1
|
Xiao W, Li P, Kong F, Kong J, Pan A, Long L, Yan X, Xiao B, Gong J, Wan L. Unraveling the Neural Circuits: Techniques, Opportunities and Challenges in Epilepsy Research. Cell Mol Neurobiol 2024; 44:27. [PMID: 38443733 PMCID: PMC10914928 DOI: 10.1007/s10571-024-01458-5] [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: 12/25/2023] [Accepted: 01/24/2024] [Indexed: 03/07/2024]
Abstract
Epilepsy, a prevalent neurological disorder characterized by high morbidity, frequent recurrence, and potential drug resistance, profoundly affects millions of people globally. Understanding the microscopic mechanisms underlying seizures is crucial for effective epilepsy treatment, and a thorough understanding of the intricate neural circuits underlying epilepsy is vital for the development of targeted therapies and the enhancement of clinical outcomes. This review begins with an exploration of the historical evolution of techniques used in studying neural circuits related to epilepsy. It then provides an extensive overview of diverse techniques employed in this domain, discussing their fundamental principles, strengths, limitations, as well as their application. Additionally, the synthesis of multiple techniques to unveil the complexity of neural circuits is summarized. Finally, this review also presents targeted drug therapies associated with epileptic neural circuits. By providing a critical assessment of methodologies used in the study of epileptic neural circuits, this review seeks to enhance the understanding of these techniques, stimulate innovative approaches for unraveling epilepsy's complexities, and ultimately facilitate improved treatment and clinical translation for epilepsy.
Collapse
Affiliation(s)
- Wenjie Xiao
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Peile Li
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Fujiao Kong
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jingyi Kong
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Lili Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiaoe Gong
- Department of Neurology, Hunan Children's Hospital, Changsha, Hunan Province, China.
| | - Lily Wan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China.
| |
Collapse
|
2
|
Sohn J. Synaptic configuration and reconfiguration in the neocortex are spatiotemporally selective. Anat Sci Int 2024; 99:17-33. [PMID: 37837522 PMCID: PMC10771605 DOI: 10.1007/s12565-023-00743-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/14/2023] [Indexed: 10/16/2023]
Abstract
Brain computation relies on the neural networks. Neurons extend the neurites such as dendrites and axons, and the contacts of these neurites that form chemical synapses are the biological basis of signal transmissions in the central nervous system. Individual neuronal outputs can influence the other neurons within the range of the axonal spread, while the activities of single neurons can be affected by the afferents in their somatodendritic fields. The morphological profile, therefore, binds the functional role each neuron can play. In addition, synaptic connectivity among neurons displays preference based on the characteristics of presynaptic and postsynaptic neurons. Here, the author reviews the "spatial" and "temporal" connection selectivity in the neocortex. The histological description of the neocortical circuitry depends primarily on the classification of cell types, and the development of gene engineering techniques allows the cell type-specific visualization of dendrites and axons as well as somata. Using genetic labeling of particular cell populations combined with immunohistochemistry and imaging at a subcellular spatial resolution, we revealed the "spatial selectivity" of cortical wirings in which synapses are non-uniformly distributed on the subcellular somatodendritic domains in a presynaptic cell type-specific manner. In addition, cortical synaptic dynamics in learning exhibit presynaptic cell type-dependent "temporal selectivity": corticocortical synapses appear only transiently during the learning phase, while learning-induced new thalamocortical synapses persist, indicating that distinct circuits may supervise learning-specific ephemeral synapse and memory-specific immortal synapse formation. The selectivity of spatial configuration and temporal reconfiguration in the neural circuitry may govern diverse functions in the neocortex.
Collapse
Affiliation(s)
- Jaerin Sohn
- Department of Systematic Anatomy and Neurobiology, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
3
|
STLV-1 Commonly Targets Neurons in the Brain of Asymptomatic Non-Human Primates. mBio 2023; 14:e0352622. [PMID: 36802226 PMCID: PMC10128043 DOI: 10.1128/mbio.03526-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
The human T-cell leukemia virus (HTLV)-1 is responsible for an aggressive neurodegenerative disease (HAM/TSP) and multiple neurological alterations. The capacity of HTLV-1 to infect central nervous system (CNS) resident cells, together with the neuroimmune-driven response, has not been well-established. Here, we combined the use of human induced pluripotent stem cells (hiPSC) and of naturally STLV-1-infected nonhuman primates (NHP) as models with which to investigate HTLV-1 neurotropism. Hence, neuronal cells obtained after hiPSC differentiation in neural polycultures were the main cell population infected by HTLV-1. Further, we report the infection of neurons with STLV-1 in spinal cord regions as well as in brain cortical and cerebellar sections of postmortem NHP. Additionally, reactive microglial cells were found in infected areas, suggesting an immune antiviral response. These results emphasize the need to develop new efficient models by which to understand HTLV-1 neuroinfection and suggest an alternative mechanism that leads to HAM/TSP.
Collapse
|
4
|
Molecular profiling of enteric nervous system cell lineages. Nat Protoc 2022; 17:1789-1817. [PMID: 35676375 DOI: 10.1038/s41596-022-00697-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 03/04/2022] [Indexed: 11/08/2022]
Abstract
The enteric nervous system (ENS) is an extensive network of enteric neurons and glial cells that is intrinsic to the gut wall and regulates almost all aspects of intestinal physiology. While considerable advancement has been made in understanding the genetic programs regulating ENS development, there is limited understanding of the molecular pathways that control ENS function in adult stages. One of the limitations in advancing the molecular characterization of the adult ENS relates to technical difficulties in purifying healthy neurons and glia from adult intestinal tissues. To overcome this, we developed novel methods for performing transcriptomic analysis of enteric neurons and glia, which are based on the isolation of fluorescently labeled nuclei. Here we provide a step-by-step protocol for the labeling of adult mouse enteric neuronal nuclei using adeno-associated-virus-mediated gene transfer, isolation of the labeled nuclei by fluorimetric analysis, RNA purification and nuclear RNA sequencing. This protocol has also been adapted for the isolation of enteric neuron and glia nuclei from myenteric plexus preparations from adult zebrafish intestine. Finally, we describe a method for visualization and quantification of RNA in myenteric ganglia: Spatial Integration of Granular Nuclear Signals (SIGNS). By following this protocol, it takes ~3 d to generate RNA and create cDNA libraries for nuclear RNA sequencing and 4 d to carry out high-resolution RNA expression analysis on ENS tissues.
Collapse
|
5
|
Ying Y, Wang JZ. Illuminating Neural Circuits in Alzheimer's Disease. Neurosci Bull 2021; 37:1203-1217. [PMID: 34089505 PMCID: PMC8353043 DOI: 10.1007/s12264-021-00716-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/06/2021] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and there is currently no cure. Neural circuit dysfunction is the fundamental mechanism underlying the learning and memory deficits in patients with AD. Therefore, it is important to understand the structural features and mechanisms underlying the deregulated circuits during AD progression, by which new tools for intervention can be developed. Here, we briefly summarize the most recently established cutting-edge experimental approaches and key techniques that enable neural circuit tracing and manipulation of their activity. We also discuss the advantages and limitations of these approaches. Finally, we review the applications of these techniques in the discovery of circuit mechanisms underlying β-amyloid and tau pathologies during AD progression, and as well as the strategies for targeted AD treatments.
Collapse
Affiliation(s)
- Yang Ying
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
6
|
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
|
7
|
Mohan S, Elhassan Taha MM, Makeen HA, Alhazmi HA, Al Bratty M, Sultana S, Ahsan W, Najmi A, Khalid A. Bioactive Natural Antivirals: An Updated Review of the Available Plants and Isolated Molecules. Molecules 2020; 25:E4878. [PMID: 33105694 PMCID: PMC7659943 DOI: 10.3390/molecules25214878] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Viral infections and associated diseases are responsible for a substantial number of mortality and public health problems around the world. Each year, infectious diseases kill 3.5 million people worldwide. The current pandemic caused by COVID-19 has become the greatest health hazard to people in their lifetime. There are many antiviral drugs and vaccines available against viruses, but they have many disadvantages, too. There are numerous side effects for conventional drugs, and active mutation also creates drug resistance against various viruses. This has led scientists to search herbs as a source for the discovery of more efficient new antivirals. According to the World Health Organization (WHO), 65% of the world population is in the practice of using plants and herbs as part of treatment modality. Additionally, plants have an advantage in drug discovery based on their long-term use by humans, and a reduced toxicity and abundance of bioactive compounds can be expected as a result. In this review, we have highlighted the important viruses, their drug targets, and their replication cycle. We provide in-depth and insightful information about the most favorable plant extracts and their derived phytochemicals against viral targets. Our major conclusion is that plant extracts and their isolated pure compounds are essential sources for the current viral infections and useful for future challenges.
