51
|
Bijoch L, Borczyk M, Czajkowski R. Bases of Jerzy Konorski's theory of synaptic plasticity. Eur J Neurosci 2019; 51:1857-1866. [PMID: 31368131 DOI: 10.1111/ejn.14532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/25/2019] [Accepted: 07/22/2019] [Indexed: 02/03/2023]
Affiliation(s)
- Lukasz Bijoch
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Borczyk
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Rafał Czajkowski
- Laboratory of Spatial Memory, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
52
|
Li SJ, Vaughan A, Sturgill JF, Kepecs A. A Viral Receptor Complementation Strategy to Overcome CAV-2 Tropism for Efficient Retrograde Targeting of Neurons. Neuron 2019; 98:905-917.e5. [PMID: 29879392 DOI: 10.1016/j.neuron.2018.05.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 03/30/2018] [Accepted: 05/17/2018] [Indexed: 12/25/2022]
Abstract
Retrogradely transported neurotropic viruses enable genetic access to neurons based on their long-range projections and have become indispensable tools for linking neural connectivity with function. A major limitation of viral techniques is that they rely on cell-type-specific molecules for uptake and transport. Consequently, viruses fail to infect variable subsets of neurons depending on the complement of surface receptors expressed (viral tropism). We report a receptor complementation strategy to overcome this by potentiating neurons for the infection of the virus of interest-in this case, canine adenovirus type-2 (CAV-2). We designed AAV vectors for expressing the coxsackievirus and adenovirus receptor (CAR) throughout candidate projection neurons. CAR expression greatly increased retrograde-labeling rates, which we demonstrate for several long-range projections, including some resistant to other retrograde-labeling techniques. Our results demonstrate a receptor complementation strategy to abrogate endogenous viral tropism and thereby facilitate efficient retrograde targeting for functional analysis of neural circuits.
Collapse
Affiliation(s)
- Shu-Jing Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | | | - Adam Kepecs
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
53
|
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: 1.8] [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
|
54
|
Ren SQ, Li Z, Lin S, Bergami M, Shi SH. Precise Long-Range Microcircuit-to-Microcircuit Communication Connects the Frontal and Sensory Cortices in the Mammalian Brain. Neuron 2019; 104:385-401.e3. [PMID: 31371111 DOI: 10.1016/j.neuron.2019.06.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 05/06/2019] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Abstract
The frontal area of the cerebral cortex provides long-range inputs to sensory areas to modulate neuronal activity and information processing. These long-range circuits are crucial for accurate sensory perception and complex behavioral control; however, little is known about their precise circuit organization. Here we specifically identified the presynaptic input neurons to individual excitatory neuron clones as a unit that constitutes functional microcircuits in the mouse sensory cortex. Interestingly, the long-range input neurons in the frontal but not contralateral sensory area are spatially organized into discrete vertical clusters and preferentially form synapses with each other over nearby non-input neurons. Moreover, the assembly of distant presynaptic microcircuits in the frontal area depends on the selective synaptic communication of excitatory neuron clones in the sensory area that provide inputs to the frontal area. These findings suggest that highly precise long-range reciprocal microcircuit-to-microcircuit communication mediates frontal-sensory area interactions in the mammalian cortex.
Collapse
Affiliation(s)
- Si-Qiang Ren
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center of Biological Structures, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhizhong Li
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Susan Lin
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Matteo Bergami
- University Hospital Cologne, CECAD Research Centre, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Song-Hai Shi
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center of Biological Structures, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
55
|
Deng K, Yang L, Xie J, Tang H, Wu GS, Luo HR. Whole-brain mapping of projection from mouse lateral septal nucleus. Biol Open 2019; 8:bio.043554. [PMID: 31208998 PMCID: PMC6679409 DOI: 10.1242/bio.043554] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The lateral septal nucleus (LS) plays a critical role in emotionality, social behavior and feeding processes, through neural connections with the hippocampus and hypothalamus. We investigated the neural circuits of LS by using herpes simplex virus 1 strain H129 (H129) and pseudorabies virus stain Bartha (PRV). Virus H129 indicates that LS directly projects to some cerebral nuclei (nucleus accumbens, bed nuclei of the stria terminalis and amygdala), part of the hypothalamus (median preoptic, paraventricular, dorsomedial nucleus and lateral area), thalamus (medial habenula, the paraventricular, parataenial and reuniens nuclei, and the medial line nuclei) and the pontine central gray. Then the LS has secondary projections to the CA3 and CA1 field of the hippocampal formation, lateral and medial preoptic area, and the mammillary body. PRV tracing shows that LS are mainly receiving primary inputs from the amygdala, hippocampus, hypothalamic, thalamus, midbrain and hindbrain, and secondary inputs from dorsal and central linear nucleus raphe, the lateral part of the superior central nucleus raphe, the ventral anterior-lateral complex, the intermediodorsal nucleus, the central medial nucleus, the rhomboid nucleus, and the submedial nucleus of the thalamus. The neural circuit data revealed here could help to understand and further research on the function of LS. Summary: We identified the sequence of projections from the lateral septal nucleus by virus tracing and expanded the data on neural circuits, which could help to understand brain function.
Collapse
Affiliation(s)
- Ke Deng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.,University of Chinese Academy of Sciences, Beijing 100039, China
| | - Lu Yang
- Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jing Xie
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.,University of Chinese Academy of Sciences, Beijing 100039, China
| | - He Tang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Gui-Sheng Wu
- Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Huai-Rong Luo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China .,Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
56
|
Smith SP, Wu G, Fooks AR, Ma J, Banyard AC. Trying to treat the untreatable: experimental approaches to clear rabies virus infection from the CNS. J Gen Virol 2019; 100:1171-1186. [PMID: 31237530 DOI: 10.1099/jgv.0.001269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rabies virus causes an invariably fatal encephalitis following the onset of clinical disease. Despite the availability of safe and effective vaccines, the clinical stages of rabies encephalitis remain untreatable, with few survivors being documented. A principal obstacle to the treatment of rabies is the neurotropic nature of the virus, with the blood-brain barrier size exclusion limit rendering the delivery of antiviral drugs and molecules to the central nervous system inherently problematic. This review focuses on efforts to try and overcome barriers to molecule delivery to treat clinical rabies and overviews current progress in the development of experimental live rabies virus vaccines that may have future applications in the treatment of clinical rabies, including the attenuation of rabies virus vectors through either the duplication or mutation of existing genes or the incorporation of non-viral elements within the genome. Rabies post-infection treatment (PIT) remains the holy grail of rabies research.
Collapse
Affiliation(s)
- Samuel P Smith
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK.,Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK
| | - Guanghui Wu
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK
| | - Anthony R Fooks
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK.,Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK.,Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Julian Ma
- Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK
| | - Ashley C Banyard
- Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK.,School of Life Sciences, University of West Sussex, Falmer, West Sussex, UK.,Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK
| |
Collapse
|
57
|
Watson TC, Obiang P, Torres-Herraez A, Watilliaux A, Coulon P, Rochefort C, Rondi-Reig L. Anatomical and physiological foundations of cerebello-hippocampal interaction. eLife 2019; 8:e41896. [PMID: 31205000 PMCID: PMC6579515 DOI: 10.7554/elife.41896] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 05/30/2019] [Indexed: 12/15/2022] Open
Abstract
Multiple lines of evidence suggest that functionally intact cerebello-hippocampal interactions are required for appropriate spatial processing. However, how the cerebellum anatomically and physiologically engages with the hippocampus to sustain such communication remains unknown. Using rabies virus as a retrograde transneuronal tracer in mice, we reveal that the dorsal hippocampus receives input from topographically restricted and disparate regions of the cerebellum. By simultaneously recording local field potential from both the dorsal hippocampus and anatomically connected cerebellar regions, we additionally suggest that the two structures interact, in a behaviorally dynamic manner, through subregion-specific synchronization of neuronal oscillations in the 6-12 Hz frequency range. Together, these results reveal a novel neural network macro-architecture through which we can understand how a brain region classically associated with motor control, the cerebellum, may influence hippocampal neuronal activity and related functions, such as spatial navigation.
Collapse
Affiliation(s)
- Thomas Charles Watson
- Neuroscience Paris Seine, Cerebellum, Navigation and Memory TeamCNRS UMR 8246, INSERM, UMR-S 1130, Sorbonne Universités, University Pierre and Marie CurieParisFrance
| | - Pauline Obiang
- Neuroscience Paris Seine, Cerebellum, Navigation and Memory TeamCNRS UMR 8246, INSERM, UMR-S 1130, Sorbonne Universités, University Pierre and Marie CurieParisFrance
| | - Arturo Torres-Herraez
- Neuroscience Paris Seine, Cerebellum, Navigation and Memory TeamCNRS UMR 8246, INSERM, UMR-S 1130, Sorbonne Universités, University Pierre and Marie CurieParisFrance
| | - Aurélie Watilliaux
- Neuroscience Paris Seine, Cerebellum, Navigation and Memory TeamCNRS UMR 8246, INSERM, UMR-S 1130, Sorbonne Universités, University Pierre and Marie CurieParisFrance
| | - Patrice Coulon
- Institut de Neurosciences de la TimoneCNRS and Aix Marseille UniversitéMarseilleFrance
| | - Christelle Rochefort
- Neuroscience Paris Seine, Cerebellum, Navigation and Memory TeamCNRS UMR 8246, INSERM, UMR-S 1130, Sorbonne Universités, University Pierre and Marie CurieParisFrance
| | - Laure Rondi-Reig
- Neuroscience Paris Seine, Cerebellum, Navigation and Memory TeamCNRS UMR 8246, INSERM, UMR-S 1130, Sorbonne Universités, University Pierre and Marie CurieParisFrance
| |
Collapse
|
58
|
Chidambaram SB, Rathipriya AG, Bolla SR, Bhat A, Ray B, Mahalakshmi AM, Manivasagam T, Thenmozhi AJ, Essa MM, Guillemin GJ, Chandra R, Sakharkar MK. Dendritic spines: Revisiting the physiological role. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:161-193. [PMID: 30654089 DOI: 10.1016/j.pnpbp.2019.01.005] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 01/04/2019] [Accepted: 01/12/2019] [Indexed: 12/11/2022]
Abstract
Dendritic spines are small, thin, specialized protrusions from neuronal dendrites, primarily localized in the excitatory synapses. Sophisticated imaging techniques revealed that dendritic spines are complex structures consisting of a dense network of cytoskeletal, transmembrane and scaffolding molecules, and numerous surface receptors. Molecular signaling pathways, mainly Rho and Ras family small GTPases pathways that converge on actin cytoskeleton, regulate the spine morphology and dynamics bi-directionally during synaptic activity. During synaptic plasticity the number and shapes of dendritic spines undergo radical reorganizations. Long-term potentiation (LTP) induction promote spine head enlargement and the formation and stabilization of new spines. Long-term depression (LTD) results in their shrinkage and retraction. Reports indicate increased spine density in the pyramidal neurons of autism and Fragile X syndrome patients and reduced density in the temporal gyrus loci of schizophrenic patients. Post-mortem reports of Alzheimer's brains showed reduced spine number in the hippocampus and cortex. This review highlights the spine morphogenesis process, the activity-dependent structural plasticity and mechanisms by which synaptic activity sculpts the dendritic spines, the structural and functional changes in spines during learning and memory using LTP and LTD processes. It also discusses on spine status in neurodegenerative diseases and the impact of nootropics and neuroprotective agents on the functional restoration of dendritic spines.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India.
