1
|
You H, Qinghan W, Kangyixin S, Jia Y, Fuqiang X, Fan J. Development of Cre-dependent retrograde trans-multisynaptic tracer based on pseudorabies virus bartha strain. Mol Brain 2025; 18:33. [PMID: 40229811 PMCID: PMC11995500 DOI: 10.1186/s13041-025-01204-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
Mapping the neural circuit of a specific neuronal subclass is central to understanding the working mechanism of the brain. Currently, numerous types of transgenic mice expressing Cre recombinase have been engineered and widely used in neuroscience. To map the multilevel inputs into the neural circuit of a specific neuronal subpopulation, a Cre-dependent retrograde trans-multisynaptic tracer must be developed. The vaccine strain of Pseudorabies virus (PRV, Bartha strain) can infect neurons and spread in a retrograde manner in the neural circuit. In this study, we engineered the genome of PRV Bartha strain to prepare two new tracers, PRV676 and PRV829, by replacing the TK gene of PRV with the Cre-dependent expression cassette of the fluorescent protein gene and the TK gene. These two tracers can separately and Cre-dependently express EGFP and mRuby3 and produce progeny viruses in vitro and in vivo, which can help to map the multilevel inputs of a specific neuronal subpopulation expressing Cre. Collectively, our work provides two new tools for neuroscience research.
Collapse
Affiliation(s)
- Hu You
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advance Technology, Translational Research Center for the Nervous System, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institutes of Advance Technology, Guangdong Provincial Medical Products Administration, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wang Qinghan
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advance Technology, Translational Research Center for the Nervous System, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institutes of Advance Technology, Guangdong Provincial Medical Products Administration, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Sun Kangyixin
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advance Technology, Translational Research Center for the Nervous System, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institutes of Advance Technology, Guangdong Provincial Medical Products Administration, Chinese Academy of Sciences, Shenzhen, 518055, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yang Jia
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advance Technology, Translational Research Center for the Nervous System, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institutes of Advance Technology, Guangdong Provincial Medical Products Administration, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xu Fuqiang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advance Technology, Translational Research Center for the Nervous System, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institutes of Advance Technology, Guangdong Provincial Medical Products Administration, Chinese Academy of Sciences, Shenzhen, 518055, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518106, China
| | - Jia Fan
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advance Technology, Translational Research Center for the Nervous System, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China.
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institutes of Advance Technology, Guangdong Provincial Medical Products Administration, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518106, China.
| |
Collapse
|
2
|
Tucker A, Baltazar A, Eisdorfer JT, Thackray JK, Vo K, Thomas H, Tandon A, Moses J, Singletary B, Gillespie T, Smith A, Pauken A, Nadella S, Pitonak M, Letchuman S, Jang J, Totty M, Jalufka FL, Aceves M, Adler AF, Maren S, Blackmon H, McCreedy DA, Abraira V, Dulin JN. Functional synaptic connectivity of engrafted spinal cord neurons with locomotor circuitry in the injured spinal cord. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.05.644402. [PMID: 40236108 PMCID: PMC11996546 DOI: 10.1101/2025.04.05.644402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Spinal cord injury (SCI) results in significant neurological deficits, with no currently available curative therapies. Neural progenitor cell (NPC) transplantation has emerged as a promising approach for neural repair, as graft-derived neurons (GDNs) can integrate into the host spinal cord and support axon regeneration. However, the mechanisms underlying functional recovery remain poorly understood. In this study, we investigate the synaptic integration of NPC-derived neurons into locomotor circuits, the projection patterns of distinct neuronal subtypes, and their potential to modulate motor circuit activity. Using transsynaptic tracing in a mouse thoracic contusion SCI model, we found that NPC-derived neurons form synaptic connections with host locomotor circuits, albeit at low frequencies. Furthermore, we mapped the axon projections of V0C and V2a interneurons, revealing distinct termination patterns within host spinal cord laminae. To assess functional integration, we employed chemogenetic activation of GDNs, which induced muscle activity in a subset of transplanted animals. However, NPC transplantation alone did not significantly improve locomotor recovery, highlighting a key challenge in the field. Our findings suggest that while GDNs can integrate into host circuits and modulate motor activity, synaptic connectivity remains a limiting factor in functional recovery. Future studies should focus on enhancing graft-host connectivity and optimizing transplantation strategies to maximize therapeutic benefits for SCI.
Collapse
|
3
|
Zhuang L, Gong J, Shen J, Zhao Y, Yang J, Liu Q, Zhang Y, Shen Q. Advances in molecular epidemiology and detection methods of pseudorabies virus. DISCOVER NANO 2025; 20:45. [PMID: 39992589 PMCID: PMC11850701 DOI: 10.1186/s11671-025-04217-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Pseudorabies (PR), a highly contagious disease caused by the pseudorabies virus (PRV), represents a significant threat to the global swine industry. Despite the success of developed countries in controlling the PRV epidemic through swine pseudorabies eradication programs, wild boars, as a potential source of infection, still require sustained attention and effective control measures. Concurrently, there has been considerable global attention directed towards cases of PRV infection in humans. In consideration of the aforementioned factors, this paper presents a comprehensive review of recent developments in the PRV genome, epidemiology, vaccine research, and molecular detection methods. The epidemiology section presents an analysis of the transmission routes, susceptible animal groups, and geographic distribution of PRV, as well as an examination of the trend of the epidemic in recent years. In the field of vaccine research, the current development of genetically engineered vaccines is emphasized, and the immunogenicity and safety of vaccines are discussed. Moreover, the molecular detection techniques utilized to identify PRV, including immunological methods, nucleic acid detection methods, biosensors, and so forth, are presented in a systematic manner. Finally, this paper presents a comprehensive discussion of the current status of PRV-related research and offers insights into future directions, with the aim of providing a foundation for the scientific prevention and control of PRV.
Collapse
Affiliation(s)
- Linlin Zhuang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 211102, People's Republic of China
| | - Jiansen Gong
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, 225125, People's Republic of China
| | - Jingyi Shen
- School of Animal Science and Food Engineering, Jinling Institute of Technology, Nanjing, 210038, People's Republic of China
| | - Ying Zhao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 211102, People's Republic of China
| | - Jianbo Yang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
| | - Qingxin Liu
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
| | - Yu Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 211102, People's Republic of China.
| | - Qiuping Shen
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China.
| |
Collapse
|
4
|
Yang J, Li L, Xu F, Jia F. Development of a reporter feline herpesvirus-1 for antiviral screening assays. Vet Res 2024; 55:167. [PMID: 39696698 DOI: 10.1186/s13567-024-01430-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/28/2024] [Indexed: 12/20/2024] Open
Abstract
Feline herpesvirus type 1 (FHV-1), a member of the Herpesviridae family, is one of the most important pathogens that causes upper respiratory tract disease in felines. Following infection, FHV-1 can spread retrogradely to the trigeminal ganglia, establishing a life-long latency. Although vaccines are available for routine feline vaccination, FHV-1 is still an agent that poses a serious threat to feline health. There are currently no specific drugs for the treatment of FHV-1. To facilitate the screening of antiviral drugs, we constructed a reporter FHV-1 virus, which expresses a secreted Gaussia luciferase (GLuc) and a bright green fluorescent protein, mNeonGreen. The reporter virus shows slower growth than does the wild-type FHV-1. The expression of the two reporter genes, Gluc and mNeonGreen, was consistent with viral propagation and remained stable during continuous passage in CRFK cells, even after twenty rounds. In addition, the known inhibitor ganciclovir was used to confirm the characteristics of the reporter virus for drug screening. We found that the reporter FHV-1 is suitable for antiviral screening assays. Overall, our work provides a useful tool for screening drugs to combat FHV-1.
Collapse
Affiliation(s)
- Jia Yang
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Li Li
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Fuqiang Xu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518107, China
| | - Fan Jia
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518107, China.
| |
Collapse
|
5
|
Walter M, Haick AK, Riley R, Massa PA, Strongin DE, Klouser LM, Loprieno MA, Stensland L, Santo TK, Roychoudhury P, Aubert M, Taylor MP, Jerome KR, Verdin E. Viral gene drive spread during herpes simplex virus 1 infection in mice. Nat Commun 2024; 15:8161. [PMID: 39289368 PMCID: PMC11408514 DOI: 10.1038/s41467-024-52395-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024] Open
Abstract
Gene drives are genetic modifications designed to propagate efficiently through a population. Most applications rely on homologous recombination during sexual reproduction in diploid organisms such as insects, but we recently developed a gene drive in herpesviruses that relies on co-infection of cells by wild-type and engineered viruses. Here, we report on a viral gene drive against human herpes simplex virus 1 (HSV-1) and show that it propagates efficiently in cell culture and during HSV-1 infection in mice. We describe high levels of co-infection and gene drive-mediated recombination in neuronal tissues during herpes encephalitis as the infection progresses from the site of inoculation to the peripheral and central nervous systems. In addition, we show evidence that a superinfecting gene drive virus could recombine with wild-type viruses during latent infection. These findings indicate that HSV-1 achieves high rates of co-infection and recombination during viral infection, a phenomenon that is currently underappreciated. Overall, this study shows that a viral gene drive could spread in vivo during HSV-1 infection, paving the way toward therapeutic applications.
Collapse
Affiliation(s)
- Marius Walter
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, US.
- Buck Institute for Research on Aging, Novato, CA, US.
| | - Anoria K Haick
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, US
| | | | - Paola A Massa
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, US
| | - Daniel E Strongin
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
| | - Lindsay M Klouser
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, US
| | - Michelle A Loprieno
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, US
| | - Laurence Stensland
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
| | - Tracy K Santo
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
| | - Pavitra Roychoudhury
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, US
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
| | - Martine Aubert
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, US
| | - Matthew P Taylor
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT, US
| | - Keith R Jerome
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, US.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US.
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, US.
| |
Collapse
|
6
|
Ambrosini AE, Borg KM, Deshmukh N, Berry MJ, Enquist LW, Hogue IB. Alpha herpesvirus exocytosis from neuron cell bodies uses constitutive secretory mechanisms, and egress and spread from axons is independent of neuronal firing activity. PLoS Pathog 2024; 20:e1012139. [PMID: 38578790 PMCID: PMC11023632 DOI: 10.1371/journal.ppat.1012139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/17/2024] [Accepted: 03/20/2024] [Indexed: 04/07/2024] Open
Abstract
Alpha herpesviruses naturally infect the peripheral nervous system, and can spread to the central nervous system, causing severe debilitating or deadly disease. Because alpha herpesviruses spread along synaptic circuits, and infected neurons exhibit altered electrophysiology and increased spontaneous activity, we hypothesized that alpha herpesviruses use activity-dependent synaptic vesicle-like regulated secretory mechanisms for egress and spread from neurons. Using live-cell fluorescence microscopy, we show that Pseudorabies Virus (PRV) particles use the constitutive Rab6 post-Golgi secretory pathway to exit from the cell body of primary neurons, independent of local calcium signaling. Some PRV particles colocalize with Rab6 in the proximal axon, but we did not detect colocalization/co-transport in the distal axon. Thus, the specific secretory mechanisms used for viral egress from axons remains unclear. To address the role of neuronal activity more generally, we used a compartmentalized neuron culture system to measure the egress and spread of PRV from axons, and pharmacological and optogenetics approaches to modulate neuronal activity. Using tetrodotoxin to silence neuronal activity, we observed no inhibition, and using potassium chloride or optogenetics to elevate neuronal activity, we also show no increase in virus spread from axons. We conclude that PRV egress from neurons uses constitutive secretory mechanisms: generally, activity-independent mechanisms in axons, and specifically, the constitutive Rab6 post-Golgi secretory pathway in cell bodies.