Collapse
MESH Headings
- Antiviral Agents/chemistry
- Antiviral Agents/classification
- Antiviral Agents/isolation & purification
- Antiviral Agents/therapeutic use
- Betacoronavirus/drug effects
- Betacoronavirus/pathogenicity
- Betacoronavirus/physiology
- COVID-19
- Coronavirus Infections/drug therapy
- Coronavirus Infections/pathology
- Coronavirus Infections/virology
- Drug Discovery
- HIV/drug effects
- HIV/pathogenicity
- HIV/physiology
- HIV Infections/drug therapy
- HIV Infections/pathology
- HIV Infections/virology
- Hepacivirus/drug effects
- Hepacivirus/pathogenicity
- Hepacivirus/physiology
- Hepatitis C, Chronic/drug therapy
- Hepatitis C, Chronic/pathology
- Hepatitis C, Chronic/virology
- Herpes Simplex/drug therapy
- Herpes Simplex/pathology
- Herpes Simplex/virology
- Humans
- Influenza, Human/drug therapy
- Influenza, Human/pathology
- Influenza, Human/virology
- Orthomyxoviridae/drug effects
- Orthomyxoviridae/pathogenicity
- Orthomyxoviridae/physiology
- Pandemics
- Phytochemicals/chemistry
- Phytochemicals/classification
- Phytochemicals/isolation & purification
- Phytochemicals/therapeutic use
- Plants, Medicinal
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/pathology
- Pneumonia, Viral/virology
- SARS-CoV-2
- Simplexvirus/drug effects
- Simplexvirus/pathogenicity
- Simplexvirus/physiology
- Virus Internalization/drug effects
- Virus Replication/drug effects
Collapse
Affiliation(s)
- Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; (M.M.E.T.); (H.A.A.); (A.K.)
| | - Manal Mohamed Elhassan Taha
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; (M.M.E.T.); (H.A.A.); (A.K.)
| | - Hafiz A. Makeen
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Hassan A. Alhazmi
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; (M.M.E.T.); (H.A.A.); (A.K.)
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (M.A.B.); (W.A.); (A.N.)
| | - Mohammed Al Bratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (M.A.B.); (W.A.); (A.N.)
| | - Shahnaz Sultana
- Department of Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Waquar Ahsan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (M.A.B.); (W.A.); (A.N.)
| | - Asim Najmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (M.A.B.); (W.A.); (A.N.)
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; (M.M.E.T.); (H.A.A.); (A.K.)
| |
Collapse
|
8
|
Viruses in connectomics: Viral transneuronal tracers and genetically modified recombinants as neuroscience research tools. J Neurosci Methods 2020; 346:108917. [PMID: 32835704 DOI: 10.1016/j.jneumeth.2020.108917] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/25/2022]
Abstract
Connectomic studies have become 'viral', as viral pathogens have been turned into irreplaceable neuroscience research tools. Highly sensitive viral transneuronal tracing technologies are available, based on the use of alpha-herpesviruses and a rhabdovirus (rabies virus), which function as self-amplifying markers by replicating in recipient neurons. These viruses highly differ with regard to host range, cellular receptors, peripheral uptake, replication, transport direction and specificity. Their characteristics, that make them useful for different purposes, will be highlighted and contrasted. Only transneuronal tracing with rabies virus is entirely specific. The neuroscientist toolbox currently include wild-type alpha-herpesviruses and rabies virus strains enabling polysynaptic tracing of neuronal networks across multiple synapses, as well as genetically modified viral tracers for dual transneuronal tracing, and complementary viral tools including defective and chimeric recombinants that function as single step or monosynaptically restricted tracers, or serve for monitoring and manipulating neuronal activity and gene expression. Methodological issues that are crucial for appropriate use of these technologies will be summarized. Among wild-type and genetically engineered viral tools, rabies virus and chimeric recombinants based on rabies virus as virus backbone are the most powerful, because of the ability of rabies virus to propagate exclusively among connected neurons unidirectionally (retrogradely), without affecting neuronal function. Understanding in depth viral properties is essential for neuroscientists who intend to exploit alpha-herpesviruses, rhabdoviruses or derived recombinants as research tools. Key knowledge will be summarized regarding their cellular receptors, intracellular trafficking and strategies to contrast host defense that explain their different pathophysiology and properties as research tools.
Collapse
|
9
|
Xu X, Holmes TC, Luo MH, Beier KT, Horwitz GD, Zhao F, Zeng W, Hui M, Semler BL, Sandri-Goldin RM. Viral Vectors for Neural Circuit Mapping and Recent Advances in Trans-synaptic Anterograde Tracers. Neuron 2020; 107:1029-1047. [PMID: 32755550 DOI: 10.1016/j.neuron.2020.07.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/23/2020] [Accepted: 07/12/2020] [Indexed: 12/17/2022]
Abstract
Viral tracers are important tools for neuroanatomical mapping and genetic payload delivery. Genetically modified viruses allow for cell-type-specific targeting and overcome many limitations of non-viral tracers. Here, we summarize the viruses that have been developed for neural circuit mapping, and we provide a primer on currently applied anterograde and retrograde viral tracers with practical guidance on experimental uses. We also discuss and highlight key technical and conceptual considerations for developing new safer and more effective anterograde trans-synaptic viral vectors for neural circuit analysis in multiple species.
Collapse
Affiliation(s)
- Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697-1275, USA; Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697-4025, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697-2715, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA.
| | - Todd C Holmes
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697-4560, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Min-Hua Luo
- State Key Laboratory of Virology, Wuhan Institute of Virology, CAS Center for Excellence in Brain Science, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697-4560, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Gregory D Horwitz
- The Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA; Department of Physiology & Biophysics, University of Washington, Seattle, WA 98195, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Fei Zhao
- School of Basic Medical Sciences, Capital Medical University, Beijing 102206, China; Chinese Institute for Brain Research (CIBR), Beijing 102206, China
| | - Wenbo Zeng
- State Key Laboratory of Virology, Wuhan Institute of Virology, CAS Center for Excellence in Brain Science, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697-4560, USA
| | - Bert L Semler
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697-4025, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Rozanne M Sandri-Goldin
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697-4025, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
10
|
A mutant vesicular stomatitis virus with reduced cytotoxicity and enhanced anterograde trans-synaptic efficiency. Mol Brain 2020; 13:45. [PMID: 32197632 PMCID: PMC7085170 DOI: 10.1186/s13041-020-00588-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/12/2020] [Indexed: 12/15/2022] Open
Abstract
Understanding the connecting structure of brain network is the basis to reveal the principle of the brain function and elucidate the mechanism of brain diseases. Trans-synaptic tracing with neurotropic viruses has become one of the most effective technologies to dissect the neural circuits. Although the retrograde trans-synaptic tracing for analyzing the input neural networks with recombinant rabies and pseudorabies virus has been broadly applied in neuroscience, viral tools for analyzing the output neural networks are still lacking. The recombinant vesicular stomatitis virus (VSV) has been used for the mapping of synaptic outputs. However, several drawbacks, including high neurotoxicity and rapid lethality in experimental animals, hinder its application in long-term studies of the structure and function of neural networks. To overcome these limitations, we generated a recombinant VSV with replication-related N gene mutation, VSV-NR7A, and examined its cytotoxicity and efficiency of trans-synaptic spreading. We found that by comparison with the wild-type tracer of VSV, the NR7A mutation endowed the virus lower rate of propagation and cytotoxicity in vitro, as well as significantly reduced neural inflammatory responses in vivo and much longer animal survival when it was injected into the nucleus of the mice brain. Besides, the spreading of the attenuated VSV was delayed when injected into the VTA. Importantly, with the reduced toxicity and extended animal survival, the number of brain regions that was trans-synaptically labeled by the mutant VSV was more than that of the wild-type VSV. These results indicated that the VSV-NR7A, could be a promising anterograde tracer that enables researchers to explore more downstream connections of a given brain region, and observe the anatomical structure and the function of the downstream circuits over a longer time window. Our work could provide an improved tool for structural and functional studies of neurocircuit.
