal models. In this study, we tested whether preventative and therapeutic vaccination could be achieved by direct injection of antigen-expressing lentiviral vector, obviating the need for ex vivo transduction of dendritic cells. Injected lentiviral vector preferentially transduced splenic dendritic cells and resulted in long-term expression. Injection of a lentiviral vector encoding an MHC class I–restricted T cell epitope of lymphocytic choriomeningitis virus (LCMV) and CD40 ligand induced an antigen-specific cytolytic CD8+ T lymphocyte response that protected the mice from infection. The injection of chronically infected mice with a lentiviral vector encoding LCMV MHC class I and II T cell epitopes and a soluble programmed cell death 1 microbody rapidly cleared the virus. Vaccination by direct injection of lentiviral vector was more effective in sterile alpha motif and HD-domain containing protein 1–knockout (SAMHD1-knockout) mice, suggesting that lentiviral vectors containing Vpx, a lentiviral protein that increases the efficiency of dendritic cell transduction by inducing the degradation of SAMHD1, would be an effective strategy for the treatment of chronic disease in humans.
Collapse
Affiliation(s)
- Takuya Tada
- Department of Medicine, NYU Grossman School of Medicine, New York, United States of America
| | - Thomas D Norton
- Department of Medicine, NYU Grossman School of Medicine, New York, United States of America
| | - Rebecca Leibowitz
- Department of Microbiology, NYU Grossman School of Medicine, New York, United States of America
| | - Nathaniel R Landau
- Department of Microbiology, NYU Grossman School of Medicine, New York, United States of America
| |
Collapse
|
8
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
9
|
Gadgil A, Raczyńska KD. U7 snRNA: A tool for gene therapy. J Gene Med 2021; 23:e3321. [PMID: 33590603 PMCID: PMC8243935 DOI: 10.1002/jgm.3321] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 01/22/2021] [Accepted: 02/09/2021] [Indexed: 12/25/2022] Open
Abstract
Most U-rich small nuclear ribonucleoproteins (snRNPs) are complexes that mediate the splicing of pre-mRNAs. U7 snRNP is an exception in that it is not involved in splicing but is a key factor in the unique 3' end processing of replication-dependent histone mRNAs. However, by introducing controlled changes in the U7 snRNA histone binding sequence and in the Sm motif, it can be used as an effective tool for gene therapy. The modified U7 snRNP (U7 Sm OPT) is thus not involved in the processing of replication-dependent histone pre-mRNA but targets splicing by inducing efficient skipping or inclusion of selected exons. U7 Sm OPT is of therapeutic importance in diseases that are an outcome of splicing defects, such as myotonic dystrophy, Duchenne muscular dystrophy, amyotrophic lateral sclerosis, β-thalassemia, HIV-1 infection and spinal muscular atrophy. The benefits of using U7 Sm OPT for gene therapy are its compact size, ability to accumulate in the nucleus without causing any toxic effects in the cells, and no immunoreactivity. The risk of transgene misregulation by using U7 Sm OPT is also low because it is involved in correcting the expression of an endogenous gene controlled by its own regulatory elements. Altogether, using U7 Sm OPT as a tool in gene therapy can ensure lifelong treatment, whereas an oligonucleotide or other drug/compound would require repeated administration. It would thus be strategic to harness these unique properties of U7 snRNP and deploy it as a tool in gene therapy.
Collapse
Affiliation(s)
- Ankur Gadgil
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of BiologyAdam Mickiewicz UniversityPoznanPoland
- Center for Advanced TechnologyAdam Mickiewicz UniversityPoznanPoland
| | - Katarzyna Dorota Raczyńska
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of BiologyAdam Mickiewicz UniversityPoznanPoland
- Center for Advanced TechnologyAdam Mickiewicz UniversityPoznanPoland
| |
Collapse
|
10
|
Abstract
Recombinant viruses are the workhorse of modern neuroscience. Whether one would like to understand a neuron's morphology, natural activity patterns, molecular composition, connectivity or behavioural and physiologic function, most studies begin with the injection of an engineered virus, often an adeno-associated virus or herpes simplex virus, among many other types. Recombinant viruses currently enable some combination of cell type-specific, circuit-selective, activity-dependent and spatiotemporally resolved transgene expression. Viruses are now used routinely to study the molecular and cellular functions of a gene within an identified cell type in the brain, and enable the application of optogenetics, chemogenetics, calcium imaging and related approaches. These advantageous properties of engineered viruses thus enable characterization of neuronal function at unprecedented resolution. However, each virus has specific advantages and disadvantages, which makes viral tool selection paramount for properly designing and executing experiments within the central nervous system. In the current Review, we discuss the key principles and uses of engineered viruses and highlight innovations that are needed moving forward.
Collapse
Affiliation(s)
- Alexander R Nectow
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
11
|
Tosolini AP, Sleigh JN. Intramuscular Delivery of Gene Therapy for Targeting the Nervous System. Front Mol Neurosci 2020; 13:129. [PMID: 32765219 PMCID: PMC7379875 DOI: 10.3389/fnmol.2020.00129] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
Virus-mediated gene therapy has the potential to deliver exogenous genetic material into specific cell types to promote survival and counteract disease. This is particularly enticing for neuronal conditions, as the nervous system is renowned for its intransigence to therapeutic targeting. Administration of gene therapy viruses into skeletal muscle, where distal terminals of motor and sensory neurons reside, has been shown to result in extensive transduction of cells within the spinal cord, brainstem, and sensory ganglia. This route is minimally invasive and therefore clinically relevant for gene therapy targeting to peripheral nerve soma. For successful transgene expression, viruses administered into muscle must undergo a series of processes, including host cell interaction and internalization, intracellular sorting, long-range retrograde axonal transport, endosomal liberation, and nuclear import. In this review article, we outline key characteristics of major gene therapy viruses—adenovirus, adeno-associated virus (AAV), and lentivirus—and summarize the mechanisms regulating important steps in the virus journey from binding at peripheral nerve terminals to nuclear delivery. Additionally, we describe how neuropathology can negatively influence these pathways, and conclude by discussing opportunities to optimize the intramuscular administration route to maximize gene delivery and thus therapeutic potential.
Collapse
Affiliation(s)
- Andrew P Tosolini
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - James N Sleigh
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,UK Dementia Research Institute, University College London, London, United Kingdom
| |
Collapse
|
12
|
Saleeba C, Dempsey B, Le S, Goodchild A, McMullan S. A Student's Guide to Neural Circuit Tracing. Front Neurosci 2019; 13:897. [PMID: 31507369 PMCID: PMC6718611 DOI: 10.3389/fnins.2019.00897] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/12/2019] [Indexed: 12/17/2022] Open
Abstract
The mammalian nervous system is comprised of a seemingly infinitely complex network of specialized synaptic connections that coordinate the flow of information through it. The field of connectomics seeks to map the structure that underlies brain function at resolutions that range from the ultrastructural, which examines the organization of individual synapses that impinge upon a neuron, to the macroscopic, which examines gross connectivity between large brain regions. At the mesoscopic level, distant and local connections between neuronal populations are identified, providing insights into circuit-level architecture. Although neural tract tracing techniques have been available to experimental neuroscientists for many decades, considerable methodological advances have been made in the last 20 years due to synergies between the fields of molecular biology, virology, microscopy, computer science and genetics. As a consequence, investigators now enjoy an unprecedented toolbox of reagents that can be directed against selected subpopulations of neurons to identify their efferent and afferent connectomes. Unfortunately, the intersectional nature of this progress presents newcomers to the field with a daunting array of technologies that have emerged from disciplines they may not be familiar with. This review outlines the current state of mesoscale connectomic approaches, from data collection to analysis, written for the novice to this field. A brief history of neuroanatomy is followed by an assessment of the techniques used by contemporary neuroscientists to resolve mesoscale organization, such as conventional and viral tracers, and methods of selecting for sub-populations of neurons. We consider some weaknesses and bottlenecks of the most widely used approaches for the analysis and dissemination of tracing data and explore the trajectories that rapidly developing neuroanatomy technologies are likely to take.
