1
|
Jaeger ECB, Vijatovic D, Deryckere A, Zorin N, Nguyen AL, Ivanian G, Woych J, Arnold RC, Gurrola AO, Shvartsman A, Barbieri F, Toma FA, Cline HT, Shay TF, Kelley DB, Yamaguchi A, Shein-Idelson M, Tosches MA, Sweeney LB. Adeno-associated viral tools to trace neural development and connectivity across amphibians. Dev Cell 2025; 60:794-812.e6. [PMID: 39603234 DOI: 10.1016/j.devcel.2024.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 09/19/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Amphibians, by virtue of their phylogenetic position, provide invaluable insights on nervous system evolution, development, and remodeling. The genetic toolkit for amphibians, however, remains limited. Recombinant adeno-associated viral vectors (AAVs) are a powerful alternative to transgenesis for labeling and manipulating neurons. Although successful in mammals, AAVs have never been shown to transduce amphibian cells efficiently. We screened AAVs in three amphibian species-the frogs Xenopus laevis and Pelophylax bedriagae and the salamander Pleurodeles waltl-and identified at least two AAV serotypes per species that transduce neurons. In developing amphibians, AAVs labeled groups of neurons generated at the same time during development. In the mature brain, AAVrg retrogradely traced long-range projections. Our study introduces AAVs as a tool for amphibian research, establishes a generalizable workflow for AAV screening in new species, and expands opportunities for cross-species comparisons of nervous system development, function, and evolution.
Collapse
Affiliation(s)
- Eliza C B Jaeger
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - David Vijatovic
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Astrid Deryckere
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Nikol Zorin
- Department of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Akemi L Nguyen
- Department of Biology, University of Utah, Salt Lake City, UT, USA
| | - Georgiy Ivanian
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Jamie Woych
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Rebecca C Arnold
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Arik Shvartsman
- Department of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | | | - Florina A Toma
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Hollis T Cline
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Timothy F Shay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Darcy B Kelley
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Ayako Yamaguchi
- Department of Biology, University of Utah, Salt Lake City, UT, USA
| | - Mark Shein-Idelson
- Department of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | - Lora B Sweeney
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| |
Collapse
|
2
|
Desmoulins LD, Molinas AJR, Dugas CM, Williams GL, Kamenetsky S, Davis RK, Derbenev AV, Zsombok A. A subset of neurons in the paraventricular nucleus of the hypothalamus directly project to liver-related premotor neurons in the ventrolateral medulla. Auton Neurosci 2025; 257:103222. [PMID: 39647176 DOI: 10.1016/j.autneu.2024.103222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/03/2024] [Accepted: 11/21/2024] [Indexed: 12/10/2024]
Abstract
Sympathetic circuits including pre-sympathetic neurons in the ventrolateral medulla (VLM) and in the paraventricular nucleus (PVN) of the hypothalamus play an important role in the regulation of hepatic glucose metabolism. Despite the importance of central regulatory pathways, specific information regarding the circuits of liver-related neurons is limited. Here, we tested the hypothesis that PVN neurons are directly connected to spinally-projecting liver-related neurons in the VLM of mice. Pseudorabies virus (PRV) was used to identify liver-related neurons and time-dependent analyses revealed the location and distribution of neurons in the PVN and ventral brainstem. Four days following PRV injection, most liver-related neurons were found in the VLM and consist of both catecholaminergic (CA) and non-CA neurons. Furthermore, in addition to PRV inoculation, a monosynaptic viral tracer was used to identify VLM-projecting PVN neurons to specifically dissect PVN-VLM connections within the liver pathway. Five days following PRV inoculation, our anatomical findings revealed that a small population of liver-related PVN neurons projected to the VLM. In addition, photo-stimulation of axonal projections from SIM1-expressing PVN neurons resulted in evoked excitatory postsynaptic currents in a subset of spinally projecting liver-related neurons in the VLM. In summary, our data demonstrate the existence of monosynaptic, glutamatergic connections between PVN neurons and pre-sympathetic liver-related neurons in the VLM. These new findings regarding the central circuits involved in the sympathetic regulation of the liver provide further information necessary for developing new strategies to improve glucose homeostasis via modulation of the autonomic nerves.
Collapse
Affiliation(s)
- Lucie D Desmoulins
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Adrien J R Molinas
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Courtney M Dugas
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Gabrielle L Williams
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Sophie Kamenetsky
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Roslyn K Davis
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Andrei V Derbenev
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA; Tulane Brain Institute, Tulane University, New Orleans, Louisiana, USA
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA; Tulane Brain Institute, Tulane University, New Orleans, Louisiana, USA.
| |
Collapse
|
3
|
Timalsina B, Lee S, Kaang BK. Advances in the labelling and selective manipulation of synapses. Nat Rev Neurosci 2024; 25:668-687. [PMID: 39174832 DOI: 10.1038/s41583-024-00851-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/24/2024]
Abstract
Synapses are highly specialized neuronal structures that are essential for neurotransmission, and they are dynamically regulated throughout the lifetime. Although accumulating evidence indicates that these structures are crucial for information processing and storage in the brain, their precise roles beyond neurotransmission are yet to be fully appreciated. Genetically encoded fluorescent tools have deepened our understanding of synaptic structure and function, but developing an ideal methodology to selectively visualize, label and manipulate synapses remains challenging. Here, we provide an overview of currently available synapse labelling techniques and describe their extension to enable synapse manipulation. We categorize these approaches on the basis of their conceptual bases and target molecules, compare their advantages and limitations and propose potential modifications to improve their effectiveness. These methods have broad utility, particularly for investigating mechanisms of synaptic function and synaptopathy.