Collapse
Affiliation(s)
- Anthony E. Ambrosini
- Department of Molecular Biology, and Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey, United States of America
| | - Kayla M. Borg
- ASU-Banner Neurodegenerative Research Center, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Nikhil Deshmukh
- Department of Molecular Biology, and Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey, United States of America
| | - Michael J. Berry
- Department of Molecular Biology, and Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey, United States of America
| | - Lynn W. Enquist
- Department of Molecular Biology, and Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey, United States of America
| | - Ian B. Hogue
- ASU-Banner Neurodegenerative Research Center, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
7
|
Zsombok A, Desmoulins LD, Derbenev AV. Sympathetic circuits regulating hepatic glucose metabolism: where we stand. Physiol Rev 2024; 104:85-101. [PMID: 37440208 PMCID: PMC11281813 DOI: 10.1152/physrev.00005.2023] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/12/2023] [Accepted: 07/10/2023] [Indexed: 07/14/2023] Open
Abstract
The prevalence of metabolic disorders, including type 2 diabetes mellitus, continues to increase worldwide. Although newer and more advanced therapies are available, current treatments are still inadequate and the search for solutions remains. The regulation of energy homeostasis, including glucose metabolism, involves an exchange of information between the nervous systems and peripheral organs and tissues; therefore, developing treatments to alter central and/or peripheral neural pathways could be an alternative solution to modulate whole body metabolism. Liver glucose production and storage are major mechanisms controlling glycemia, and the autonomic nervous system plays an important role in the regulation of hepatic functions. Autonomic nervous system imbalance contributes to excessive hepatic glucose production and thus to the development and progression of type 2 diabetes mellitus. At cellular levels, change in neuronal activity is one of the underlying mechanisms of autonomic imbalance; therefore, modulation of the excitability of neurons involved in autonomic outflow governance has the potential to improve glycemic status. Tissue-specific subsets of preautonomic neurons differentially control autonomic outflow; therefore, detailed information about neural circuits and properties of liver-related neurons is necessary for the development of strategies to regulate liver functions via the autonomic nerves. This review provides an overview of our current understanding of the hypothalamus-ventral brainstem-liver pathway involved in the sympathetic regulation of the liver, outlines strategies to identify organ-related neurons, and summarizes neuronal plasticity during diabetic conditions with a particular focus on liver-related neurons in the paraventricular nucleus.
Collapse
Affiliation(s)
- Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, United States
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana, United States
| | - Lucie D Desmoulins
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, United States
| | - Andrei V Derbenev
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, United States
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana, United States
| |
Collapse
|
8
|
Rashid M, Kondoh K, Palfalvi G, Nakajima KI, Minokoshi Y. Inhibition of high-fat diet-induced inflammatory responses in adipose tissue by SF1-expressing neurons of the ventromedial hypothalamus. Cell Rep 2023; 42:112627. [PMID: 37339627 DOI: 10.1016/j.celrep.2023.112627] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 03/27/2023] [Accepted: 05/24/2023] [Indexed: 06/22/2023] Open
Abstract
Inflammation and thermogenesis in white adipose tissue (WAT) at different sites influence the overall effects of obesity on metabolic health. In mice fed a high-fat diet (HFD), inflammatory responses are less pronounced in inguinal WAT (ingWAT) than in epididymal WAT (epiWAT). Here we show that ablation and activation of steroidogenic factor 1 (SF1)-expressing neurons in the ventromedial hypothalamus (VMH) oppositely affect the expression of inflammation-related genes and the formation of crown-like structures by infiltrating macrophages in ingWAT, but not in epiWAT, of HFD-fed mice, with these effects being mediated by sympathetic nerves innervating ingWAT. In contrast, SF1 neurons of the VMH preferentially regulated the expression of thermogenesis-related genes in interscapular brown adipose tissue (BAT) of HFD-fed mice. These results suggest that SF1 neurons of the VMH differentially regulate inflammatory responses and thermogenesis among various adipose tissue depots and restrain inflammation associated with diet-induced obesity specifically in ingWAT.
Collapse
Affiliation(s)
- Misbah Rashid
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan
| | - Kunio Kondoh
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan.
| | - Gergo Palfalvi
- Division of Evolutionary Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | - Ken-Ichiro Nakajima
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan
| | - Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan.
| |
Collapse
|
9
|
Soriano JE, Hudelle R, Squair JW, Mahe L, Amir S, Gautier M, Puchalt VP, Barraud Q, Phillips AA, Courtine G. Longitudinal interrogation of sympathetic neural circuits and hemodynamics in preclinical models. Nat Protoc 2023; 18:340-373. [PMID: 36418397 DOI: 10.1038/s41596-022-00764-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 05/27/2022] [Indexed: 11/24/2022]
Abstract
Neurological disorders, including spinal cord injury, result in hemodynamic instability due to the disruption of supraspinal projections to the sympathetic circuits located in the spinal cord. We recently developed a preclinical model that allows the identification of the topology and dynamics through which sympathetic circuits modulate hemodynamics, supporting the development of a neuroprosthetic baroreflex that precisely controls blood pressure in rats, monkeys and humans with spinal cord injuries. Here, we describe the continuous monitoring of arterial blood pressure and sympathetic nerve activity over several months in preclinical models of chronic neurological disorders using commercially available telemetry technologies, as well as optogenetic and neuronal tract-tracing procedures specifically adapted to the sympathetic circuitry. Using a blueprint to construct a negative-pressure chamber, the approach enables the reproduction, in rats, of well-controlled and reproducible episodes of hypotension-mimicking orthostatic challenges already used in humans. Blood pressure variations can thus be directly induced and linked to the molecular, functional and anatomical properties of specific neurons in the brainstem, spinal cord and ganglia. Each procedure can be completed in under 2 h, while the construction of the negative-pressure chamber requires up to 1 week. With training, individuals with a basic understanding of cardiovascular physiology, engineering or neuroscience can collect longitudinal recordings of hemodynamics and sympathetic nerve activity over several months.
Collapse
Affiliation(s)
- Jan Elaine Soriano
- Neuro-X Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.,Department of Physiology and Pharmacology, Clinical Neurosciences, Cardiac Sciences, Hotchkiss Brain Institute, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Rémi Hudelle
- Neuro-X Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Jordan W Squair
- Neuro-X Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.,Department of Physiology and Pharmacology, Clinical Neurosciences, Cardiac Sciences, Hotchkiss Brain Institute, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Lois Mahe
- Neuro-X Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Suje Amir
- Neuro-X Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Matthieu Gautier
- Neuro-X Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Victor Perez Puchalt
- Neuro-X Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Quentin Barraud
- Neuro-X Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Aaron A Phillips
- Department of Physiology and Pharmacology, Clinical Neurosciences, Cardiac Sciences, Hotchkiss Brain Institute, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| | - Gregoire Courtine
- Neuro-X Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland. .,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland. .,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.
| |
Collapse
|
10
|
Gao Y, Sridhar A, Bernard N, He B, Zhang H, Pirotte S, Desmecht S, Vancsok C, Boutier M, Suárez NM, Davison AJ, Donohoe O, Vanderplasschen AFC. Virus-induced interference as a means for accelerating fitness-based selection of cyprinid herpesvirus 3 single-nucleotide variants in vitro and in vivo. Virus Evol 2023; 9:vead003. [PMID: 36816049 PMCID: PMC9936792 DOI: 10.1093/ve/vead003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/16/2022] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
Cyprinid herpesvirus 3 (CyHV-3) is the archetype of fish alloherpesviruses and is advantageous to research because, unlike many herpesviruses, it can be studied in the laboratory by infection of the natural host (common and koi carp). Previous studies have reported a negative correlation among CyHV-3 strains between viral growth in vitro (in cell culture) and virulence in vivo (in fish). This suggests the existence of genovariants conferring enhanced fitness in vitro but reduced fitness in vivo and vice versa. Here, we identified the syncytial plaque formation in vitro as a common trait of CyHV-3 strains adapted to cell culture. A comparison of the sequences of virion transmembrane protein genes in CyHV-3 strains, and the use of various recombinant viruses, demonstrated that this trait is linked to a single-nucleotide polymorphism (SNP) in the open reading frame (ORF) 131 coding sequence (C225791T mutation) that results in codon 183 encoding either an alanine (183A) or a threonine (183T) residue. In experiments involving infections with recombinant viruses differing only by this SNP, the 183A genovariant associated with syncytial plaque formation was the more fit in vitro but the less fit in vivo. In experiments involving coinfection with both viruses, the more fit genovariant contributed to the purifying selection of the less fit genovariant by outcompeting it. In addition, this process appeared to be accelerated by viral stimulation of interference at a cellular level and stimulation of resistance to superinfection at a host level. Collectively, this study illustrates how the fundamental biological properties of some viruses and their hosts may have a profound impact on the degree of diversity that arises within viral populations.
Collapse
Affiliation(s)
- Yuan Gao
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège B-4000, Belgium
| | - Arun Sridhar
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège B-4000, Belgium
| | - Noah Bernard
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège B-4000, Belgium
| | - Bo He
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège B-4000, Belgium
| | - Haiyan Zhang
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège B-4000, Belgium
| | - Sébastien Pirotte
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège B-4000, Belgium
| | - Salomé Desmecht
- Laboratory of Animal Genomics, GIGA-Medical Genomics, GIGA-Institute, University of Liège, Liège B-4000, Belgium
| | - Catherine Vancsok
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège B-4000, Belgium
| | - Maxime Boutier
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège B-4000, Belgium
| | - Nicolás M Suárez
- MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK
| | - Andrew J Davison
- MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK
| | - Owen Donohoe
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège B-4000, Belgium.,Bioscience Research Institute, Technological University of the Shannon, Midlands Midwest, Athlone, Co. Westmeath N37HD68, Ireland
| | - Alain F C Vanderplasschen
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège B-4000, Belgium
| |
Collapse
|
11
|
O'Dell DE, Smith-Bell CA, Enquist LW, Engel EA, Schreurs BG. Eyeblink tract tracing with two strains of herpes simplex virus 1. Brain Res 2022; 1793:148040. [PMID: 35932812 DOI: 10.1016/j.brainres.2022.148040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Neuroinvasive herpes simplex-1 (HSV-1) isolates including H129 and McIntyre cross at or near synapses labeling higher-order neurons directly connected to infected cells. H129 spreads predominately in the anterograde direction while McIntyre strains spread only in the retrograde direction. However, it is unknown if neurons are functional once infected with derivatives of H129 or McIntyre. NEW METHOD We describe a previously unpublished HSV-1 recombinant derived from H129 (HSV-373) expressing mCherry fluorescent reporters and one new McIntyre recombinant (HSV-780) expressing the mCherry fluorophore and demonstrate how infections affect neuron viability. RESULTS AND COMPARISON WITH EXISTING METHODS Each recombinant virus behaved similarly and spread to the target 4 days post-infection. We tested H129 recombinant infected neurons for neurodegeneration using Fluoro-jade C and found them to be necrotic as a result of viral infection. We performed dual inoculations with both HSV-772 and HSV-780 to identify cells comprising both the anterograde pathway and the retrograde pathway, respectively, of our circuit of study. We examined the presence of postsynaptic marker PSD-95, which plays a role in synaptic plasticity, in HSV-772 infected and in dual-infected rats (HSV-772 and HSV-780). PSD-95 reactivity decreased in HSV-772-infected neurons and dual-infected tissue had no PSD-95 reactivity. CONCLUSIONS Infection by these new recombinant viruses traced the circuit of interest but functional studies of the cells comprising the pathway were not possible because viral-infected neurons died as a result of necrosis or were stripped of PSD-95 by the time the viral labels reached the target.
Collapse
Affiliation(s)
- Deidre E O'Dell
- Department of Neuroscience, Rockefeller Neuroscience Institute, United States; West Virginia University, Morgantown, WV 26505, United States.
| | - Carrie A Smith-Bell
- Department of Neuroscience, Rockefeller Neuroscience Institute, United States; West Virginia University, Morgantown, WV 26505, United States
| | - Lynn W Enquist
- Department of Molecular Biology, United States; Princeton Neuroscience Institute, United States; Princeton University, Princeton, NJ 08544, United States
| | - Esteban A Engel
- Princeton Neuroscience Institute, United States; Princeton University, Princeton, NJ 08544, United States
| | - Bernard G Schreurs
- Department of Neuroscience, Rockefeller Neuroscience Institute, United States; West Virginia University, Morgantown, WV 26505, United States.
| |
Collapse
|
12
|
Zheng DJ, Okobi DE, Shu R, Agrawal R, Smith SK, Long MA, Phelps SM. Mapping the vocal circuitry of Alston's singing mouse with pseudorabies virus. J Comp Neurol 2022; 530:2075-2099. [PMID: 35385140 PMCID: PMC11987554 DOI: 10.1002/cne.25321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/06/2022] [Accepted: 03/07/2022] [Indexed: 11/11/2022]
Abstract
Vocalizations are often elaborate, rhythmically structured behaviors. Vocal motor patterns require close coordination of neural circuits governing the muscles of the larynx, jaw, and respiratory system. In the elaborate vocalization of Alston's singing mouse (Scotinomys teguina) each note of its rapid, frequency-modulated trill is accompanied by equally rapid modulation of breath and gape. To elucidate the neural circuitry underlying this behavior, we introduced the polysynaptic retrograde neuronal tracer pseudorabies virus (PRV) into the cricothyroid and digastricus muscles, which control frequency modulation and jaw opening, respectively. Each virus singly labels ipsilateral motoneurons (nucleus ambiguus for cricothyroid, and motor trigeminal nucleus for digastricus). We find that the two isogenic viruses heavily and bilaterally colabel neurons in the gigantocellular reticular formation, a putative central pattern generator. The viruses also show strong colabeling in compartments of the midbrain including the ventrolateral periaqueductal gray and the parabrachial nucleus, two structures strongly implicated in vocalizations. In the forebrain, regions important to social cognition and energy balance both exhibit extensive colabeling. This includes the paraventricular and arcuate nuclei of the hypothalamus, the lateral hypothalamus, preoptic area, extended amygdala, central amygdala, and the bed nucleus of the stria terminalis. Finally, we find doubly labeled neurons in M1 motor cortex previously described as laryngeal, as well as in the prelimbic cortex, which indicate these cortical regions play a role in vocal production. The progress of both viruses is broadly consistent with vertebrate-general patterns of vocal circuitry, as well as with circuit models derived from primate literature.