Collapse
Affiliation(s)
- Christine Saleeba
- Neurobiology of Vital Systems Node, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
- The School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Bowen Dempsey
- CNRS, Hindbrain Integrative Neurobiology Laboratory, Neuroscience Paris-Saclay Institute (Neuro-PSI), Université Paris-Saclay, Gif-sur-Yvette, France
| | - Sheng Le
- Neurobiology of Vital Systems Node, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Ann Goodchild
- Neurobiology of Vital Systems Node, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Simon McMullan
- Neurobiology of Vital Systems Node, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
13
|
Evans TA, Barkauskas DS, Silver J. Intravital imaging of immune cells and their interactions with other cell types in the spinal cord: Experiments with multicolored moving cells. Exp Neurol 2019; 320:112972. [PMID: 31234058 DOI: 10.1016/j.expneurol.2019.112972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/25/2019] [Accepted: 06/04/2019] [Indexed: 12/25/2022]
Abstract
Intravital imaging of the immune system is a powerful technique for studying biology of the immune response in the spinal cord using a variety of disease models ranging from traumatic injury to autoimmune disorders. Here, we will discuss specific technical aspects as well as many intriguing biological phenomena that have been revealed with the use of intravital imaging for investigation of the immune system in the spinal cord. We will discuss surgical techniques for exposing and stabilizing the spine that are critical for obtaining images, visualizing immune and CNS cells with genetically expressed fluorescent proteins, fluorescent labeling techniques and briefly discuss some of the challenges of image analysis.
Collapse
Affiliation(s)
- Teresa A Evans
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
| | | | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
14
|
Palma-Chavez A, Konar-Nié M, Órdenes P, Maurelia F, Elizondo-Vega R, Oyarce K, López S, Rojas J, Steinberg X, García-Robles MA, Sepúlveda FJ. Glucose Increase DAGLα Levels in Tanycytes and Its Inhibition Alters Orexigenic and Anorexigenic Neuropeptides Expression in Response to Glucose. Front Endocrinol (Lausanne) 2019; 10:647. [PMID: 31620093 PMCID: PMC6763563 DOI: 10.3389/fendo.2019.00647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022] Open
Abstract
The endocannabinoid system (ECS) is composed of a group of Gi-coupled protein receptors and enzymes, producing and degrading the endocannabinoids, 2-arachidonoylglycerol (2-AG) and N-arachidonoyl-ethanolamine (AEA). Endocannabinoid-mediated signaling modulates brain functions, such as pain, mood, memory, and feeding behavior. The activation of the ECS is associated with overeating and obesity; however, the expression of components of this system has been only partially studied in the hypothalamus, a critical region implicated in feeding behavior. Within this brain region, anorexigenic, and orexigenic neurons of the arcuate nucleus (ARC) are in close contact with tanycytes, glial radial-like cells that line the lateral walls and floor of the third ventricle (3V). The specific function of tanycytes and the effects of metabolic signals generated by them on adjacent neurons is starting to be elucidated. We have proposed that the ECS within tanycytes modulates ARC neurons, thus modifying food intake. Here, we evaluated the expression and the loss of function of the 2-AG-producing enzyme, diacylglycerol lipase-alpha (DAGLα). Using Western blot and immunohistochemistry analyses in basal hypothalamus sections of adult rats under several glycemic conditions, we confirm that DAGLα is strongly expressed at the basal hypothalamus in glial and neuronal cells, increasing further in response to greater extracellular glucose levels. Using a DAGLα-inhibiting adenovirus (shRNA), suppression of DAGLα expression in tanycytes altered the usual response to intracerebroventricular glucose in terms of neuropeptides produced by neurons of the ARC. Thus, these results strongly suggest that the tanycytes could generate 2-AG, which modulates the function of anorexigenic and orexigenic neurons.
Collapse
Affiliation(s)
- Alejandra Palma-Chavez
- Laboratorio de Biología Celular, Departamento de Biología Celular, Universidad de Concepcion, Concepción, Chile
- Laboratorio de Bioquímica y Biología Celular, Departamento de Bioquímica y Biología Molecular, Universidad de Concepción, Concepción, Chile
| | - Macarena Konar-Nié
- Laboratorio de Biología Celular, Departamento de Biología Celular, Universidad de Concepcion, Concepción, Chile
| | - Patricio Órdenes
- Laboratorio de Biología Celular, Departamento de Biología Celular, Universidad de Concepcion, Concepción, Chile
| | - Felipe Maurelia
- Laboratorio de Bioquímica y Biología Celular, Departamento de Bioquímica y Biología Molecular, Universidad de Concepción, Concepción, Chile
| | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Universidad de Concepcion, Concepción, Chile
| | - Karina Oyarce
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
| | - Sergio López
- Laboratorio de Biología Celular, Departamento de Biología Celular, Universidad de Concepcion, Concepción, Chile
| | - Joaquin Rojas
- Centro de Estudios Avanzados para la Vida (CREAV), Universidad de Concepción, Concepción, Chile
| | - Ximena Steinberg
- Laboratorio de Bioquímica y Biología Celular, Departamento de Bioquímica y Biología Molecular, Universidad de Concepción, Concepción, Chile
| | - María A. García-Robles
- Laboratorio de Biología Celular, Departamento de Biología Celular, Universidad de Concepcion, Concepción, Chile
- Centro de Estudios Avanzados para la Vida (CREAV), Universidad de Concepción, Concepción, Chile
- *Correspondence: María A. García-Robles
| | - Fernando J. Sepúlveda
- Laboratorio de Biología Celular, Departamento de Biología Celular, Universidad de Concepcion, Concepción, Chile
- Laboratorio de Bioquímica y Biología Celular, Departamento de Bioquímica y Biología Molecular, Universidad de Concepción, Concepción, Chile
- Fernando J. Sepúlveda
| |
Collapse
|
15
|
Kitada M, Wakao S, Dezawa M. Intracellular signaling similarity reveals neural stem cell-like properties of ependymal cells in the adult rat spinal cord. Dev Growth Differ 2018; 60:326-340. [DOI: 10.1111/dgd.12546] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 01/19/2023]
Affiliation(s)
- Masaaki Kitada
- Department of Stem Cell Biology and Histology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Shohei Wakao
- Department of Stem Cell Biology and Histology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology; Tohoku University Graduate School of Medicine; Sendai Japan
| |
Collapse
|
16
|
Tobita T, Kiyozumi D, Ikawa M. Placenta-specific gene manipulation using lentiviral vector and its application. Placenta 2017; 59 Suppl 1:S37-S43. [PMID: 28988726 DOI: 10.1016/j.placenta.2017.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 09/20/2017] [Accepted: 09/25/2017] [Indexed: 12/16/2022]
Abstract
The placenta is an essential organ for embryo development in the uterus of eutherian mammals. Large contributions in unveiling molecular mechanisms and physiological functions underlying placental formation were made by analyzing mutant and transgenic animals. However, it had been difficult to elucidate whether the placental defects observed in such animals originate from the placenta itself or from the fetus, as both placental and fetal genomes are modified. Therefore strategies to modify the placental genome without affecting the "fetal genome" had been needed. Through the ingenious use of lentiviral (LV) vectors, placenta-specific modification is now possible. Lentivirus is a genus of retroviruses that use reverse-transcriptase to convert its single-strand RNA genome to double-strand DNA and integrate into the host genome. Previous studies showed that when LV vectors were used to transduce embryos at the 2-cell stage, the viral genome is systemically introduced into host genome. Interestingly, by delaying the timing of transduction to the blastocyst stage, the transgene is expressed specifically in the placenta as a consequence of trophectoderm-specific viral transduction. This review summarizes the development of the LV vector-mediated placenta-specific gene manipulation technology and its application in placental research over the past decade. A perspective for future application of LV vectors to further placenta research, especially in combination with next generation genome editing technologies, is also presented.