| | - A G Rathipriya
- Food and Brain Research Foundation, Chennai, Tamil Nadu, India
| | - Srinivasa Rao Bolla
- Department of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, Damam, Saudi Arabia
| | - Abid Bhat
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Arehally Marappa Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Thamilarasan Manivasagam
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, India
| | - Arokiasamy Justin Thenmozhi
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
| | - Gilles J Guillemin
- Neuropharmacology Group, Faculty of Medicine and Health Sciences, Deb Bailey MND Research Laboratory, Macquarie University, Sydney, NSW 2109, Australia
| | - Ramesh Chandra
- Department of Chemistry, Ambedkar Centre for BioMedical Research, Delhi University, Delhi 110007, India
| | - Meena Kishore Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, 107, Wiggins Road, Saskatoon, SK S7N 5C9, Canada.
| |
Collapse
|
59
|
Ugolini G, Prevosto V, Graf W. Ascending vestibular pathways to parietal areas MIP and LIPv and efference copy inputs from the medial reticular formation: Functional frameworks for body representations updating and online movement guidance. Eur J Neurosci 2019; 50:2988-3013. [DOI: 10.1111/ejn.14426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 03/25/2019] [Accepted: 04/04/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Gabriella Ugolini
- Paris‐Saclay Institute of Neuroscience (UMR9197) CNRS ‐ Université Paris‐Sud Université Paris‐Saclay Gif‐sur‐Yvette France
| | - Vincent Prevosto
- Paris‐Saclay Institute of Neuroscience (UMR9197) CNRS ‐ Université Paris‐Sud Université Paris‐Saclay Gif‐sur‐Yvette France
- Department of Biomedical Engineering Pratt School of Engineering Durham North Carolina
- Department of Neurobiology Duke School of Medicine Duke University Durham North Carolina
| | - Werner Graf
- Department of Physiology and Biophysics Howard University Washington District of Columbia
| |
Collapse
|
60
|
Lama Z, Gaudin Y, Blondel D, Lagaudrière-Gesbert C. Kinase inhibitors tyrphostin 9 and rottlerin block early steps of rabies virus cycle. Antiviral Res 2019; 168:51-60. [PMID: 31071352 DOI: 10.1016/j.antiviral.2019.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/07/2019] [Accepted: 04/29/2019] [Indexed: 11/26/2022]
Abstract
Rabies virus (RABV) is a neurotropic virus that causes fatal encephalitis in humans and animals and still kills up to 59,000 people worldwide every year. To date, only preventive or post-exposure vaccination protects against the disease but therapeutics are missing. After screening a library of 80 kinases inhibitors, we identified two compounds as potent inhibitors of RABV infection: tyrphostin 9 and rottlerin. Mechanism of action studies show that both inhibitors interfere with an early step of viral cycle and can prevent viral replication. In presence of tyrphostin 9, the viral entry through endocytosis is disturbed leading to improper delivery of viral particles in cytoplasm, whereas rottlerin is inhibiting the transcription, most likely by decreasing intracellular ATP concentration, and therefore the replication of the viral genome.
Collapse
Affiliation(s)
- Zoé Lama
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Yves Gaudin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Danielle Blondel
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Cécile Lagaudrière-Gesbert
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France.
| |
Collapse
|
61
|
Yeo SH, Kyle V, Blouet C, Jones S, Colledge WH. Mapping neuronal inputs to Kiss1 neurons in the arcuate nucleus of the mouse. PLoS One 2019; 14:e0213927. [PMID: 30917148 PMCID: PMC6436706 DOI: 10.1371/journal.pone.0213927] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
The normal function of the mammalian reproductive axis is strongly influenced by physiological, metabolic and environmental factors. Kisspeptin neuropeptides, encoded by the Kiss1 gene, are potent regulators of the mammalian reproductive axis by stimulating gonadodropin releasing hormone secretion from the hypothalamus. To understand how the reproductive axis is modulated by higher order neuronal inputs we have mapped the afferent circuits into arcuate (ARC) Kiss1 neurons. We used a transgenic mouse that expresses the CRE recombinase in Kiss1 neurons for conditional viral tracing with genetically modified viruses. CRE-mediated activation of these viruses in Kiss1 neurons allows the virus to move transynaptically to label neurons with primary or secondary afferent inputs into the Kiss1 neurons. Several regions of the brain showed synaptic connectivity to arcuate Kiss1 neurons including proopiomelanocortin neurons in the ARC itself, kisspeptin neurons in the anteroventral periventricular nucleus, vasopressin neurons in the supraoptic and suprachiasmatic nuclei, thyrotropin releasing neurons in the paraventricular nucleus and unidentified neurons in other regions including the subfornical organ, amygdala, interpeduncular nucleus, ventral premammilary nucleus, basal nucleus of stria terminalis and the visual, somatosensory and piriform regions of the cortex. These data provide an insight into how the activity of Kiss1 neurons may be regulated by metabolic signals and provide a detailed neuroanatomical map for future functional studies.
Collapse
Affiliation(s)
- Shel-Hwa Yeo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Victoria Kyle
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Clemence Blouet
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Susan Jones
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - William Henry Colledge
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
62
|
Li S, Quan T, Xu C, Huang Q, Kang H, Chen Y, Li A, Fu L, Luo Q, Gong H, Zeng S. Optimization of Traced Neuron Skeleton Using Lasso-Based Model. Front Neuroanat 2019; 13:18. [PMID: 30846931 PMCID: PMC6393391 DOI: 10.3389/fnana.2019.00018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/01/2019] [Indexed: 11/30/2022] Open
Abstract
Reconstruction of neuronal morphology from images involves mainly the extraction of neuronal skeleton points. It is an indispensable step in the quantitative analysis of neurons. Due to the complex morphology of neurons, many widely used tracing methods have difficulties in accurately acquiring skeleton points near branch points or in structures with tortuosity. Here, we propose two models to solve these problems. One is based on an L1-norm minimization model, which can better identify tortuous structure, namely, a local structure with large curvature skeleton points; the other detects an optimized branch point by considering the combination patterns of all neurites that link to this point. We combined these two models to achieve optimized skeleton detection for a neuron. We validate our models in various datasets including MOST and BigNeuron. In addition, we demonstrate that our method can optimize the traced skeletons from large-scale images. These characteristics of our approach indicate that it can reduce manual editing of traced skeletons and help to accelerate the accurate reconstruction of neuronal morphology.
Collapse
Affiliation(s)
- Shiwei Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China
| | - Tingwei Quan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China.,School of Mathematics and Economics, Hubei University of Education, Hubei, China
| | - Cheng Xu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China
| | - Qing Huang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China
| | - Hongtao Kang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China
| | - Yijun Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China
| | - Ling Fu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China
| | - Shaoqun Zeng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Hubei, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Hubei, China
| |
Collapse
|
63
|
Sun L, Tang Y, Yan K, Yu J, Zou Y, Xu W, Xiao K, Zhang Z, Li W, Wu B, Hu Z, Chen K, Fu ZF, Dai J, Cao G. Differences in neurotropism and neurotoxicity among retrograde viral tracers. Mol Neurodegener 2019; 14:8. [PMID: 30736827 PMCID: PMC6368820 DOI: 10.1186/s13024-019-0308-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/13/2019] [Indexed: 01/01/2023] Open
Abstract
Background Neurotropic virus-based tracers have been extensively applied in mapping and manipulation of neural circuits. However, their neurotropic and neurotoxic properties remain to be fully characterized. Methods Through neural circuit tracing, we systematically compared the neurotropism discrepancy among different multi-trans-synaptic and mono-synaptic retrograde viral tracers including pseudorabies virus (PRV), rabies virus (RV), and the newly engineered retro adeno-associated virus (rAAV2-retro) tracers. The (single-cell) RNA sequencing analysis was utilized for seeking possible attribution to neurotropism discrepancy and comparing cell toxicity caused by viral infection between glycoprotein-deleted RV (RV-∆G) and rAAV2-retro. Viral toxicity induced microglia activation and neuronal protein change were evaluated by immunohistochemistry. Results Multi-trans-synaptic retrograde viral tracers, PRV and RV, exhibit differential neurotropism when they were used for central neural circuit tracing from popliteal lymph nodes. Mono-synaptic retrograde tracers, including RV-∆G and rAAV2-retro, displayed discrepant neurotropic property, when they were applied to trace the inputs of lateral hypothalamic area and medial preoptic nucleus. rAAV2-retro demonstrated preference in cerebral cortex, whereas RV-∆G prefers to label basal ganglia and hypothalamus. Remarkably, we detected a distinct preference for specific cortical layer of rAAV2-retro in layer 5 and RV-∆G in layer 6 when they were injected into dorsal lateral geniculate nucleus to label corticothalamic neurons in primary visual cortex. Complementation of TVA receptor gene in RV-resistant neurons enabled EnvA-pseudotyped RV infection, supporting receptors attribution to viral neurotropism. Furthermore, both RV-∆G and rAAV2-retro exerted neurotoxic influence at the injection sites and retrogradely labeled sites, while the changes were more profound for RV-∆G infection. Finally, we demonstrated a proof-of-concept strategy for more comprehensive high-order circuit tracing of a specific target nucleus by combining rAAV2-retro, RV, and rAAV tracers. Conclusions Different multi-trans-synaptic and mono-synaptic retrograde viral tracers exhibited discrepant neurotropism within certain brain regions, even cortical layer preference. More neurotoxicity was observed under RV-∆G infection as compared with rAAV2-retro. By combining rAAV2-retro, RV, and rAAV tracers, high-order circuit tracing can be achieved. Our findings provide important reference for appropriate application of viral tracers to delineate the landscape and dissect the function of neural network. Electronic supplementary material The online version of this article (10.1186/s13024-019-0308-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leqiang Sun
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yajie Tang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Keji Yan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jinsong Yu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yanyan Zou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Weize Xu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ke Xiao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhihui Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Weiming Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Beili Wu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhe Hu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Kening Chen
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhen F Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.,Departments of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Jinxia Dai
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China. .,Bio-Medical Center, Huazhong Agricultural University, Wuhan, 430070, China. .,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China. .,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, 430070, China.