Collapse
Affiliation(s)
- Da-Jiang Zheng
- Department of Integrative Biology, The University of Texas at Austin, Austin TX 78712, USA
| | - Daniel E. Okobi
- Department of Neurology, University of California Los Angeles, Los Angeles CA 90095, USA
| | - Ryan Shu
- Department of Integrative Biology, The University of Texas at Austin, Austin TX 78712, USA
| | - Rania Agrawal
- Department of Integrative Biology, The University of Texas at Austin, Austin TX 78712, USA
| | - Samantha K. Smith
- Department of Integrative Biology, The University of Texas at Austin, Austin TX 78712, USA
| | - Michael A. Long
- NYU Neuroscience Institute and Department of Otolaryngology, New York University Langone Medical Center, New York NY 10016, USA
| | - Steven M. Phelps
- Department of Integrative Biology, The University of Texas at Austin, Austin TX 78712, USA
| |
Collapse
|
13
|
DeFinis JH, Hou S. Dual-Pseudorabies Viral Tracing for Spinal Tyrosine Hydroxylase Interneurons Involved in Segmental Micturition Reflex Circuitry in Spinal Cord Injured Rats. Neurotrauma Rep 2022; 2:660-668. [PMID: 35018366 PMCID: PMC8742299 DOI: 10.1089/neur.2021.0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Traumatic spinal cord injury (SCI) often leads to urinary dysfunction. Although an involuntary micturition reflex can be established to elicit voiding with time, complications arise in the form of bladder hyper-reflexia and detrusor-sphincter dyssynergia that cause incontinence and inefficient expulsion of urine. To date, the neuronal mechanisms that underlie regulation of micturition after SCI are not well understood. We recently observed an increase of a population of tyrosine hydroxylase (TH)+ cells in the rat lumbosacral cord post-SCI, which contribute to the sustention of a low level of dopamine that modulates the recovered bladder reflex. To identify whether spinal TH+ cells are involved in the micturition reflex pathway post-SCI, two isoforms of the trans-synaptic retrograde tracer, pseudorabies virus encoding green fluorescent protein (GFP; PRV-152) or red fluorescent protein (RFP; PRV-614), were injected into the bladder detrusor or the external urethral sphincter (EUS), respectively, 3 weeks after a spinal cord transection at the 10th thoracic level (T10) in rats. Immunohistochemistry was performed to examine infected TH+ cells in the caudal cord at both 48 and 72 h post-injection. As a result, double-labeled TH+/GFP+ and TH+/RFP+ cells could be found in the superficial dorsal horn, parasympathetic nuclei, and dorsal gray commissure (lamina X) at both time points. More importantly, a shared population of TH+ interneurons (TH+/GFP+/RFP+) exists between bladder and EUS circuitry. These results suggest that spinal TH+ interneurons may coordinate activity of the bladder and EUS that occurs during micturition reflexes post-SCI.
Collapse
Affiliation(s)
- Jaclyn H DeFinis
- Marion Murray Spinal Cord Research Center, Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Shaoping Hou
- Marion Murray Spinal Cord Research Center, Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Fortino TA, Randelman ML, Hall AA, Singh J, Bloom DC, Engel E, Hoh DJ, Hou S, Zholudeva LV, Lane MA. Transneuronal tracing to map connectivity in injured and transplanted spinal networks. Exp Neurol 2022; 351:113990. [DOI: 10.1016/j.expneurol.2022.113990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/09/2021] [Accepted: 01/20/2022] [Indexed: 11/24/2022]
|
15
|
McEwan S, Kwon H, Tahiri A, Shanmugarajah N, Cai W, Ke J, Huang T, Belton A, Singh B, Wang L, Pang ZP, Dirice E, Engel EA, El Ouaamari A. Deconstructing the origins of sexual dimorphism in sensory modulation of pancreatic β cells. Mol Metab 2021; 53:101260. [PMID: 34023484 PMCID: PMC8258979 DOI: 10.1016/j.molmet.2021.101260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/29/2021] [Accepted: 05/17/2021] [Indexed: 01/02/2023] Open
Abstract
The regulation of glucose-stimulated insulin secretion and glucose excursion has a sensory component that operates in a sex-dependent manner. OBJECTIVE Here, we aim to dissect the basis of the sexually dimorphic interaction between sensory neurons and pancreatic β cells and its overall impact on insulin release and glucose homeostasis. METHODS We used viral retrograde tracing techniques, surgical and chemodenervation models, and primary cell-based co-culture systems to uncover the biology underlying sex differences in sensory modulation of pancreatic β-cell activity. RESULTS Retrograde transsynaptic labeling revealed a sex difference in the density of sensory innervation in the pancreas. The number of sensory neurons emanating from the dorsal root and nodose ganglia that project in the pancreas is higher in male than in female mice. Immunostaining and confocal laser scanning microscopy confirmed the higher abundance of peri-islet sensory axonal tracts in the male pancreas. Capsaicin-induced sensory chemodenervation concomitantly enhanced glucose-stimulated insulin secretion and glucose clearance in male mice. These metabolic benefits were blunted when mice were orchidectomized prior to the ablation of sensory nerves. Interestingly, orchidectomy also lowered the density of peri-islet sensory neurons. In female mice, capsaicin treatment did not affect glucose-induced insulin secretion nor glucose excursion and ovariectomy did not modify these outcomes. Interestingly, same- and opposite-sex sensory-islet co-culture paradigms unmasked the existence of potential gonadal hormone-independent mechanisms mediating the male-female difference in sensory modulation of islet β-cell activity. CONCLUSION Taken together, these data suggest that the sex-biased nature of the sensory control of islet β-cell activity is a result of a combination of neurodevelopmental inputs, sex hormone-dependent mechanisms and the potential action of somatic molecules encoded by the sex chromosome complement.
Collapse
Affiliation(s)
- Sara McEwan
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Hyokjoon Kwon
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Azeddine Tahiri
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Nivetha Shanmugarajah
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, 11568, USA
| | - Weikang Cai
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, 11568, USA
| | - Jin Ke
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Tianwen Huang
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ariana Belton
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Bhagat Singh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Le Wang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Zhiping P. Pang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Ercument Dirice
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - Esteban A. Engel
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Abdelfattah El Ouaamari
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,Corresponding author. Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
16
|
Karnup S, De Groat W. Mapping of spinal interneurons involved in regulation of the lower urinary tract in juvenile male rats. IBRO Rep 2020; 9:115-131. [PMID: 32775758 PMCID: PMC7394742 DOI: 10.1016/j.ibror.2020.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/03/2020] [Indexed: 12/23/2022] Open
Abstract
Coordination between the urinary bladder (BL) and external urethral sphincter (EUS) is necessary for storage and elimination of urine. In rats interneuronal circuits at two levels of the spinal cord (i.e., L6-S1 and L3-L4) play an important role in this coordination. In the present experiments retrograde trans-synaptic transport of pseudorabies virus (PRV) encoding fluorescent markers (GFP and RFP) was used to trace these circuits. To examine the relative localization of EUS-related and BL-related interneuronal populations we injected PRV-GFP into the EUS and PRV-RFP into the BL wall. The PRV infected populations of spinal interneurons were localized primarily in the dorsal commissure (DCM) of L6/S1 and in a hypothesized lumbar spinal coordinating center (LSCC) in L3/L4 above and lateral to central canal (CC). At both sites colocalization of markers occurred in a substantial number of labeled interneurons indicating concomitant involvement of these double-labelled neurons in the EUS- and BL-circuits and suggesting their role in EUS-BL coordination. Intense GFP or RFP fluorescent was detected in a subpopulation of cells at both sites suggesting that they were infected earlier and therefore likely to represent first order, primary interneurons that directly synapse with output neurons. Larger numbers of weakly fluorescent neurons that likely represent second order interneurons were also identified. Within the population of EUS-related first order interneurons only 3-8 % exhibited positive immunoreaction for an early transcription factor Pax2 specific to GABAergic and glycinergic inhibitory neurons suggesting that the majority of interneurons in DCM and LSCC projecting directly to the EUS motoneurons are excitatory.
Collapse
Key Words
- BCM, bulbocavernosus muscle
- BL, bladder
- B_G, bright green
- B_R, bright red
- CC -, central canal
- DCM, dorsal commissure
- DSD, detrusor-sphincter-dyssynergia
- EMG, electromyogram
- EUS, external urethral sphincter
- EUS-MN, motoneuron of the external urethral sphincter
- GFP, green fluorescent protein
- IML, intermediolateral nucleus
- IN, interneuron
- LSCC, lumbar spinal coordinating center
- LUT, lower urinary tract
- Lf, lateral funiculus
- PPN, propriospinal neuron
- PRV, pseudorabies virus
- Pseudorabies virus
- RFP, red fluorescent protein
- SC, spinal cord
- SCI, spinal cord injury
- SPPN, spinal parasympathetic preganglionic neuron
- Spinal cord
- Transsynaptic tracing
- VMf, ventro-medial funiculus
- W_G, weak/moderate green
- W_R, weak/moderate red
- pIN, primary interneuron
- sIN, secondary interneuron
Collapse
Affiliation(s)
- S.V. Karnup
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15213, United States
| | - W.C. De Groat
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15213, United States
| |
Collapse
|
17
|
Abstract
Recombinant viruses are the workhorse of modern neuroscience. Whether one would like to understand a neuron's morphology, natural activity patterns, molecular composition, connectivity or behavioural and physiologic function, most studies begin with the injection of an engineered virus, often an adeno-associated virus or herpes simplex virus, among many other types. Recombinant viruses currently enable some combination of cell type-specific, circuit-selective, activity-dependent and spatiotemporally resolved transgene expression. Viruses are now used routinely to study the molecular and cellular functions of a gene within an identified cell type in the brain, and enable the application of optogenetics, chemogenetics, calcium imaging and related approaches. These advantageous properties of engineered viruses thus enable characterization of neuronal function at unprecedented resolution. However, each virus has specific advantages and disadvantages, which makes viral tool selection paramount for properly designing and executing experiments within the central nervous system. In the current Review, we discuss the key principles and uses of engineered viruses and highlight innovations that are needed moving forward.
Collapse
Affiliation(s)
- Alexander R Nectow
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
18
|
Muller PA, Matheis F, Schneeberger M, Kerner Z, Jové V, Mucida D. Microbiota-modulated CART + enteric neurons autonomously regulate blood glucose. Science 2020; 370:314-321. [PMID: 32855216 PMCID: PMC7886298 DOI: 10.1126/science.abd6176] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/18/2020] [Indexed: 12/19/2022]
Abstract
The gut microbiota affects tissue physiology, metabolism, and function of both the immune and nervous systems. We found that intrinsic enteric-associated neurons (iEANs) in mice are functionally adapted to the intestinal segment they occupy; ileal and colonic neurons are more responsive to microbial colonization than duodenal neurons. Specifically, a microbially responsive subset of viscerofugal CART+ neurons, enriched in the ileum and colon, modulated feeding and glucose metabolism. These CART+ neurons send axons to the prevertebral ganglia and are polysynaptically connected to the liver and pancreas. Microbiota depletion led to NLRP6- and caspase 11-dependent loss of CART+ neurons and impaired glucose regulation. Hence, iEAN subsets appear to be capable of regulating blood glucose levels independently from the central nervous system.