Collapse
Affiliation(s)
- Binod Timalsina
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Sangkyu Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Bong-Kiun Kaang
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, South Korea.
| |
Collapse
|
4
|
Bai L, Xu J, Zeng L, Zhang L, Zhou F. A review of HSV pathogenesis, vaccine development, and advanced applications. MOLECULAR BIOMEDICINE 2024; 5:35. [PMID: 39207577 PMCID: PMC11362470 DOI: 10.1186/s43556-024-00199-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Herpes simplex virus (HSV), an epidemic human pathogen threatening global public health, gains notoriety for its complex pathogenesis that encompasses lytic infection of mucosal cells, latent infection within neurons, and periodic reactivation. This intricate interplay, coupled with HSV's sophisticated immune evasion strategies, gives rise to various diseases, including genital lesions, neonatal encephalitis, and cancer. Despite more than 70 years of relentless research, an effective preventive or therapeutic vaccine against HSV has yet to emerge, primarily due to the limited understanding of virus-host interactions, which in turn impedes the identification of effective vaccine targets. However, HSV's unique pathological features, including its substantial genetic load capacity, high replicability, transmissibility, and neurotropism, render it a promising candidate for various applications, spanning oncolytic virotherapy, gene and immune therapies, and even as an imaging tracer in neuroscience. In this review, we comprehensively update recent breakthroughs in HSV pathogenesis and immune evasion, critically summarize the progress made in vaccine candidate development, and discuss the multifaceted applications of HSV as a biological tool. Importantly, we highlight both success and challenges, emphasizing the critical need for intensified research into HSV, with the aim of providing deeper insights that can not only advance HSV treatment strategies but also broaden its application horizons.
Collapse
Affiliation(s)
- Lan Bai
- International Biomed-X Research Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jiuzhi Xu
- Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Linghui Zeng
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, China.
| | - Long Zhang
- International Biomed-X Research Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| | - Fangfang Zhou
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, China.
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
5
|
Krause TB, Cepko CL. Abortive and productive infection of CNS cell types following in vivo delivery of VSV. Proc Natl Acad Sci U S A 2024; 121:e2406421121. [PMID: 39159381 PMCID: PMC11363278 DOI: 10.1073/pnas.2406421121] [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: 03/29/2024] [Accepted: 07/11/2024] [Indexed: 08/21/2024] Open
Abstract
Viral infection is frequently assayed by ongoing expression of viral genes. These assays fail to identify cells that have been exposed to the virus but limit or inhibit viral replication. To address this limitation, we used a dual-labeling vesicular stomatitis virus (DL-VSV), which has a deletion of the viral glycoprotein gene, to allow evaluation of primary infection outcomes. This virus encodes Cre, which can stably mark any cell with even a minimal level of viral gene expression. Additionally, the virus encodes GFP, which distinguishes cells with higher levels of viral gene expression, typically due to genome replication. Stereotactic injections of DL-VSV into the murine brain showed that different cell types had very different responses to the virus. Almost all neurons hosted high levels of viral gene expression, while glial cells varied in their responses. Astrocytes (Sox9+) were predominantly productively infected, while oligodendrocytes (Sox10+) were largely abortively infected. Microglial cells (Iba1+) were primarily uninfected. Furthermore, we monitored the early innate immune response to viral infection and identified unique patterns of interferon (IFN) induction. Shortly after infection, microglia were the main producers of IFNb, whereas later, oligodendrocytes were the main producers. IFNb+ cells were primarily abortively infected regardless of cell type. Last, we investigated whether IFN signaling had any impact on the outcome of primary infection and did not observe significant changes, suggesting that intrinsic factors are likely responsible for determining the outcome of primary infection.
Collapse
Affiliation(s)
- Tyler B Krause
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Constance L. Cepko
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
- Howard Hughes Medical Institute, Chevy Chase, MD20815
| |
Collapse
|
6
|
Xu P, Peng J, Yuan T, Chen Z, He H, Wu Z, Li T, Li X, Wang L, Gao L, Yan J, Wei W, Li CT, Luo ZG, Chen Y. High-throughput mapping of single-neuron projection and molecular features by retrograde barcoded labeling. eLife 2024; 13:e85419. [PMID: 38390967 PMCID: PMC10914349 DOI: 10.7554/elife.85419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/22/2024] [Indexed: 02/24/2024] Open
Abstract
Deciphering patterns of connectivity between neurons in the brain is a critical step toward understanding brain function. Imaging-based neuroanatomical tracing identifies area-to-area or sparse neuron-to-neuron connectivity patterns, but with limited throughput. Barcode-based connectomics maps large numbers of single-neuron projections, but remains a challenge for jointly analyzing single-cell transcriptomics. Here, we established a rAAV2-retro barcode-based multiplexed tracing method that simultaneously characterizes the projectome and transcriptome at the single neuron level. We uncovered dedicated and collateral projection patterns of ventromedial prefrontal cortex (vmPFC) neurons to five downstream targets and found that projection-defined vmPFC neurons are molecularly heterogeneous. We identified transcriptional signatures of projection-specific vmPFC neurons, and verified Pou3f1 as a marker gene enriched in neurons projecting to the lateral hypothalamus, denoting a distinct subset with collateral projections to both dorsomedial striatum and lateral hypothalamus. In summary, we have developed a new multiplexed technique whose paired connectome and gene expression data can help reveal organizational principles that form neural circuits and process information.
Collapse
Affiliation(s)
- Peibo Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired TechnologyShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jian Peng
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech UniversityShanghaiChina
| | - Tingli Yuan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired TechnologyShanghaiChina
| | - Zhaoqin Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired TechnologyShanghaiChina
| | - Hui He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired TechnologyShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ziyan Wu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired TechnologyShanghaiChina
| | - Ting Li
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech UniversityShanghaiChina
| | - Xiaodong Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired TechnologyShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Luyue Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of ScienceShanghaiChina
| | - Le Gao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired TechnologyShanghaiChina
| | - Jun Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired TechnologyShanghaiChina
- Shanghai Center for Brain Science and Brain-Inspired Intelligence TechnologyShanghaiChina
- School of Future Technology, University of Chinese Academy of SciencesBeijingChina
| | - Wu Wei
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of ScienceShanghaiChina
- Lingang LaboratoryShanghaiChina
| | - Chengyu T Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired TechnologyShanghaiChina
- Shanghai Center for Brain Science and Brain-Inspired Intelligence TechnologyShanghaiChina
- School of Future Technology, University of Chinese Academy of SciencesBeijingChina
- Lingang LaboratoryShanghaiChina
| | - Zhen-Ge Luo
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech UniversityShanghaiChina
| | - Yuejun Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired TechnologyShanghaiChina
- Shanghai Center for Brain Science and Brain-Inspired Intelligence TechnologyShanghaiChina
| |
Collapse
|
7
|
Xu F, Liu Q. Virus-Based Neural Circuit Tracing. ADVANCES IN NEUROBIOLOGY 2024; 41:113-131. [PMID: 39589712 DOI: 10.1007/978-3-031-69188-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Neural circuits provide an anatomical basis for functional networks. Therefore, dissecting the structure of neural circuits is an indispensable prerequisite to understanding how the brain functions. Knowing how the neural circuits organize and function under physiological conditions and their progressive alterations under pathophysiological conditions are key to understanding the underlying circuit mechanism of diseases, thus finding cures for the diseases. Recombinant neurotropic viruses are important tools for neural circuit tracing with many advantages over non-viral tracers: they allow for anterograde, retrograde, and trans-synaptic delivery of tracers in a cell-type-specific, circuit-selective manner. We herein summarize the recent developments in the viral tools for neural circuit tracing, discuss the key principles of using viral tools in neuroscience research, and highlight innovations for developing and optimizing viral tools for neural circuit tracing across diverse animal species, including nonhuman primates.