Collapse
|
11
|
Cong W, Shi Y, Qi Y, Wu J, Gong L, He M. Viral approaches to study the mammalian brain: Lineage tracing, circuit dissection and therapeutic applications. J Neurosci Methods 2020; 335:108629. [PMID: 32045571 DOI: 10.1016/j.jneumeth.2020.108629] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/01/2020] [Accepted: 02/04/2020] [Indexed: 02/09/2023]
Abstract
Viral vectors are widely used to study the development, function and pathology of neural circuits in the mammalian brain. Their flexible payloads with customizable choices of tool genes allow versatile applications ranging from lineage tracing, circuit mapping and functional interrogation, to translational and therapeutic applications. Different applications have distinct technological requirements, therefore, often utilize different types of virus. This review introduces the most commonly used viruses for these applications and some recent advances in improving the resolution and throughput of lineage tracing, the efficacy and selectivity of circuit tracing and the specificity of cell type targeting.
Collapse
Affiliation(s)
- Wei Cong
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Shi
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanqing Qi
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinyun Wu
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ling Gong
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Miao He
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Li J, Liu T, Dong Y, Kondoh K, Lu Z. Trans-synaptic Neural Circuit-Tracing with Neurotropic Viruses. Neurosci Bull 2019; 35:909-920. [PMID: 31004271 PMCID: PMC6754522 DOI: 10.1007/s12264-019-00374-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/15/2018] [Indexed: 12/19/2022] Open
Abstract
A central objective in deciphering the nervous system in health and disease is to define the connections of neurons. The propensity of neurotropic viruses to spread among synaptically-linked neurons makes them ideal for mapping neural circuits. So far, several classes of viral neuronal tracers have become available and provide a powerful toolbox for delineating neural networks. In this paper, we review the recent developments of neurotropic viral tracers and highlight their unique properties in revealing patterns of neuronal connections.
Collapse
Affiliation(s)
- Jiamin Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Taian Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yun Dong
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Kunio Kondoh
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institute of Natural Sciences, Myodaiji, Okazaki, Aichi, 444-8585, Japan.
- Japan Science and Technology Agency, PRESTO, Myodaiji, Okazaki, Aichi, 444-8585, Japan.
| | - Zhonghua Lu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
13
|
Beier KT. Hitchhiking on the neuronal highway: Mechanisms of transsynaptic specificity. J Chem Neuroanat 2019; 99:9-17. [PMID: 31075318 PMCID: PMC6701464 DOI: 10.1016/j.jchemneu.2019.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/20/2019] [Accepted: 05/06/2019] [Indexed: 01/28/2023]
Abstract
Transsynaptic viral tracers are an invaluable neuroanatomical tool to define neuronal circuit connectivity across single or multiple synapses. There are variants that label either inputs or outputs of defined starter populations, most of which are based on the herpes and rabies viruses. However, we still have an incomplete understanding of the factors influencing specificity of neuron-neuron transmission and labeling of inputs vs. outputs. This article will touch on three topics: First, how specific are the directional transmission patterns of these viruses? Second, what are the properties that confer synaptic specificity of viral transmission? Lastly, what can we learn from this specificity, and can we use it to devise better transsynaptic tracers?
Collapse
Affiliation(s)
- Kevin T Beier
- Department of Physiology and Biophysics, Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, 92697, United States.
| |
Collapse
|
14
|
Zheng N, Su P, Liu Y, Wang H, Nie B, Fang X, Xu Y, Lin K, Lv P, He X, Guo Y, Shan B, Manyande A, Wang J, Xu F. Detection of neural connections with ex vivo MRI using a ferritin-encoding trans-synaptic virus. Neuroimage 2019; 197:133-142. [DOI: 10.1016/j.neuroimage.2019.04.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/06/2019] [Accepted: 04/11/2019] [Indexed: 12/11/2022] Open
|
15
|
Yamaguchi A, Woller DJ, Rodrigues P. Development of an Acute Method to Deliver Transgenes Into the Brains of Adult Xenopus laevis. Front Neural Circuits 2018; 12:92. [PMID: 30416430 PMCID: PMC6213920 DOI: 10.3389/fncir.2018.00092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 10/03/2018] [Indexed: 01/06/2023] Open
Abstract
The central vocal pathway of the African clawed frog, Xenopus laevis, is a powerful vertebrate model to understand mechanisms underlying central pattern generation. However, fast and efficient methods of introducing exogenous genes into the neurons of adult X. laevis are currently not available. Here, we systematically tested methods of transgene delivery into adult X. laevis neurons. Although successfully used for tadpole neurons for over a decade, electroporation was not efficient in transfecting adult neurons. Similarly, adeno-associated virus (AAV) was not reliable, and lentivirus (LV) failed to function as viral vector in adult Xenopus neurons. In contrast, vesicular stomatitis virus (VSV) was a fast and robust vector for adult X. laevis neurons. Although toxic to the host cells, VSV appears to be less virulent to frog neurons than they are to mice neurons. At a single cell level, infected neurons showed normal physiological properties up to 7 days post infection and vocal circuits that included infected neurons generated normal fictive vocalizations up to 9 days post infection. The relatively long time window during which the physiology of VSV-infected neurons can be studied presents an ideal condition for the use of optogenetic tools. We showed that VSV does not gain entry into myelinated axons, but is taken up by both the soma and axon terminal; this is an attractive feature that drives transgene expression in projection neurons. Previous studies showed that VSVs can spread across synapses in anterograde or retrograde directions depending on the types of glycoprotein that are encoded. However, rVSV did not spread across synapses in the Xenopus central nervous system. The successful use of VSV as a transgene vector in amphibian brains not only allows us to exploit the full potential of the genetic tools to answer questions central to understanding central pattern generation, but also opens the door to other research programs that focus on non-genetic model organisms to address unique questions.
Collapse
Affiliation(s)
- Ayako Yamaguchi
- Department of Biology, University of Utah, Salt Lake City, UT, United States
| | - Diana J Woller
- Department of Biology, University of Utah, Salt Lake City, UT, United States
| | - Paulo Rodrigues
- Department of Biology, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
16
|
Shiromani PJ, Peever JH. New Neuroscience Tools That Are Identifying the Sleep-Wake Circuit. Sleep 2017; 40:3059391. [PMID: 28329204 DOI: 10.1093/sleep/zsx032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The complexity of the brain is yielding to technology. In the area of sleep neurobiology, conventional neuroscience tools such as lesions, cell recordings, c-Fos, and axon-tracing methodologies have been instrumental in identifying the complex and intermingled populations of sleep- and arousal-promoting neurons that orchestrate and generate wakefulness, NREM, and REM sleep. In the last decade, new technologies such as optogenetics, chemogenetics, and the CRISPR-Cas system have begun to transform how biologists understand the finer details associated with sleep-wake regulation. These additions to the neuroscience toolkit are helping to identify how discrete populations of brain cells function to trigger and shape the timing and transition into and out of different sleep-wake states, and how glia partner with neurons to regulate sleep. Here, we detail how some of the newest technologies are being applied to understand the neural circuits underlying sleep and wake.
Collapse
Affiliation(s)
- Priyattam J Shiromani
- Ralph H. Johnson Veterans Administration Medical Center, Research Service, Charleston, SC
| | - John H Peever
- Centre for Biological Timing and Cognition, Department Cell and Systems Biology, and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Chikungunya, Influenza, Nipah, and Semliki Forest Chimeric Viruses with Vesicular Stomatitis Virus: Actions in the Brain. J Virol 2017; 91:JVI.02154-16. [PMID: 28077641 DOI: 10.1128/jvi.02154-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/02/2017] [Indexed: 02/02/2023] Open
Abstract
Recombinant vesicular stomatitis virus (VSV)-based chimeric viruses that include genes from other viruses show promise as vaccines and oncolytic viruses. However, the critical safety concern is the neurotropic nature conveyed by the VSV glycoprotein. VSVs that include the VSV glycoprotein (G) gene, even in most recombinant attenuated strains, can still show substantial adverse or lethal actions in the brain. Here, we test 4 chimeric viruses in the brain, including those in which glycoprotein genes from Nipah, chikungunya (CHIKV), and influenza H5N1 viruses were substituted for the VSV glycoprotein gene. We also test a virus-like vesicle (VLV) in which the VSV glycoprotein gene is expressed from a replicon encoding the nonstructural proteins of Semliki Forest virus. VSVΔG-CHIKV, VSVΔG-H5N1, and VLV were all safe in the adult mouse brain, as were VSVΔG viruses expressing either the Nipah F or G glycoprotein. In contrast, a complementing pair of VSVΔG viruses expressing Nipah G and F glycoproteins were lethal within the brain within a surprisingly short time frame of 2 days. Intranasal inoculation in postnatal day 14 mice with VSVΔG-CHIKV or VLV evoked no adverse response, whereas VSVΔG-H5N1 by this route was lethal in most mice. A key immune mechanism underlying the safety of VSVΔG-CHIKV, VSVΔG-H5N1, and VLV in the adult brain was the type I interferon response; all three viruses were lethal in the brains of adult mice lacking the interferon receptor, suggesting that the viruses can infect and replicate and spread in brain cells if not blocked by interferon-stimulated genes within the brain.IMPORTANCE Vesicular stomatitis virus (VSV) shows considerable promise both as a vaccine vector and as an oncolytic virus. The greatest limitation of VSV is that it is highly neurotropic and can be lethal within the brain. The neurotropism can be mostly attributed to the VSV G glycoprotein. Here, we test 4 chimeric viruses of VSV with glycoprotein genes from Nipah, chikungunya, and influenza viruses and nonstructural genes from Semliki Forest virus. Two of the four, VSVΔG-CHIKV and VLV, show substantially attenuated neurotropism and were safe in the healthy adult mouse brain. VSVΔG-H5N1 was safe in the adult brain but lethal in the younger brain. VSVΔG Nipah F+G was even more neurotropic than wild-type VSV, evoking a rapid lethal response in the adult brain. These results suggest that while chimeric VSVs show promise, each must be tested with both intranasal and intracranial administration to ensure the absence of lethal neurotropism.