Collapse
Affiliation(s)
- Paul A Muller
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA.
| | - Fanny Matheis
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA
| | - Marc Schneeberger
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Zachary Kerner
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA
| | - Veronica Jové
- Laboratory of Neurogenetics and Behavior, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
19
|
Ueno M, Nakamura Y, Nakagawa H, Niehaus JK, Maezawa M, Gu Z, Kumanogoh A, Takebayashi H, Lu QR, Takada M, Yoshida Y. Olig2-Induced Semaphorin Expression Drives Corticospinal Axon Retraction After Spinal Cord Injury. Cereb Cortex 2020; 30:5702-5716. [PMID: 32564090 DOI: 10.1093/cercor/bhaa142] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/02/2020] [Accepted: 05/03/2020] [Indexed: 12/24/2022] Open
Abstract
Axon regeneration is limited in the central nervous system, which hinders the reconstruction of functional circuits following spinal cord injury (SCI). Although various extrinsic molecules to repel axons following SCI have been identified, the role of semaphorins, a major class of axon guidance molecules, has not been thoroughly explored. Here we show that expression of semaphorins, including Sema5a and Sema6d, is elevated after SCI, and genetic deletion of either molecule or their receptors (neuropilin1 and plexinA1, respectively) suppresses axon retraction or dieback in injured corticospinal neurons. We further show that Olig2+ cells are essential for SCI-induced semaphorin expression, and that Olig2 binds to putative enhancer regions of the semaphorin genes. Finally, conditional deletion of Olig2 in the spinal cord reduces the expression of semaphorins, alleviating the axon retraction. These results demonstrate that semaphorins function as axon repellents following SCI, and reveal a novel transcriptional mechanism for controlling semaphorin levels around injured neurons to create zones hostile to axon regrowth.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata 951-8585, Japan.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan
| | - Yuka Nakamura
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata 951-8585, Japan.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Hiroshi Nakagawa
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama 484-8506, Japan.,Department of Molecular Neuroscience, WPI Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | - Jesse K Niehaus
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan
| | - Mari Maezawa
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Zirong Gu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
| | - Qing Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Masahiko Takada
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama 484-8506, Japan
| | - Yutaka Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Neural Connectivity Development in Physiology and Disease Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
20
|
Wang YB, de Lartigue G, Page AJ. Dissecting the Role of Subtypes of Gastrointestinal Vagal Afferents. Front Physiol 2020; 11:643. [PMID: 32595525 PMCID: PMC7300233 DOI: 10.3389/fphys.2020.00643] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/20/2020] [Indexed: 12/22/2022] Open
Abstract
Gastrointestinal (GI) vagal afferents convey sensory signals from the GI tract to the brain. Numerous subtypes of GI vagal afferent have been identified but their individual roles in gut function and feeding regulation are unclear. In the past decade, technical approaches to selectively target vagal afferent subtypes and to assess their function has significantly progressed. This review examines the classification of GI vagal afferent subtypes and discusses the current available techniques to study vagal afferents. Investigating the distribution of GI vagal afferent subtypes and understanding how to access and modulate individual populations are essential to dissect their fundamental roles in the gut-brain axis.
Collapse
Affiliation(s)
- Yoko B Wang
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Guillaume de Lartigue
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States.,Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, United States
| | - Amanda J Page
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
21
|
Wang F, Shen J, Jiang S, Qiu Y, Ye X, Wang C, Liang C, Xu W. The Recognition of the Distribution Features of Corticospinal Neurons by a Retrograde Trans-synaptic Tracing to Elucidate the Clinical Application of Contralateral Middle Trunk Transfer. Neuroscience 2019; 424:86-101. [PMID: 31678345 DOI: 10.1016/j.neuroscience.2019.09.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 11/29/2022]
Abstract
Corticospinal neurons (CSNs) undertake direct cortical outputs to the spinal cord and innervate the upper limb through the brachial plexus. Our previous study has shown that the contralateral middle trunk transfer to the paralyzed upper extremity due to cerebral injury can reconstruct the functional cerebral cortex and improve the function of the paralyzed upper extremity. To interpret the cortical reconstruction and the motor improvement after the middle trunk transfer, we explored the distribution of CSNs connecting to the middle, upper, and lower trunk of the brachial plexus by retrograde trans-neuronal tracing using pseudorabies virus (PRV-EGFP or PRV-mRFP). We show that, rather than an individual specific area, these CSNs labelled by each trunk of the brachial plexus were widespread and mainly assembled within the primary motor cortex (M1), secondary motor cortex (M2), primary somatosensory cortex (S1), and slightly within the secondary somatosensory cortex (S2). The three trunk-labelled CSNs were intermingled in these cortices, and mostly connected to more than two trunks, especially the middle trunk-labelled CSNs with higher proportion of co-labelled neurons. Our findings revealed the distribution features of CSNs connecting to the adjacent spinal nerves that innervate the upper limb, which can improve our understanding of the corticospinal circuits associated with motor improvement and the functional cortical reconstruction after the middle trunk transfer.
Collapse
Affiliation(s)
- Fei Wang
- Department of Hand and Upper Extremity Surgery, Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 200040, China
| | - Jun Shen
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Su Jiang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yanqun Qiu
- Department of Hand and Upper Extremity Surgery, Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 200040, China
| | - Xuan Ye
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chengpan Wang
- Department of Anatomy and Histology & Embryology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chunmin Liang
- Department of Hand and Upper Extremity Surgery, Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 200040, China; Department of Anatomy and Histology & Embryology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wendong Xu
- Department of Hand and Upper Extremity Surgery, Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 200040, China; Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China; Institutes of Brain Science, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center of Brain Science, Fudan University, Shanghai 200032, China; Priority Among Priorities of Shanghai Municipal Clinical Medicine Center, Shanghai 200040, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
22
|
Li J, Liu T, Dong Y, Kondoh K, Lu Z. Trans-synaptic Neural Circuit-Tracing with Neurotropic Viruses. Neurosci Bull 2019; 35:909-920. [PMID: 31004271 PMCID: PMC6754522 DOI: 10.1007/s12264-019-00374-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/15/2018] [Indexed: 12/19/2022] Open
Abstract
A central objective in deciphering the nervous system in health and disease is to define the connections of neurons. The propensity of neurotropic viruses to spread among synaptically-linked neurons makes them ideal for mapping neural circuits. So far, several classes of viral neuronal tracers have become available and provide a powerful toolbox for delineating neural networks. In this paper, we review the recent developments of neurotropic viral tracers and highlight their unique properties in revealing patterns of neuronal connections.
Collapse
Affiliation(s)
- Jiamin Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Taian Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yun Dong
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Kunio Kondoh
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institute of Natural Sciences, Myodaiji, Okazaki, Aichi, 444-8585, Japan.
- Japan Science and Technology Agency, PRESTO, Myodaiji, Okazaki, Aichi, 444-8585, Japan.
| | - Zhonghua Lu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
23
|
Saleeba C, Dempsey B, Le S, Goodchild A, McMullan S. A Student's Guide to Neural Circuit Tracing. Front Neurosci 2019; 13:897. [PMID: 31507369 PMCID: PMC6718611 DOI: 10.3389/fnins.2019.00897] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/12/2019] [Indexed: 12/17/2022] Open
Abstract
The mammalian nervous system is comprised of a seemingly infinitely complex network of specialized synaptic connections that coordinate the flow of information through it. The field of connectomics seeks to map the structure that underlies brain function at resolutions that range from the ultrastructural, which examines the organization of individual synapses that impinge upon a neuron, to the macroscopic, which examines gross connectivity between large brain regions. At the mesoscopic level, distant and local connections between neuronal populations are identified, providing insights into circuit-level architecture. Although neural tract tracing techniques have been available to experimental neuroscientists for many decades, considerable methodological advances have been made in the last 20 years due to synergies between the fields of molecular biology, virology, microscopy, computer science and genetics. As a consequence, investigators now enjoy an unprecedented toolbox of reagents that can be directed against selected subpopulations of neurons to identify their efferent and afferent connectomes. Unfortunately, the intersectional nature of this progress presents newcomers to the field with a daunting array of technologies that have emerged from disciplines they may not be familiar with. This review outlines the current state of mesoscale connectomic approaches, from data collection to analysis, written for the novice to this field. A brief history of neuroanatomy is followed by an assessment of the techniques used by contemporary neuroscientists to resolve mesoscale organization, such as conventional and viral tracers, and methods of selecting for sub-populations of neurons. We consider some weaknesses and bottlenecks of the most widely used approaches for the analysis and dissemination of tracing data and explore the trajectories that rapidly developing neuroanatomy technologies are likely to take.
Collapse
Affiliation(s)
- Christine Saleeba
- Neurobiology of Vital Systems Node, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
- The School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Bowen Dempsey
- CNRS, Hindbrain Integrative Neurobiology Laboratory, Neuroscience Paris-Saclay Institute (Neuro-PSI), Université Paris-Saclay, Gif-sur-Yvette, France
| | - Sheng Le
- Neurobiology of Vital Systems Node, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Ann Goodchild
- Neurobiology of Vital Systems Node, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Simon McMullan
- Neurobiology of Vital Systems Node, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
24
|
Zheng T, Feng Z, Wang X, Jiang T, Jin R, Zhao P, Luo T, Gong H, Luo Q, Yuan J. Review of micro-optical sectioning tomography (MOST): technology and applications for whole-brain optical imaging [Invited]. BIOMEDICAL OPTICS EXPRESS 2019; 10:4075-4096. [PMID: 31452996 PMCID: PMC6701528 DOI: 10.1364/boe.10.004075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/20/2019] [Accepted: 06/25/2019] [Indexed: 05/14/2023]
Abstract
Elucidating connectivity and functionality at the whole-brain level is one of the most challenging research goals in neuroscience. Various whole-brain optical imaging technologies with submicron lateral resolution have been developed to reveal the fine structures of brain-wide neural and vascular networks at the mesoscopic level. Among them, micro-optical sectioning tomography (MOST) is attracting increasing attention, as a variety of technological variations and solutions tailored toward different biological applications have been optimized. Here, we summarize the recent development of MOST technology in whole-brain imaging and anticipate future improvements.
Collapse
Affiliation(s)
- Ting Zheng
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Britton Chance Center and MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Equal contribution
| | - Zhao Feng
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Britton Chance Center and MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Equal contribution
| | - Xiaojun Wang
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Britton Chance Center and MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Tao Jiang
- HUST–Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, Jiangsu 215000, China
| | - Rui Jin
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Britton Chance Center and MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Peilin Zhao
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Britton Chance Center and MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Ting Luo
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Britton Chance Center and MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Hui Gong
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Britton Chance Center and MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- HUST–Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, Jiangsu 215000, China
| | - Qingming Luo
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Britton Chance Center and MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- HUST–Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, Jiangsu 215000, China
| | - Jing Yuan
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Britton Chance Center and MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- HUST–Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, Jiangsu 215000, China
| |
Collapse
|
25
|
Jia F, Lv P, Miao H, Shi X, Mei H, Li L, Xu X, Tao S, Xu F. Optimization of the Fluorescent Protein Expression Level Based on Pseudorabies Virus Bartha Strain for Neural Circuit Tracing. Front Neuroanat 2019; 13:63. [PMID: 31281245 PMCID: PMC6597954 DOI: 10.3389/fnana.2019.00063] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/04/2019] [Indexed: 11/22/2022] Open
Abstract
Mapping the neural circuits facilitates understanding the brain’s working mechanism. Pseudorabies virus (PRV; Bartha stain) as a tracer can infect neurons and retrogradely transport in neural circuits. To illuminate the network, tracers expressing reporter genes at a high level are needed. In this study, we optimized the expression level of reporter genes and constructed two new retrograde trans-multisynaptic tracers PRV531 and PRV724, which separately express more robust green and red fluorescent proteins than the existing retrograde tracers PRV152 and PRV614. PRV531 and PRV724 can be used for mapping the neural circuit of the central nervous system (CNS) and the peripheral nervous system (PNS). Overall, our work adds two valuable tracers to the toolbox for mapping neural circuits.
Collapse
Affiliation(s)
- Fan Jia
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China.,Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Pei Lv
- Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Huan Miao
- Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Xiangwei Shi
- University of the Chinese Academy of Sciences, Beijing, China
| | - Hongjun Mei
- Department of Orthopaedics, The Fifth Hospital of Wuhan, Wuhan, China
| | - Li Li
- Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Xiaoqin Xu
- Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Sijue Tao
- Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Fuqiang Xu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China.,Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China.,University of the Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
26
|
Rim A, Nacira L, Jihene N, Said S, Khaled M, Ahmed R, Abdeljelil G. Viral interference between H9N2-low pathogenic avian influenza virus and avian infectious bronchitis virus vaccine strain H120 in vivo. Comp Immunol Microbiol Infect Dis 2019; 65:219-225. [PMID: 31300117 PMCID: PMC7112602 DOI: 10.1016/j.cimid.2019.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/10/2019] [Accepted: 06/10/2019] [Indexed: 12/20/2022]
Abstract
AIV and IBV co-infection led to decreased growth of both viruses. During super-infection, the second virus decreased the growth of the first virus. ELISA antibody titers, depending on the experimental conditions. Histopathological findings showed important lesions.