Collapse
Affiliation(s)
- Tomohiro Tobita
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan; Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Daiji Kiyozumi
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan; Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
17
|
Imaoka T, Date I, Miyoshi Y, Ono T, Furuta T, Asari S, Ohmoto T, Yasuda T, Tsuda M. Preliminary Results of Gene Transfer to Central Nervous System by Continuous Injection of Dna-Liposome Complex. Cell Transplant 2017. [DOI: 10.1177/096368979500401s07] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Takashi Imaoka
- Departments of Neurological Surgery, Okayama University Medical School, 2-5-1 Shikata-cho, Okayama 700, Japan
| | - Isao Date
- Departments of Neurological Surgery, Okayama University Medical School, 2-5-1 Shikata-cho, Okayama 700, Japan
| | - Yasuyuki Miyoshi
- Departments of Neurological Surgery, Okayama University Medical School, 2-5-1 Shikata-cho, Okayama 700, Japan
| | - Takeshi Ono
- Departments of Neurological Surgery, Okayama University Medical School, 2-5-1 Shikata-cho, Okayama 700, Japan
| | - Tomohisa Furuta
- Departments of Neurological Surgery, Okayama University Medical School, 2-5-1 Shikata-cho, Okayama 700, Japan
| | - Shoji Asari
- Departments of Neurological Surgery, Okayama University Medical School, 2-5-1 Shikata-cho, Okayama 700, Japan
| | - Takashi Ohmoto
- Departments of Neurological Surgery, Okayama University Medical School, 2-5-1 Shikata-cho, Okayama 700, Japan
| | - Tatsuji Yasuda
- Departments of Cell Chemistry, Institute of Cellular and Molecular Biology, Okayama University Medical School, 2-5-1 Shikata-cho, Okayama 700, Japan
| | - Masaaki Tsuda
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Okayama University, Tsushima-naka, Okayama 700, Japan
| |
Collapse
|
18
|
Choudhury SR, Hudry E, Maguire CA, Sena-Esteves M, Breakefield XO, Grandi P. Viral vectors for therapy of neurologic diseases. Neuropharmacology 2017; 120:63-80. [PMID: 26905292 PMCID: PMC5929167 DOI: 10.1016/j.neuropharm.2016.02.013] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/07/2016] [Accepted: 02/15/2016] [Indexed: 12/21/2022]
Abstract
Neurological disorders - disorders of the brain, spine and associated nerves - are a leading contributor to global disease burden with a shockingly large associated economic cost. Various treatment approaches - pharmaceutical medication, device-based therapy, physiotherapy, surgical intervention, among others - have been explored to alleviate the resulting extent of human suffering. In recent years, gene therapy using viral vectors - encoding a therapeutic gene or inhibitory RNA into a "gutted" viral capsid and supplying it to the nervous system - has emerged as a clinically viable option for therapy of brain disorders. In this Review, we provide an overview of the current state and advances in the field of viral vector-mediated gene therapy for neurological disorders. Vector tools and delivery methods have evolved considerably over recent years, with the goal of providing greater and safer genetic access to the central nervous system. Better etiological understanding of brain disorders has concurrently led to identification of improved therapeutic targets. We focus on the vector technology, as well as preclinical and clinical progress made thus far for brain cancer and various neurodegenerative and neurometabolic disorders, and point out the challenges and limitations that accompany this new medical modality. Finally, we explore the directions that neurological gene therapy is likely to evolve towards in the future. This article is part of the Special Issue entitled "Beyond small molecules for neurological disorders".
Collapse
Affiliation(s)
- Sourav R Choudhury
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Eloise Hudry
- Alzheimer's Disease Research Unit, Harvard Medical School & Massachusetts General Hospital, Charlestown, MA 02129, USA.
| | - Casey A Maguire
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Miguel Sena-Esteves
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Xandra O Breakefield
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Paola Grandi
- Department of Neurological Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15219, USA.
| |
Collapse
|
19
|
Kaliberov SA, Kaliberova LN, Yan H, Kapoor V, Hallahan DE. Retargeted adenoviruses for radiation-guided gene delivery. Cancer Gene Ther 2016; 23:303-14. [PMID: 27492853 PMCID: PMC5031535 DOI: 10.1038/cgt.2016.32] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/21/2016] [Accepted: 06/30/2016] [Indexed: 11/25/2022]
Abstract
The combination of radiation with radiosensitizing gene delivery or oncolytic viruses promises to provide an advantage that could improve the therapeutic results for glioblastoma. X-rays can induce significant molecular changes in cancer cells. We isolated the GIRLRG peptide that binds to radiation-inducible 78 kDa glucose-regulated protein (GRP78), which is overexpressed on the plasma membranes of irradiated cancer cells and tumor-associated microvascular endothelial cells. The goal of our study was to improve tumor-specific adenovirus-mediated gene delivery by selectively targeting the adenovirus binding to this radiation-inducible protein. We employed an adenoviral fiber replacement approach to conduct a study of the targeting utility of GRP78-binding peptide. We have developed fiber-modified adenoviruses encoding the GRP78-binding peptide inserted into the fiber-fibritin. We have evaluated the reporter gene expression of fiber-modified adenoviruses in vitro using a panel of glioma cells and a human D54MG tumor xenograft model. The obtained results demonstrated that employment of the GRP78-binding peptide resulted in increased gene expression in irradiated tumors following infection with fiber-modified adenoviruses, compared with untreated tumor cells. These studies demonstrate the feasibility of adenoviral retargeting using the GRP78-binding peptide that selectively recognizes tumor cells responding to radiation treatment.
Collapse
Affiliation(s)
- S A Kaliberov
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA.,Biologic Therapeutics Center, Washington University School of Medicine, St Louis, MO, USA
| | - L N Kaliberova
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA.,Biologic Therapeutics Center, Washington University School of Medicine, St Louis, MO, USA
| | - H Yan
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA.,Biologic Therapeutics Center, Washington University School of Medicine, St Louis, MO, USA
| | - V Kapoor
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - D E Hallahan
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA.,Biologic Therapeutics Center, Washington University School of Medicine, St Louis, MO, USA.,Siteman Cancer Center, St Louis, MO, USA
| |
Collapse
|
20
|
Craig AJ, Housley GD. Evaluation of Gene Therapy as an Intervention Strategy to Treat Brain Injury from Stroke. Front Mol Neurosci 2016; 9:34. [PMID: 27252622 PMCID: PMC4877374 DOI: 10.3389/fnmol.2016.00034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 05/06/2016] [Indexed: 01/01/2023] Open
Abstract
Stroke is a leading cause of death and disability, with a lack of treatments available to prevent cell death, regenerate damaged cells and pathways, or promote neurogenesis. The extended period of hours to weeks over which tissue damage continues to occur makes this disorder a candidate for gene therapy. This review highlights the development of gene therapy in the area of stroke, with the evolution of viral administration, in experimental stroke models, from pre-injury to clinically relevant timeframes of hours to days post-stroke. The putative therapeutic proteins being examined include anti-apoptotic, pro-survival, anti-inflammatory, and guidance proteins, targeting multiple pathways within the complex pathology, with promising results. The balance of findings from animal models suggests that gene therapy provides a viable translational platform for treatment of ischemic brain injury arising from stroke.
Collapse
Affiliation(s)
- Amanda J Craig
- Translational Neuroscience Facility & Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney NSW, Australia
| | - Gary D Housley
- Translational Neuroscience Facility & Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney NSW, Australia
| |
Collapse
|
21
|
Kamran N, Candolfi M, Baker GJ, Ayala MM, Dzaman M, Lowenstein PR, Castro MG. Gene Therapy for the Treatment of Neurological Disorders: Central Nervous System Neoplasms. Methods Mol Biol 2016; 1382:467-82. [PMID: 26611605 PMCID: PMC4677484 DOI: 10.1007/978-1-4939-3271-9_31] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults with a median survival of 16.2-21.2 months post diagnosis (Stupp et al., N Engl J Med 352(10): 987-996, 2005). Because of its location, complete surgical resection is impossible; additionally because GBM is also resistant to chemotherapeutic and radiotherapy approaches, development of novel therapies is urgently needed. In this chapter we describe the development of preclinical animal models and a conditionally cytotoxic and immune-stimulatory gene therapy strategy that successfully causes tumor regression in several rodent GBM models.