Collapse
Affiliation(s)
- Fuqiang Xu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, China.
| | - Qing Liu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, China
| |
Collapse
|
8
|
Mei H, Tan J, Hu Y, Shi X, Liu Y, Jia F, Xu F. Developing a trans-multisynaptic tracer to map the neural circuit of recovered sciatic nerve after treatment with nerve growth factor. IBRO Neurosci Rep 2023; 15:235-241. [PMID: 37841085 PMCID: PMC10570716 DOI: 10.1016/j.ibneur.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/19/2023] [Accepted: 09/30/2023] [Indexed: 10/17/2023] Open
Abstract
Nerve growth factor (NGF) has been shown to support the survival and differentiation of neurons. In this study, we first developed a retrograde trans-multisynaptic tracer PRV580 expressing the mCherry fluorescent protein based on pseudorabies virus Bartha strain to map the neural circuit of sciatic nerve. Secondly, the newly developed PRV580 was used to map the neural circuit of the recovering sciatic nerve upon treatment with NGF. Our results showed that red signals from PRV580 were observed in various brain regions. Among these regions, many areas of the pyramidal system and the extra-pyramidal system had been mapped, accounting for as much as 56.8 % of the total inputs. Furthermore, we found that NGF could significantly increase the ratio of total input (29.05 %) compared to PBS (3.65 %), indicating that NGF indeed can aid in the repair of injured sciatic nerve. These findings indicated that NGF has therapeutic ability for the treatment of peripheral nerve injuries and virus-based tracers can be used to monitor the recovery.
Collapse
Affiliation(s)
- Hongjun Mei
- Department of Orthopaedics, The Fifth hospital of Wuhan, Wuhan 430050, China
| | - Junfeng Tan
- Department of Orthopaedics, The Fifth hospital of Wuhan, Wuhan 430050, China
| | - You Hu
- Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xiangwei Shi
- Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, 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
| | - Yang Liu
- Department of Orthopaedics, The Fifth hospital of Wuhan, Wuhan 430050, China
| | - Fan Jia
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, 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
| | - Fuqiang Xu
- Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, 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
| |
Collapse
|
9
|
Xiao H, Hu H, Guo Y, Li J, Wen L, Zeng WB, Wang M, Luo MH, Hu Z. Construction and characterization of a synthesized herpes simplex virus H129-Syn-G2. Virol Sin 2023:S1995-820X(23)00026-3. [PMID: 36940800 DOI: 10.1016/j.virs.2023.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/15/2023] [Indexed: 03/23/2023] Open
Abstract
Herpes simplex virus type 1 (HSV-1) causes lifelong infections worldwide, and currently there is no efficient cure or vaccine. HSV-1-derived tools, such as neuronal circuit tracers and oncolytic viruses, have been used extensively; however, further genetic engineering of HSV-1 is hindered by its complex genome structure. In the present study, we designed and constructed a synthetic platform for HSV-1 based on H129-G4. The complete genome was constructed from 10 fragments through 3 rounds of synthesis using transformation-associated recombination (TAR) in yeast, and was named H129-Syn-G2. The H129-Syn-G2 genome contained two copies of the gfp gene and was transfected into cells to rescue the virus. According to growth curve assay and electron microscopy results, the synthetic viruses exhibited more optimized growth properties and similar morphogenesis compared to the parental virus. This synthetic platform will facilitate further manipulation of the HSV-1 genome for the development of neuronal circuit tracers, oncolytic viruses, and vaccines.
Collapse
Affiliation(s)
- Han Xiao
- State Key laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Hengrui Hu
- State Key laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, 430071, China
| | - Yijia Guo
- State Key laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiang Li
- State Key laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, 430071, China
| | - Le Wen
- State Key laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, 430071, China
| | - Wen-Bo Zeng
- State Key laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, 430071, China.
| | - Manli Wang
- State Key laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China.
| | - Min-Hua Luo
- State Key laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China.
| | - Zhihong Hu
- State Key laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, 430071, China.
| |
Collapse
|
10
|
Liu M, Xie F, Dai J, Zhang J, Yuan K, Wang N. Brain-wide inputs to the non-lemniscal inferior colliculus in mice. Neurosci Lett 2023; 793:136976. [PMID: 36427816 DOI: 10.1016/j.neulet.2022.136976] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/27/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022]
Abstract
The inferior colliculus (IC) is the hub along the auditory pathway. Although it is fundamentally an auditory structure, the neurons in the IC, especially its non-lemniscal part also respond to multimodal stimuli. However, the sources of these non-auditory inputs are unclear. In this study, we injected the rAAV2-retro virus, a virus with efficient retrograde function, into the non-lemniscal IC of the Ai14 reporter line. The majority of cortical and subcortical brain areas, including cognitive, motor, somatosensory, auditory, and visual-related regions were revealed. The quantified whole brain input data have showed that the non-lemniscal IC received a higher proportion of inputs from ipsilateral cortical brain regions. The non-lemniscal IC integrates different multimodal patterns, for the dorsal cortex (ICD) receives primarily auditory inputs, and the external cortex (ICE) receives primarily auditory and somatosensory inputs. These findings demonstrate that auditory integration is shaped by a network of multi-sensory connections in the non-lemniscal IC subregions.