The interaction between a low pathogenic avian influenza virus (A/CK/TUN/145/2012), a H9N2 Tunisian isolate, and a vaccine strain (H120) of avian infectious bronchitis, administered simultaneously or sequentially three days apart to chicks during 20 days, was evaluated using ELISA antibody levels, quantitative reverse transcription–polymerase chain reaction (qRT-PCR) analyses and histopathology examination. First, the in vivo replication interference of avian influenza virus (AIV) and infectious bronchitis virus (IBV) was evaluated using qRT-PCR to detect accurately either AIV or IBV genomes or viral copy numbers during dual infections. Second, we have determined the amount of specific antibodies in sera of chick’s infected with AIV alone, IBV alone, mixed AIV + IBV, IBV then AIV or AIV IBV 3 days later using an ELISA test. Finally, histopathological analyses of internal organs from inoculated chicks were realized. Quantitative results of AIV and IBV co-infection showed that interferences between the two viruses yielded decreased viral growth. However, in the case of super-infection, the second virus, either AIV or IBV, induced a decrease in the growth of the first inoculated virus. According to our results, vaccine application was safe and do not interfere with AIV H9N2 infection, and does not enhance such infection. In conclusion, co-infection of chicks with AIV and IBV, simultaneously or sequentially, affected the clinical signs, the virus replication dynamics as well as the internal organ integrity. The results proposed that infection with heterologous virus may result in temporary competition for cell receptors or competent cells for replication, most likely interferon-mediated.
Collapse
Affiliation(s)
- Aouini Rim
- University Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Epidemiology and Veterinary Microbiology,13 Place Pasteur, 1002 Tunis-Belvedere, Tunisia; University of Carthage, Faculty of Sciences of Bizerte, 7021 Zarzouna-Bizerte, Tunisia.
| | - Laamiri Nacira
- University Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Epidemiology and Veterinary Microbiology,13 Place Pasteur, 1002 Tunis-Belvedere, Tunisia; University of Carthage, Faculty of Sciences of Bizerte, 7021 Zarzouna-Bizerte, Tunisia.
| | - Nsiri Jihene
- University Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Epidemiology and Veterinary Microbiology,13 Place Pasteur, 1002 Tunis-Belvedere, Tunisia.
| | - Salhi Said
- University Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Epidemiology and Veterinary Microbiology,13 Place Pasteur, 1002 Tunis-Belvedere, Tunisia; University of Carthage, Faculty of Sciences of Bizerte, 7021 Zarzouna-Bizerte, Tunisia.
| | - Miled Khaled
- University Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Epidemiology and Veterinary Microbiology,13 Place Pasteur, 1002 Tunis-Belvedere, Tunisia.
| | - Rejab Ahmed
- National School of Veterinary Medicine of Sidi-Thabet, 2020 Sidi-Thabet, Tunisia.
| | - Ghram Abdeljelil
- University Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Epidemiology and Veterinary Microbiology,13 Place Pasteur, 1002 Tunis-Belvedere, Tunisia.
| |
Collapse
|
27
|
Doslikova B, Tchir D, McKinty A, Zhu X, Marks DL, Baracos VE, Colmers WF. Convergent neuronal projections from paraventricular nucleus, parabrachial nucleus, and brainstem onto gastrocnemius muscle, white and brown adipose tissue in male rats. J Comp Neurol 2019; 527:2826-2842. [PMID: 31045239 DOI: 10.1002/cne.24710] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 01/06/2023]
Abstract
When energy balance is altered by aerobic exercise, starvation, and cold exposure, for example, there appears to be coordination of the responses of skeletal muscle, white adipose (WAT), and brown adipose (BAT) tissues. We hypothesized that WAT, BAT, and skeletal muscle may share an integrated regulation by the central nervous system (CNS); specifically, that neurons in brain regions associated with energy balance would possess neuroanatomical connections to permit coordination of multiple, complementary responses in these downstream tissues. To study this, we used trans-neuronal viral retrograde tract tracing, using isogenic strains of pseudorabies virus (PRV) with distinct fluorescent reporters (either eGFP or mRFP), injected pairwise into male rat gastrocnemius, subcutaneous WAT and interscapular BAT, coupled with neurochemical characterization of specific cell populations for cocaine- and amphetamine-related transcript (CART), oxytocin (OX), corticotrophin releasing hormone (CRH) and calcitonin gene-related peptide (CGRP). Cells in the paraventricular (PVN) and parabrachial (PBN) nuclei and brainstem showed dual projections to muscle + WAT, muscle + BAT, and WAT + BAT. Dual PRV-labeled cells were found in parvocellular, magnocellular and descending/pre-autonomic regions of the PVN, and multiple structural divisions of the PBN and brainstem. In most PBN subdivisions, more than 50% of CGRP cells dually projected to muscle + WAT and muscle + BAT. Similarly, 31-68% of CGRP cells projected both to WAT + BAT. However, dual PRV-labeled cells in PVN only occasionally expressed OX or CRH but not CART. These studies reveal for the first time both separate and shared outflow circuitries among skeletal muscle and subcutaneous WAT and BAT.
Collapse
Affiliation(s)
- Barbora Doslikova
- Department of Pharmacology, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Devan Tchir
- Department of Pharmacology, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Amanda McKinty
- Department of Pharmacology, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Xinxia Zhu
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon
| | - Vickie E Baracos
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - William F Colmers
- Department of Pharmacology, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
28
|
Wee NKY, Lorenz MR, Bekirov Y, Jacquin MF, Scheller EL. Shared Autonomic Pathways Connect Bone Marrow and Peripheral Adipose Tissues Across the Central Neuraxis. Front Endocrinol (Lausanne) 2019; 10:668. [PMID: 31611846 PMCID: PMC6776593 DOI: 10.3389/fendo.2019.00668] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/16/2019] [Indexed: 12/31/2022] Open
Abstract
Bone marrow adipose tissue (BMAT) is increased in both obesity and anorexia. This is unique relative to white adipose tissue (WAT), which is generally more attuned to metabolic demand. It suggests that there may be regulatory pathways that are common to both BMAT and WAT and also those that are specific to BMAT alone. The central nervous system (CNS) is a key mediator of adipose tissue function through sympathetic adrenergic neurons. Thus, we hypothesized that central autonomic pathways may be involved in BMAT regulation. To test this, we first quantified the innervation of BMAT by tyrosine hydroxylase (TH) positive nerves within the metaphysis and diaphysis of the tibia of B6 and C3H mice. We found that many of the TH+ axons were concentrated around central blood vessels in the bone marrow. However, there were also areas of free nerve endings which terminated in regions of BMAT adipocytes. Overall, the proportion of nerve-associated BMAT adipocytes increased from proximal to distal along the length of the tibia (from ~3-5 to ~14-24%), regardless of mouse strain. To identify the central pathways involved in BMAT innervation and compare to peripheral WAT, we then performed retrograde viral tract tracing with an attenuated pseudorabies virus (PRV) to infect efferent nerves from the tibial metaphysis (inclusive of BMAT) and inguinal WAT (iWAT) of C3H mice. PRV positive neurons were identified consistently from both injection sites in the intermediolateral horn of the spinal cord, reticular formation, rostroventral medulla, solitary tract, periaqueductal gray, locus coeruleus, subcoeruleus, Barrington's nucleus, and hypothalamus. We also observed dual-PRV infected neurons within the majority of these regions. Similar tracings were observed in pons, midbrain, and hypothalamic regions from B6 femur and tibia, demonstrating that these results persist across mouse strains and between skeletal sites. Altogether, this is the first quantitative report of BMAT autonomic innervation and reveals common central neuroanatomic pathways, including putative "command" neurons, involved in coordinating multiple aspects of sympathetic output and facilitation of parallel processing between bone marrow/BMAT and peripheral adipose tissue.
Collapse
Affiliation(s)
- Natalie K. Y. Wee
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Department of Reconstructive Sciences, UConn Health, Farmington, CT, United States
| | - Madelyn R. Lorenz
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Yusuf Bekirov
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Mark F. Jacquin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
- *Correspondence: Erica L. Scheller
| |
Collapse
|
29
|
Han W, Tellez LA, Perkins MH, Perez IO, Qu T, Ferreira J, Ferreira TL, Quinn D, Liu ZW, Gao XB, Kaelberer MM, Bohórquez DV, Shammah-Lagnado SJ, de Lartigue G, de Araujo IE. A Neural Circuit for Gut-Induced Reward. Cell 2018; 175:665-678.e23. [PMID: 30245012 PMCID: PMC6195474 DOI: 10.1016/j.cell.2018.08.049] [Citation(s) in RCA: 404] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 07/08/2018] [Accepted: 08/16/2018] [Indexed: 12/31/2022]
Abstract
The gut is now recognized as a major regulator of motivational and emotional states. However, the relevant gut-brain neuronal circuitry remains unknown. We show that optical activation of gut-innervating vagal sensory neurons recapitulates the hallmark effects of stimulating brain reward neurons. Specifically, right, but not left, vagal sensory ganglion activation sustained self-stimulation behavior, conditioned both flavor and place preferences, and induced dopamine release from Substantia nigra. Cell-specific transneuronal tracing revealed asymmetric ascending pathways of vagal origin throughout the CNS. In particular, transneuronal labeling identified the glutamatergic neurons of the dorsolateral parabrachial region as the obligatory relay linking the right vagal sensory ganglion to dopamine cells in Substantia nigra. Consistently, optical activation of parabrachio-nigral projections replicated the rewarding effects of right vagus excitation. Our findings establish the vagal gut-to-brain axis as an integral component of the neuronal reward pathway. They also suggest novel vagal stimulation approaches to affective disorders.
Collapse
Affiliation(s)
- Wenfei Han
- The John B. Pierce Laboratory, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luis A Tellez
- The John B. Pierce Laboratory, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Matthew H Perkins
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isaac O Perez
- The John B. Pierce Laboratory, New Haven, CT, USA; Section of Neurobiology of Oral Sensations, FES-Iztacala, National Autonomous University of Mexico, Mexico City, Mexico
| | - Taoran Qu
- The John B. Pierce Laboratory, New Haven, CT, USA
| | - Jozelia Ferreira
- The John B. Pierce Laboratory, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Department of Anatomy, Biomedical Sciences Institute, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Tatiana L Ferreira
- The John B. Pierce Laboratory, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Mathematics, Computing and Cognition Center, Federal University of ABC, São Bernardo do Campo, São Paulo, Brazil
| | | | - Zhong-Wu Liu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Xiao-Bing Gao
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Diego V Bohórquez
- Department of Medicine, Duke University, Durham, NC, USA; Department of Neurobiology, Duke University, Durham, NC, USA
| | - Sara J Shammah-Lagnado
- Department of Physiology and Biophysics, Biomedical Sciences Institute, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Guillaume de Lartigue
- The John B. Pierce Laboratory, New Haven, CT, USA; Department of Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ivan E de Araujo
- The John B. Pierce Laboratory, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Physiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
30
|
Hogue IB, Card JP, Rinaman L, Staniszewska Goraczniak H, Enquist LW. Characterization of the neuroinvasive profile of a pseudorabies virus recombinant expressing the mTurquoise2 reporter in single and multiple injection experiments. J Neurosci Methods 2018; 308:228-239. [PMID: 30098326 PMCID: PMC6294127 DOI: 10.1016/j.jneumeth.2018.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/29/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Viral transneuronal tracing has become a well established technology used to define the synaptic architecture of polysynaptic neural networks. NEW METHOD In this report we define the neuroinvasive profile and reporter expression of a new recombinant of the Bartha strain of pseudorabies virus (PRV). The new recombinant, PRV-290, expresses the mTurquoise2 fluorophor and is designed to complement other isogenic recombinants of Bartha that express different reporters of infection. Results & Comparison with Existing Methods: PRV-290 was injected either alone or in combination with isogenic recombinants of PRV that express enhanced green fluorescent protein (EGFP; PRV-152) or monomeric red fluorescent protein (mRFP; PRV-614). Circuits previously defined using PRV-152 and PRV-614 were used for the analysis. The data demonstrate that PRV-290 is a retrograde transneuronal tracer with temporal kinetics similar to those of its isogenic recombinants. Stable expression of the diffusible mTurquoise2 reporter filled infected neurons, with the extent and intensity of labeling increasing with advancing post inoculation survival. In multiple injection experiments, PRV-290 established productive infections in neurons also replicating PRV-152 and/or PRV-614. This novel demonstration of three recombinants infecting individual neurons represents an important advance in the technology. CONCLUSION Collectively, these data demonstrate that PRV-290 is a valuable addition to the viral tracer toolbox for transneuronal tracing of neural circuitry.