Collapse
Affiliation(s)
- Neha Kamran
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5689, USA
- Department of Cell and Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5689, USA
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Piso 10, C1121ABG, Buenos Aires, Argentina
| | - Gregory J Baker
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5689, USA
- Department of Cell and Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5689, USA
| | - Mariela Moreno Ayala
- Instituto de Investigaciones Biomédicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Piso 10, C1121ABG, Buenos Aires, Argentina
| | - Marta Dzaman
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5689, USA
- Department of Cell and Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5689, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5689, USA
- Department of Cell and Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5689, USA
| | - Maria G Castro
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5689, USA.
- Department of Cell and Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5689, USA.
| |
Collapse
|
22
|
Garanina EE, Mukhamedshina YO, Salafutdinov II, Kiyasov AP, Lima LM, Reis HJ, Palotás A, Islamov RR, Rizvanov AA. Construction of recombinant adenovirus containing picorna-viral 2A-peptide sequence for the co-expression of neuro-protective growth factors in human umbilical cord blood cells. Spinal Cord 2015; 54:423-30. [PMID: 26439843 DOI: 10.1038/sc.2015.162] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/18/2015] [Accepted: 07/31/2015] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN Experimental study. OBJECTIVE Several neuro-degenerative disorders such as Alzheimer's dementia, Parkinson's disease and amyotrophic lateral sclerosis (ALS) are associated with genetic mutations, and replacing or disrupting defective sequences might offer therapeutic benefits. Single gene delivery has so far failed to achieve significant clinical improvements in humans, leading to the advent of co-expression of multiple therapeutic genes. Co-transfection using two or more individual constructs might inadvertently result in disproportionate delivery of the products into the cells. To prevent this, and in order to rule out interference among the many promoters with varying strength, expressing multiple proteins in equimolar amounts can be achieved by linking open reading frames under the control of only one promoter. SETTING Kazan, Russian Federation. METHODS Here we describe a strategy for adeno-viral co-expression of vascular endothelial growth factor (VEGF) and fibroblast growth factor 2 (FGF2) interconnected through picorna-viral 2A-amino-acid sequence in transfected human umbilical cord blood mono-nuclear cells (hUCB-MCs). RESULTS Presence of both growth factors, as well as absence of immune response to 2A-antigen, was demonstrated after 28-52 days. Following injection of hUCB-MCs into ALS transgenic mice, co-expression of VEGF and FGF2, as well as viable xeno-transplanted cells, were observed in the spinal cord after 1 month. CONCLUSION These results suggest that recombinant adeno-virus containing 2A-sequences could serve as a promising alternative in regenerative medicine for the delivery of therapeutic molecules to treat neurodegenerative diseases, such as ALS.
Collapse
Affiliation(s)
- E E Garanina
- Kazan Federal University, Kazan, Russian Federation
| | | | | | - A P Kiyasov
- Kazan Federal University, Kazan, Russian Federation
| | - L M Lima
- Universidade Federal de Viçosa, Viçosa, Brazil
| | - H J Reis
- Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - A Palotás
- Kazan Federal University, Kazan, Russian Federation.,Asklepios-Med (Private Medical Practice and Research Center), Szeged, Hungary
| | - R R Islamov
- Kazan State Medical University, Kazan, Russian Federation
| | - A A Rizvanov
- Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
23
|
Nassi JJ, Cepko CL, Born RT, Beier KT. Neuroanatomy goes viral! Front Neuroanat 2015; 9:80. [PMID: 26190977 PMCID: PMC4486834 DOI: 10.3389/fnana.2015.00080] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 05/25/2015] [Indexed: 02/03/2023] Open
Abstract
The nervous system is complex not simply because of the enormous number of neurons it contains but by virtue of the specificity with which they are connected. Unraveling this specificity is the task of neuroanatomy. In this endeavor, neuroanatomists have traditionally exploited an impressive array of tools ranging from the Golgi method to electron microscopy. An ideal method for studying anatomy would label neurons that are interconnected, and, in addition, allow expression of foreign genes in these neurons. Fortuitously, nature has already partially developed such a method in the form of neurotropic viruses, which have evolved to deliver their genetic material between synaptically connected neurons while largely eluding glia and the immune system. While these characteristics make some of these viruses a threat to human health, simple modifications allow them to be used in controlled experimental settings, thus enabling neuroanatomists to trace multi-synaptic connections within and across brain regions. Wild-type neurotropic viruses, such as rabies and alpha-herpes virus, have already contributed greatly to our understanding of brain connectivity, and modern molecular techniques have enabled the construction of recombinant forms of these and other viruses. These newly engineered reagents are particularly useful, as they can target genetically defined populations of neurons, spread only one synapse to either inputs or outputs, and carry instructions by which the targeted neurons can be made to express exogenous proteins, such as calcium sensors or light-sensitive ion channels, that can be used to study neuronal function. In this review, we address these uniquely powerful features of the viruses already in the neuroanatomist's toolbox, as well as the aspects of their biology that currently limit their utility. Based on the latter, we consider strategies for improving viral tracing methods by reducing toxicity, improving control of transsynaptic spread, and extending the range of species that can be studied.
Collapse
Affiliation(s)
- Jonathan J Nassi
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies La Jolla, CA, USA
| | - Constance L Cepko
- Department of Genetics, Harvard Medical School Boston, MA, USA ; Department of Ophthalmology, Howard Hughes Medical Institute, Harvard Medical School Boston, MA, USA
| | - Richard T Born
- Department of Neurobiology, Harvard Medical School Boston, MA, USA ; Center for Brain Science, Harvard University Cambridge, MA, USA
| | - Kevin T Beier
- Department of Psychiatry and Behavioral Sciences and Department of Biology, Stanford University Stanford, CA, USA
| |
Collapse
|
24
|
Serguera C, Bemelmans AP. Gene therapy of the central nervous system: general considerations on viral vectors for gene transfer into the brain. Rev Neurol (Paris) 2014; 170:727-38. [PMID: 25459120 DOI: 10.1016/j.neurol.2014.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/13/2014] [Accepted: 09/10/2014] [Indexed: 02/04/2023]
Abstract
The last decade has nourished strong doubts on the beneficial prospects of gene therapy for curing fatal diseases. However, this climate of reservation is currently being transcended by the publication of several successful clinical protocols, restoring confidence in the appropriateness of therapeutic gene transfer. A strong sign of this present enthusiasm for gene therapy by clinicians and industrials is the market approval of the therapeutic viral vector Glybera, the first commercial product in Europe of this class of drug. This new field of medicine is particularly attractive when considering therapies for a number of neurological disorders, most of which are desperately waiting for a satisfactory treatment. The central nervous system is indeed a very compliant organ where gene transfer can be stable and successful if provided through an appropriate strategy. The purpose of this review is to present the characteristics of the most efficient virus-derived vectors used by researchers and clinicians to genetically modify particular cell types or whole regions of the brain. In addition, we discuss major issues regarding side effects, such as genotoxicity and immune response associated to the use of these vectors.