Collapse
Affiliation(s)
- Mengting Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Fenghua Xie
- Department of Biomedical Engineering, School of Medicine, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Jinsheng Dai
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Juan Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Kexin Yuan
- Department of Biomedical Engineering, School of Medicine, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| | - Ningyu Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
11
|
Xiong F, Yang H, Song YG, Qin HB, Zhang QY, Huang X, Jing W, Deng M, Liu Y, Liu Z, Shen Y, Han Y, Lu Y, Xu X, Holmes TC, Luo M, Zhao F, Luo MH, Zeng WB. An HSV-1-H129 amplicon tracer system for rapid and efficient monosynaptic anterograde neural circuit tracing. Nat Commun 2022; 13:7645. [PMID: 36496505 PMCID: PMC9741617 DOI: 10.1038/s41467-022-35355-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Monosynaptic viral tracers are essential tools for dissecting neuronal connectomes and for targeted delivery of molecular sensors and effectors. Viral toxicity and complex multi-injection protocols are major limiting application barriers. To overcome these barriers, we developed an anterograde monosynaptic H129Amp tracer system based on HSV-1 strain H129. The H129Amp tracer system consists of two components: an H129-dTK-T2-pacFlox helper which assists H129Amp tracer's propagation and transneuronal monosynaptic transmission. The shared viral features of tracer/helper allow for simultaneous single-injection and subsequent high expression efficiency from multiple-copy of expression cassettes in H129Amp tracer. These improvements of H129Amp tracer system shorten experiment duration from 28-day to 5-day for fast-bright monosynaptic tracing. The lack of toxic viral genes in the H129Amp tracer minimizes toxicity in postsynaptic neurons, thus offering the potential for functional anterograde mapping and long-term tracer delivery of genetic payloads. The H129Amp tracer system is a powerful tracing tool for revealing neuronal connectomes.
Collapse
Affiliation(s)
- Feng Xiong
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China ,grid.9227.e0000000119573309Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Hong Yang
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Yi-Ge Song
- grid.33199.310000 0004 0368 7223Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hai-Bin Qin
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Qing-Yang Zhang
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Xian Huang
- grid.33199.310000 0004 0368 7223Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Jing
- grid.33199.310000 0004 0368 7223Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manfei Deng
- grid.33199.310000 0004 0368 7223Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Liu
- grid.410717.40000 0004 0644 5086National Institute of Biological Sciences, Beijing, China
| | - Zhixiang Liu
- grid.410717.40000 0004 0644 5086National Institute of Biological Sciences, Beijing, China
| | - Yin Shen
- grid.49470.3e0000 0001 2331 6153Eye Center, Renmin Hospital, Wuhan University, Wuhan, China
| | - Yunyun Han
- grid.49470.3e0000 0001 2331 6153Eye Center, Renmin Hospital, Wuhan University, Wuhan, China
| | - Youming Lu
- grid.33199.310000 0004 0368 7223Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangmin Xu
- grid.266093.80000 0001 0668 7243Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA USA ,grid.266093.80000 0001 0668 7243Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, CA USA
| | - Todd C. Holmes
- grid.266093.80000 0001 0668 7243Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, CA USA ,grid.266093.80000 0001 0668 7243Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA USA
| | - Minmin Luo
- grid.410717.40000 0004 0644 5086National Institute of Biological Sciences, Beijing, China ,grid.510934.a0000 0005 0398 4153Chinese Institute for Brain Research, Beijing, China
| | - Fei Zhao
- grid.510934.a0000 0005 0398 4153Chinese Institute for Brain Research, Beijing, China ,grid.24696.3f0000 0004 0369 153XSchool of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Min-Hua Luo
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China ,grid.9227.e0000000119573309Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China ,grid.266093.80000 0001 0668 7243Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, CA USA
| | - Wen-Bo Zeng
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
12
|
Liu Q, Wu Y, Wang H, Jia F, Xu F. Viral Tools for Neural Circuit Tracing. Neurosci Bull 2022; 38:1508-1518. [PMID: 36136267 PMCID: PMC9723069 DOI: 10.1007/s12264-022-00949-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/09/2022] [Indexed: 10/14/2022] Open
Abstract
Neural circuits provide an anatomical basis for functional networks. Therefore, dissecting the structure of neural circuits is essential to understanding how the brain works. Recombinant neurotropic viruses are important tools for neural circuit tracing with many advantages over non-viral tracers: they allow for anterograde, retrograde, and trans-synaptic delivery of tracers in a cell type-specific, circuit-selective manner. In this review, we summarize the recent developments in the viral tools for neural circuit tracing, discuss the key principles of using viral tools in neuroscience research, and highlight innovations for developing and optimizing viral tools for neural circuit tracing across diverse animal species, including nonhuman primates.
Collapse
Affiliation(s)
- Qing Liu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huadong Wang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fan Jia
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fuqiang Xu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
13
|
Arias A, Manubens-Gil L, Dierssen M. Fluorescent transgenic mouse models for whole-brain imaging in health and disease. Front Mol Neurosci 2022; 15:958222. [PMID: 36211979 PMCID: PMC9538927 DOI: 10.3389/fnmol.2022.958222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022] Open
Abstract
A paradigm shift is occurring in neuroscience and in general in life sciences converting biomedical research from a descriptive discipline into a quantitative, predictive, actionable science. Living systems are becoming amenable to quantitative description, with profound consequences for our ability to predict biological phenomena. New experimental tools such as tissue clearing, whole-brain imaging, and genetic engineering technologies have opened the opportunity to embrace this new paradigm, allowing to extract anatomical features such as cell number, their full morphology, and even their structural connectivity. These tools will also allow the exploration of new features such as their geometrical arrangement, within and across brain regions. This would be especially important to better characterize brain function and pathological alterations in neurological, neurodevelopmental, and neurodegenerative disorders. New animal models for mapping fluorescent protein-expressing neurons and axon pathways in adult mice are key to this aim. As a result of both developments, relevant cell populations with endogenous fluorescence signals can be comprehensively and quantitatively mapped to whole-brain images acquired at submicron resolution. However, they present intrinsic limitations: weak fluorescent signals, unequal signal strength across the same cell type, lack of specificity of fluorescent labels, overlapping signals in cell types with dense labeling, or undetectable signal at distal parts of the neurons, among others. In this review, we discuss the recent advances in the development of fluorescent transgenic mouse models that overcome to some extent the technical and conceptual limitations and tradeoffs between different strategies. We also discuss the potential use of these strains for understanding disease.