Collapse
Affiliation(s)
- Ian B Hogue
- Department of Molecular Biology, Neuroscience Institute, Princeton University, Princeton, NJ, 08544, United States.
| | - J Patrick Card
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, United States; Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306, United States.
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306, United States
| | | | - Lynn W Enquist
- Department of Molecular Biology, Neuroscience Institute, Princeton University, Princeton, NJ, 08544, United States
| |
Collapse
|
31
|
Aouini R, Laamiri N, Ghram A. Viral interference between low pathogenic avian influenza H9N2 and avian infectious bronchitis viruses in vitro and in ovo. J Virol Methods 2018; 259:92-99. [PMID: 29940196 PMCID: PMC7119724 DOI: 10.1016/j.jviromet.2018.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Low pathogenic avian influenza (LPAI) H9N2 and infectious bronchitis virus (IBV) are important pathogens of poultry, causing important economic losses for the sector. Replication interference between these two viruses was described using cell cultures (CC) and embryonated chicken eggs (ECE). Chicken embryo lung (CEL) and ECE were simultaneously or sequentially infected with IBV vaccine strain (H120) and LPAIV-H9N2 (A/Ck/TUN/145/2012) to evaluate viral interactionsin vitro and in ovo, respectively. Real-time RT-PCR was developed to specifically quantify both AIV and IBV genomes as well as viral gene copy numbers during mixed infections. The amount of IL-1 beta, in supernatants of co-infected cell cultures, was determined using an ELISA assay. RESULTS Quantitative results of AIV and IBV co-infection showed that interferences between the two viruses yielded decreased viral growth. However, in the case of super-infection, the second virus, either AIV or IBV, induced a decrease in the growth of the first inoculated virus. CONCLUSION It appears that either AIV or IBV has a negative impact on the other virus growth when they are inoculated simultaneously or sequentially. The ELISA results showed that higher level of secreted IL-1beta varies, depending on the viral interference conditions between both viruses, during mixed infections.
Collapse
Affiliation(s)
- Rim Aouini
- University Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Epidemiology and Veterinary Microbiology, 13 Place Pasteur, Tunis, Belvedere, 1002, Tunisia; University of Carthage, Faculty of Sciences of Bizerte, 7021, Zarzouna, Bizerte, Tunisia.
| | - Nacira Laamiri
- University Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Epidemiology and Veterinary Microbiology, 13 Place Pasteur, Tunis, Belvedere, 1002, Tunisia; University of Carthage, Faculty of Sciences of Bizerte, 7021, Zarzouna, Bizerte, Tunisia.
| | - Abdeljelil Ghram
- University Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Epidemiology and Veterinary Microbiology, 13 Place Pasteur, Tunis, Belvedere, 1002, Tunisia.
| |
Collapse
|
32
|
Changes in membrane properties of rat deep cerebellar nuclear projection neurons during acquisition of eyeblink conditioning. Proc Natl Acad Sci U S A 2018; 115:E9419-E9428. [PMID: 30154170 DOI: 10.1073/pnas.1808539115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Previous studies have shown changes in membrane properties of neurons in rat deep cerebellar nuclei (DCN) as a function of development, but due to technical difficulties in obtaining viable DCN slices from adult animals, it remains unclear whether there are learning-related alterations in the membrane properties of DCN neurons in adult rats. This study was designed to record from identified DCN cells in cerebellar slices from postnatal day 25-26 (P25-26) rats that had a relatively mature sensory nervous system and were able to acquire learning as a result of tone-shock eyeblink conditioning (EBC) and to document resulting changes in electrophysiological properties. After electromyographic electrode implantation at P21 and inoculation with a fluorescent pseudorabies virus (PRV-152) at P22-23, rats received either four sessions of paired delay EBC or unpaired stimulus presentations with a tone conditioned stimulus and a shock unconditioned stimulus or sat in the training chamber without stimulus presentations. Compared with rats given unpaired stimuli or no stimulus presentations, rats given paired EBC showed an increase in conditioned responses across sessions. Whole-cell recordings of both fluorescent and nonfluorescent DCN projection neurons showed that delay EBC induced significant changes in membrane properties of evoked DCN action potentials including a reduced after-hyperpolarization amplitude and shortened latency. Similar findings were obtained in hyperpolarization-induced rebound spikes of DCN neurons. In sum, delay EBC produced significant changes in the membrane properties of juvenile rat DCN projection neurons. These learning-specific changes in DCN excitability have not previously been reported in any species or task.
Collapse
|
33
|
Lyu C, Wang S, Sun M, Tang Y, Peng J, Tian Z, Cai X. Deletion of pseudorabies virus US2 gene enhances viral titers in a porcine cerebral cortex primary culture system. Virus Genes 2018. [PMID: 29541931 DOI: 10.1007/s11262-018-1552-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pseudorabies virus (PRV) is a neurotropic virus with the ability to infect peripheral sensory ganglia. The transport of PRV from the peripheral to the central nervous system can cause lethal encephalitis in young piglets. However, the pathogenicity of PRV in the cerebral cortex remains poorly understood. In the present study, we developed a porcine cerebral cortex primary culture system (PCCS) using cerebral cortex tissue dissected from a 3-day-old piglet to investigate the pathogenicity of wild-type (WT) and US2 deleted (ΔUS2) PRV in the CNS in vitro. Immunofluorescence assays revealed cell bodies and neurites as the cellular locations infected by PRV. Growth kinetic analysis showed a persistent increase in WT and ΔUS2 viral titers in PCCS from 4 to 24 h post infection (hpi), thus indicating that US2 deletion did not disrupt viral growth. However, the mean plaque size was significantly higher in ΔUS2 PRV than in WT PRV in infected Vero cells. The viral titers and DNA levels of ΔUS2 PRV were significantly higher at 8 hpi than at 4 hpi, whereas those of WT showed no significant difference between the two time points in PCCS. Morphological investigation revealed induction of massive amounts of bouton-like swellings (varicosities) along the axon shaft in both WT and ΔUS2 PRV-infected neurons in the PCCS. Our data suggest that PRV US2 gene deletion enhances viral titers in PCCS but does not affect the varicosities induced by the viral infection.
Collapse
Affiliation(s)
- Chuang Lyu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Haping Road No. 678, Xiang Fang District, Harbin, 150069, Heilongjiang, People's Republic of China
| | - Shuwen Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Haping Road No. 678, Xiang Fang District, Harbin, 150069, Heilongjiang, People's Republic of China
| | - Mingxia Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Haping Road No. 678, Xiang Fang District, Harbin, 150069, Heilongjiang, People's Republic of China
| | - Yandong Tang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Haping Road No. 678, Xiang Fang District, Harbin, 150069, Heilongjiang, People's Republic of China
| | - Jinmei Peng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Haping Road No. 678, Xiang Fang District, Harbin, 150069, Heilongjiang, People's Republic of China
| | - Zhijun Tian
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Haping Road No. 678, Xiang Fang District, Harbin, 150069, Heilongjiang, People's Republic of China
| | - Xuehui Cai
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Haping Road No. 678, Xiang Fang District, Harbin, 150069, Heilongjiang, People's Republic of China.
| |
Collapse
|
34
|
Chen N, Chopp M, Xiong Y, Qian JY, Lu M, Zhou D, He L, Liu Z. Subacute intranasal administration of tissue plasminogen activator improves stroke recovery by inducing axonal remodeling in mice. Exp Neurol 2018. [PMID: 29518364 DOI: 10.1016/j.expneurol.2018.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In addition to thrombolysis, tissue plasminogen activator (tPA) can evoke neurorestorative processes. We therefore investigated the therapeutic effect of subacute intranasal administration of tPA post stroke on neurological recovery and on corticospinal innervation in mice. A transgenic mouse line, in which the pyramidal neurons and corticospinal tract (CST) axons are specifically labeled by yellow fluorescent protein (YFP) was employed. Adult CST-YFP mice were subjected to right unilateral middle cerebral artery occlusion (MCAo), and were randomly divided into groups treated with saline or tPA intranasally in the subacute phase. Pseudorabies virus (PRV)-614-monomeric red fluorescent protein (RFP) was injected into the left forelimb. The cervical spinal cord and brain were processed for fluorescent microscopy to detect YFP and RFP labeling. Primary embryonic neurons were cultured with tPA at different concentrations. Neurite length and branch numbers were then measured. In vivo, subacute tPA treatment significantly enhanced functional recovery (p < 0.05), and increased CST density in the denervated gray matter, and in the numbers of PRV-labeled neurons in bilateral cortices. The behavioral performance was significantly correlated with axonal density in the denervated spinal cord. In vitro, both neurite length and branch numbers significantly increased with concentration of tPA (p < 0.05). Our results demonstrate that tPA dose-dependently increases neurite outgrowth and branching of cultured cortical neurons. Subacute intranasal administration of tPA may provide enhance neurological recovery after stroke by promoting CST axonal remodeling.
Collapse
Affiliation(s)
- Ning Chen
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China; Department of Neurology, Henry Ford Hospital, Detrot, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detrot, MI, United States; Department of Physics, Oakland University, Rochester, MI, United States
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, United States
| | - Jian-Yong Qian
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China
| | - Mei Lu
- Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, MI, United States
| | - Dong Zhou
- Department of Neurology, Henry Ford Hospital, Detrot, MI, United States
| | - Li He
- Department of Neurology, Henry Ford Hospital, Detrot, MI, United States.
| | - Zhongwu Liu
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
35
|
Skilled Movements Require Non-apoptotic Bax/Bak Pathway-Mediated Corticospinal Circuit Reorganization. Neuron 2017; 94:626-641.e4. [PMID: 28472660 DOI: 10.1016/j.neuron.2017.04.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/04/2017] [Accepted: 04/13/2017] [Indexed: 12/15/2022]
Abstract
Early postnatal mammals, including human babies, can perform only basic motor tasks. The acquisition of skilled behaviors occurs later, requiring anatomical changes in neural circuitry to support the development of coordinated activation or suppression of functionally related muscle groups. How this circuit reorganization occurs during postnatal development remains poorly understood. Here we explore the connectivity between corticospinal (CS) neurons in the motor cortex and muscles in mice. Using trans-synaptic viral and electrophysiological assays, we identify the early postnatal reorganization of CS circuitry for antagonistic muscle pairs. We further show that this synaptic rearrangement requires the activity-dependent, non-apoptotic Bax/Bak-caspase signaling cascade. Adult Bax/Bak mutant mice exhibit aberrant co-activation of antagonistic muscle pairs and skilled grasping deficits but normal reaching and retrieval behaviors. Our findings reveal key cellular and molecular mechanisms driving postnatal motor circuit reorganization and the resulting impacts on muscle activation patterns and the execution of skilled movements.
Collapse
|
36
|
Herr AE, Hain KS, Taylor MP. Limitations on the Multiplicity of Cellular Infection During Human Alphaherpesvirus Disease. CURRENT CLINICAL MICROBIOLOGY REPORTS 2017. [DOI: 10.1007/s40588-017-0071-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
37
|
Gene Expression Profiling with Cre-Conditional Pseudorabies Virus Reveals a Subset of Midbrain Neurons That Participate in Reward Circuitry. J Neurosci 2017; 37:4128-4144. [PMID: 28283558 DOI: 10.1523/jneurosci.3193-16.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/06/2017] [Accepted: 02/27/2017] [Indexed: 01/29/2023] Open
Abstract
The mesolimbic dopamine pathway receives inputs from numerous regions of the brain as part of a neural system that detects rewarding stimuli and coordinates a behavioral response. The capacity to simultaneously map and molecularly define the components of this complex multisynaptic circuit would thus advance our understanding of the determinants of motivated behavior. To accomplish this, we have constructed pseudorabies virus (PRV) strains in which viral propagation and fluorophore expression are activated only after exposure to Cre recombinase. Once activated in Cre-expressing neurons, the virus serially labels chains of presynaptic neurons. Dual injection of GFP and mCherry tracing viruses simultaneously illuminates nigrostriatal and mesolimbic circuitry and shows no overlap, demonstrating that PRV transmission is confined to synaptically connected neurons. To molecularly profile mesolimbic dopamine neurons and their presynaptic inputs, we injected Cre-conditional GFP virus into the NAc of (anti-GFP) nanobody-L10 transgenic mice and immunoprecipitated translating ribosomes from neurons infected after retrograde tracing. Analysis of purified RNA revealed an enrichment of transcripts expressed in neurons of the dorsal raphe nuclei and lateral hypothalamus that project to the mesolimbic dopamine circuit. These studies identify important inputs to the mesolimbic dopamine pathway and further show that PRV circuit-directed translating ribosome affinity purification can be broadly applied to identify molecularly defined neurons comprising complex, multisynaptic circuits.SIGNIFICANCE STATEMENT The mesolimbic dopamine circuit integrates signals from key brain regions to detect and respond to rewarding stimuli. To further define this complex multisynaptic circuit, we constructed a panel of Cre recombinase-activated pseudorabies viruses (PRVs) that enabled retrograde tracing of neural inputs that terminate on Cre-expressing neurons. Using these viruses and Retro-TRAP (translating ribosome affinity purification), a previously reported molecular profiling method, we developed a novel technique that provides anatomic as well as molecular information about the neural components of polysynaptic circuits. We refer to this new method as PRV-Circuit-TRAP (PRV circuit-directed TRAP). Using it, we have identified major projections to the mesolimbic dopamine circuit from the lateral hypothalamus and dorsal raphe nucleus and defined a discrete subset of transcripts expressed in these projecting neurons, which will allow further characterization of this important pathway. Moreover, the method we report is general and can be applied to the study of other neural circuits.