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China. .,Bio-Medical Center, Huazhong Agricultural University, Wuhan, 430070, China. .,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China. .,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, 430070, China.
| |
Collapse
|
64
|
Li S, Quan T, Zhou H, Yin F, Li A, Fu L, Luo Q, Gong H, Zeng S. Identifying Weak Signals in Inhomogeneous Neuronal Images for Large-Scale Tracing of Sparsely Distributed Neurites. Neuroinformatics 2019; 17:497-514. [PMID: 30635864 PMCID: PMC6841657 DOI: 10.1007/s12021-018-9414-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tracing neurites constitutes the core of neuronal morphology reconstruction, a key step toward neuronal circuit mapping. Modern optical-imaging techniques allow observation of nearly complete mouse neuron morphologies across brain regions or even the whole brain. However, high-level automation reconstruction of neurons, i.e., the reconstruction with a few of manual edits requires discrimination of weak foreground points from the inhomogeneous background. We constructed an identification model, where empirical observations made from neuronal images were summarized into rules for designing feature vectors that to classify foreground and background, and a support vector machine (SVM) was used to learn these feature vectors. We embedded this constructed SVM classifier into a previously developed tool, SparseTracer, to obtain SparseTracer-Learned Feature Vector (ST-LFV). ST-LFV can trace sparsely distributed neurites with weak signals (contrast-to-noise ratio < 1.5) against an inhomogeneous background in datasets imaged by widely used light-microscopy techniques like confocal microscopy and two-photon microscopy. Moreover, 12 sub-blocks were extracted from different brain regions. The average recall and precision rates were 99% and 97%, respectively. These results indicated that ST-LFV is well suited for weak signal identification with varying image characteristics. We also applied ST-LFV to trace long-range neurites from images where neurites are sparsely distributed but their image intensities are weak in some cases. When tracing this long-range neurites, manual edit was required once to obtain results equivalent to the ground truth, compared with 20 times of manual edits required by SparseTracer. This improvement in the level of automatic reconstruction indicates that ST-LFV has the potential to rapidly reconstruct sparsely distributed neurons at the large scale.
Collapse
Affiliation(s)
- Shiwei Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Tingwei Quan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China. .,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China. .,School of Mathematics and Economics, Hubei University of Education, Wuhan, 430205, Hubei, China.
| | - Hang Zhou
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - FangFang Yin
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Ling Fu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Shaoqun Zeng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| |
Collapse
|
65
|
Dedic N, Chen A, Deussing JM. The CRF Family of Neuropeptides and their Receptors - Mediators of the Central Stress Response. Curr Mol Pharmacol 2018; 11:4-31. [PMID: 28260504 PMCID: PMC5930453 DOI: 10.2174/1874467210666170302104053] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 11/26/2015] [Accepted: 08/03/2016] [Indexed: 12/12/2022]
Abstract
Background: Dysregulated stress neurocircuits, caused by genetic and/or environmental changes, underlie the development of many neuropsychiatric disorders. Corticotropin-releasing factor (CRF) is the major physiological activator of the hypothalamic-pituitary-adrenal (HPA) axis and conse-quently a primary regulator of the mammalian stress response. Together with its three family members, urocortins (UCNs) 1, 2, and 3, CRF integrates the neuroendocrine, autonomic, metabolic and behavioral responses to stress by activating its cognate receptors CRFR1 and CRFR2. Objective: Here we review the past and current state of the CRF/CRFR field, ranging from pharmacologi-cal studies to genetic mouse models and virus-mediated manipulations. Results: Although it is well established that CRF/CRFR1 signaling mediates aversive responses, includ-ing anxiety and depression-like behaviors, a number of recent studies have challenged this viewpoint by revealing anxiolytic and appetitive properties of specific CRF/CRFR1 circuits. In contrast, the UCN/CRFR2 system is less well understood and may possibly also exert divergent functions on physiol-ogy and behavior depending on the brain region, underlying circuit, and/or experienced stress conditions. Conclusion: A plethora of available genetic tools, including conventional and conditional mouse mutants targeting CRF system components, has greatly advanced our understanding about the endogenous mecha-nisms underlying HPA system regulation and CRF/UCN-related neuronal circuits involved in stress-related behaviors. Yet, the detailed pathways and molecular mechanisms by which the CRF/UCN-system translates negative or positive stimuli into the final, integrated biological response are not completely un-derstood. The utilization of future complementary methodologies, such as cell-type specific Cre-driver lines, viral and optogenetic tools will help to further dissect the function of genetically defined CRF/UCN neurocircuits in the context of adaptive and maladaptive stress responses.
Collapse
Affiliation(s)
- Nina Dedic
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| |
Collapse
|
66
|
A circuit from hippocampal CA2 to lateral septum disinhibits social aggression. Nature 2018; 564:213-218. [PMID: 30518859 PMCID: PMC6364572 DOI: 10.1038/s41586-018-0772-0] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 10/12/2018] [Indexed: 01/06/2023]
Abstract
Although the hippocampus is known to be important for declarative memory, how hippocampal output regulates motivated behaviors, such as social aggression, is less well understood. Here we report that hippocampal CA2 pyramidal neurons, which are important for social memory, promote social aggression. This action depends on CA2 output to the lateral septum that is selectively enhanced immediately prior to attack. Activation of lateral septum by CA2 recruits a circuit that disinhibits a subnucleus of the ventro-medial hypothalamus known to trigger attack. The social hormone arginine-vasopressin enhances social aggression by acting on arginine-vasopressin 1b receptors on CA2 presynaptic terminals in lateral septum to facilitate excitatory synaptic transmission. In this manner, release of vasopressin in lateral septum, driven by an animal’s internal state, may serve as a modulatory control that determines whether CA2 activity leads to declarative memory of a social encounter or proceeds to promote motivated social aggression.
Collapse
|
67
|
Murabe N, Mori T, Fukuda S, Isoo N, Ohno T, Mizukami H, Ozawa K, Yoshimura Y, Sakurai M. Higher primate-like direct corticomotoneuronal connections are transiently formed in a juvenile subprimate mammal. Sci Rep 2018; 8:16536. [PMID: 30410053 PMCID: PMC6224497 DOI: 10.1038/s41598-018-34961-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/22/2018] [Indexed: 12/30/2022] Open
Abstract
The corticospinal (CS) tract emerged and evolved in mammals, and is essentially involved in voluntary movement. Over its phylogenesis, CS innervation gradually invaded to the ventral spinal cord, eventually making direct connections with spinal motoneurons (MNs) in higher primates. Despite its importance, our knowledge of the origin of the direct CS-MN connections is limited; in fact, there is controversy as to whether these connections occur in subprimate mammals, such as rodents. Here we studied the retrograde transsynaptic connection between cortical neurons and MNs in mice by labeling the cells with recombinant rabies virus. On postnatal day 14 (P14), we found that CS neurons make direct connections with cervical MNs innervating the forearm muscles. Direct connections were also detected electrophysiologically in whole cell recordings from identified MNs retrogradely-labeled from their target muscles and optogenetic CS stimulation. In contrast, few, if any, lumbar MNs innervating hindlimbs showed direct connections on P18. Moreover, the direct CS-MN connections observed on P14 were later eliminated. The transient CS-MN cells were distributed predominantly in the M1 and S1 areas. These findings provide insight into the ontogeny and phylogeny of the CS projection and appear to settle the controversy about direct CS-MN connections in subprimate mammals.
Collapse
Affiliation(s)
- Naoyuki Murabe
- Department of Physiology, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Takuma Mori
- Division of Visual Information Processing, National Institute for Physiological Sciences, National Institutes for Natural Sciences, Okazaki, 444-8585, Japan.,Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan
| | - Satoshi Fukuda
- Department of Physiology, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Noriko Isoo
- Department of Physiology, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Takae Ohno
- Department of Physiology, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Keiya Ozawa
- Division of Genetic Therapeutics, Jichi Medical University, Tochigi, 329-0498, Japan.,Research Hospital, Institute of Medical Science, Tokyo University, Tokyo, 108-8639, Japan
| | - Yumiko Yoshimura
- Division of Visual Information Processing, National Institute for Physiological Sciences, National Institutes for Natural Sciences, Okazaki, 444-8585, Japan.,Department of Physiological Sciences, Graduate University for Advanced Studies, Okazaki, 444-8585, Japan
| | - Masaki Sakurai
- Department of Physiology, Teikyo University School of Medicine, Tokyo, 173-8605, Japan.
| |
Collapse
|
68
|
|
69
|
Ontogenesis of the pinealo-retinal neuronal connection in albino rats. Neurosci Lett 2018; 665:189-194. [DOI: 10.1016/j.neulet.2017.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/29/2017] [Accepted: 12/03/2017] [Indexed: 12/19/2022]
|
70
|
Yeo SH, Colledge WH. The Role of Kiss1 Neurons As Integrators of Endocrine, Metabolic, and Environmental Factors in the Hypothalamic-Pituitary-Gonadal Axis. Front Endocrinol (Lausanne) 2018; 9:188. [PMID: 29755406 PMCID: PMC5932150 DOI: 10.3389/fendo.2018.00188] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/06/2018] [Indexed: 01/06/2023] Open
Abstract
Kisspeptin-GPR54 signaling in the hypothalamus is required for reproduction and fertility in mammals. Kiss1 neurons are key regulators of gonadotropin-releasing hormone (GnRH) release and modulation of the hypothalamic-pituitary-gonadal (HPG) axis. Arcuate Kiss1 neurons project to GnRH nerve terminals in the median eminence, orchestrating the pulsatile secretion of luteinizing hormone (LH) through the intricate interaction between GnRH pulse frequency and the pituitary gonadotrophs. Arcuate Kiss1 neurons, also known as KNDy neurons in rodents and ruminants because of their co-expression of neurokinin B and dynorphin represent an ideal hub to receive afferent inputs from other brain regions in response to physiological and environmental changes, which can regulate the HPG axis. This review will focus on studies performed primarily in rodent and ruminant species to explore potential afferent inputs to Kiss1 neurons with emphasis on the arcuate region but also considering the rostral periventricular region of the third ventricle (RP3V). Specifically, we will discuss how these inputs can be modulated by hormonal, metabolic, and environmental factors to control gonadotropin secretion and fertility. We also summarize the methods and techniques that can be used to study functional inputs into Kiss1 neurons.