Collapse
Affiliation(s)
- Adrian Arias
- Department of System Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Linus Manubens-Gil
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Mara Dierssen
- Department of System Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| |
Collapse
|
14
|
Huang Y, Zhang Y, He Z, Manyande A, Wu D, Feng M, Xiang H. The connectome from the cerebral cortex to skeletal muscle using viral transneuronal tracers: a review. Am J Transl Res 2022; 14:4864-4879. [PMID: 35958450 PMCID: PMC9360884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 06/22/2022] [Indexed: 06/15/2023]
Abstract
Connectomics has developed from an initial observation under an electron microscope to the present well-known medical imaging research approach. The emergence of the most popular transneuronal tracers has further advanced connectomics research. Researchers use the virus trans-nerve tracing method to trace the whole brain, mark the brain nerve circuit and nerve connection structure, and construct a complete nerve conduction pathway. This review assesses current methods of studying cortical to muscle connections using viral neuronal tracers and demonstrates their application in disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Yan Huang
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
- Department of Interventional Therapy, The First Affiliated Hospital of Dalian Medical UniversityDalian 116000, Liaoning, P. R. China
| | - Yunhua Zhang
- Hubei Provincial Hospital of Traditional Chinese MedicineWuhan 430061, Hubei, P. R. China
- Clinical Medical College of Hubei University of Chinese MedicineWuhan 430061, Hubei, P. R. China
- Hubei Province Academy of Traditional Chinese MedicineWuhan 430061, Hubei, P. R. China
| | - Zhigang He
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| | - Anne Manyande
- School of Human and Social Sciences, University of West LondonLondon, UK
| | - Duozhi Wu
- Department of Anesthesiology, Hainan General HospitalHaikou 570311, Hainan, P. R. China
| | - Maohui Feng
- Department of Gastrointestinal Surgery, Wuhan Peritoneal Cancer Clinical Medical Research Center, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study CenterWuhan 430071, Hubei, P. R. China
| | - Hongbing Xiang
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| |
Collapse
|
15
|
Zheng N, Li M, Wu Y, Kaewborisuth C, Li Z, Gui Z, Wu J, Cai A, Lin K, Su KP, Xiang H, Tian X, Manyande A, Xu F, Wang J. A novel technology for in vivo detection of cell type-specific neural connection with AQP1-encoding rAAV2-retro vector and metal-free MRI. Neuroimage 2022; 258:119402. [PMID: 35732245 DOI: 10.1016/j.neuroimage.2022.119402] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 06/18/2022] [Accepted: 06/18/2022] [Indexed: 01/10/2023] Open
Abstract
A mammalian brain contains numerous neurons with distinct cell types for complex neural circuits. Virus-based circuit tracing tools are powerful in tracking the interaction among the different brain regions. However, detecting brain-wide neural networks in vivo remains challenging since most viral tracing systems rely on postmortem optical imaging. We developed a novel approach that enables in vivo detection of brain-wide neural connections based on metal-free magnetic resonance imaging (MRI). The recombinant adeno-associated virus (rAAV) with retrograde ability, the rAAV2-retro, encoding the human water channel aquaporin 1 (AQP1) MRI reporter gene was generated to label neural connections. The mouse was micro-injected with the virus at the Caudate Putamen (CPU) region and subjected to detection with Diffusion-weighted MRI (DWI). The prominent structure of the CPU-connected network was clearly defined. In combination with a Cre-loxP system, rAAV2-retro expressing Cre-dependent AQP1 provides a CPU-connected network of specific type neurons. Here, we established a sensitive, metal-free MRI-based strategy for in vivo detection of cell type-specific neural connections in the whole brain, which could visualize the dynamic changes of neural networks in rodents and potentially in non-human primates.
Collapse
Affiliation(s)
- Ning Zheng
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Mei Li
- The Brain Cognition and Brain Disease Institute (BCBDI), NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yang Wu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Challika Kaewborisuth
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand
| | - Zhen Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhu Gui
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinfeng Wu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Aoling Cai
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Kangguang Lin
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kuan-Pin Su
- Department of Psychiatry, China Medical University Hospital, Taichung City, Taiwan, China
| | - Hongbing Xiang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuebi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, Middlesex, TW8 9GA, UK
| | - Fuqiang Xu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China; The Brain Cognition and Brain Disease Institute (BCBDI), NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China; Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China.
| |
Collapse
|
16
|
Zhang Y, Wang J, Li J, Chen Y, Sun J, Lu Z, Li Y, Liu T. Functional analysis of mutations endowing rAAV2-retro with retrograde tracing capacity. Neurosci Lett 2022; 784:136746. [PMID: 35718237 DOI: 10.1016/j.neulet.2022.136746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/24/2022] [Accepted: 06/14/2022] [Indexed: 10/18/2022]
Abstract
Recombinant adeno-associated viruses (rAAVs) are widespread vectors in neuroscience research. However, the nearly absent retrograde access to projection neurons hampers their application in functional dissection of neural circuits and in therapeutic intervention. Recently, engineering of the AAV2 capsid has generated an AAV variant, called rAAV2-retro, with exceptional retrograde functionality. This variant comprises a 10-mer peptide insertion at residue 587 and two point mutations (LADQDYTKTA + V708I + N382D). Here, we evaluated the contribution of each mutation to retrograde transport in prefrontal cortex -striatum and amygdala-striatum pathways, respectively. Results showed that disruption of the inserted decapeptide almost completely abolishes the retrograde access to neurons projecting to striatum. Eliminating N382D has little effect on the retrograde functionality. Restoring another mutation V708I, however, even improves its performance in amygdala-striatum pathway. Parallel comparison within same animal further confirms this conflicting effect of V708I. These results demonstrate a pivotal role of decapeptide insertion in gaining the capacity of retrograde transport and highlight a neural circuit-dependent contribution of V708I. It suggests constant and custom engineering of rAAV2-retro might be required to tackle the challenge of tremendous neuronal heterogeneity.