Collapse
Affiliation(s)
- C Serguera
- CEA, DSV, I(2)BM, Molecular Imaging Research Center (MIRCen) and CNRS, CEA URA 2210, 18, route du Panorama, 92265 Fontenay-aux-Roses, France
| | - A-P Bemelmans
- CEA, DSV, I(2)BM, Molecular Imaging Research Center (MIRCen) and CNRS, CEA URA 2210, 18, route du Panorama, 92265 Fontenay-aux-Roses, France.
| |
Collapse
|
25
|
Electroporation markedly improves Sleeping Beauty transposon-induced tumorigenesis in mice. Cancer Gene Ther 2014; 21:333-9. [PMID: 24992966 DOI: 10.1038/cgt.2014.33] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/31/2014] [Accepted: 06/02/2014] [Indexed: 11/08/2022]
Abstract
The Sleeping Beauty (SB) transposon system is an important tool for genetic studies. It is used to insert a gene of interest into the host chromosome, thus enabling permanent gene expression. However, this system is less useful in higher eukaryotes because the transposition frequency is low. Efforts to improve the efficacy of the SB transposon system have focused on the method of gene delivery, but although electroporation has recently attracted much attention as an in vivo gene delivery tool, the simultaneous use of electroporation and the SB transposon system has not been studied for gene transfer in mice. In this study, electroporation was used in a model of SB transposon-induced insertional tumorigenesis. Electroporation increased the rate of tumor development to three times that of the control group. There was no difference in phenotype between tumors induced with the SB transposon system alone and those induced by the SB transposon and electroporation. Electroporation therefore may be an efficient means of improving the efficacy of gene transfer via the SB transposon system.
Collapse
|
26
|
Pancreatic cancer induced by in vivo electroporation-enhanced sleeping beauty transposon gene delivery system in mouse. Pancreas 2014; 43:614-8. [PMID: 24713671 DOI: 10.1097/mpa.0000000000000102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The aim of this study was to establish a pancreatic tumor model of mouse using the electroporation-enhanced Sleeping Beauty (SB) transposon system. METHODS The SB transposon system was used in conjunction with electroporation to deliver oncogenes, c-Myc and HRAS, and shRNA against p53 into the mouse pancreas to induce tumors. Oncogenes (c-Myc and HRAS) and shRNA against p53 gene were directly injected into the pancreas of the mouse along with in vivo electroporation applied on the injection site. The tumors were identified grossly and confirmed using animal positron emission tomographic imaging. The tumors were then characterized using histological and immunohistochemical techniques. The expression of the targeted genes (c-Myc, HRAS, and p53) was analyzed by a real-time quantitative polymerase chain reaction. RESULTS Pancreatic tumors were successfully induced. The tumor phenotype was a sarcomatoid carcinoma, which was verified through immunohistochemistry. Some cysts or duct-like structures suggested to be metaplastic acinar cells were visible in the induced tumor. CONCLUSIONS The SB transposon enhanced with electroporation can readily generate pancreatic tumors in the mice, and thus, this model serves as a valuable resource for the mouse models of pancreatic cancer.
Collapse
|
27
|
Salinas S, Zussy C, Loustalot F, Henaff D, Menendez G, Morton PE, Parsons M, Schiavo G, Kremer EJ. Disruption of the coxsackievirus and adenovirus receptor-homodimeric interaction triggers lipid microdomain- and dynamin-dependent endocytosis and lysosomal targeting. J Biol Chem 2013; 289:680-95. [PMID: 24273169 DOI: 10.1074/jbc.m113.518365] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The coxsackievirus and adenovirus receptor (CAR) serves as a docking factor for some adenovirus (AdV) types and group B coxsackieviruses. Its role in AdV internalization is unclear as studies suggest that its intracellular domain is dispensable for some AdV infection. We previously showed that in motor neurons, AdV induced CAR internalization and co-transport in axons, suggesting that CAR was linked to endocytic and long-range transport machineries. Here, we characterized the mechanisms of CAR endocytosis in neurons and neuronal cells. We found that CAR internalization was lipid microdomain-, actin-, and dynamin-dependent, and subsequently followed by CAR degradation in lysosomes. Moreover, ligands that disrupted the homodimeric CAR interactions in its D1 domains triggered an internalization cascade involving sequences in its intracellular tail.
Collapse
Affiliation(s)
- Sara Salinas
- From the Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, Universités de Montpellier I & II, Montpellier, France
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Piersanti S, Astrologo L, Licursi V, Costa R, Roncaglia E, Gennetier A, Ibanes S, Chillon M, Negri R, Tagliafico E, Kremer EJ, Saggio I. Differentiated neuroprogenitor cells incubated with human or canine adenovirus, or lentiviral vectors have distinct transcriptome profiles. PLoS One 2013; 8:e69808. [PMID: 23922808 PMCID: PMC3724896 DOI: 10.1371/journal.pone.0069808] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 06/13/2013] [Indexed: 12/13/2022] Open
Abstract
Several studies have demonstrated the potential for vector-mediated gene transfer to the brain. Helper-dependent (HD) human (HAd) and canine (CAV-2) adenovirus, and VSV-G-pseudotyped self-inactivating HIV-1 vectors (LV) effectively transduce human brain cells and their toxicity has been partly analysed. However, their effect on the brain homeostasis is far from fully defined, especially because of the complexity of the central nervous system (CNS). With the goal of dissecting the toxicogenomic signatures of the three vectors for human neurons, we transduced a bona fide human neuronal system with HD-HAd, HD-CAV-2 and LV. We analysed the transcriptional response of more than 47,000 transcripts using gene chips. Chip data showed that HD-CAV-2 and LV vectors activated the innate arm of the immune response, including Toll-like receptors and hyaluronan circuits. LV vector also induced an IFN response. Moreover, HD-CAV-2 and LV vectors affected DNA damage pathways--but in opposite directions--suggesting a differential response of the p53 and ATM pathways to the vector genomes. As a general response to the vectors, human neurons activated pro-survival genes and neuron morphogenesis, presumably with the goal of re-establishing homeostasis. These data are complementary to in vivo studies on brain vector toxicity and allow a better understanding of the impact of viral vectors on human neurons, and mechanistic approaches to improve the therapeutic impact of brain-directed gene transfer.
Collapse
Affiliation(s)
- Stefania Piersanti
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
| | - Letizia Astrologo
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
| | - Valerio Licursi
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
| | - Rossella Costa
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
| | - Enrica Roncaglia
- Dipartimento di Scienze Biomediche, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Aurelie Gennetier
- Institut de Génétique Moléculaire de Montpellier, University Montpellier I & II, Montpellier, France
| | - Sandy Ibanes
- Institut de Génétique Moléculaire de Montpellier, University Montpellier I & II, Montpellier, France
| | - Miguel Chillon
- Istituto Pasteur Fondazione Cenci Bolognetti, Roma, Italy
| | - Rodolfo Negri
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Roma, Italy
| | - Enrico Tagliafico
- Dipartimento di Scienze Biomediche, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, University Montpellier I & II, Montpellier, France
| | - Isabella Saggio
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Roma, Italy
- Istituto di Biologia e Patologia Molecolari del CNR, Roma, Italy
- * E-mail:
| |
Collapse
|
29
|
Tada T, Nguyen JB, Hitoshi Y, Watson NP, Dunn JF, Ohara S, Nagano S, Kosai KI, Israel MA. Diffuse encephaloventriculitis and substantial leukoencephalopathy after intraventricular administration of recombinant adenovirus. Neurol Res 2013; 27:378-86. [PMID: 15949235 DOI: 10.1179/016164105x22075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES The use of recombinant adenovirus as a vehicle for gene transfer into ependymal cells is a potential therapeutic tool for the treatment of various neural disorders. However, gene transfer into the ependymal cells of the ventricular wall is associated with high-level expression of the transferred gene, which declines rapidly. The purpose of this study is to understand the cause of this early decline in gene expression. METHODS Different doses of adenovirus-expressing beta-galactosidase (Ad-beta-gal) were injected into the lateral brain ventricle of C57BL/6 mice, and the brains were observed histologically and with magnetic resonance (MR) imaging for a month. RESULTS Inoculation of the lateral ventricle with more than 1 x 10(8) viral particles (2.6 x 10(6) pfu) resulted in a rapid decline of beta -gal expression. MR imaging indicated gradual ventriculomegaly and histological analysis showed the loss of the ependymal cells from the ventricular wall, lymphocytes infiltration near the wall, degeneration of myelinated fibers and apoptosis in the external capsule. Reactive astrocytes proliferated in the external capsule 17 days following inoculation. To avoid this irreversible brain atrophy, the inoculated adenovirus should be reduced to less than 1 x 10(7) particles (2.6 x 10(5) pfu) in mice. DISCUSSION Our results indicate the presence of a unique and diffuse immune response of the brain; therefore, the clinical use of recombinant virus for intraventricular gene transfer must be carefully evaluated.