Collapse
|
71
|
Singh R, Singh KP, Cherian S, Saminathan M, Kapoor S, Manjunatha Reddy GB, Panda S, Dhama K. Rabies - epidemiology, pathogenesis, public health concerns and advances in diagnosis and control: a comprehensive review. Vet Q 2017. [PMID: 28643547 DOI: 10.1080/01652176.2017.1343516] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Rabies is a zoonotic, fatal and progressive neurological infection caused by rabies virus of the genus Lyssavirus and family Rhabdoviridae. It affects all warm-blooded animals and the disease is prevalent throughout the world and endemic in many countries except in Islands like Australia and Antarctica. Over 60,000 peoples die every year due to rabies, while approximately 15 million people receive rabies post-exposure prophylaxis (PEP) annually. Bite of rabid animals and saliva of infected host are mainly responsible for transmission and wildlife like raccoons, skunks, bats and foxes are main reservoirs for rabies. The incubation period is highly variable from 2 weeks to 6 years (avg. 2-3 months). Though severe neurologic signs and fatal outcome, neuropathological lesions are relatively mild. Rabies virus exploits various mechanisms to evade the host immune responses. Being a major zoonosis, precise and rapid diagnosis is important for early treatment and effective prevention and control measures. Traditional rapid Seller's staining and histopathological methods are still in use for diagnosis of rabies. Direct immunofluoroscent test (dFAT) is gold standard test and most commonly recommended for diagnosis of rabies in fresh brain tissues of dogs by both OIE and WHO. Mouse inoculation test (MIT) and polymerase chain reaction (PCR) are superior and used for routine diagnosis. Vaccination with live attenuated or inactivated viruses, DNA and recombinant vaccines can be done in endemic areas. This review describes in detail about epidemiology, transmission, pathogenesis, advances in diagnosis, vaccination and therapeutic approaches along with appropriate prevention and control strategies.
Collapse
Affiliation(s)
- Rajendra Singh
- a Division of Pathology , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| | - Karam Pal Singh
- b Centre for Animal Disease Research and Diagnosis (CADRAD) , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| | - Susan Cherian
- a Division of Pathology , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| | - Mani Saminathan
- a Division of Pathology , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| | - Sanjay Kapoor
- c Department of Veterinary Microbiology , LLR University of Veterinary and Animal Sciences , Hisar , Haryana , India
| | - G B Manjunatha Reddy
- d ICAR-National Institute of Veterinary Epidemiology and Disease Informatics , Bengaluru , Karnataka , India
| | - Shibani Panda
- a Division of Pathology , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| | - Kuldeep Dhama
- a Division of Pathology , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| |
Collapse
|
72
|
Porres CP, Grothe B, Felmy F. Breakdown of Excitability by Attenuated PRV-152 Infection in Auditory Brainstem Neurons of Mongolian Gerbils. Neuroscience 2017; 367:1-9. [PMID: 29069619 DOI: 10.1016/j.neuroscience.2017.10.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/12/2017] [Accepted: 10/16/2017] [Indexed: 11/27/2022]
Abstract
Pseudorabies virus (PRV), a neurovirulent α-herpesvirus, spreads between neurons at synaptic connections. PRV-infected neurons have been shown to exhibit functional deficits with the attenuated PRV152 Bartha strain negatively influencing neuronal functioning in in vitro model systems. However, the impact of this attenuated PRV152 Bartha strain on the native central nervous system has not been fully explored. Using a combination of in vivo stereotactic injections and post-hoc in vitro whole-cell recordings, we investigated the functional impact of PRV152 Bartha in the auditory system of juvenile Mongolian gerbils. The specificity of this virus strain to spread exclusively trans-synaptically in a retrograde fashion and the well-defined structure of the ascending auditory brainstem pathways allowed us to determine the physiological alterations in primary and secondary infected neurons. We find at primary and secondary infections sites, the inferior colliculus (IC) and dorsal nucleus of the lateral lemniscus respectively, a reduced excitability of infected cells. The loss of excitability is manifested by an increase in current threshold and a loss of action potential generation. The minor changes in the approximated passive membrane parameters induced by the infection cannot explain the full loss in excitability, indicating that channel densities and properties have changed. This impact on neuronal functioning might contribute to the lethal neurovirulent effects of PRV viruses as vital neuronal circuits might cease activity. Since the detrimental effects of the attenuated PRV152 Bartha strain are reduced compared to wild-type strains, it comprises an excellent tool to study the neuropathological mechanisms of viral infections.
Collapse
Affiliation(s)
- Christian P Porres
- Department Biology II, Division of Neurobiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Benedikt Grothe
- Department Biology II, Division of Neurobiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Felix Felmy
- Department Biology II, Division of Neurobiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany; Institute of Zoology, University of Veterinary Medicine Hannover, 30599 Hannover, Germany.
| |
Collapse
|
73
|
Abstract
Magnetic resonance imaging, positron emission tomography, and optical imaging have emerged as key tools to understand brain function and neurological disorders in preclinical mouse models. They offer the unique advantage of monitoring individual structural and functional changes over time. What remained unsolved until recently was to generate whole-brain microscopy data which can be correlated to the 3D in vivo neuroimaging data. Conventional histological sections are inappropriate especially for neuronal tracing or the unbiased screening for molecular targets through the whole brain. As part of the European Society for Molecular Imaging (ESMI) meeting 2016 in Utrecht, the Netherlands, we addressed this issue in the Molecular Neuroimaging study group meeting. Presentations covered new brain clearing methods, light sheet microscopes for large samples, and automatic registration of microscopy to in vivo imaging data. In this article, we summarize the discussion; give an overview of the novel techniques; and discuss the practical needs, benefits, and limitations.
Collapse
|
74
|
3D reconstruction and standardization of the rat facial nucleus for precise mapping of vibrissal motor networks. Neuroscience 2017; 368:171-186. [PMID: 28958919 PMCID: PMC5798596 DOI: 10.1016/j.neuroscience.2017.09.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/21/2017] [Accepted: 09/17/2017] [Indexed: 12/16/2022]
Abstract
The rodent facial nucleus (FN) comprises motoneurons (MNs) that control the facial musculature. In the lateral part of the FN, populations of vibrissal motoneurons (vMNs) innervate two groups of muscles that generate movements of the whiskers. Vibrissal MNs thus represent the terminal point of the neuronal networks that generate rhythmic whisking during exploratory behaviors and that modify whisker movements based on sensory-motor feedback during tactile-based perception. Here, we combined retrograde tracer injections into whisker-specific muscles, with large-scale immunohistochemistry and digital reconstructions to generate an average model of the rat FN. The model incorporates measurements of the FN geometry, its cellular organization and a whisker row-specific map formed by vMNs. Furthermore, the model provides a digital 3D reference frame that allows registering structural data - obtained across scales and animals - into a common coordinate system with a precision of ∼60 µm. We illustrate the registration method by injecting replication competent rabies virus into the muscle of a single whisker. Retrograde transport of the virus to vMNs enabled reconstruction of their dendrites. Subsequent trans-synaptic transport enabled mapping the presynaptic neurons of the reconstructed vMNs. Registration of these data to the FN reference frame provides a first account of the morphological and synaptic input variability within a population of vMNs that innervate the same muscle.
Collapse
|
75
|
Hasse JM, Briggs F. Corticogeniculate feedback sharpens the temporal precision and spatial resolution of visual signals in the ferret. Proc Natl Acad Sci U S A 2017; 114:E6222-E6230. [PMID: 28698363 PMCID: PMC5544308 DOI: 10.1073/pnas.1704524114] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The corticogeniculate (CG) pathway connects the visual cortex with the visual thalamus (LGN) in the feedback direction and enables the cortex to directly influence its own input. Despite numerous investigations, the role of this feedback circuit in visual perception remained elusive. To probe the function of CG feedback in a causal manner, we selectively and reversibly manipulated the activity of CG neurons in anesthetized ferrets in vivo using a combined viral-infection and optogenetics approach to drive expression of channelrhodopsin2 (ChR2) in CG neurons. We observed significant increases in temporal precision and spatial resolution of LGN neuronal responses to drifting grating and white noise stimuli when CG neurons expressing ChR2 were light activated. Enhancing CG feedback reduced visually evoked response latencies, increased spike-timing precision, and reduced classical receptive field size. Increased precision among LGN neurons led to increased spike-timing precision among granular layer V1 neurons as well. Together, our findings suggest that the function of CG feedback is to control the timing and precision of thalamic responses to incoming visual signals.