Collapse
Affiliation(s)
- Yujing Zhang
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, 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 Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingyi Wang
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, 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 Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiamin Li
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, 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 Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yefei Chen
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, 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 Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China
| | - Jing Sun
- Department of Anesthesiology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen 518055, China
| | - Zhonghua Lu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, 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 Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China
| | - Yuantao Li
- Department of Anesthesiology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen 518055, China.
| | - Taian Liu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, 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 Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China.
| |
Collapse
|
17
|
Yan M, Yu W, Lv Q, Lv Q, Bo T, Chen X, Liu Y, Zhan Y, Yan S, Shen X, Yang B, Hu Q, Yu J, Qiu Z, Feng Y, Zhang XY, Wang H, Xu F, Wang Z. Mapping brain-wide excitatory projectome of primate prefrontal cortex at submicron resolution and comparison with diffusion tractography. eLife 2022; 11:72534. [PMID: 35593765 PMCID: PMC9122499 DOI: 10.7554/elife.72534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Resolving trajectories of axonal pathways in the primate prefrontal cortex remains crucial to gain insights into higher-order processes of cognition and emotion, which requires a comprehensive map of axonal projections linking demarcated subdivisions of prefrontal cortex and the rest of brain. Here, we report a mesoscale excitatory projectome issued from the ventrolateral prefrontal cortex (vlPFC) to the entire macaque brain by using viral-based genetic axonal tracing in tandem with high-throughput serial two-photon tomography, which demonstrated prominent monosynaptic projections to other prefrontal areas, temporal, limbic, and subcortical areas, relatively weak projections to parietal and insular regions but no projections directly to the occipital lobe. In a common 3D space, we quantitatively validated an atlas of diffusion tractography-derived vlPFC connections with correlative green fluorescent protein-labeled axonal tracing, and observed generally good agreement except a major difference in the posterior projections of inferior fronto-occipital fasciculus. These findings raise an intriguing question as to how neural information passes along long-range association fiber bundles in macaque brains, and call for the caution of using diffusion tractography to map the wiring diagram of brain circuits. In the brain is a web of interconnected nerve cells that send messages to one another via spindly projections called axons. These axons join together at junctions called synapses to create circuits of nerve cells which connect neighboring or distant brain regions. Notably, long-range neural connections underpin higher-order cognitive skills (such as planning and emotion regulation) which make humans distinct from our primate relatives. Only by untangling these far-reaching networks can researchers begin to delineate what sets the human brain apart from other species. Researchers deploy a range of imaging techniques to map neural networks: scanning entire brains using MRI machines, or imaging thin slices of fluorescently labelled brain tissue using powerful microscopes. However, tracing long-range axons at a high resolution is challenging, and has stirred up debate about whether some neural tracts, such as the inferior fronto-occipital fasciculus, are present in all primates or only humans. To address these discrepancies, Yan, Yu et al. employed a two-pronged approach to map neural circuits in the brains of macaques. First, two techniques – called viral tracing and two-photon microscopy – were used to create a three-dimensional, fine-grain map showing how the ventrolateral prefrontal cortex (vlPFC), which regulates complex behaviors, connects to the rest of the brain. This revealed prominent axons from the vlPFC projecting via a single synapse to distant brain regions involved in higher-order functions, such as encoding memories and processing emotion. However, there were no direct, monosynaptic connections between the vlPFC and the occipital lobe, the brain’s visual processing center at the back of the head. Next, Yan, Yu et al. used a specialized MRI scanner to create an atlas of neural circuits connected to the vlPFC, and compared these results to a technique tracing axons stained with a fluorescent dye. In general, there was good agreement between the two methods, except for major differences in the rear-end projections that typically form the inferior fronto-occipital fasciculus. This suggests that this long-range neural pathway exists in monkeys, but it connects via multiple synapses instead of a single junction as was previously thought. The findings of Yan, Yu et al. provide new insights on the far-reaching neural pathways connecting distant parts of the macaque brain. It also suggests that atlases of neural circuits from whole brain scans should be taken with caution and validated using neural tracing experiments.
Collapse
Affiliation(s)
- Mingchao Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenwen Yu
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Qian Lv
- School of Psychological and Cognitive Sciences; Beijing Key Laboratory of Behavior and Mental Health; IDG/McGovern Institute for Brain Research; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Qiming Lv
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Tingting Bo
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyu Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yilin Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yafeng Zhan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shengyao Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiangyu Shen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Baofeng Yang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Qiming Hu
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Jiangli Yu
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Zilong Qiu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yuanjing Feng
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Xiao-Yong Zhang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - He Wang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Fuqiang Xu
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Zheng Wang
- School of Psychological and Cognitive Sciences; Beijing Key Laboratory of Behavior and Mental Health; IDG/McGovern Institute for Brain Research; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
18
|
Feng Y, Li Y, Shen PP, Wang B. Gene-Modified Stem Cells for Spinal Cord Injury: a Promising Better Alternative Therapy. Stem Cell Rev Rep 2022; 18:2662-2682. [PMID: 35587330 DOI: 10.1007/s12015-022-10387-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2022] [Indexed: 12/18/2022]
Abstract
Stem cell therapy holds great promise for the treatment of spinal cord injury (SCI), which can reverse neurodegeneration and promote tissue regeneration via its pluripotency and ability to secrete neurotrophic factors. Although various stem cell-based approaches have shown certain therapeutic effects when applied to the treatment of SCI, their clinical efficacies have been disappointing. Thus, it is an urgent need to further enhance the neurological benefits of stem cells through bioengineering strategies including genetic engineering. In this review, we summarize the progress of stem cell therapy for SCI and the prospect of genetically modified stem cells, focusing on the genome editing tools and functional molecules involved in SCI repair, trying to provide a deeper understanding of genetically modified stem cell therapy and more applicable clinical strategies for SCI repair.
Collapse
Affiliation(s)
- Yirui Feng
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yu Li
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, Jiangsu Province, China
| | - Ping-Ping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and the Comprehensive Cancer Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Bin Wang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.