Collapse
Affiliation(s)
- Tsuyoshi Tada
- Department of Neurosurgery, Shinshu University School of Medicine, Asahi 3-1-1, Matsumoto 390-8621, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kondo S, Maekawa A, Saito I, Kanegae Y. [Recent progress in adenovirus vectors: focusing on VA-deleted AdV]. Uirusu 2013; 63:155-164. [PMID: 25366050 DOI: 10.2222/jsv.63.155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
First-generation adenovirus vectors (FG-AdVs) are widely used because transduction efficiency of the vectors is very high. However, severe immune responses especially to the liver have been a serious problem of this vector. We succeeded to identify a viral protein that cause the immune responses and reported ''low-inflammatory AdVs'' that mostly solve this problem. However, to develop the ultimate form of this vector, it is necessary to remove virus-associated RNA (VA RNA) genes from the AdV vector genome. VA RNAs are transcribed by polymerase III; they are not essential for viral growth but have important roles to make appropriate circumstances for this virus. Large amount of VA RNAs are required in the late phase to support viral growth. Hence it is difficult to establish 293 cell lines that can support replication of AdVs lacking VA RNA genes (VA-deleted AdVs) supplying sufficient amount of VA RNA in trans. Recently we have developed a method for efficient production of VA-deleted AdVs and succeeded to obtain a high titer of VA-deleted AdVs. Then we construct VA-deleted AdVs expressing shRNA that knockdown the replication of hepatitis C virus (HCV). In fact, VA-deleted AdVs expressing these shRNAs suppressed HCV replication more effectively than conventional FG-AdV. Therefore, we showed that VA RNAs expressed from FG-AdVs probably compete with shRNA in the maturation pathway and reduce the effect of shRNAs. We think that VA-deleted AdV may substitute for current FG-AdVs and become a standard AdV.
Collapse
Affiliation(s)
- Saki Kondo
- Laboratory of Molecular Genetics, Institute of Medical Science, University of Tokyo
| | | | | | | |
Collapse
|
31
|
Biomaterial-Based Vectors for Targeted Delivery of Nucleic Acids to the Nervous System. DRUG DELIVERY SYSTEMS: ADVANCED TECHNOLOGIES POTENTIALLY APPLICABLE IN PERSONALISED TREATMENT 2013. [DOI: 10.1007/978-94-007-6010-3_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
32
|
A new finding concerning adenoviral-mediated gene transfer: A high-level, cell-specific transgene expression in the neural stem cells of adult mice. J Virol Methods 2012; 186:1-6. [DOI: 10.1016/j.jviromet.2012.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 07/05/2012] [Accepted: 07/09/2012] [Indexed: 01/28/2023]
|
33
|
Sivasubramaniam S, Fooks A, Lee J, Stacey G, Jennings A. Gene delivery into neuronal and glial cells by using a replication-deficient adenovirus vector: prospects for neurological gene therapy. Cytotechnology 2012; 24:253-9. [PMID: 22358769 DOI: 10.1023/a:1007904429698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have used a recombinant adenovirus vector (E1-) expressing β-galactosidase to explore a novel mechanism with which to transfer genes into cells of the central nervous system (CNS). The replication-deficient adenovirus vector expressing β-galactosidase (RAd35) was propagated on a permissive helper cell line (293 cells). High level protein expression from the human cytomegalovirus immediate early promoter (hCMV IE) was obtained in a target cell population of RAd35 infected cultured neuronal and glial cell lines. Light microscopy showed that over 50% of the glial cells studied expressed β-galactosidase. Following retinoic acid treatment, RAd35 infected cell lines ND7/23, NG108 and NTera2, showed β-galactosidase expression in up to 90% of the cells. In addition, these cells showed morphological evidence of differentiation into neurons. This pattern of β-galactosidase expression was also observed in primary rat cerebella granule neuron cultures. In vivo studies were performed in Balb/c mice following direct intracranial injections of RAd35 into the brain. Cell sections showed a localised staining in the brain at the site of injection of the virus. Non-replicating adenovirus vectors are therefore highly efficient systems for delivering a transgene into brain cells. However, their broad cell tropism may limit their applications for genetic disorders in which a specific cell type is to be targeted for gene therapy. To address this problem, we have constructed adenovirus vectors which contain specific neuronal promoters and are currently assessing in vitro expression.
Collapse
|
34
|
Transcellular targeting of fiber- and hexon-modified adenovirus vectors across the brain microvascular endothelial cells in vitro. PLoS One 2012; 7:e45977. [PMID: 23029348 PMCID: PMC3459940 DOI: 10.1371/journal.pone.0045977] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 08/23/2012] [Indexed: 12/23/2022] Open
Abstract
In central nervous system (CNS)-directed gene therapy, efficient targeting of brain parenchyma through the vascular route is prevented by the endothelium and the epithelium of the blood-brain and the blood-cerebrospinal fluid barriers, respectively. In this study, we evaluated the feasibility of the combined genetic and chemical adenovirus capsid modification technology to enable transcellular delivery of targeted adenovirus (Ad) vectors across the blood-brain barrier (BBB) in vitro models. As a proof-of-principle ligand, maleimide-activated full-length human transferrin (hTf) was covalently attached to cysteine-modified Ad serotype 5 vectors either to its fiber or hexon protein. In transcytosis experiments, hTf-coupled vectors were shown to be redirected across the BBB models, the transcytosis activity of the vectors being dependent on the location of the capsid modification and the in vitro model used. The transduction efficiency of hTf-targeted vectors decreased significantly in confluent, polarized cells, indicating that the intracellular route of the vectors differed between unpolarized and polarized cells. After transcellular delivery the majority of the hTf-modified vectors remained intact and partly capable of gene transfer. Altogether, our results demonstrate that i) covalent attachment of a ligand to Ad capsid can mediate transcellular targeting across the cerebral endothelium in vitro, ii) the attachment site of the ligand influences its transcytosis efficiency and iii) combined genetic/chemical modification of Ad vector can be used as a versatile platform for the development of Ad vectors for transcellular targeting.
Collapse
|
35
|
Urban A, Rossier J. Genetic targeting of specific neuronal cell types in the cerebral cortex. PROGRESS IN BRAIN RESEARCH 2012; 196:163-92. [PMID: 22341326 DOI: 10.1016/b978-0-444-59426-6.00009-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Understanding the structure and function of cortical circuits requires the identification of and control over specific cell types in the cortex. To address these obstacles, recent optogenetic approaches have been developed. The capacity to activate, silence, or monitor specific cell types by combining genetics, virology, and optics will decipher the role of specific groups of neurons within circuits with a spatiotemporal resolution that overcomes standard approaches. In this review, the various strategies for selective genetic targeting of a defined neuronal population are discussed as well as the pros and cons of the use of transgenic animals and recombinant viral vectors for the expression of transgenes in a specific set of neurons.