Collapse
|
18
|
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: 120] [Impact Index Per Article: 12.0] [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
|
19
|
de Sousa AA, Dos Reis RR, de Lima CM, de Oliveira MA, Fernandes TN, Gomes GF, Diniz DG, Magalhães NM, Diniz CG, Sosthenes MCK, Bento-Torres J, Diniz JAP, Vasconcelos PFDC, Diniz CWP. Three-dimensional morphometric analysis of microglial changes in a mouse model of virus encephalitis: age and environmental influences. Eur J Neurosci 2015; 42:2036-50. [PMID: 25980955 DOI: 10.1111/ejn.12951] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 04/16/2015] [Accepted: 05/13/2015] [Indexed: 02/05/2023]
Abstract
Many RNA virus CNS infections cause neurological disease. Because Piry virus has a limited human pathogenicity and exercise reduces activation of microglia in aged mice, possible influences of environment and aging on microglial morphology and behavior in mice sublethal encephalitis were investigated. Female albino Swiss mice were raised either in standard (S) or in enriched (EE) cages from age 2 to 6 months (young - Y), or from 2 to 16 months (aged - A). After behavioral tests, mice nostrils were instilled with Piry-virus-infected or with normal brain homogenates. Brain sections were immunolabeled for virus antigens or microglia at 8 days post-infection (dpi), when behavioral changes became apparent, and at 20 and 40 dpi, after additional behavioral testing. Young infected mice from standard (SYPy) and enriched (EYPy) groups showed similar transient impairment in burrowing activity and olfactory discrimination, whereas aged infected mice from both environments (EAPy, SAPy) showed permanent reduction in both tasks. The beneficial effects of an enriched environment were smaller in aged than in young mice. Six-hundred and forty microglial cells, 80 from each group were reconstructed. An unbiased, stereological sampling approach and multivariate statistical analysis were used to search for microglial morphological families. This procedure allowed distinguishing between microglial morphology of infected and control subjects. More severe virus-associated microglial changes were observed in young than in aged mice, and EYPy seem to recover microglial homeostatic morphology earlier than SYPy . Because Piry-virus encephalitis outcomes were more severe in aged mice, it is suggested that the reduced inflammatory response in those individuals may aggravate encephalitis outcomes.
Collapse
Affiliation(s)
- Aline A de Sousa
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Renata R Dos Reis
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Camila M de Lima
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Marcus A de Oliveira
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | | | - Giovanni F Gomes
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Daniel G Diniz
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Nara M Magalhães
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Cristovam G Diniz
- Instituto Federal de Educação, Ciência e Tecnologia do Pará, Bragança, Pará, Brazil
| | - Marcia C K Sosthenes
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - João Bento-Torres
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - José Antonio P Diniz
- Instituto Evandro Chagas (IEC), Departamento de Arbovirologia e Febres Hemorrágicas, Ananindeua, Pará, Brazil
| | - Pedro F da C Vasconcelos
- Instituto Evandro Chagas (IEC), Departamento de Arbovirologia e Febres Hemorrágicas, Ananindeua, Pará, Brazil
| | - Cristovam Wanderley P Diniz
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil.,Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| |
Collapse
|
20
|
Mundell NA, Beier KT, Pan YA, Lapan SW, Göz Aytürk D, Berezovskii VK, Wark AR, Drokhlyansky E, Bielecki J, Born RT, Schier AF, Cepko CL. Vesicular stomatitis virus enables gene transfer and transsynaptic tracing in a wide range of organisms. J Comp Neurol 2015; 523:1639-63. [PMID: 25688551 PMCID: PMC4458151 DOI: 10.1002/cne.23761] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/03/2015] [Accepted: 02/10/2015] [Indexed: 12/20/2022]
Abstract
Current limitations in technology have prevented an extensive analysis of the connections among neurons, particularly within nonmammalian organisms. We developed a transsynaptic viral tracer originally for use in mice, and then tested its utility in a broader range of organisms. By engineering the vesicular stomatitis virus (VSV) to encode a fluorophore and either the rabies virus glycoprotein (RABV‐G) or its own glycoprotein (VSV‐G), we created viruses that can transsynaptically label neuronal circuits in either the retrograde or anterograde direction, respectively. The vectors were investigated for their utility as polysynaptic tracers of chicken and zebrafish visual pathways. They showed patterns of connectivity consistent with previously characterized visual system connections, and revealed several potentially novel connections. Further, these vectors were shown to infect neurons in several other vertebrates, including Old and New World monkeys, seahorses, axolotls, and Xenopus. They were also shown to infect two invertebrates, Drosophila melanogaster, and the box jellyfish, Tripedalia cystophora, a species previously intractable for gene transfer, although no clear evidence of transsynaptic spread was observed in these species. These vectors provide a starting point for transsynaptic tracing in most vertebrates, and are also excellent candidates for gene transfer in organisms that have been refractory to other methods. J. Comp. Neurol. 523:1639–1663, 2015. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nathan A Mundell
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, 02115.,Department of Ophthalmology, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, 02115
| | - Kevin T Beier
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, 02115.,Department of Ophthalmology, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, 02115
| | - Y Albert Pan
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, 01238
| | - Sylvain W Lapan
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, 02115.,Department of Ophthalmology, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, 02115
| | - Didem Göz Aytürk
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, 02115.,Department of Ophthalmology, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, 02115
| | | | - Abigail R Wark
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, 02115
| | - Eugene Drokhlyansky
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, 02115.,Department of Ophthalmology, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, 02115
| | - Jan Bielecki
- Department of Ecology, Evolution and Marine Biology, University of California, Santa Barbara, Santa Barbara, California, 93106
| | - Richard T Born
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, 02115
| | - Alexander F Schier
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, 01238
| | - Constance L Cepko
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, 02115.,Department of Ophthalmology, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, 02115
| |
Collapse
|
21
|
Wollmann G, Paglino JC, Maloney PR, Ahmadi SA, van den Pol AN. Attenuation of vesicular stomatitis virus infection of brain using antiviral drugs and an adeno-associated virus-interferon vector. Virology 2014; 475:1-14. [PMID: 25462341 DOI: 10.1016/j.virol.2014.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/04/2014] [Accepted: 10/20/2014] [Indexed: 12/21/2022]
Abstract
Vesicular stomatitis virus (VSV) shows promise as a vaccine-vector and oncolytic virus. However, reports of neurotoxicity of VSV remain a concern. We compared 12 antiviral compounds to control infection of VSV-CT9-M51 and VSV-rp30 using murine and human brain cultures, and in vivo mouse models. Inhibition of replication, cytotoxicity and infectivity was strongest with ribavirin and IFN-α and to some extent with mycophenolic acid, chloroquine, and adenine 9-β-d-arabinofuranoside. To generate continuous IFN exposure, we made an adeno-associated virus vector expressing murine IFN; AAV-mIFN-β protected mouse brain cells from VSV, as did a combination of IFN, ribavirin and chloroquine. Intracranial AAV-mIFN-β protected the brain against VSV-CT9-M51. In SCID mice bearing human glioblastoma, AAV-mIFN-β moderately enhanced survival. VSV-CT9-M51 doubled median survival when administered after AAV-mIFN-β; some surviving mice showed complete tumor destruction. Together, these data suggest that AAV-IFN or IFN with ribavirin and chloroquine provide an optimal anti-virus combination against VSV in the brain.