| |
Collapse
|
19
|
A novel H129-based anterograde monosynaptic tracer exhibits features of strong labeling intensity, high tracing efficiency, and reduced retrograde labeling. Mol Neurodegener 2022; 17:6. [PMID: 35012591 PMCID: PMC8744342 DOI: 10.1186/s13024-021-00508-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/09/2021] [Indexed: 12/05/2022] Open
Abstract
Background Viral tracers are important tools for mapping brain connectomes. The feature of predominant anterograde transneuronal transmission offers herpes simplex virus-1 (HSV-1) strain H129 (HSV1-H129) as a promising candidate to be developed as anterograde viral tracers. In our earlier studies, we developed H129-derived anterograde polysynaptic tracers and TK deficient (H129-dTK) monosynaptic tracers. However, their broad application is limited by some intrinsic drawbacks of the H129-dTK tracers, such as low labeling intensity due to TK deficiency and potential retrograde labeling caused by axon terminal invasion. The glycoprotein K (gK) of HSV-1 plays important roles in virus entry, egress, and virus-induced cell fusion. Its deficiency severely disables virus egress and spread, while only slightly limits viral genome replication and expression of viral proteins. Therefore, we created a novel H129-derived anterograde monosynaptic tracer (H129-dgK) by targeting gK, which overcomes the limitations of H129-dTK. Methods Using our established platform and pipeline for developing viral tracers, we generated a novel tracer by deleting the gK gene from the H129-G4. The gK-deleted virus (H129-dgK-G4) was reconstituted and propagated in the Vero cell expressing wildtype H129 gK (gKwt) or the mutant gK (gKmut, A40V, C82S, M223I, L224V, V309M), respectively. Then the obtained viral tracers of gKmut pseudotyped and gKwt coated H129-dgK-G4 were tested in vitro and in vivo to characterize their tracing properties. Results H129-dgK-G4 expresses high levels of fluorescent proteins, eliminating the requirement of immunostaining for imaging detection. Compared to the TK deficient monosynaptic tracer H129-dTK-G4, H129-dgK-G4 labeled neurons with 1.76-fold stronger fluorescence intensity, and visualized 2.00-fold more postsynaptic neurons in the downstream brain regions. gKmut pseudotyping leads to a 77% decrease in retrograde labeling by reducing axon terminal invasion, and thus dramatically improves the anterograde-specific tracing of H129-dgK-G4. In addition, assisted by the AAV helper trans-complementarily expressing gKwt, H129-dgK-G4 allows for mapping monosynaptic connections and quantifying the circuit connectivity difference in the Alzheimer’s disease and control mouse brains. Conclusions gKmut pseudotyped H129-dgK-G4, a novel anterograde monosynaptic tracer, overcomes the limitations of H129-dTK tracers, and demonstrates desirable features of strong labeling intensity, high tracing efficiency, and improved anterograde specificity. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-021-00508-6.
Collapse
|
20
|
Birnie MT, Levis SC, Mahler SV, Baram TZ. Developmental Trajectories of Anhedonia in Preclinical Models. Curr Top Behav Neurosci 2022; 58:23-41. [PMID: 35156184 DOI: 10.1007/7854_2021_299] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This chapter discusses how the complex concept of anhedonia can be operationalized and studied in preclinical models. It provides information about the development of anhedonia in the context of early-life adversity, and the power of preclinical models to tease out the diverse molecular, epigenetic, and network mechanisms that are responsible for anhedonia-like behaviors.Specifically, we first discuss the term anhedonia, reviewing the conceptual components underlying reward-related behaviors and distinguish anhedonia pertaining to deficits in motivational versus consummatory behaviors. We then describe the repertoire of experimental approaches employed to study anhedonia-like behaviors in preclinical models, and the progressive refinement over the past decade of both experimental instruments (e.g., chemogenetics, optogenetics) and conceptual constructs (salience, valence, conflict). We follow with an overview of the state of current knowledge of brain circuits, nodes, and projections that execute distinct aspects of hedonic-like behaviors, as well as neurotransmitters, modulators, and receptors involved in the generation of anhedonia-like behaviors. Finally, we discuss the special case of anhedonia that arises following early-life adversity as an eloquent example enabling the study of causality, mechanisms, and sex dependence of anhedonia.Together, this chapter highlights the power, potential, and limitations of using preclinical models to advance our understanding of the origin and mechanisms of anhedonia and to discover potential targets for its prevention and mitigation.
Collapse
Affiliation(s)
- Matthew T Birnie
- Departments of Anatomy/Neurobiology and Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Sophia C Levis
- Departments of Anatomy/Neurobiology and Neurobiology/Behavior, University of California-Irvine, Irvine, CA, USA
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Anatomy/Neurobiology and Pediatrics, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
21
|
Zingg B, Dong HW, Tao HW, Zhang LI. Application of AAV1 for Anterograde Transsynaptic Circuit Mapping and Input-Dependent Neuronal Cataloging. Curr Protoc 2022; 2:e339. [PMID: 35044725 PMCID: PMC8852298 DOI: 10.1002/cpz1.339] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Viruses that spread transsynaptically provide a powerful means to study interconnected circuits in the brain. Here we describe the use of adeno-associated virus, serotype 1 (AAV1), as a tool to achieve robust, anterograde transsynaptic spread in a variety of unidirectional pathways. A protocol for performing intracranial AAV1 injections in mice is presented, along with additional guidance for planning experiments, sourcing materials, and optimizing the approach to achieve the most successful outcomes. By following the methods presented here, researchers will be able to reveal postsynaptically connected neurons downstream of a given AAV1 injection site and access these input-defined cells for subsequent mapping, recording, and manipulation to characterize their anatomical and functional properties. © 2022 Wiley Periodicals LLC. Basic Protocol: Stereotaxic injection of AAV1 for anterograde transsynaptic spread.
Collapse
Affiliation(s)
- Brian Zingg
- Department of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Hong-Wei Dong
- Department of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Huizhong Whit Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Li I. Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
22
|
Sun S, Zhu Z, He T, Chen F, Wang X, Zhang X, Li M, Li Y, Sun Y, He Q, Li X, Wang M. A study of adeno-associated virus in cortical-thalamostriatal pathway. Brain Res 2021; 1773:147698. [PMID: 34655617 DOI: 10.1016/j.brainres.2021.147698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/29/2022]
Abstract
The cortical-thalamostriatal pathway constitutes the cortico-basal ganglia circuit and plays a critical role in the control of movement. Emerging evidence shows that center median/parafascicular (CM/Pf) neurons are lost in Parkinson's disease (PD) patients with motor deficits and CM/Pf neurons send massive and topographically organized projections to specific regions of the dorsal striatum, but provide only minor inputs to the cerebral cortex. However, anatomical connectivity in the cortical-thalamostriatal pathway are poorly understood at present. In the present study, we used a neural tracing method with adeno-associated virus (AAV) to monitor the cortical-thalamostriatal connectivity in rats. We found that parafascicular nucleus (PF) not only project directly to the striatum but send minor inputs to the cortical regions. It was manifested by green fluorescent protein (GFP) expressing fibers observed in dorsolateral striatum (DLS) and the primary motor cortex (M1) after adeno-associated virus serotype 2/9 (AAV2/9)-GFP injection into PF and GFP expressing cells observed in PF after injection AAV2/retro-GFP into M1. And the PF also receive projections from the DLS and it was demonstrated by GFP expressing fibers in PF after AAV2/9-GFP injection into DLS and GFP expressing cells in DLS after injection AAV2/retro-GFP into PF. Histological and behavioral analysis revealed that AAV vector transduction cause damage in neurons on the injection sites and also damage motor activity of rats suggesting caution in clinical application.