Collapse
Affiliation(s)
- Alan Urban
- Laboratoire de Neurobiologie et Diversité Cellulaire, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7637, Ecole Supérieure de Physique et de Chimie Industrielles, Paris, France.
| | | |
Collapse
|
36
|
|
37
|
Pfrieger FW, Slezak M. Genetic approaches to study glial cells in the rodent brain. Glia 2011; 60:681-701. [PMID: 22162024 DOI: 10.1002/glia.22283] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 11/18/2011] [Indexed: 01/02/2023]
Abstract
The development, function, and pathology of the brain depend on interactions of neurons and different types of glial cells, namely astrocytes, oligodendrocytes, microglia, and ependymal cells. Understanding neuron-glia interactions in vivo requires dedicated experimental approaches to manipulate each cell type independently. In this review, we first summarize techniques that allow for cell-specific gene modification including targeted mutagenesis and viral transduction. In the second part, we describe the genetic models that allow to target the main glial cell types in the central nervous system. The existing arsenal of approaches to study glial cells in vivo and its expansion in the future are key to understand neuron-glia interactions under normal and pathologic conditions.
Collapse
Affiliation(s)
- Frank W Pfrieger
- CNRS UPR 3212, University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI), 67084 Strasbourg, France.
| | | |
Collapse
|
38
|
Louboutin JP, Reyes BAS, Agrawal L, Van Bockstaele EJ, Strayer DS. Intracisternal rSV40 administration provides effective pan-CNS transgene expression. Gene Ther 2011; 19:114-8. [PMID: 21614027 DOI: 10.1038/gt.2011.75] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Potential genetic treatments for many generalized central nervous system (CNS) diseases require transgene expression throughout the CNS. Using oxidant stress and apoptosis caused by HIV-1 envelope gp120 as a model, we studied pan-CNS neuroprotective gene delivery into the cisterna magna (CM). Recombinant SV40 vectors carrying Cu/Zn superoxide dismutase or glutathione peroxidase were injected into rat CMs following intraperitoneal administration of mannitol. Sustained transgene expression was seen in neurons throughout the CNS. On challenge, 8 weeks later with gp120 injected into the caudate putamen, significant neuroprotection was documented. Thus, intracisternal administration of antioxidant-carrying rSV40 vectors may be useful in treating widespread CNS diseases such as HIV-1-associated neurocognitive disorders characterized by oxidative stress.
Collapse
Affiliation(s)
- J-P Louboutin
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| | | | | | | | | |
Collapse
|
39
|
Louboutin JP, Reyes BAS, Van Bockstaele EJ, Strayer DS. Gene transfer to the cerebellum. THE CEREBELLUM 2011; 9:587-97. [PMID: 20700772 DOI: 10.1007/s12311-010-0202-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There are several diseases for which gene transfer therapy to the cerebellum might be practicable. In these studies, we used recombinant Tag-deleted SV40-derived vectors (rSV40s) to study gene delivery targeting the cerebellum. These vectors transduce neurons and microglia very effectively in vitro and in vivo, and so we tested them to evaluate gene transfer to the cerebellum in vivo. Using a rSV40 vector carrying human immunodeficiency virus (HIV)-Nef with a C-terminal FLAG epitope, we characterized the distribution, duration, and cell types transduced. Rats received test and control vectors by stereotaxic injection into the cerebellum. Transgene expression was assessed 1, 2, and 4 weeks later by immunostaining of serial brain sections. FLAG epitope-expressing cells were seen, at all times after vector administration, principally detected in the Purkinje cells of the cerebellum, identified as immunopositive for calbindin. Occasional microglial cells were tranduced; transgene expression was not detected in astrocytes or oligodendrocytes. No inflammatory or other reaction was detected at any time. Thus, SV40-derived vectors can deliver effective, safe, and durable transgene expression to the cerebellum.
Collapse
Affiliation(s)
- Jean-Pierre Louboutin
- Department of Pathology, Jefferson Medical College, Thomas Jefferson University, 1020 Locust Street, Room 255, Philadelphia, PA 19107, USA.
| | | | | | | |
Collapse
|
40
|
Nomura T, Göritz C, Catchpole T, Henkemeyer M, Frisén J. EphB signaling controls lineage plasticity of adult neural stem cell niche cells. Cell Stem Cell 2011; 7:730-43. [PMID: 21112567 DOI: 10.1016/j.stem.2010.11.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 07/22/2010] [Accepted: 08/27/2010] [Indexed: 11/29/2022]
Abstract
Stem cells remain in specialized niches over the lifespan of the organism in many organs to ensure tissue homeostasis and enable regeneration. How the niche is maintained is not understood, but is probably as important as intrinsic stem cell self-renewal capacity for tissue integrity. We here demonstrate a high degree of phenotypic plasticity of the two main niche cell types, ependymal cells and astrocytes, in the neurogenic lateral ventricle walls in the adult mouse brain. In response to a lesion, astrocytes give rise to ependymal cells and ependymal cells give rise to niche astrocytes. We identify EphB2 forward signaling as a key pathway regulating niche cell plasticity. EphB2 acts downstream of Notch and is required for the maintenance of ependymal cell characteristics, thereby inhibiting the transition from ependymal cell to astrocyte. Our results show that niche cell identity is actively maintained and that niche cells retain a high level of plasticity.
Collapse
Affiliation(s)
- Tadashi Nomura
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
41
|
Comparison of the efficacy of four viral vectors for transducing hypothalamic magnocellular neurosecretory neurons in the rat supraoptic nucleus. J Neurosci Methods 2011; 197:238-48. [PMID: 21392530 DOI: 10.1016/j.jneumeth.2011.02.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 02/09/2011] [Accepted: 02/24/2011] [Indexed: 02/08/2023]
Abstract
Since transgenes were first cloned into recombinant adenoviruses almost 30 years ago, a variety of viral vectors have become important tools in genetic research. Viruses adeptly transport genetic material into eukaryotic cells, and replacing all or part of the viral genome with genes of interest or silencing sequences creates a method of gene expression modulation in which the timing and location of manipulations can be specific. The hypothalamo-neurohypophyseal system (HNS), consisting of the paraventricular (PVN) and supraoptic (SON) nuclei in the hypothalamus, regulates fluid balance homeostasis and is highly plastic, yet tightly regulated by extracellular fluid (ECF) osmolality and volume. Its reversible plasticity and physiological relevance make it a good system for studying interactions between gene expression and physiology. Here, four viral vectors were compared for their ability to transduce magnocellular neurosecretory neurons (MNCs) of the SON in adult rats. The vectors included an adenovirus, a lentivirus (HIV) and two serotypes of adeno-associated viruses (AAV5 and AAV2). Though adenovirus and AAV2 vectors have previously been used to transduce SON neurons, HIV and AAV5 have not. All four vectors transduced MNCs, but the AAV vectors were the most effective, transducing large numbers of MNCs, with minimal or no glial transduction. The AAV vectors were injected using a convection enhanced delivery protocol to maximize dispersal through the tissue, resulting in the transduction of neurons throughout the anterior to posterior length of the SON (∼1.5mm). AAV5, but not AAV2, showed some selectivity for SON neurons relative to those in the surrounding hypothalamus.
Collapse
|
42
|
Louboutin JP, Marusich E, Fisher-Perkins J, Dufour JP, Bunnell BA, Strayer DS. Gene transfer to the rhesus monkey brain using SV40-derived vectors is durable and safe. Gene Ther 2011; 18:682-91. [DOI: 10.1038/gt.2011.13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
43
|
Development of advanced therapies based on viral vector-mediated overexpression of therapeutic molecules and knockdown of disease-related genes for Parkinson’s disease. Ther Deliv 2011; 2:37-50. [DOI: 10.4155/tde.10.95] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The last decade witnessed the translation of several gene-based therapeutic approaches from experimental studies to early clinical trials. Studies targeting the treatment of Parkinson’s disease (PD) were among the forefront of trials in the CNS. In this article, we overview three major strategies for the treatment of PD: the enzyme-replacement strategies are based on well-defined principles of functional restoration and are well suited for treatment of patients with advanced disease who would typically experience complications due to side effects of pharmacotherapy. Neurotrophic factor delivery, on the other hand, aims to delay the disability and eventually modifiy disease progression. Finally, we present an outlook to a completely new way of interfering with the disease process, which is taking advantage of recently discovered RNAi mechanisms in cells. Gene therapy is now becoming a reality in the clinics and developments in the next decade will help uncover the true potential of this approach for not only the treatment of PD patients, but also many other neurological disorders.