Collapse
|
38
|
Tombácz D, Balázs Z, Csabai Z, Moldován N, Szűcs A, Sharon D, Snyder M, Boldogkői Z. Characterization of the Dynamic Transcriptome of a Herpesvirus with Long-read Single Molecule Real-Time Sequencing. Sci Rep 2017; 7:43751. [PMID: 28256586 PMCID: PMC5335617 DOI: 10.1038/srep43751] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/26/2017] [Indexed: 11/09/2022] Open
Abstract
Herpesvirus gene expression is co-ordinately regulated and sequentially ordered during productive infection. The viral genes can be classified into three distinct kinetic groups: immediate-early, early, and late classes. In this study, a massively parallel sequencing technique that is based on PacBio Single Molecule Real-time sequencing platform, was used for quantifying the poly(A) fraction of the lytic transcriptome of pseudorabies virus (PRV) throughout a 12-hour interval of productive infection on PK-15 cells. Other approaches, including microarray, real-time RT-PCR and Illumina sequencing are capable of detecting only the aggregate transcriptional activity of particular genomic regions, but not individual herpesvirus transcripts. However, SMRT sequencing allows for a distinction between transcript isoforms, including length- and splice variants, as well as between overlapping polycistronic RNA molecules. The non-amplified Isoform Sequencing (Iso-Seq) method was used to analyse the kinetic properties of the lytic PRV transcripts and to then classify them accordingly. Additionally, the present study demonstrates the general utility of long-read sequencing for the time-course analysis of global gene expression in practically any organism.
Collapse
Affiliation(s)
- Dóra Tombácz
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. u. 4., Szeged, H-6720, Hungary
| | - Zsolt Balázs
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. u. 4., Szeged, H-6720, Hungary
| | - Zsolt Csabai
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. u. 4., Szeged, H-6720, Hungary
| | - Norbert Moldován
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. u. 4., Szeged, H-6720, Hungary
| | - Attila Szűcs
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. u. 4., Szeged, H-6720, Hungary
| | - Donald Sharon
- Department of Genetics, School of Medicine, Stanford University, 300 Pasteur Dr., Stanford, CA 94305-5120, USA
| | - Michael Snyder
- Department of Genetics, School of Medicine, Stanford University, 300 Pasteur Dr., Stanford, CA 94305-5120, USA
| | - Zsolt Boldogkői
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Somogyi B. u. 4., Szeged, H-6720, Hungary
| |
Collapse
|
39
|
Bruce KD, Zsombok A, Eckel RH. Lipid Processing in the Brain: A Key Regulator of Systemic Metabolism. Front Endocrinol (Lausanne) 2017; 8:60. [PMID: 28421037 PMCID: PMC5378716 DOI: 10.3389/fendo.2017.00060] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/17/2017] [Indexed: 12/25/2022] Open
Abstract
Metabolic disorders, particularly aberrations in lipid homeostasis, such as obesity, type 2 diabetes mellitus, and hypertriglyceridemia often manifest together as the metabolic syndrome (MetS). Despite major advances in our understanding of the pathogenesis of these disorders, the prevalence of the MetS continues to rise. It is becoming increasingly apparent that intermediary metabolism within the central nervous system is a major contributor to the regulation of systemic metabolism. In particular, lipid metabolism within the brain is tightly regulated to maintain neuronal structure and function and may signal nutrient status to modulate metabolism in key peripheral tissues such as the liver. There is now a growing body of evidence to suggest that fatty acid (FA) sensing in hypothalamic neurons via accumulation of FAs or FA metabolites may signal nutritional sufficiency and may decrease hepatic glucose production, lipogenesis, and VLDL-TG secretion. In addition, recent studies have highlighted the existence of liver-related neurons that have the potential to direct such signals through parasympathetic and sympathetic nervous system activity. However, to date whether these liver-related neurons are FA sensitive remain to be determined. The findings discussed in this review underscore the importance of the autonomic nervous system in the regulation of systemic metabolism and highlight the need for further research to determine the key features of FA neurons, which may serve as novel therapeutic targets for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Kimberley D. Bruce
- University of Colorado School of Medicine, Division of Endocrinology, Metabolism and Diabetes, Aurora, CO, USA
- *Correspondence: Kimberley D. Bruce,
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Robert H. Eckel
- University of Colorado School of Medicine, Division of Endocrinology, Metabolism and Diabetes, Aurora, CO, USA
| |
Collapse
|
40
|
Silencing spinal interneurons inhibits immune suppressive autonomic reflexes caused by spinal cord injury. Nat Neurosci 2016; 19:784-7. [PMID: 27089020 PMCID: PMC4882232 DOI: 10.1038/nn.4289] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/21/2016] [Indexed: 12/19/2022]
Abstract
Spinal cord injury (SCI) at high spinal levels (e.g., above thoracic level 5) causes systemic immune suppression; however, the underlying mechanisms are unknown. Here, we show that profound plasticity develops within spinal autonomic circuitry below the injury, creating a sympathetic anti-inflammatory reflex, and that chemogenetic silencing of this reflex circuitry blocks post-SCI immune suppression. These data provide new insights and potential therapeutic options for limiting the devastating consequences of post-traumatic autonomic hyperreflexia and post-injury immune suppression.
Collapse
|
41
|
Criddle A, Thornburg T, Kochetkova I, DePartee M, Taylor MP. gD-Independent Superinfection Exclusion of Alphaherpesviruses. J Virol 2016; 90:4049-58. [PMID: 26842480 PMCID: PMC4810564 DOI: 10.1128/jvi.00089-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 01/29/2016] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Many viruses have the capacity to prevent a cell from being infected by a second virus, often termed superinfection exclusion. Alphaherpesviruses, including the human pathogen herpes simplex virus 1 (HSV-1) and the animal herpesvirus pseudorabies virus (PRV), encode a membrane-bound glycoprotein, gD, that can interfere with subsequent virion entry. We sought to characterize the timing and mechanism of superinfection exclusion during HSV-1 and PRV infection. To this end, we utilized recombinant viruses expressing fluorescent protein (FP) markers of infection that allowed the visualization of viral infections by microscopy and flow cytometry as well as the differentiation of viral progeny. Our results demonstrated the majority of HSV-1- and PRV-infected cells establish superinfection exclusion by 2 h postinfection. The modification of viral infections by virion inactivation and phosphonoacetic acid, cycloheximide, and actinomycin D treatments indicated new protein synthesis is needed to establish superinfection exclusion. Primary infection with gene deletion PRV recombinants identified that new gD expression is not required to establish superinfection exclusion of a secondary viral inoculum. We also identified the timing of coinfection events during axon-to-cell spread, with most occurring within a 2-h window, suggesting a role for cellular superinfection exclusion during neuroinvasive spread of infection. In summary, we have characterized a gD-independent mechanism of superinfection exclusion established by two members of the alphaherpesvirus family and identified a potential role of exclusion during the pathogenic spread of infection. IMPORTANCE Superinfection exclusion is a widely observed phenomenon initiated by a primary viral infection to prevent further viruses from infecting the same cell. The capacity for alphaherpesviruses to infect the same cell impacts rates of interviral recombination and disease. Interviral recombination allows genome diversification, facilitating the development of resistance to antiviral therapeutics and evasion of vaccine-mediated immune responses. Our results demonstrate superinfection exclusion occurs early, through a gD-independent process, and is important in the directed spread of infection. Identifying when and where in an infected host viral genomes are more likely to coinfect the same cell and generate viral recombinants will enhance the development of effective antiviral therapies and interventions.
Collapse
Affiliation(s)
- A Criddle
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, USAUniversity of California, Irvine
| | - T Thornburg
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, USAUniversity of California, Irvine
| | - I Kochetkova
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, USAUniversity of California, Irvine
| | - M DePartee
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, USAUniversity of California, Irvine
| | - M P Taylor
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, USAUniversity of California, Irvine
| |
Collapse
|
42
|
Niu H, He X, Zhou T, Shi X, Zhang Q, Zhang Z, Qiao Y, Xu F, Hu M. Neural circuits containing olfactory neurons are involved in the prepulse inhibition of the startle reflex in rats. Front Behav Neurosci 2015; 9:74. [PMID: 25859195 PMCID: PMC4373374 DOI: 10.3389/fnbeh.2015.00074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 03/08/2015] [Indexed: 11/24/2022] Open
Abstract
Many neuropsychiatric disorders, such as schizophrenia, have been associated with olfactory dysfunction and abnormalities in the prepulse inhibition (PPI) response to a startle reflex. However, whether these two abnormalities could be related is unclear. The present investigations were designed to determine whether theblockage of olfactory sensory input by zinc sulfate infusion in the olfactory naris (0.5 ml, 0.17 M, ZnE) can disturb the PPI response. Furthermore, a bilateral microinjection of lidocaine/MK801 in the olfactory bulb (OB) was administered to examine whether the blockage of olfactory sensory input could impair the PPI response. To identify the neural projection between olfaction and PPI-related areas, trans-synaptic retrograde tracing with the recombinant pseudorabies virus (PRV) was used. Our results demonstrated that blockage of olfactory sensory input could disturb olfactory behavior. In the function studies, we demonstrated that blockage of olfactory sensory input could impair the pre-pulse inhibition of the startle response following decreased c-Fos expression in relevant brain regions during the PPI responses. Furthermore, similar and more robust findings indicated that blockage of olfactory sensory input by microinjection of lidocaine/MK801 in the OB could impair the PPI response. In the circuit-level studies, we demonstrated that trans-synaptic retrograde tracing with PRV exhibited a large portion of labeled neurons in several regions of the olfactory cortices from the pedunculopontine tegmental nucleus (PPTg). Thus, these data suggest that the olfactory system participates in the PPI regulating fields and plays a role in the pre-pulse inhibition of the startle response in rats.