Collapse
Affiliation(s)
- J Michael Hasse
- Department of Physiology & Neurobiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756
- Program in Experimental and Molecular Medicine, Dartmouth College, Hanover, NH 03755
| | - Farran Briggs
- Department of Physiology & Neurobiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756;
- Program in Experimental and Molecular Medicine, Dartmouth College, Hanover, NH 03755
| |
Collapse
|
76
|
Ohara S, Sota Y, Sato S, Tsutsui KI, Iijima T. Increased transgene expression level of rabies virus vector for transsynaptic tracing. PLoS One 2017; 12:e0180960. [PMID: 28700657 PMCID: PMC5507306 DOI: 10.1371/journal.pone.0180960] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 06/23/2017] [Indexed: 12/27/2022] Open
Abstract
Viral vectors that can infect neurons transsynaptically and can strongly express foreign genes are useful for investigating the organization of neural circuits. We previously developed a propagation-competent rabies virus (RV) vector based on a highly attenuated HEP-Flury strain (rHEP5.0-CVSG), which selectively infects neurons and propagates between synaptically connected neurons in a retrograde direction. Its relatively low level of transgene expression, however, makes immunostaining necessary to visualize the morphological features of infected neurons. To increase the transgene expression level of this RV vector, in this study we focused on two viral proteins: the large protein (L) and matrix protein (M). We first attempted to enhance the expression of L, which is a viral RNA polymerase, by deleting the extra transcription unit and shortening the intergenic region between the G and L genes. This viral vector (rHEP5.0-GctL) showed increased transgene expression level with efficient transsynaptic transport. We next constructed an RV vector with a rearranged gene order (rHEP5.0-GML) with the aim to suppress the expression of M, which plays a regulatory role in virus RNA synthesis. Although this vector showed high transgene expression level, the efficiency of transsynaptic transport was low. To further evaluate the usability of rHEP5.0-GctL as a transsynaptic tracer, we inserted a fluorescent timer as a transgene, which changes the color of its fluorescence from blue to red over time. This viral vector enabled us the differentiation of primary infected neurons from secondary infected neurons in terms of the fluorescence wavelength. We expect this propagation-competent RV vector to be useful for elucidating the complex organization of the central nervous system.
Collapse
Affiliation(s)
- Shinya Ohara
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Yasuhiro Sota
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Sho Sato
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Ken-Ichiro Tsutsui
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Toshio Iijima
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
- * E-mail:
| |
Collapse
|
77
|
Nikolic J, Le Bars R, Lama Z, Scrima N, Lagaudrière-Gesbert C, Gaudin Y, Blondel D. Negri bodies are viral factories with properties of liquid organelles. Nat Commun 2017; 8:58. [PMID: 28680096 PMCID: PMC5498545 DOI: 10.1038/s41467-017-00102-9] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/31/2017] [Indexed: 11/23/2022] Open
Abstract
Replication of Mononegavirales occurs in viral factories which form inclusions in the host-cell cytoplasm. For rabies virus, those inclusions are called Negri bodies (NBs). We report that NBs have characteristics similar to those of liquid organelles: they are spherical, they fuse to form larger structures, and they disappear upon hypotonic shock. Their liquid phase is confirmed by FRAP experiments. Live-cell imaging indicates that viral nucleocapsids are ejected from NBs and transported along microtubules to form either new virions or secondary viral factories. Coexpression of rabies virus N and P proteins results in cytoplasmic inclusions recapitulating NBs properties. This minimal system reveals that an intrinsically disordered domain and the dimerization domain of P are essential for Negri bodies-like structures formation. We suggest that formation of liquid viral factories by phase separation is common among Mononegavirales and allows specific recruitment and concentration of viral proteins but also the escape to cellular antiviral response. Negative strand RNA viruses, such as rabies virus, induce formation of cytoplasmic inclusions for genome replication. Here, Nikolic et al. show that these so-called Negri bodies (NBs) have characteristics of liquid organelles and they identify the minimal protein domains required for NB formation.
Collapse
Affiliation(s)
- Jovan Nikolic
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Romain Le Bars
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Zoé Lama
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Nathalie Scrima
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Cécile Lagaudrière-Gesbert
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Yves Gaudin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France.
| | - Danielle Blondel
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France.
| |
Collapse
|
78
|
Oswald M, Geissler S, Goepferich A. Targeting the Central Nervous System (CNS): A Review of Rabies Virus-Targeting Strategies. Mol Pharm 2017; 14:2177-2196. [DOI: 10.1021/acs.molpharmaceut.7b00158] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Mira Oswald
- Chemical & Pharmaceutical Development, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Simon Geissler
- Chemical & Pharmaceutical Development, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Achim Goepferich
- Department for Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 94030 Regensburg, Germany
| |
Collapse
|
79
|
Prevosto V, Graf W, Ugolini G. The control of eye movements by the cerebellar nuclei: polysynaptic projections from the fastigial, interpositus posterior and dentate nuclei to lateral rectus motoneurons in primates. Eur J Neurosci 2017; 45:1538-1552. [DOI: 10.1111/ejn.13546] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/09/2017] [Accepted: 02/17/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Vincent Prevosto
- Paris-Saclay Institute of Neuroscience (UMR9197) CNRS; Université Paris-Sud; Université Paris-Saclay; Bât 32 CNRS 1 av de la Terrasse 91198 Gif-sur-Yvette France
- Department of Biomedical Engineering; Pratt School of Engineering; Duke University; Durham NC USA
- Department of Neurobiology; Duke School of Medicine; Duke University; Durham NC USA
| | - Werner Graf
- Department of Physiology and Biophysics; Howard University; Washington DC USA
| | - Gabriella Ugolini
- Paris-Saclay Institute of Neuroscience (UMR9197) CNRS; Université Paris-Sud; Université Paris-Saclay; Bât 32 CNRS 1 av de la Terrasse 91198 Gif-sur-Yvette France
| |
Collapse
|
80
|
Teklu GG, Hailu TG, Eshetu GR. High Incidence of Human Rabies Exposure in Northwestern Tigray, Ethiopia: A Four-Year Retrospective Study. PLoS Negl Trop Dis 2017; 11:e0005271. [PMID: 28060935 PMCID: PMC5245898 DOI: 10.1371/journal.pntd.0005271] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/19/2017] [Accepted: 12/19/2016] [Indexed: 11/28/2022] Open
Abstract
Background Rabies is a fatal zoonotic disease that has been known in Ethiopia for centuries in society as “Mad Dog Disease”. It is an important disease with veterinary and public health significance in the North western zone of Tigray where previous studies have not been conducted. Frequent occurrence of outbreaks in the area led the researchers to carry out a four year retrospective study to estimate the incidence of human rabies exposure in Northwestern Tigray, Ethiopia. Methodology A referent study was conducted on human rabies exposure cases recorded from 2012 to 2015 at Suhul hospital, Shire Endaselase, Northwestern Tigray, Ethiopia. Exposure cases included in this research constituted victims bitten by unprovoked dogs and who received post exposure prophylaxis (PEP) at the hospital. Two thousand one hundred eighty human rabies exposure cases retrieved from the rabies case database were included in this study. Principal findings The majority of the exposed cases were males (1363/2180, 63%). Age wise, the most exposed age group was ≥15 years in all the study years: 166 (58%), 335 (65%), 492 (66%) and 394 (63%) in 2012, 2013, 2014 and 2015, respectively. Similarly, exposure cases for human rabies increased with age in both males and females across the study years. The incidence of human rabies exposure cases calculated per 100,000 populations was 35.8, 63.0, 89.8 and 73.1 in 2012, 2013, 2014 and 2015, respectively. Binary logistic regression analysis revealed that being male was a risk for human rabies exposure in all the study years. Conclusion The study discovered the highest annual human rabies exposure incidence in Ethiopia. This suggests an urgent need for synergistic efforts of human and animal health sectors to implement prevention and control strategies in this area. Rabies is a deadly disease of human and animals. The disease has been recognized in Ethiopia for centuries, but its impact remained underestimated. This limitation masks the true magnitude of rabies incidence and has been a stumbling block for its prevention and control. The aim of the study was to determine the incidence of human rabies exposure in Northwestern Tigray, Ethiopia, where studies have not been conducted, although outbreaks of the disease were common. A health facility-based four year referent study was conducted on human rabies exposure cases recorded from 2012 to 2015 at Suhul hospital, located in Shire Endaselase, Northwestern Zone of Tigray. Majority of the exposed cases were males. The most exposed age group was ≥15 years in all the study years. Similarly, exposure cases increased with age in both male and female individuals across the study years. Incidence of human rabies exposure cases per 100,000 populations also showed a continuous increment with the highest being recorded in 2014. Moreover, the study discovered the highest annual human rabies exposure incidence in Ethiopia. This suggests an urgent need for synergistic efforts of human and animal health sectors to implement prevention and control strategies.
Collapse
Affiliation(s)
- Gebreyohans Gebru Teklu
- Department of Animal Sciences, College of Agriculture, Aksum University, Shire Campus, Shire, Ethiopia
- * E-mail:
| | | | - Gebremedhin Romha Eshetu
- Department of Animal Production and Technology, College of Agriculture and Environmental Science, Adigrat University, Adigrat, Ethiopia
| |
Collapse
|
81
|
Zingg B, Chou XL, Zhang ZG, Mesik L, Liang F, Tao HW, Zhang LI. AAV-Mediated Anterograde Transsynaptic Tagging: Mapping Corticocollicular Input-Defined Neural Pathways for Defense Behaviors. Neuron 2017; 93:33-47. [PMID: 27989459 PMCID: PMC5538794 DOI: 10.1016/j.neuron.2016.11.045] [Citation(s) in RCA: 472] [Impact Index Per Article: 67.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/22/2016] [Accepted: 11/23/2016] [Indexed: 11/26/2022]
Abstract
To decipher neural circuits underlying brain functions, viral tracers are widely applied to map input and output connectivity of neuronal populations. Despite the successful application of retrograde transsynaptic viruses for identifying presynaptic neurons of transduced neurons, analogous anterograde transsynaptic tools for tagging postsynaptically targeted neurons remain under development. Here, we discovered that adeno-associated viruses (AAV1 and AAV9) exhibit anterograde transsynaptic spread properties. AAV1-Cre from transduced presynaptic neurons effectively and specifically drives Cre-dependent transgene expression in selected postsynaptic neuronal targets, thus allowing axonal tracing and functional manipulations of the latter input-defined neuronal population. Its application in superior colliculus (SC) reveals that SC neuron subpopulations receiving corticocollicular projections from auditory and visual cortex specifically drive flight and freezing, two different types of defense behavior, respectively. Together with an intersectional approach, AAV-mediated anterograde transsynaptic tagging can categorize neurons by their inputs and molecular identity, and allow forward screening of distinct functional neural pathways embedded in complex brain circuits.