Collapse
Affiliation(s)
- Guido Wollmann
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Justin C Paglino
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Patrick R Maloney
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Sebastian A Ahmadi
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Anthony N van den Pol
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States.
| |
Collapse
|
22
|
Wouterlood FG, Bloem B, Mansvelder HD, Luchicchi A, Deisseroth K. A fourth generation of neuroanatomical tracing techniques: exploiting the offspring of genetic engineering. J Neurosci Methods 2014; 235:331-48. [PMID: 25107853 DOI: 10.1016/j.jneumeth.2014.07.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 07/28/2014] [Accepted: 07/29/2014] [Indexed: 11/18/2022]
Abstract
The first three generations of neuroanatomical tract-tracing methods include, respectively, techniques exploiting degeneration, retrograde cellular transport and anterograde cellular transport. This paper reviews the most recent development in third-generation tracing, i.e., neurochemical fingerprinting based on BDA tracing, and continues with an emerging tracing technique called here 'selective fluorescent protein expression' that in our view belongs to an entirely new 'fourth-generation' class. Tracing techniques in this class lean on gene expression technology designed to 'label' projections exclusively originating from neurons expressing a very specific molecular phenotype. Genetically engineered mice that express cre-recombinase in a neurochemically specific neuronal population receive into a brain locus of interest an injection of an adeno-associated virus (AAV) carrying a double-floxed promoter-eYFP DNA sequence. After transfection this sequence is expressed only in neurons metabolizing recombinase protein. These particular neurons promptly start manufacturing the fluorescent protein which then accumulates and labels to full detail all the neuronal processes, including fibers and terminal arborizations. All other neurons remain optically 'dark'. The AAV is not replicated by the neurons, prohibiting intracerebral spread of 'infection'. The essence is that the fiber projections of discrete subpopulations of neurochemically specific neurons can be traced in full detail. One condition is that the transgenic mouse strain is recombinase-perfect. We illustrate selective fluorescent protein expression in parvalbumin-cre (PV-cre) mice and choline acetyltransferase-cre (ChAT-cre) mice. In addition we compare this novel tracing technique with observations in brains of native PV mice and ChAT-GFP mice. We include a note on tracing techniques using viruses.
Collapse
Affiliation(s)
- Floris G Wouterlood
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, Vrije University Medical Center, Amsterdam, The Netherlands.
| | - Bernard Bloem
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands; Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Antonio Luchicchi
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Karl Deisseroth
- Bioengineering Department, James E. Clark Center, Stanford University, Stanford, CA, USA
| |
Collapse
|
23
|
An anterograde rabies virus vector for high-resolution large-scale reconstruction of 3D neuron morphology. Brain Struct Funct 2014; 220:1369-79. [PMID: 24723034 PMCID: PMC4409643 DOI: 10.1007/s00429-014-0730-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 02/07/2014] [Indexed: 12/19/2022]
Abstract
Glycoprotein-deleted rabies virus (RABV ∆G) is a powerful tool for the analysis of neural circuits. Here, we demonstrate the utility of an anterograde RABV ∆G variant for novel neuroanatomical approaches involving either bulk or sparse neuronal populations. This technology exploits the unique features of RABV ∆G vectors, namely autonomous, rapid high-level expression of transgenes, and limited cytotoxicity. Our vector permits the unambiguous long-range and fine-scale tracing of the entire axonal arbor of individual neurons throughout the brain. Notably, this level of labeling can be achieved following infection with a single viral particle. The vector is effective over a range of ages (>14 months) aiding the studies of neurodegenerative disorders or aging, and infects numerous cell types in all brain regions tested. Lastly, it can also be readily combined with retrograde RABV ∆G variants. Together with other modern technologies, this tool provides new possibilities for the investigation of the anatomy and physiology of neural circuits.
Collapse
|
24
|
Wickersham IR, Sullivan HA, Seung HS. Axonal and subcellular labelling using modified rabies viral vectors. Nat Commun 2014; 4:2332. [PMID: 23945836 PMCID: PMC5939574 DOI: 10.1038/ncomms3332] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 07/19/2013] [Indexed: 01/03/2023] Open
Abstract
An important aspect of any neural circuit is the placement of its output synapses, at levels ranging from macroscopic to subcellular. The many new molecular tools for locating and manipulating synapses are limited by the viral vectors available for delivering them. Adeno-associated viruses are the best current means of labeling and manipulating axons and synapses, but they have never expressed more than one transgene highly enough to label fine axonal structure while also labeling or perturbing synapses. Their slow expression also makes them incompatible with retrograde and transsynaptic vectors, preventing powerful combinatorial experiments. Here we show that deletion-mutant rabies virus can be specifically targeted to cells local to an injection site, brightly labeling axons even when coexpressing two other transgenes. We demonstrate several novel capabilities: simultaneously labeling axons and presynaptic terminals, labeling both dendrites and postsynaptic densities, and simultaneously labeling a region’s inputs and outputs using coinjected vectors.
Collapse
Affiliation(s)
- Ian R Wickersham
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
25
|
Khan AM. Controlling feeding behavior by chemical or gene-directed targeting in the brain: what's so spatial about our methods? Front Neurosci 2013; 7:182. [PMID: 24385950 PMCID: PMC3866545 DOI: 10.3389/fnins.2013.00182] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 09/20/2013] [Indexed: 12/26/2022] Open
Abstract
Intracranial chemical injection (ICI) methods have been used to identify the locations in the brain where feeding behavior can be controlled acutely. Scientists conducting ICI studies often document their injection site locations, thereby leaving kernels of valuable location data for others to use to further characterize feeding control circuits. Unfortunately, this rich dataset has not yet been formally contextualized with other published neuroanatomical data. In particular, axonal tracing studies have delineated several neural circuits originating in the same areas where ICI injection feeding-control sites have been documented, but it remains unclear whether these circuits participate in feeding control. Comparing injection sites with other types of location data would require careful anatomical registration between the datasets. Here, a conceptual framework is presented for how such anatomical registration efforts can be performed. For example, by using a simple atlas alignment tool, a hypothalamic locus sensitive to the orexigenic effects of neuropeptide Y (NPY) can be aligned accurately with the locations of neurons labeled by anterograde tracers or those known to express NPY receptors or feeding-related peptides. This approach can also be applied to those intracranial "gene-directed" injection (IGI) methods (e.g., site-specific recombinase methods, RNA expression or interference, optogenetics, and pharmacosynthetics) that involve viral injections to targeted neuronal populations. Spatial alignment efforts can be accelerated if location data from ICI/IGI methods are mapped to stereotaxic brain atlases to allow powerful neuroinformatics tools to overlay different types of data in the same reference space. Atlas-based mapping will be critical for community-based sharing of location data for feeding control circuits, and will accelerate our understanding of structure-function relationships in the brain for mammalian models of obesity and metabolic disorders.
Collapse
Affiliation(s)
- Arshad M. Khan
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El PasoEl Paso, TX, USA
- Neurobiology Section, Department of Biological Sciences, University of Southern CaliforniaLos Angeles, CA, USA
| |
Collapse
|
26
|
Hyrc KL, Minta A, Escamilla PR, Chan PPL, Meshik XA, Goldberg MP. Synthesis and properties of Asante Calcium Red--a novel family of long excitation wavelength calcium indicators. Cell Calcium 2013; 54:320-33. [PMID: 24017967 DOI: 10.1016/j.ceca.2013.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 07/29/2013] [Accepted: 08/01/2013] [Indexed: 10/26/2022]
Abstract
Although many synthetic calcium indicators are available, a search for compounds with improved characteristics continues. Here, we describe the synthesis and properties of Asante Calcium Red-1 (ACR-1) and its low affinity derivative (ACR-1-LA) created by linking BAPTA to seminaphthofluorescein. The indicators combine a visible light (450-540 nm) excitation with deep-red fluorescence (640 nm). Upon Ca2+ binding, the indicators raise their fluorescence with longer excitation wavelengths producing higher responses. Although the changes occur without any spectral shifts, it is possible to ratio Ca(2+)-dependent (640 nm) and quasi-independent (530 nm) emission when using visible (< 490 nm) or multiphoton (∼780 nm) excitation. Therefore, both probes can be used as single wavelength or, less dynamic, ratiometric indicators. Long indicator emission might allow easy [Ca2+]i measurement in GFP expressing cells. The indicators bind Ca2+ with either high (Kd = 0.49 ± 0.07 μM; ACR-1) or low affinity (Kd = 6.65 ± 0.13 μM; ACR-1-LA). Chelating Zn2+ (Kd = 0.38 ± 0.02 nM) or Mg2+ (Kd∼5mM) slightly raises and binding Co2+ quenches dye fluorescence. New indicators are somewhat pH-sensitive (pKa = 6.31 ± 0.07), but fairly resistant to bleaching. The probes are rather dim, which combined with low AM ester loading efficiency, might complicate in situ imaging. Despite potential drawbacks, ACR-1 and ACR-1-LA are promising new calcium indicators.