Collapse
Affiliation(s)
- Haichen Niu
- Department of Genetics, Xuzhou Medical College Xuzhou, China ; The Institute of Audiology and Speech Science, Xuzhou Medical Collage Xuzhou, China
| | - Xiaobin He
- Key Laboratory of Magnetic Resonance in Biological Systems and State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China ; Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology Wuhan, China
| | - Ting Zhou
- Department of Genetics, Xuzhou Medical College Xuzhou, China
| | - Xi Shi
- The Institute of Audiology and Speech Science, Xuzhou Medical Collage Xuzhou, China
| | - Qiang Zhang
- Department of Genetics, Xuzhou Medical College Xuzhou, China
| | - Zhijian Zhang
- Key Laboratory of Magnetic Resonance in Biological Systems and State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China
| | - Yuehua Qiao
- The Institute of Audiology and Speech Science, Xuzhou Medical Collage Xuzhou, China
| | - Fuqiang Xu
- Key Laboratory of Magnetic Resonance in Biological Systems and State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China ; Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology Wuhan, China
| | - Min Hu
- Department of Ophthalmology, The Second People's Hospital of Yunnan Province Kunming, China
| |
Collapse
|
43
|
Xu H, Boychuk JA, Boychuk CR, Uteshev VV, Smith BN. Nicotine enhances inhibition of mouse vagal motor neurons by modulating excitability of premotor GABAergic neurons in the nucleus tractus solitarii. J Neurophysiol 2014; 113:1165-74. [PMID: 25429117 DOI: 10.1152/jn.00614.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The caudal nucleus of the solitary tract (NTS) serves as the site of the first synapse for visceral sensory inputs to the central nervous system. The NTS sends functional projections to multiple brain nuclei, with gastric-related projections primarily targeting the dorsal motor nucleus of the vagus (DMV). Previous studies have demonstrated that the majority of caudal NTS neurons that project to the DMV respond robustly to nicotine and express nicotinic acetylcholine receptors (nAChRs). However, the cytochemical identity and relationship with specific viscera of DMV-projecting, nicotine-responsive caudal NTS neurons have not been determined. The present study used transgenic mice that express enhanced green fluorescent protein (EGFP) under a GAD67 promoter in a subset of GABAergic neurons, in vivo retrograde pseudorabies viral labeling to identify gastric-related vagal complex neurons, and patch-clamp electrophysiology in acute brain stem slices to test the hypothesis that gastric-related and GABAergic inhibitory synaptic input to the DMV from the caudal NTS is under a robust modulatory control by nAChRs. Our results suggest that activation of nAChRs in the caudal NTS, but not DMV, potentiates GABAergic, but not glutamatergic, input to the DMV. Gastric-related caudal NTS and DMV neurons are directly involved in this nicotine-sensitive circuitry. Understanding the central patterns of nicotinic modulation of visceral sensory-motor circuitry may help develop therapeutic interventions to restore autonomic homeostasis in patients with autonomic impairments.
Collapse
Affiliation(s)
- Hong Xu
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| | - Jeffery A Boychuk
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| | - Carie R Boychuk
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| | - Victor V Uteshev
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas
| | - Bret N Smith
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| |
Collapse
|
44
|
Characterization of a replication-incompetent pseudorabies virus mutant lacking the sole immediate early gene IE180. mBio 2014; 5:e01850. [PMID: 25389174 PMCID: PMC4235210 DOI: 10.1128/mbio.01850-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The alphaherpesvirus pseudorabies virus (PRV) encodes a single immediate early gene called IE180. The IE180 protein is a potent transcriptional activator of viral genes involved in DNA replication and RNA transcription. A PRV mutant with both copies of IE180 deleted was constructed 20 years ago (S. Yamada and M. Shimizu, Virology 199:366–375, 1994, doi:10.1006/viro.1994.1134), but propagation of the mutant depended on complementing cell lines that expressed the toxic IE180 protein constitutively. Recently, Oyibo et al. constructed a novel set of PRV IE180 mutants and a stable cell line with inducible IE180 expression (H. Oyibo, P. Znamenskiy, H. V. Oviedo, L. W. Enquist, A. Zador, Front. Neuroanat. 8:86, 2014, doi:10.3389/fnana.2014.00086), which we characterized further here. These mutants failed to replicate new viral genomes, synthesize immediate early, early, or late viral proteins, and assemble infectious virions. The PRV IE180-null mutant did not form plaques in epithelial cell monolayers and could not spread from primary infected neurons to second-order neurons in culture. PRV IE180-null mutants lacked the property of superinfection exclusion. When PRV IE180-null mutants infected cells first, subsequent superinfecting viruses were not blocked in cell entry and formed replication compartments in epithelial cells, fibroblasts, and neurons. Cells infected with PRV IE180-null mutants survived as long as uninfected cells in culture while expressing a fluorescent reporter gene. Transcomplementation with IE180 in epithelial cells restored all mutant phenotypes to wild type. The conditional expression of PRV IE180 protein enables the propagation of replication-incompetent PRV IE180-null mutants and will facilitate construction of long-term single-cell-infecting PRV mutants for precise neural circuit tracing and high-capacity gene delivery vectors. Pseudorabies virus (PRV) is widely used for neural tracing in animal models. The virus replicates and spreads between synaptically connected neurons. Current tracing strains of PRV are cytotoxic and kill infected cells. Infected cells exclude superinfection with a second virus, limiting multiple virus infections in circuit tracing. By removing the only immediate early gene of PRV (called IE180), the mutant virus will not replicate or spread in epithelial cells, fibroblasts, or neurons. The wild-type phenotype can be restored by transcomplementation of infected cells with IE180. The PRV IE180-null mutant can express fluorescent reporters for weeks in cells with no toxicity; infected cells survive as long as uninfected cells. Infection with the mutant virus allows superinfection of the same cell with a second virus that can enter and replicate. The PRV IE180-null mutant will permit conditional long-term tracing in animals and is a high-capacity vector for gene delivery.
Collapse
|
45
|
Bartness TJ, Liu Y, Shrestha YB, Ryu V. Neural innervation of white adipose tissue and the control of lipolysis. Front Neuroendocrinol 2014; 35:473-93. [PMID: 24736043 PMCID: PMC4175185 DOI: 10.1016/j.yfrne.2014.04.001] [Citation(s) in RCA: 242] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/10/2014] [Accepted: 04/04/2014] [Indexed: 01/22/2023]
Abstract
White adipose tissue (WAT) is innervated by the sympathetic nervous system (SNS) and its activation is necessary for lipolysis. WAT parasympathetic innervation is not supported. Fully-executed SNS-norepinephrine (NE)-mediated WAT lipolysis is dependent on β-adrenoceptor stimulation ultimately hinging on hormone sensitive lipase and perilipin A phosphorylation. WAT sympathetic drive is appropriately measured electrophysiologically and neurochemically (NE turnover) in non-human animals and this drive is fat pad-specific preventing generalizations among WAT depots and non-WAT organs. Leptin-triggered SNS-mediated lipolysis is weakly supported, whereas insulin or adenosine inhibition of SNS/NE-mediated lipolysis is strongly supported. In addition to lipolysis control, increases or decreases in WAT SNS drive/NE inhibit and stimulate white adipocyte proliferation, respectively. WAT sensory nerves are of spinal-origin and sensitive to local leptin and increases in sympathetic drive, the latter implicating lipolysis. Transsynaptic viral tract tracers revealed WAT central sympathetic and sensory circuits including SNS-sensory feedback loops that may control lipolysis.
Collapse
Affiliation(s)
- Timothy J Bartness
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302-4010, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA.
| | - Yang Liu
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302-4010, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA; Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yogendra B Shrestha
- Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vitaly Ryu
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302-4010, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA; Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
46
|
Evidence for multiple sensory circuits in the brain arising from the respiratory system: an anterograde viral tract tracing study in rodents. Brain Struct Funct 2014; 220:3683-99. [PMID: 25158901 DOI: 10.1007/s00429-014-0883-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 08/20/2014] [Indexed: 01/07/2023]
Abstract
Complex sensations accompany the activation of sensory neurons within the respiratory system, yet little is known about the organization of sensory pathways in the brain that mediate these sensations. In the present study, we employ anterograde viral neuroanatomical tract tracing with isogenic self-reporting recombinants of HSV-1 strain H129 to map the higher brain regions in receipt of vagal sensory neurons arising from the trachea versus the lungs, and single-cell PCR to characterize the phenotype of sensory neurons arising from these two divisions of the respiratory tree. The results suggest that the upper and lower airways are predominantly innervated by sensory neurons derived from the somatic jugular and visceral nodose cranial ganglia, respectively. This coincides with central circuitry that is predominately somatic-like, arising from the trachea, and visceral-like, arising from the lungs. Although some convergence of sensory pathways was noted in preautonomic cell groups, this was notably absent in thalamic and cortical regions. These data support the notion that distinct afferent subtypes, via distinct central circuits, subserve sensations arising from the upper versus lower airways. The findings may explain why sensations arising from different levels of the respiratory tree are qualitatively and quantitatively unique.
Collapse
|
47
|
Abstract
Our ability to understand the function of the nervous system is dependent upon defining the connections of its constituent neurons. Development of methods to define connections within neural networks has always been a growth industry in the neurosciences. Transneuronal spread of neurotropic viruses currently represents the best means of defining synaptic connections within neural networks. The method exploits the ability of viruses to invade neurons, replicate, and spread through the intimate synaptic connections that enable communication among neurons. Since the method was first introduced in the 1970s, it has benefited from an increased understanding of the virus life cycle, the function of viral genome, and the ability to manipulate the viral genome in support of directional spread of virus and the expression of transgenes. In this unit, we review these advances in viral tracing technology and the way in which they may be applied for functional dissection of neural networks.
Collapse
Affiliation(s)
- J Patrick Card
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
48
|
Xiang HB, Liu C, Liu TT, Xiong J. Central circuits regulating the sympathetic outflow to lumbar muscles in spinally transected mice by retrograde transsynaptic transport. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:2987-2997. [PMID: 25031717 PMCID: PMC4097212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 04/23/2014] [Indexed: 06/03/2023]
Abstract
Despite considerable interest in the mechanisms that control the hyperalgesia associated with muscle inflammation, the CNS descending pathways that coordinate autonomic circuits regulating lumbar muscles are not adequately understood. Here we used both pseudorabies virus (PRV)-614 retrograde transsynaptic tracing and spinally transected method in 33 C57BL/6J mice to map the polysynaptic pathways between lumbar muscle and CNS. Tissues were processed for dual-label immunocytochemical detection between PRV-614 and tryptophan hydroxylase (TPH) or tyrosine hydroxylase (TH)-expressing neurons in CNS. In intact mice, PRV-614 was transported to the intermediolateral column (IML) and ventral horn (VH) of spinal cord, with subsequent transport to many brain regions, including the medullary raphe nuclei, rostral ventrolateral medulla (RVLM), A5 cell group regions (A5), locus coeruleus (LC), the medullary and pontine reticular formation nucleus (MRN and PRN), paraventricular nucleus of the hypothalamus (PVN), and other central sites. However, PRV-614 in spinally transected mice produced retrograde infection of IML, with subsequent transport to main brain regions that have been shown to contribute to regulating sympathetic circuits, including RVLM, Lateral paragigantocellular reticular nucleus (LPGi), A5, LC, and PVN, whereas PRV-614 labeling in VH and MRN was eliminated in almost every case. In above five brain regions, dual-labeling immunocytochemistry showed coexpression of PRV-614/TPH and PRV-614/TH immunoreactive (IR) neurons involved in these regulatory circuits. Our results reveal a hierarchical organization of central autonomic circuits controlling the lumbar muscles, thus providing neuroanatomical substrates for the central catecholaminergic and serotonergic system to regulate the lumbar muscles.
Collapse
Affiliation(s)
- Hong-Bing Xiang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, PR China
| | - Cheng Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, PR China
| | - Tao-Tao Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, PR China
| | - Jun Xiong
- Hepatobiliary Surgery Center, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, PR China
| |
Collapse
|
49
|
Identification of CNS neurons with polysynaptic connections to both the sympathetic and parasympathetic innervation of the submandibular gland. Brain Struct Funct 2014; 220:2103-20. [DOI: 10.1007/s00429-014-0781-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/14/2014] [Indexed: 10/25/2022]
|
50
|
The neuroinvasive profiles of H129 (herpes simplex virus type 1) recombinants with putative anterograde-only transneuronal spread properties. Brain Struct Funct 2014; 220:1395-420. [PMID: 24585022 DOI: 10.1007/s00429-014-0733-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 02/11/2014] [Indexed: 10/25/2022]
Abstract
The use of viruses as transneuronal tracers has become an increasingly powerful technique for defining the synaptic organization of neural networks. Although a number of recombinant alpha herpesviruses are known to spread selectively in the retrograde direction through neural circuits only one strain, the H129 strain of herpes simplex virus type 1, is reported to selectively spread in the anterograde direction. However, it is unclear from the literature whether there is an absolute block or an attenuation of retrograde spread of H129. Here, we demonstrate efficient anterograde spread, and temporally delayed retrograde spread, of H129 and three novel recombinants. In vitro studies revealed no differences in anterograde and retrograde spread of parental H129 and its recombinants through superior cervical ganglion neurons. In vivo injections of rat striatum revealed a clear bias of anterograde spread, although evidence of deficient retrograde transport was also present. Evidence of temporally delayed retrograde transneuronal spread of H129 in the retina was observed following injection of the lateral geniculate nucleus. The data also demonstrated that three novel recombinants efficiently express unique fluorescent reporters and have the capacity to infect the same neurons in dual infection paradigms. From these experiments we conclude that H129 and its recombinants not only efficiently infect neurons through anterograde transneuronal passage, but also are capable of temporally delayed retrograde transneuronal spread. In addition, the capacity to produce dual infection of projection targets following anterograde transneuronal passage provides an important addition to viral transneuronal tracing technology.
Collapse
|