Collapse
Affiliation(s)
- Brian Zingg
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90033, USA
| | - Xiao-Lin Chou
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90033, USA
| | - Zheng-Gang Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lukas Mesik
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90033, USA
| | - Feixue Liang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Huizhong Whit Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
82
|
Feng MH, He ZG, Liu BW, Li ZX, Wu DZ, Liu SG, Xiang HB. Parafascicular nucleus circuits: Implications for the alteration of gastrointestinal functions during epileptogenesis. Epilepsy Behav 2016; 64:295-298. [PMID: 27773642 DOI: 10.1016/j.yebeh.2016.07.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 07/14/2016] [Indexed: 11/16/2022]
Affiliation(s)
- Mao-Hui Feng
- Department of Oncology, Wuhan Peritoneal Cancer Clinical Medical Research Center, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, No. 169 Donghu Road, Wuhan, Hubei 430071, PR China.
| | - Zhi-Gang He
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Bao-Wen Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Zhi-Xiao Li
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Duo-Zhi Wu
- Department of Anesthesiology, People's Hospital of Hainan Province, Haikou, Hainan 570311, PR China.
| | - San-Guang Liu
- Department of Hepatobiliary Surgery, The Second Hospital, Hebei Medical University, Shijiazhuang 050000, Hebei, PR China.
| | - Hong-Bing Xiang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| |
Collapse
|
83
|
Corticospinal Inputs to Primate Motoneurons Innervating the Forelimb from Two Divisions of Primary Motor Cortex and Area 3a. J Neurosci 2016; 36:2605-16. [PMID: 26937002 DOI: 10.1523/jneurosci.4055-15.2016] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Previous anatomical work in primates has suggested that only corticospinal axons originating in caudal primary motor cortex ("new M1") and area 3a make monosynaptic cortico-motoneuronal connections with limb motoneurons. By contrast, the more rostral "old M1" is proposed to control motoneurons disynaptically via spinal interneurons. In six macaque monkeys, we examined the effects from focal stimulation within old and new M1 and area 3a on 135 antidromically identified motoneurons projecting to the upper limb. EPSPs with segmental latency shorter than 1.2 ms were classified as definitively monosynaptic; these were seen only after stimulation within new M1 or at the new M1/3a border (incidence 6.6% and 1.3%, respectively; total n = 27). However, most responses had longer latencies. Using measures of the response facilitation after a second stimulus compared with the first, and the reduction in response latency after a third stimulus compared with the first, we classified these late responses as likely mediated by either long-latency monosynaptic (n = 108) or non-monosynaptic linkages (n = 108). Both old and new M1 generated putative long-latency monosynaptic and non-monosynaptic effects; the majority of responses from area 3a were non-monosynaptic. Both types of responses from new M1 had significantly greater amplitude than those from old M1. We suggest that slowly conducting corticospinal fibers from old M1 generate weak late monosynaptic effects in motoneurons. These may represent a stage in control of primate motoneurons by the cortex intermediate between disynaptic output via an interposed interneuron seen in nonprimates and the fast direct monosynaptic connections present in new M1. SIGNIFICANCE STATEMENT The corticospinal tract in Old World primates makes monosynaptic connections to motoneurons; previous anatomical work suggests that these connections come only from corticospinal tract (CST) neurons in the subdivision of primary motor cortex within the central sulcus ("new M1") and area 3a. Here, we show using electrophysiology that cortico-motoneuronal connections from fast conducting CST fibers are indeed made exclusively from new M1 and its border with 3a. However, we also show that all parts of M1 and 3a have cortico-motoneuronal connections over more slowly conducting CST axons, as well as exert disynaptic effects on motoneurons via interposed interneurons. Differences between old and new M1 are thus more subtle than previously thought.
Collapse
|
84
|
Motor, cognitive, and affective areas of the cerebral cortex influence the adrenal medulla. Proc Natl Acad Sci U S A 2016; 113:9922-7. [PMID: 27528671 DOI: 10.1073/pnas.1605044113] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Modern medicine has generally viewed the concept of "psychosomatic" disease with suspicion. This view arose partly because no neural networks were known for the mind, conceptually associated with the cerebral cortex, to influence autonomic and endocrine systems that control internal organs. Here, we used transneuronal transport of rabies virus to identify the areas of the primate cerebral cortex that communicate through multisynaptic connections with a major sympathetic effector, the adrenal medulla. We demonstrate that two broad networks in the cerebral cortex have access to the adrenal medulla. The larger network includes all of the cortical motor areas in the frontal lobe and portions of somatosensory cortex. A major component of this network originates from the supplementary motor area and the cingulate motor areas on the medial wall of the hemisphere. These cortical areas are involved in all aspects of skeletomotor control from response selection to motor preparation and movement execution. The second, smaller network originates in regions of medial prefrontal cortex, including a major contribution from pregenual and subgenual regions of anterior cingulate cortex. These cortical areas are involved in higher-order aspects of cognition and affect. These results indicate that specific multisynaptic circuits exist to link movement, cognition, and affect to the function of the adrenal medulla. This circuitry may mediate the effects of internal states like chronic stress and depression on organ function and, thus, provide a concrete neural substrate for some psychosomatic illness.
Collapse
|
85
|
Ruigrok TJH, van Touw S, Coulon P. Caveats in Transneuronal Tracing with Unmodified Rabies Virus: An Evaluation of Aberrant Results Using a Nearly Perfect Tracing Technique. Front Neural Circuits 2016; 10:46. [PMID: 27462206 PMCID: PMC4939302 DOI: 10.3389/fncir.2016.00046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/15/2016] [Indexed: 12/23/2022] Open
Abstract
Apart from the genetically engineered, modified, strains of rabies virus (RABV), unmodified ‘fixed’ virus strains of RABV, such as the ‘French’ subtype of CVS11, are used to examine synaptically connected networks in the brain. This technique has been shown to have all the prerequisite characteristics for ideal tracing as it does not metabolically affect infected neurons within the time span of the experiment, it is transferred transneuronally in one direction only and to all types of neurons presynaptic to the infected neuron, number of transneuronal steps can be precisely controlled by survival time and it is easily detectable with a sensitive technique. Here, using the ‘French’ CVS 11 subtype of RABV in Wistar rats, we show that some of these characteristics may not be as perfect as previously indicated. Using injection of RABV in hind limb muscles, we show that RABV-infected spinal motoneurons may already show lysis 1 or 2 days after infection. Using longer survival times we were able to establish that Purkinje cells may succumb approximately 3 days after infection. In addition, some neurons seem to resist infection, as we noted that the number of RABV-infected inferior olivary neurons did not progress in the same rate as other infected neurons. Furthermore, in our hands, we noted that infection of Purkinje cells did not result in expected transneuronal labeling of cell types that are presynaptic to Purkinje cells such as molecular layer interneurons and granule cells. However, these cell types were readily infected when RABV was injected directly in the cerebellar cortex. Conversely, neurons in the cerebellar nuclei that project to the inferior olive did not take up RABV when this was injected in the inferior olive, whereas these cells could be infected with RABV via a transneuronal route. These results suggest that viral entry from the extracellular space depends on other factors or mechanisms than those used for retrograde transneuronal transfer. We conclude that transneuronal tracing with RABV may result in unexpected results, as not all properties of RABV seem to be ubiquitously valid.
Collapse
Affiliation(s)
- Tom J H Ruigrok
- Department of Neuroscience, Erasmus Medical Center Rotterdam, Netherlands
| | - Sven van Touw
- Department of Neuroscience, Erasmus Medical Center Rotterdam, Netherlands
| | - Patrice Coulon
- Equipe P3M - UMR 7298, Institut de Neurosciences de la Timone, Aix-Marseille Université, CNRS Marseille, France
| |
Collapse
|
86
|
Treweek JB, Gradinaru V. Extracting structural and functional features of widely distributed biological circuits with single cell resolution via tissue clearing and delivery vectors. Curr Opin Biotechnol 2016; 40:193-207. [PMID: 27393829 DOI: 10.1016/j.copbio.2016.03.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 03/10/2016] [Accepted: 03/15/2016] [Indexed: 12/13/2022]
Abstract
The scientific community has learned a great deal from imaging small and naturally transparent organisms such as nematodes and zebrafish. The consequences of genetic mutations on their organ development and survival can be visualized easily and with high-throughput at the organism-wide scale. In contrast, three-dimensional information is less accessible in mammalian subjects because the heterogeneity of light-scattering tissue elements renders their organs opaque. Likewise, genetically labeling desired circuits across mammalian bodies is prohibitively slow and costly via the transgenic route. Emerging breakthroughs in viral vector engineering, genome editing tools, and tissue clearing can render larger opaque organisms genetically tractable and transparent for whole-organ cell phenotyping, tract tracing and imaging at depth.
Collapse
Affiliation(s)
- Jennifer Brooke Treweek
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
87
|
Ellis EM, Gauvain G, Sivyer B, Murphy GJ. Shared and distinct retinal input to the mouse superior colliculus and dorsal lateral geniculate nucleus. J Neurophysiol 2016; 116:602-10. [PMID: 27169509 PMCID: PMC4982907 DOI: 10.1152/jn.00227.2016] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/10/2016] [Indexed: 11/23/2022] Open
Abstract
Our results suggest that the mouse superior colliculus (SC) has access to input from most of the retinal ganglion cells (RGCs) that innervate the dorsal lateral geniculate nucleus (dLGN). By comparison, a number of RGC types appear to innervate the SC but not the dLGN; these RGCs generally exhibit more transient responses and respond best to small stimuli. The mammalian retina conveys the vast majority of information about visual stimuli to two brain regions: the dorsal lateral geniculate nucleus (dLGN) and the superior colliculus (SC). The degree to which retinal ganglion cells (RGCs) send similar or distinct information to the two areas remains unclear despite the important constraints that different patterns of RGC input place on downstream visual processing. To resolve this ambiguity, we injected a glycoprotein-deficient rabies virus coding for the expression of a fluorescent protein into the dLGN or SC; rabies virus labeled a smaller fraction of RGCs than lipophilic dyes such as DiI but, crucially, did not label RGC axons of passage. Approximately 80% of the RGCs infected by rabies virus injected into the dLGN were colabeled with DiI injected into the SC, suggesting that many dLGN-projecting RGCs also project to the SC. However, functional characterization of RGCs revealed that the SC receives input from several classes of RGCs that largely avoid the dLGN, in particular RGCs in which 1) sustained changes in light intensity elicit transient changes in firing rate and/or 2) a small range of stimulus sizes or temporal fluctuations in light intensity elicit robust activity. Taken together, our results illustrate several unexpected asymmetries in the information that the mouse retina conveys to two major downstream targets and suggest that differences in the output of dLGN and SC neurons reflect, at least in part, differences in the functional properties of RGCs that innervate the SC but not the dLGN.