Collapse
Affiliation(s)
- Krzysztof L Hyrc
- The Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA; Alafi Neuroimaging Laboratory, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Beier KT, Saunders AB, Oldenburg IA, Sabatini BL, Cepko CL. Vesicular stomatitis virus with the rabies virus glycoprotein directs retrograde transsynaptic transport among neurons in vivo. Front Neural Circuits 2013; 7:11. [PMID: 23403489 PMCID: PMC3566411 DOI: 10.3389/fncir.2013.00011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 01/20/2013] [Indexed: 12/24/2022] Open
Abstract
Defining the connections among neurons is critical to our understanding of the structure and function of the nervous system. Recombinant viruses engineered to transmit across synapses provide a powerful approach for the dissection of neuronal circuitry in vivo. We recently demonstrated that recombinant vesicular stomatitis virus (VSV) can be endowed with anterograde or retrograde transsynaptic tracing ability by providing the virus with different glycoproteins. Here we extend the characterization of the transmission and gene expression of recombinant VSV (rVSV) with the rabies virus glycoprotein (RABV-G), and provide examples of its activity relative to the anterograde transsynaptic tracer form of rVSV. rVSV with RABV-G was found to drive strong expression of transgenes and to spread rapidly from neuron to neuron in only a retrograde manner. Depending upon how the RABV-G was delivered, VSV served as a polysynaptic or monosynaptic tracer, or was able to define projections through axonal uptake and retrograde transport. In animals co-infected with rVSV in its anterograde form, rVSV with RABV-G could be used to begin to characterize the similarities and differences in connections to different areas. rVSV with RABV-G provides a flexible, rapid, and versatile tracing tool that complements the previously described VSV-based anterograde transsynaptic tracer.
Collapse
Affiliation(s)
- Kevin T Beier
- Department of Genetics and Department of Ophthalmology, Harvard Medical School, Harvard University and Howard Hughes Medical Institute Boston, MA, USA
| | | | | | | | | |
Collapse
|
28
|
Polymer Complexes in Biological Applications. FROM SINGLE MOLECULES TO NANOSCOPICALLY STRUCTURED MATERIALS 2013. [DOI: 10.1007/12_2013_229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
Ponzio TA, Fields RL, Rashid OM, Salinas YD, Lubelski D, Gainer H. Cell-type specific expression of the vasopressin gene analyzed by AAV mediated gene delivery of promoter deletion constructs into the rat SON in vivo. PLoS One 2012; 7:e48860. [PMID: 23155418 PMCID: PMC3498266 DOI: 10.1371/journal.pone.0048860] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 10/02/2012] [Indexed: 11/23/2022] Open
Abstract
The magnocellular neurons (MCNs) in the supraoptic nucleus (SON) of the hypothalamus selectively express either oxytocin (Oxt) or vasopressin (Avp) neuropeptide genes. In this paper we examine the cis-regulatory domains in the Avp gene promoter that are responsible for its cell-type specific expression. AAV vectors that contain various Avp gene promoter deletion constructs using EGFP as the reporter were stereotaxically injected into the rat SON. Two weeks following the injection immunohistochemical assays of EGFP expression from these constructs were done to determine whether the expressed EGFP reporter co-localizes with either the Oxt- or Avp-immunoreactivity in the MCNs. The results identify three major enhancer domains located at −2.0 to −1.5 kbp, −1.5 to −950 bp, and −950 to −543 bp in the Avp gene promoter that regulate the expression in Avp MCNs. The results also show that cell–type specific expression in Avp MCNs is maintained in constructs containing at least 288 bp of the promoter region upstream of the transcription start site, but this specificity is lost at 116 bp and below. Based on these data, we hypothesize that the −288 bp to −116 bp domain contains an Avp MCN specific activator and a possible repressor that inhibits expression in Oxt-MCNs, thereby leading to the cell-type specific expression of the Avp gene only in the Avp-MCNs.
Collapse
Affiliation(s)
| | | | | | | | | | - Harold Gainer
- Laboratory of Neurochemistry, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
30
|
Highly attenuated recombinant vesicular stomatitis virus VSV-12'GFP displays immunogenic and oncolytic activity. J Virol 2012; 87:1019-34. [PMID: 23135719 DOI: 10.1128/jvi.01106-12] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vesicular stomatitis virus (VSV) has shown considerable promise both as an immunization vector and as an oncolytic virus. In both applications, an important concern is the safety profile of the virus. To generate a highly attenuated virus, we added two reporter genes to the 3' end of the VSV genome, thereby shifting the NPMGL genes from positions 1 to 5 to positions 3 to 7. The resulting virus (VSV-12'GFP) was highly attenuated, generating smaller plaques than four other attenuated VSVs. In one-step growth curves, VSV-12'GFP displayed the slowest growth kinetics. The mechanism of attenuation appears to be due to reduced expression of VSV genes downstream of the reporter genes, as suggested by a 10.4-fold reduction in L-protein RNA transcript. Although attenuated, VSV-12'GFP was highly effective at generating an immune response, indicated by a high-titer antibody response against the green fluorescent protein (GFP) expressed by the virus. Although VSV-12'GFP was more attenuated than other VSVs on both normal and cancer cells, it nonetheless showed a greater level of infection of human cancer cells (glioma and melanoma) than of normal cells, and this effect was magnified in glioma by interferon application, indicating selective oncolysis. Intravenous VSV-12'GFP selectively infected human gliomas implanted into SCID mice subcutaneously or intracranially. All postnatal day 16 mice given intranasal VSV-12'GFP survived, whereas only 10% of those given VSV-G/GFP survived, indicating reduced neurotoxicity. Intratumoral injection of tumors with VSV-12'GFP dramatically suppressed tumor growth and enhanced survival. Together these data suggest this recombinant virus merits further study for its oncolytic and vaccine potential.
Collapse
|
31
|
Abstract
Classical methods for studying neuronal circuits are fairly low throughput. Transsynaptic viruses, particularly the pseudorabies (PRV) and rabies virus (RABV), and more recently vesicular stomatitis virus (VSV), for studying circuitry, is becoming increasingly popular. These higher throughput methods use viruses that transmit between neurons in either the anterograde or retrograde direction. Recently, a modified RABV for monosynaptic retrograde tracing was developed. (Figure 1A). In this method, the glycoprotein (G) gene is deleted from the viral genome, and resupplied only in targeted neurons. Infection specificity is achieved by substituting a chimeric G, composed of the extracellular domain of the ASLV-A glycoprotein and the cytoplasmic domain of the RABV-G (A/RG), for the normal RABV-G(1). This chimeric G specifically infects cells expressing the TVA receptor(1). The gene encoding TVA can been delivered by various methods(2-8). Following RABV-G infection of a TVA-expressing neuron, the RABV can transmit to other, synaptically connected neurons in a retrograde direction by nature of its own G which was co-delivered with the TVA receptor. This technique labels a relatively large number of inputs (5-10%)(2) onto a defined cell type, providing a sampling of all of the inputs onto a defined starter cell type. We recently modified this technique to use VSV as a transsynaptic tracer(9). VSV has several advantages, including the rapidity of gene expression. Here we detail a new viral tracing system using VSV useful for probing microcircuitry with increased resolution. While the original published strategies by Wickersham et al.(4) and Beier et al.(9) permit labeling of any neurons that project onto initially-infected TVA-expressing-cells, here VSV was engineered to transmit only to TVA-expressing cells (Figure 1B). The virus is first pseudotyped with RABV-G to permit infection of neurons downstream of TVA-expressing neurons. After infecting this first population of cells, the virus released can only infect TVA-expressing cells. Because the transsynaptic viral spread is limited to TVA-expressing cells, presence of absence of connectivity from defined cell types can be explored with high resolution. An experimental flow chart of these experiments is shown in Figure 2. Here we show a model circuit, that of direction-selectivity in the mouse retina. We examine the connectivity of starburst amacrine cells (SACs) to retinal ganglion cells (RGCs).