Collapse
|
44
|
Lewis TB, Glasgow JN, Glandon AM, Curiel DT, Standaert DG. Transduction of brain dopamine neurons by adenoviral vectors is modulated by CAR expression: rationale for tropism modified vectors in PD gene therapy. PLoS One 2010; 5. [PMID: 20862245 PMCID: PMC2941453 DOI: 10.1371/journal.pone.0012672] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 08/18/2010] [Indexed: 01/01/2023] Open
Abstract
Background Gene-based therapy is a new paradigm for the treatment of Parkinson disease (PD) and offers considerable promise for precise targeting and flexibility to impact multiple pathobiological processes for which small molecule agents are not available. Some success has been achieved utilizing adeno-associated virus for this approach, but it is likely that the characteristics of this vector system will ultimately create barriers to progress in clinical therapy. Adenovirus (Ad) vector overcomes limitations in payload size and targeting. The cellular tropism of Ad serotype 5 (Ad5)–based vectors is regulated by the Ad attachment protein binding to its primary cellular receptor, the coxsackie and adenovirus receptor (CAR). Many clinically relevant tissues are refractory to Ad5 infection due to negligible CAR levels but can be targeted by tropism-modified, CAR-independent forms of Ad. Our objective was to evaluate the role of CAR protein in transduction of dopamine (DA) neurons in vivo. Methodology/Principal Findings Ad5 was delivered to the substantia nigra (SN) in wild type (wt) and CAR transgenic animals. Cellular tropism was assessed by immunohistochemistry (IHC) in the SN and striatal terminals. CAR expression was assessed by western blot and IHC. We found in wt animals, Ad5 results in robust transgene expression in astrocytes and other non-neuronal cells but poor infection of DA neurons. In contrast, in transgenic animals, Ad5 infects SNc neurons resulting in expression of transduced protein in their striatal terminals. Western blot showed low CAR expression in the ventral midbrain of wt animals compared to transgenic animals. Interestingly, hCAR protein localizes with markers of post-synaptic structures, suggesting synapses are the point of entry into dopaminergic neurons in transgenic animals. Conclusions/Significance These findings demonstrate that CAR deficiency limits infection of wild type DA neurons by Ad5 and provide a rationale for the development of tropism-modified, CAR-independent Ad-vectors for use in gene therapy of human PD.
Collapse
Affiliation(s)
- Travis B. Lewis
- Department of Cell Biology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, and Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Joel N. Glasgow
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, and Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Anya M. Glandon
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - David T. Curiel
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, and Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - David G. Standaert
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
45
|
Lacar B, Young SZ, Platel JC, Bordey A. Imaging and recording subventricular zone progenitor cells in live tissue of postnatal mice. Front Neurosci 2010; 4:43. [PMID: 20700392 PMCID: PMC2918349 DOI: 10.3389/fnins.2010.00043] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2010] [Accepted: 06/08/2010] [Indexed: 01/30/2023] Open
Abstract
The subventricular zone (SVZ) is one of two regions where neurogenesis persists in the postnatal brain. The SVZ, located along the lateral ventricle, is the largest neurogenic zone in the brain that contains multiple cell populations including astrocyte-like cells and neuroblasts. Neuroblasts migrate in chains to the olfactory bulb where they differentiate into interneurons. Here, we discuss the experimental approaches to record the electrophysiology of these cells and image their migration and calcium activity in acute slices. Although these techniques were in place for studying glial cells and neurons in mature networks, the SVZ raises new challenges due to the unique properties of SVZ cells, the cellular diversity, and the architecture of the region. We emphasize different methods, such as the use of transgenic mice and in vivo electroporation that permit identification of the different SVZ cell populations for patch clamp recording or imaging. Electroporation also permits genetic labeling of cells using fluorescent reporter mice and modification of the system using either RNA interference technology or floxed mice. In this review, we aim to provide conceptual and technical details of the approaches to perform electrophysiological and imaging studies of SVZ cells.
Collapse
Affiliation(s)
- Benjamin Lacar
- Department of Neurosurgery, Yale University School of MedicineNew Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew Haven, CT, USA
| | - Stephanie Z. Young
- Department of Neurosurgery, Yale University School of MedicineNew Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew Haven, CT, USA
| | - Jean-Claude Platel
- Department of Neurosurgery, Yale University School of MedicineNew Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew Haven, CT, USA
| | - Angélique Bordey
- Department of Neurosurgery, Yale University School of MedicineNew Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew Haven, CT, USA
| |
Collapse
|
46
|
Noninvasive imaging of lipid nanoparticle-mediated systemic delivery of small-interfering RNA to the liver. Mol Ther 2010; 18:1657-66. [PMID: 20628357 DOI: 10.1038/mt.2010.147] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Mouse models with liver-specific expression of firefly luciferase were developed that enable a noninvasive and longitudinal assessment of small-interfering RNA (siRNA)-mediated gene silencing in hepatocytes of live animals via bioluminescence imaging. Using these models, a set of lipid nanoparticles (LNPs) with different compositions of cationic lipids, polyethylene glycol (PEG), and cholesterol, were tested for their abilities in delivering a luciferase siRNA to the liver via systemic administration. A dose-dependent luciferase knockdown by LNP/siRNA assemblies was measured by in vivo bioluminescence imaging, which correlated well with the results from parallel ex vivo analyses of luciferase mRNA and protein levels in the liver. RNA interference (RNAi)-mediated target silencing was further confirmed by the detection of RNAi-specific target mRNA cleavage. A single dose of LNP02L at 3 mg/kg (siRNA) caused 90% reduction of luciferase expression and the target repression lasted for at least 10 days. With identical components, LNPs containing 2% PEG are more potent than those with 5.4% PEG. Our results demonstrate that these liver-luciferase mouse models provide a powerful tool for a high-throughput evaluation of hepatic delivery platforms by noninvasive imaging and that the molar ratio of PEG lipid can affect the efficacy of LNPs in silencing liver targets via systemic administration.
Collapse
|
47
|
Abstract
AbstractThe work of Sinden et al. suggests that it may be possible to produce improvement in the “highest” areas of brain function by transplanting brain tissue. What appears to be the limiting factor is not the complexity of the mental process under consideration but the discreteness of the lesion which causes the impairment and the appropriateness and accuracy of placement of the grafted tissue.
Collapse
|
48
|
Abstract
AbstractIn spite of Stein and Glasier's justifiable conclusion that initial optimism concerning the immediate clinical applicability of neural transplantation was premature, there exists much experimental evidence to support the potential for incorporating this procedure into a therapeutic arsenal in the future. To realize this potential will require continued evolution of our knowledge at multiple levels of the clinical and basic neurosciences.
Collapse
|
49
|
Abstract
AbstractThe concept of structure, operation, and functionality, as they may be understood by clinicians or researchers using neural transplantation techniques, are briefly defined. Following Stein & Glasier, we emphasize that the question of whether an intracerebral graft is really functional should be addressed not only in terms of what such a graft does in a given brain structure, but also in terms of what it does at the level of the organism.
Collapse
|
50
|
The NGF superfamily of neurotrophins: Potential treatment for Alzheimer's and Parkinson's disease. Behav Brain Sci 2010. [DOI: 10.1017/s0140525x00037432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AbstractStein & Glasier suggest embryonic neural tissue grafts as a potential treatment strategy for Alzheimer's and Parkinson's disease. As an alternative, we suggest that the family of nerve growth factor-related neurotrophins and their trk (tyrosine kinase) receptors underlie cholinergic basal forebrain (CBF) and dopaminergic substantia nigra neuron degeneration in these diseases, respectively. Therefore, treatment approaches for these disorders could utilize neurotrophins.
Collapse
|