Collapse
Affiliation(s)
- Erika M Ellis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia; and
| | - Gregory Gauvain
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia; and
| | - Benjamin Sivyer
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia; and Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia
| | - Gabe J Murphy
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia; and
| |
Collapse
|
88
|
Yao Q, Pho H, Kirkness J, Ladenheim EE, Bi S, Moran TH, Fuller DD, Schwartz AR, Polotsky VY. Localizing Effects of Leptin on Upper Airway and Respiratory Control during Sleep. Sleep 2016; 39:1097-106. [PMID: 26951402 DOI: 10.5665/sleep.5762] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/01/2016] [Indexed: 12/30/2022] Open
Abstract
STUDY OBJECTIVES Obesity hypoventilation and obstructive sleep apnea are common complications of obesity linked to defects in respiratory pump and upper airway neural control. Leptin-deficient ob/ob mice have impaired ventilatory control and inspiratory flow limitation during sleep, which are both reversed with leptin. We aimed to localize central nervous system (CNS) site(s) of leptin action on respiratory and upper airway neuroventilatory control. METHODS We localized the effect of leptin to medulla versus hypothalamus by administering intracerbroventricular leptin (10 μg/2 μL) versus vehicle to the lateral (n = 14) versus fourth ventricle (n = 11) of ob/ob mice followed by polysomnographic recording. Analyses were stratified for effects on respiratory (nonflow-limited breaths) and upper airway (inspiratory flow limitation) functions. CNS loci were identified by (1) leptin-induced signal transducer and activator of transcription 3 (STAT3) phosphorylation and (2) projections of respiratory and upper airway motoneurons with a retrograde transsynaptic tracer (pseudorabies virus). RESULTS Both routes of leptin administration increased minute ventilation during nonflow-limited breathing in sleep. Phrenic motoneurons were synaptically coupled to the nucleus of the solitary tract, which also showed STAT3 phosphorylation, but not to the hypothalamus. Inspiratory flow limitation and obstructive hypopneas were attenuated by leptin administration to the lateral but not to the fourth cerebral ventricle. Upper airway motoneurons were synaptically coupled with the dorsomedial hypothalamus, which exhibited STAT3 phosphorylation. CONCLUSIONS Leptin relieves upper airway obstruction in sleep apnea by activating the forebrain, possibly in the dorsomedial hypothalamus. In contrast, leptin upregulates ventilatory control through hindbrain sites of action, possibly in the nucleus of the solitary tract.
Collapse
Affiliation(s)
- Qiaoling Yao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Physiology; Preclinical School, Xinjiang Medical University, Xinjiang, China
| | - Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jason Kirkness
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ellen E Ladenheim
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sheng Bi
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Timothy H Moran
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - David D Fuller
- Departments of Physical Therapy, University of Florida, Gainesville, FL.,McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Alan R Schwartz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
89
|
Reardon TR, Murray AJ, Turi GF, Wirblich C, Croce KR, Schnell MJ, Jessell TM, Losonczy A. Rabies Virus CVS-N2c(ΔG) Strain Enhances Retrograde Synaptic Transfer and Neuronal Viability. Neuron 2016; 89:711-24. [PMID: 26804990 DOI: 10.1016/j.neuron.2016.01.004] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/22/2015] [Accepted: 12/24/2015] [Indexed: 12/20/2022]
Abstract
Virally based transsynaptic tracing technologies are powerful experimental tools for neuronal circuit mapping. The glycoprotein-deletion variant of the SAD-B19 vaccine strain rabies virus (RABV) has been the reagent of choice in monosynaptic tracing, since it permits the mapping of synaptic inputs to genetically marked neurons. Since its introduction, new helper viruses and reagents that facilitate complementation have enhanced the efficiency of SAD-B19(ΔG) transsynaptic transfer, but there has been little focus on improvements to the core RABV strain. Here we generate a new deletion mutant strain, CVS-N2c(ΔG), and examine its neuronal toxicity and efficiency in directing retrograde transsynaptic transfer. We find that by comparison with SAD-B19(ΔG), the CVS-N2c(ΔG) strain exhibits a reduction in neuronal toxicity and a marked enhancement in transsynaptic neuronal transfer. We conclude that the CVS-N2c(ΔG) strain provides a more effective means of mapping neuronal circuitry and of monitoring and manipulating neuronal activity in vivo in the mammalian CNS.
Collapse
Affiliation(s)
- Thomas R Reardon
- Departments of Neuroscience and Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University, New York, NY 10032, USA
| | - Andrew J Murray
- Departments of Neuroscience and Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University, New York, NY 10032, USA.
| | - Gergely F Turi
- Department of Neuroscience, Columbia University, New York, NY 10032, USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Katherine R Croce
- Departments of Neuroscience and Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University, New York, NY 10032, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Thomas M Jessell
- Departments of Neuroscience and Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University, New York, NY 10032, USA; Kavli Institute for Brain Science, Columbia University, New York, NY 10032, USA.
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY 10032, USA; Kavli Institute for Brain Science, Columbia University, New York, NY 10032, USA
| |
Collapse
|
90
|
Sarkar A, Marchetto MC, Gage FH. Synaptic activity: An emerging player in schizophrenia. Brain Res 2015; 1656:68-75. [PMID: 26723567 DOI: 10.1016/j.brainres.2015.12.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 12/02/2015] [Accepted: 12/15/2015] [Indexed: 01/15/2023]
Abstract
Schizophrenia is a polygenic disorder with a complex etiology. While the genetic and molecular underpinnings of the disease are poorly understood, variations in genes encoding synaptic pathways are consistently implicated. Although its impact is still an open question, a deficit in synaptic activity provides an attractive model to explain the cognitive etiology of schizophrenia. Recent advances in high-throughput imaging and functional studies bring new hope for the application of in vitro disease modeling with patient-derived neurons to empirically ascertain the extent to which these synaptic pathways are involved in the disease. In addition, the emergent avenue of research targeted to probe neuronal connections is revealing critical insight into circuitry and may influence how we think about psychiatric disorders in the near future. This article is part of a Special Issue entitled SI: Exploiting human neurons.
Collapse
Affiliation(s)
- Anindita Sarkar
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Maria C Marchetto
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Fred H Gage
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
91
|
Zhang Y, Zhao S, Rodriguez E, Takatoh J, Han BX, Zhou X, Wang F. Identifying local and descending inputs for primary sensory neurons. J Clin Invest 2015; 125:3782-94. [PMID: 26426077 DOI: 10.1172/jci81156] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 07/23/2015] [Indexed: 12/25/2022] Open
Abstract
Primary pain and touch sensory neurons not only detect internal and external sensory stimuli, but also receive inputs from other neurons. However, the neuronal derived inputs for primary neurons have not been systematically identified. Using a monosynaptic rabies viruses-based transneuronal tracing method combined with sensory-specific Cre-drivers, we found that sensory neurons receive intraganglion, intraspinal, and supraspinal inputs, the latter of which are mainly derived from the rostroventral medulla (RVM). The viral-traced central neurons were largely inhibitory but also consisted of some glutamatergic neurons in the spinal cord and serotonergic neurons in the RVM. The majority of RVM-derived descending inputs were dual GABAergic and enkephalinergic (opioidergic). These inputs projected through the dorsolateral funiculus and primarily innervated layers I, II, and V of the dorsal horn, where pain-sensory afferents terminate. Silencing or activation of the dual GABA/enkephalinergic RVM neurons in adult animals substantially increased or decreased behavioral sensitivity, respectively, to heat and mechanical stimuli. These results are consistent with the fact that both GABA and enkephalin can exert presynaptic inhibition of the sensory afferents. Taken together, this work provides a systematic view of and a set of tools for examining peri- and extrasynaptic regulations of pain-afferent transmission.
Collapse
|
92
|
Sato S, Ohara S, Tsutsui KI, Iijima T. Effects of G-gene Deletion and Replacement on Rabies Virus Vector Gene Expression. PLoS One 2015; 10:e0128020. [PMID: 26023771 PMCID: PMC4449044 DOI: 10.1371/journal.pone.0128020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 04/21/2015] [Indexed: 12/26/2022] Open
Abstract
The glycoprotein-gene (G gene) -deleted rabies virus (RV) vector is a powerful tool to examine the function and structure of neural circuits. We previously reported that the deletion of the G gene enhances the transgene expression level of the RV vector. However, the mechanism of this enhancement remains to be clarified. We presume that there are two possible factors for this enhancement. The first factor is the glycoprotein of RV, which shows cytotoxicity; thus, may cause a dysfunction in the translation process of infected cells. The second possible factor is the enhanced expression of the L gene, which encodes viral RNA polymerase. In the RV, it is known that the gene expression level is altered depending on the position of the gene. Since G-gene deletion displaces the L gene in the genome, the expression of the L gene and viral transcription may be enhanced. In this study, we compared the transgene expression level and viral transcription of three recombinant RV vectors. The effect of glycoprotein was examined by comparing the viral gene expression of G-gene-intact RV and G-gene-replaced RV. Despite the fact that the L-gene transcription level of these two RV vectors was similar, the G-gene-replaced RV vector showed higher viral transcription and transgene expression level than the G-gene-intact RV vector. To examine the effect of the position of the L gene, we compared the viral gene expression of the G-gene-deleted RV and G-gene-replaced RV. The G-gene-deleted RV vector showed higher L-gene transcription, viral transcription, and transgene expression level than the G-gene-replaced RV vector. These results indicate that G-gene deletion enhances the transgene expression level through at least two factors, the absence of glycoprotein and enhancement of L-gene expression. These findings enable investigators to design a useful viral vector that shows a controlled desirable transgene expression level in applications.