Collapse
Affiliation(s)
- Kevin Beier
- Department of Genetics, Harvard Medical School, USA
| | | |
Collapse
|
32
|
Du Z, Whitt MA, Baumann J, Garner JM, Morton CL, Davidoff AM, Pfeffer LM. Inhibition of type I interferon-mediated antiviral action in human glioma cells by the IKK inhibitors BMS-345541 and TPCA-1. J Interferon Cytokine Res 2012; 32:368-77. [PMID: 22509977 DOI: 10.1089/jir.2012.0002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The nuclear factor-kappa B (NFκB) signal transduction pathway plays an important role in immunity, inflammation, cell growth, and survival. Since dysregulation of this pathway results in high, constitutive NFκB activation in various cancers and immune disorders, the development of specific drugs to target this pathway has become a focus for treating these diseases. NFκB regulates various aspects of the cellular response to interferon (IFN). However, the role of the upstream regulator of the NFκB signaling pathway, the inhibitor of κB kinase (IKK) complex, on IFN function has not been examined. In the present study, we examined the effects of 2 IKK inhibitors, N-(1,8-Dimethylimidazo[1,2-a]quinoxalin-4-yl)-1,2-ethanediamine hydrochloride (BMS-345541) and 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA-1), on IFN action in several human glioma cell lines. IKK inhibitors inhibit glioma cell proliferation, as well as TNF-induced RelA (p65) nuclear translocation and NFκB-dependent IL8 gene expression. Importantly, BMS-345541 and TPCA-1 differentially inhibit IFN-induced gene expression, completely suppressing MX1 and GBP1 gene expression, while having only a minor effect on ISG15 expression. Furthermore, these IKK inhibitors displayed marked differences in blocking IFN-induced antiviral action against cytopathic effects and replication of vesicular stomatitis virus (VSV) and encephalomyocarditis virus (EMCV). Our results show that the IKK complex plays an important function in IFN-induced gene expression and antiviral activity. Since VSV and EMCV are oncolytic viruses used in cancer therapy, our results indicate the potential synergy in combining IKK inhibitors with oncolytic viruses.
Collapse
Affiliation(s)
- Ziyun Du
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
The supraoptic nucleus (SON) is a particularly good model for the study of cell-type specific gene expression because it contains two distinct neuronal phenotypes, the oxytocin (OT) and vasopressin (AVP) synthesising magnocellular neurones (MCNs). The MCNs are found in approximately equal numbers and selectively express either the OT or the AVP gene in approximately 97% of the MCN population in the SON. An unresolved issue has been to determine what mechanisms are responsible for the highly selective regulation of the cell-type specific expression of OT and AVP genes in the MCNs. Previous attempts to address this question have used various bioinformatic and molecular approaches, which included using heterologous cell lines to study the putative cis-elements in the OT and AVP genes, and the use of OT and/or AVP transgenes in transgenic rodents. The data from all of the above studies identified a region < 0.6 kbp upstream of OT exon I and approximately 3 kb upstream of AVP exon I as being sufficient to produce cell-specific expression of the OT and AVP genes, respectively, although they failed to identify the specific cis-domains responsible for the MCN-specific gene expression. An alternative experimental approach to perform promoter deletion analysis in vivo (i.e. to use stereotaxic viral vector gene transfer into the SON to further dissect the cis-elements in the OT and AVP genes) will be described here. This in vivo method uses adeno-associated viral (AAV) vectors expressing OT-promoter deletion constructs and utilises the enhanced green fluorescent protein (EGFP) as the reporter. The AAV constructs are stereotaxically injected into the rat brain above the SON and, 2 weeks post injection, the rats are sacrificed and assayed for EGFP expression. Using this method, it has been possible to identify specific regions upstream of the transcription start site in the OT and AVP gene promoters that are responsible for conferring the cell-type specificity of the OT and AVP gene expression in the SON.
Collapse
Affiliation(s)
- H Gainer
- Section on Molecular Neuroscience, Laboratory of Neurochemistry, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
34
|
Fields RL, Ponzio TA, Kawasaki M, Gainer H. Cell-type specific oxytocin gene expression from AAV delivered promoter deletion constructs into the rat supraoptic nucleus in vivo. PLoS One 2012; 7:e32085. [PMID: 22363799 PMCID: PMC3283729 DOI: 10.1371/journal.pone.0032085] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 01/23/2012] [Indexed: 12/13/2022] Open
Abstract
The magnocellular neurons (MCNs) in the hypothalamus selectively express either oxytocin (OXT) or vasopressin (AVP) neuropeptide genes, a property that defines their phenotypes. Here we examine the molecular basis of this selectivity in the OXT MCNs by stereotaxic microinjections of adeno-associated virus (AAV) vectors that contain various OXT gene promoter deletion constructs using EGFP as the reporter into the rat supraoptic nucleus (SON). Two weeks following injection of the AAVs, immunohistochemical assays of EGFP expression from these constructs were done to determine whether the EGFP reporter co-localizes with either the OXT- or AVP-immunoreactivity in the MCNs. The results show that the key elements in the OT gene promoter that regulate the cell-type specific expression the SON are located -216 to -100 bp upstream of the transcription start site. We hypothesize that within this 116 bp domain a repressor exists that inhibits expression specifically in AVP MCNs, thereby leading to the cell-type specific expression of the OXT gene only in the OXT MCNs.
Collapse
Affiliation(s)
| | | | | | - Harold Gainer
- Laboratory of Neurochemistry, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
35
|
New technologies for imaging synaptic partners. Curr Opin Neurobiol 2012; 22:121-7. [PMID: 22221865 DOI: 10.1016/j.conb.2011.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 11/30/2011] [Accepted: 12/01/2011] [Indexed: 12/25/2022]
Abstract
Understanding the brain will require unraveling its synaptic circuitry, but methods that can reliably identify connected neurons are often excruciatingly slow. Although light microscopy can provide much higher throughput, synapses are smaller than the diffraction limit and cannot readily be assigned to particular presynaptic and postsynaptic cells without specialized labeling methods. Here we review the ongoing development of techniques that allow direct imaging of neural networks by specifically marking connected cells or their synapses.
Collapse
|
36
|
Anterograde or retrograde transsynaptic labeling of CNS neurons with vesicular stomatitis virus vectors. Proc Natl Acad Sci U S A 2011; 108:15414-9. [PMID: 21825165 DOI: 10.1073/pnas.1110854108] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
To understand how the nervous system processes information, a map of the connections among neurons would be of great benefit. Here we describe the use of vesicular stomatitis virus (VSV) for tracing neuronal connections in vivo. We made VSV vectors that used glycoprotein (G) genes from several other viruses. The G protein from lymphocytic choriomeningitis virus endowed VSV with the ability to spread transsynaptically, specifically in an anterograde direction, whereas the rabies virus glycoprotein gave a specifically retrograde transsynaptic pattern. The use of an avian G protein fusion allowed specific targeting of cells expressing an avian receptor, which allowed a demonstration of monosynaptic anterograde tracing from defined cells. Synaptic connectivity of pairs of virally labeled cells was demonstrated by using slice cultures and electrophysiology. In vivo infections of several areas in the mouse brain led to the predicted patterns of spread for anterograde or retrograde tracers.
Collapse
|
37
|
de Sousa AA, Reis R, Bento-Torres J, Trévia N, Lins NADA, Passos A, Santos Z, Diniz JAP, Vasconcelos PFDC, Cunningham C, Perry VH, Picanço Diniz CW. Influence of enriched environment on viral encephalitis outcomes: behavioral and neuropathological changes in albino Swiss mice. PLoS One 2011; 6:e15597. [PMID: 21264301 PMCID: PMC3019164 DOI: 10.1371/journal.pone.0015597] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 11/13/2010] [Indexed: 11/25/2022] Open
Abstract
An enriched environment has previously been described as enhancing natural killer cell activity of recognizing and killing virally infected cells. However, the effects of environmental enrichment on behavioral changes in relation to virus clearance and the neuropathology of encephalitis have not been studied in detail. We tested the hypothesis that environmental enrichment leads to less CNS neuroinvasion and/or more rapid viral clearance in association with T cells without neuronal damage. Stereology-based estimates of activated microglia perineuronal nets and neurons in CA3 were correlated with behavioral changes in the Piry rhabdovirus model of encephalitis in the albino Swiss mouse. Two-month-old female mice maintained in impoverished (IE) or enriched environments (EE) for 3 months were behaviorally tested. After the tests, an equal volume of Piry virus (IEPy, EEPy)-infected or normal brain homogenates were nasally instilled. Eight days post-instillation (dpi), when behavioral changes became apparent, brains were fixed and processed to detect viral antigens, activated microglia, perineuronal nets, and T lymphocytes by immuno- or histochemical reactions. At 20 or 40 dpi, the remaining animals were behaviorally tested and processed for the same markers. In IEPy mice, burrowing activity decreased and recovered earlier (8–10 dpi) than open field (20–40 dpi) but remained unaltered in the EEPy group. EEPy mice presented higher T-cell infiltration, less CNS cell infection by the virus and/or faster virus clearance, less microgliosis, and less damage to the extracellular matrix than IEPy. In both EEPy and IEPy animals, CA3 neuronal number remained unaltered. The results suggest that an enriched environment promotes a more effective immune response to clear CNS virus and not at the cost of CNS damage.
Collapse
Affiliation(s)
- Aline Andrade de Sousa
- Universidade Federal do Pará (UFPA), Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Belém, Brazil
| | - Renata Reis
- Universidade Federal do Pará (UFPA), Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Belém, Brazil
| | - João Bento-Torres
- Universidade Federal do Pará (UFPA), Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Belém, Brazil
| | - Nonata Trévia
- Universidade Federal do Pará (UFPA), Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Belém, Brazil
| | - Nara Alves de Almeida Lins
- Universidade Federal do Pará (UFPA), Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Belém, Brazil
| | - Aline Passos
- Universidade Federal do Pará (UFPA), Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Belém, Brazil
| | - Zaire Santos
- Universidade Federal do Pará (UFPA), Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Belém, Brazil
| | | | - Pedro Fernando da Costa Vasconcelos
- Instituto Evandro Chagas (IEC), Departamento de Arbovirologia e Febres Hemorrágicas, Ananindeua, Brazil
- Departamento de Patologia, Universidade do Estado do Pará, Belém, Brazil
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Victor Hugh Perry
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Cristovam Wanderley Picanço Diniz
- Universidade Federal do Pará (UFPA), Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Belém, Brazil
- * E-mail:
| |
Collapse
|
38
|
Wickersham IR, Sullivan HA, Seung HS. Production of glycoprotein-deleted rabies viruses for monosynaptic tracing and high-level gene expression in neurons. Nat Protoc 2010; 5:595-606. [PMID: 20203674 DOI: 10.1038/nprot.2009.248] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recombinant rabies viruses rendered replication-deficient by the deletion of their envelope glycoprotein gene are useful tools for neuroscientists, permitting (1) extraordinarily high transgene expression levels within neurons, (2) retrograde infection of projection neurons through their axon terminals, (3) targeted infection of genetically specified neurons and (4) monosynaptic tracing of neuronal inputs. Here we present a detailed protocol for the production of high-titer and high-purity viral stocks, from initial generation of infectious virus from cDNA through amplification on complementing cell lines, pseudotyping if desired, purification by ultracentrifugation and titering. The procedure requires 3-4 weeks to complete.
Collapse
Affiliation(s)
- Ian R Wickersham
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | | | |
Collapse
|
39
|
Davis JN, van den Pol AN. Viral mutagenesis as a means for generating novel proteins. J Virol 2010; 84:1625-30. [PMID: 19906913 PMCID: PMC2812339 DOI: 10.1128/jvi.01747-09] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Accepted: 11/04/2009] [Indexed: 11/20/2022] Open
Abstract
We demonstrate that a mutation-prone virus engineered to express a foreign gene is an expedient means for generating novel mutant nonviral proteins in mammalian cells. Using vesicular stomatitis virus to express a gene coding for a fluorescent DsRed protein, a number of green mutant variants including a new variant not previously described were rapidly isolated from infected cells, sequenced, and cloned. Similar methods may be useful in the development of physiologically sensitive fluorescent reporter proteins and directed evolution or mutagenesis of proteins in general.
Collapse
Affiliation(s)
- John N. Davis
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Anthony N. van den Pol
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
40
|
Heldt SA, Ressler KJ. The Use of Lentiviral Vectors and Cre/loxP to Investigate the Function of Genes in Complex Behaviors. Front Mol Neurosci 2009; 2:22. [PMID: 20011219 PMCID: PMC2790954 DOI: 10.3389/neuro.02.022.2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 10/11/2009] [Indexed: 11/13/2022] Open
Abstract
The use of conventional knockout technologies has proved valuable for understanding the role of key genes and proteins in development, disease states, and complex behaviors. However, these strategies are limited in that they produce broad changes in gene function throughout the neuroaxis and do little to identify the effects of such changes on neural circuits thought to be involved in distinct functions. Because the molecular functions of genes often depend on the specific neuronal circuit in which they are expressed, restricting gene manipulation to specific brain regions and times may be more useful for understanding gene functions. Conditional gene manipulation strategies offer a powerful alternative. In this report we briefly describe two conditional gene strategies that are increasingly being used to investigate the role of genes in behavior – the Cre/loxP recombination system and lentiviral vectors. Next, we summarize a number of recent experiments which have used these techniques to investigate behavior after spatial and/or temporal and gene manipulation. These conditional gene targeting strategies provide useful tools to study the endogenous mechanisms underlying complex behaviors and to model disease states resulting from aberrant gene expression.
Collapse
Affiliation(s)
- Scott A Heldt
- Howard Hughes Medical Institute, Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University Atlanta, GA, USA
| | | |
Collapse
|
41
|
Some attenuated variants of vesicular stomatitis virus show enhanced oncolytic activity against human glioblastoma cells relative to normal brain cells. J Virol 2009; 84:1563-73. [PMID: 19906910 DOI: 10.1128/jvi.02040-09] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Vesicular stomatitis virus (VSV) has been shown in laboratory studies to be effective against a variety of tumors, including malignant brain tumors. However, attenuation of VSV may be necessary to balance the potential toxicity toward normal cells, particularly when targeting brain tumors. Here we compared 10 recombinant VSV variants resulting from different attenuation strategies. Attenuations included gene shifting (VSV-p1-GFP/RFP), M protein mutation (VSV-M51), G protein cytoplasmic tail truncations (VSV-CT1/CT9), G protein deletions (VSV-dG-GFP/RFP), and combinations thereof (VSV-CT9-M51). Using in vitro viability and replication assays, the VSV variants were grouped into three categories, based on their antitumor activity and non-tumor-cell attenuation. In the first group, wild-type-based VSV-G/GFP, tumor-adapted VSV-rp30, and VSV-CT9 showed a strong antitumor profile but also retained some toxicity toward noncancer control cells. The second group, VSV-CT1, VSV-dG-GFP, and VSV-dG-RFP, had significantly diminished toxicity toward normal cells but showed little oncolytic action. The third group displayed a desired combination of diminished general toxicity and effective antitumor action; this group included VSV-M51, VSV-CT9-M51, VSV-p1-GFP, and VSV-p1-RFP. A member of the last group, VSV-p1-GFP, was then compared in vivo against wild-type-based VSV-G/GFP. Intranasal inoculation of young, postnatal day 16 mice with VSV-p1-GFP showed no adverse neurological effects, whereas VSV-G/GFP was associated with high lethality (80%). Using an intracranial tumor xenograft model, we further demonstrated that attenuated VSV-p1-GFP targets and kills human U87 glioblastoma cells after systemic application. We concluded that some, but not all, attenuated VSV mutants display a favorable oncolytic profile and merit further investigation.
Collapse
|
42
|
Peripheral immunization blocks lethal actions of vesicular stomatitis virus within the brain. J Virol 2009; 83:11540-9. [PMID: 19726512 DOI: 10.1128/jvi.02558-08] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vesicular stomatitis virus (VSV) is the prototype virus for 75 or more negative-strand RNA viruses in the rhabdovirus family. Some of these viruses, including VSV, can cause neurological impairment or death upon brain infection. VSV has shown promise in the prevention and treatment of disease as a vaccine vector and an oncolytic virus, but infection of the brain remains a concern. Three VSV variants, the wild-type-related VSV-G/GFP and two attenuated viruses, VSV-CT1 and VSV-CT9-M51, were compared for neuroinvasiveness and neuromorbidity. In nonimmunized mice, direct VSV-G/GFP injection into the brain invariably resulted in lethal encephalitis; in contrast, partial survival was seen after direct injection of the attenuated VSV strains. In addition, both attenuated VSV strains showed significantly reduced neuroinvasiveness after intranasal inoculation of young postnatal day 16 mice. Of the three tested variants, VSV-CT9-M51 generated the lowest degree of neuropathology. Despite its attenuated state, peripheral inoculations of VSV-CT9-M51 targeted and killed human glioblastoma implanted into the mouse brain. Importantly, we show here that intranasal or intramuscular immunization prevents the lethal effects of subsequent VSV-G/GFP, VSV-CT1, and VSV-CT9-M51 injections into the brain. These results indicate that attenuated recombinant viruses show reduced neurovirulence and that peripheral immunization blocks the lethal actions of all VSVs tested.
Collapse
|