Collapse
Affiliation(s)
- Sho Sato
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Shinya Ohara
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Ken-Ichiro Tsutsui
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Toshio Iijima
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
- * E-mail:
| |
Collapse
|
93
|
Csáki Á, Vígh B, Boldogkői Z, Vereczki V, Szél Á, Köves K. Is a neuronal chain between the pineal body and the retina in rats and hamsters? Transneural tracing studies. Neurosci Lett 2015; 588:1-6. [DOI: 10.1016/j.neulet.2014.12.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 12/03/2014] [Accepted: 12/19/2014] [Indexed: 11/27/2022]
|
94
|
Matthews DW, Deschênes M, Furuta T, Moore JD, Wang F, Karten HJ, Kleinfeld D. Feedback in the brainstem: an excitatory disynaptic pathway for control of whisking. J Comp Neurol 2015; 523:921-42. [PMID: 25503925 DOI: 10.1002/cne.23724] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 11/03/2014] [Accepted: 12/08/2014] [Indexed: 12/30/2022]
Abstract
Sensorimotor processing relies on hierarchical neuronal circuits to mediate sensory-driven behaviors. In the mouse vibrissa system, trigeminal brainstem circuits are thought to mediate the first stage of vibrissa scanning control via sensory feedback that provides reflexive protraction in response to stimulation. However, these circuits are not well defined. Here we describe a complete disynaptic sensory receptor-to-muscle circuit for positive feedback in vibrissa movement. We identified a novel region of trigeminal brainstem, spinal trigeminal nucleus pars muralis, which contains a class of vGluT2+ excitatory projection neurons involved in vibrissa motor control. Complementary single- and dual-labeling with traditional and virus tracers demonstrate that these neurons both receive primary inputs from vibrissa sensory afferent fibers and send monosynaptic connections to facial nucleus motoneurons that directly innervate vibrissa musculature. These anatomical results suggest a general role of disynaptic architecture in fast positive feedback for motor output that drives active sensation.
Collapse
Affiliation(s)
- David W Matthews
- Graduate Program in Neuroscience, University of California, San Diego, La Jolla, CA, 92093, USA; Department of Physics, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | | | | | | | | | | |
Collapse
|
95
|
Taylor MP, Enquist LW. Axonal spread of neuroinvasive viral infections. Trends Microbiol 2015; 23:283-8. [PMID: 25639651 DOI: 10.1016/j.tim.2015.01.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/31/2014] [Accepted: 01/07/2015] [Indexed: 02/05/2023]
Abstract
Neuroinvasive viral infections invade the nervous system, often eliciting serious disease and death. Members of four viral families are both neuroinvasive and capable of transmitting progeny virions or virion components within the long neuronal extensions known as axons. Axons provide physical structures that enable viral infection to spread within the host while avoiding extracellular immune responses. Technological advances in the analysis of in vivo neural circuits, neuronal culturing, and live imaging of fluorescent fusion proteins have enabled an unprecedented view into the steps of virion assembly, transport, and egress involved in axonal spread. In this review we summarize the literature supporting anterograde (axon to cell) spread of viral infection, describe the various strategies of virion transport, and discuss the effects of spread on populations of neuroinvasive viruses.
Collapse
Affiliation(s)
- Matthew P Taylor
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59718, USA.
| | - Lynn W Enquist
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
96
|
Osakada F, Takahashi M. Challenges in retinal circuit regeneration: linking neuronal connectivity to circuit function. Biol Pharm Bull 2015; 38:341-57. [PMID: 25757915 DOI: 10.1248/bpb.b14-00771] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tremendous progress has been made in retinal regeneration, as exemplified by successful transplantation of retinal pigment epithelia and photoreceptor cells in the adult retina, as well as by generation of retinal tissue from embryonic stem cells and induced pluripotent cells. However, it remains unknown how new photoreceptors integrate within retinal circuits and contribute to vision restoration. There is a large gap in our understanding, at both the cellular and behavioral levels, of the functional roles of new neurons in the adult retina. This gap largely arises from the lack of appropriate methods for analyzing the organization and function of new neurons at the circuit level. To bridge this gap and understand the functional roles of new neurons in living animals, it will be necessary to identify newly formed connections, correlate them with function, manipulate their activity, and assess the behavioral outcome of these manipulations. Recombinant viral vectors are powerful tools not only for controlling gene expression and reprogramming cells, but also for tracing cell fates and neuronal connectivity, monitoring biological functions, and manipulating the physiological state of a specific cell population. These virus-based approaches, combined with electrophysiology and optical imaging, will provide circuit-level insight into neural regeneration and facilitate new strategies for achieving vision restoration in the adult retina. Herein, we discuss challenges and future directions in retinal regeneration research.
Collapse
Affiliation(s)
- Fumitaka Osakada
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University; Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, California 92037, USA; PRESTO, Japan Science and Technology Agency, Saitama 332-0012, Japan.
| | | |
Collapse
|
97
|
Genome Sequence of the Anterograde-Spread-Defective Herpes Simplex Virus 1 Strain MacIntyre. GENOME ANNOUNCEMENTS 2014; 2:2/6/e01161-14. [PMID: 25395637 PMCID: PMC4241663 DOI: 10.1128/genomea.01161-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
We used paired-end Illumina deep sequencing and de novo assembly to determine the genome sequence of herpes simplex virus 1 (HSV-1) strain MacIntyre (aka McIntyre). The MacIntyre strain originated from the brain of a patient with lethal HSV encephalitis and has a unique limitation in its neuronal spread, moving solely in the retrograde direction.
Collapse
|
98
|
Anterograde glycoprotein-dependent transport of newly generated rabies virus in dorsal root ganglion neurons. J Virol 2014; 88:14172-83. [PMID: 25275124 DOI: 10.1128/jvi.02254-14] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Rabies virus (RABV) spread is widely accepted to occur only by retrograde axonal transport. However, examples of anterograde RABV spread in peripheral neurons such as dorsal root ganglion (DRG) neurons indicated a possible bidirectional transport by an uncharacterized mechanism. Here, we analyzed the axonal transport of fluorescence-labeled RABV in DRG neurons by live-cell microscopy. Both entry-related retrograde transport of RABV after infection at axon endings and postreplicative transport of newly formed virus were visualized in compartmentalized DRG neuron cultures. Whereas entry-related transport at 1.5 μm/s occurred only retrogradely, after 2 days of infection, multiple particles were observed in axons moving in both the anterograde and retrograde directions. The dynamics of postreplicative retrograde transport (1.6 μm/s) were similar to those of entry-related retrograde transport. In contrast, anterograde particle transport at 3.4 μm/s was faster, indicating active particle transport. Interestingly, RABV missing the glycoproteins did not move anterogradely within the axon. Thus, anterograde RABV particle transport depended on the RABV glycoprotein. Moreover, colocalization of green fluorescent protein (GFP)-labeled ribonucleoproteins (RNPs) and glycoprotein in distal axonal regions as well as cotransport of labeled RNPs with membrane-anchored mCherry reporter confirmed that either complete enveloped virus particles or vesicle associated RNPs were transported. Our data show that anterograde RABV movement in peripheral DRG neurons occurs by active motor protein-dependent transport. We propose two models for postreplicative long-distance transport in peripheral neurons: either transport of complete virus particles or cotransport of RNPs and G-containing vesicles through axons to release virus at distal sites of infected DRG neurons. IMPORTANCE Rabies virus retrograde axonal transport by dynein motors supports virus spread over long distances and lethal infection of the central nervous system. Though active rabies virus transport has been widely accepted to be unidirectional, evidence for anterograde spread in peripheral neurons supports the hypothesis that in some neurons RABV also enters the anterograde pathway by so-far unknown mechanisms. By live microscopy we visualized fast anterograde axonal transport of rabies virus. The velocities exceeded those of retrograde movements, suggesting that active, most likely kinesin-dependent transport machineries are involved. Dependency of anterograde transport on the expression of virus glycoprotein G and cotransport with vesicles further suggest that complete enveloped virus particles or cotransport of virus ribonucleoprotein and G-containing vesicles occurred. These data provide the first insight in the mechanism of anterograde rabies virus transport and substantially contribute to the understanding of RABV replication and spread of newly formed virus in peripheral neurons.
Collapse
|
99
|
Watakabe A, Takaji M, Kato S, Kobayashi K, Mizukami H, Ozawa K, Ohsawa S, Matsui R, Watanabe D, Yamamori T. Simultaneous visualization of extrinsic and intrinsic axon collaterals in Golgi-like detail for mouse corticothalamic and corticocortical cells: a double viral infection method. Front Neural Circuits 2014; 8:110. [PMID: 25278843 PMCID: PMC4166322 DOI: 10.3389/fncir.2014.00110] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/22/2014] [Indexed: 11/21/2022] Open
Abstract
Here we present a novel tracing technique to stain projection neurons in Golgi-like detail by double viral infection. We used retrograde lentiviral vectors and adeno-associated viral vectors (AAV) to drive “TET-ON/TET-OFF system” in neurons connecting two regions. Using this method, we successfully labeled the corticothalamic (CT) cells of the mouse somatosensory barrel field (S1BF) and motor cortex (M1) in their entirety. We also labeled contra- and ipsilaterally-projecting corticocortical (CC) cells of M1 by targeting contralateral M1 or ipsilateral S1 for retrograde infection. The strength of this method is that we can observe the morphology of specific projection neuron subtypes en masse. We found that the group of CT cells extends their dendrites and intrinsic axons extensively below but not within the thalamorecipient layer in both S1BF and M1, suggesting that the primary target of this cell type is not layer 4. We also found that both ipsi- and contralateral targeting CC cells in M1 commonly exhibit widespread collateral extensions to contralateral M1 (layers 1–6), bilateral S1 and S2 (layers 1, 5 and 6), perirhinal cortex (layers 1, 2/3, 5, and 6), striatum and claustrum. These findings not only strengthened the previous findings of single cell tracings but also extended them by enabling cross-area comparison of CT cells or comparison of CC cells of two different labeling.
Collapse
Affiliation(s)
- Akiya Watakabe
- Division of Brain Biology, National Institute for Basic Biology Okazaki, Japan
| | - Masafumi Takaji
- Division of Brain Biology, National Institute for Basic Biology Okazaki, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine Fukushima, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine Fukushima, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University Shimotsuke, Japan
| | - Keiya Ozawa
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University Shimotsuke, Japan
| | - Sonoko Ohsawa
- Division of Brain Biology, National Institute for Basic Biology Okazaki, Japan
| | - Ryosuke Matsui
- Department of Molecular and Systems Biology, Graduate School of Biostudies, Kyoto University Kyoto, Japan
| | - Dai Watanabe
- Department of Molecular and Systems Biology, Graduate School of Biostudies, Kyoto University Kyoto, Japan
| | - Tetsuo Yamamori
- Division of Brain Biology, National Institute for Basic Biology Okazaki, Japan
| |
Collapse
|
100
|
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: 4.6] [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
|