Collapse
Affiliation(s)
- Shuang Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Zhiwei Zhu
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Tianqi He
- Qilu Hospital, Shandong University, Jinan 250014, China
| | - Feiyu Chen
- School of International Education, Qilu University of Technology, Jinan 250014, China
| | - Xiaojun Wang
- The First Hospital Affiliated with Shandong First Medicine University, Jinan 250014, China
| | - Xiao Zhang
- Editorial Department of Journal, Shandong Jianzhu University, Jinan 250014, China
| | - Min Li
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Yuchuan Li
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Yue Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Qin He
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Xiuhua Li
- The First Hospital Affiliated with Shandong First Medicine University, Jinan 250014, China.
| | - Min Wang
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China.
| |
Collapse
|
23
|
Beier KT. The Serendipity of Viral Trans-Neuronal Specificity: More Than Meets the Eye. Front Cell Neurosci 2021; 15:720807. [PMID: 34671244 PMCID: PMC8521040 DOI: 10.3389/fncel.2021.720807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/07/2021] [Indexed: 12/25/2022] Open
Abstract
Trans-neuronal viruses are frequently used as neuroanatomical tools for mapping neuronal circuits. Specifically, recombinant one-step rabies viruses (RABV) have been instrumental in the widespread application of viral circuit mapping, as these viruses have enabled labs to map the direct inputs onto defined cell populations. Within the neuroscience community, it is widely believed that RABV spreads directly between neurons via synaptic connections, a hypothesis based principally on two observations. First, the virus labels neurons in a pattern consistent with known anatomical connectivity. Second, few glial cells appear to be infected following RABV injections, despite the fact that glial cells are abundant in the brain. However, there is no direct evidence that RABV can actually be transmitted through synaptic connections. Here we review the immunosubversive mechanisms that are critical to RABV’s success for infiltration of the central nervous system (CNS). These include interfering with and ultimately killing migratory T cells while maintaining levels of interferon (IFN) signaling in the brain parenchyma. Finally, we critically evaluate studies that support or are against synaptically-restricted RABV transmission and the implications of viral-host immune responses for RABV transmission in the brain.
Collapse
Affiliation(s)
- Kevin Thomas Beier
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
24
|
Pisano TJ, Dhanerawala ZM, Kislin M, Bakshinskaya D, Engel EA, Hansen EJ, Hoag AT, Lee J, de Oude NL, Venkataraju KU, Verpeut JL, Hoebeek FE, Richardson BD, Boele HJ, Wang SSH. Homologous organization of cerebellar pathways to sensory, motor, and associative forebrain. Cell Rep 2021; 36:109721. [PMID: 34551311 PMCID: PMC8506234 DOI: 10.1016/j.celrep.2021.109721] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/06/2021] [Accepted: 08/25/2021] [Indexed: 12/31/2022] Open
Abstract
Cerebellar outputs take polysynaptic routes to reach the rest of the brain, impeding conventional tracing. Here, we quantify pathways between the cerebellum and forebrain by using transsynaptic tracing viruses and a whole-brain analysis pipeline. With retrograde tracing, we find that most descending paths originate from the somatomotor cortex. Anterograde tracing of ascending paths encompasses most thalamic nuclei, especially ventral posteromedial, lateral posterior, mediodorsal, and reticular nuclei. In the neocortex, sensorimotor regions contain the most labeled neurons, but we find higher densities in associative areas, including orbital, anterior cingulate, prelimbic, and infralimbic cortex. Patterns of ascending expression correlate with c-Fos expression after optogenetic inhibition of Purkinje cells. Our results reveal homologous networks linking single areas of the cerebellar cortex to diverse forebrain targets. We conclude that shared areas of the cerebellum are positioned to provide sensory-motor information to regions implicated in both movement and nonmotor function.
Collapse
Affiliation(s)
- Thomas J Pisano
- Neuroscience Institute, Washington Road, Princeton University, Princeton, NJ 08544, USA
| | - Zahra M Dhanerawala
- Neuroscience Institute, Washington Road, Princeton University, Princeton, NJ 08544, USA
| | - Mikhail Kislin
- Neuroscience Institute, Washington Road, Princeton University, Princeton, NJ 08544, USA
| | - Dariya Bakshinskaya
- Neuroscience Institute, Washington Road, Princeton University, Princeton, NJ 08544, USA
| | - Esteban A Engel
- Neuroscience Institute, Washington Road, Princeton University, Princeton, NJ 08544, USA
| | - Ethan J Hansen
- WWAMI Medical Education, University of Idaho, Moscow, ID 83844, USA
| | - Austin T Hoag
- Neuroscience Institute, Washington Road, Princeton University, Princeton, NJ 08544, USA
| | - Junuk Lee
- Neuroscience Institute, Washington Road, Princeton University, Princeton, NJ 08544, USA
| | - Nina L de Oude
- Department of Neuroscience, Erasmus MC, 3000 DR Rotterdam, the Netherlands
| | | | - Jessica L Verpeut
- Neuroscience Institute, Washington Road, Princeton University, Princeton, NJ 08544, USA
| | - Freek E Hoebeek
- Department for Developmental Origins of Disease, Brain Center and Wilhelmina Childrens Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ben D Richardson
- WWAMI Medical Education, University of Idaho, Moscow, ID 83844, USA; Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Henk-Jan Boele
- Neuroscience Institute, Washington Road, Princeton University, Princeton, NJ 08544, USA; Department of Neuroscience, Erasmus MC, 3000 DR Rotterdam, the Netherlands.
| | - Samuel S-H Wang
- Neuroscience Institute, Washington Road, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
25
|
Kuljis DA, Micheva KD, Ray A, Wegner W, Bowman R, Madison DV, Willig KI, Barth AL. Gephyrin-Lacking PV Synapses on Neocortical Pyramidal Neurons. Int J Mol Sci 2021; 22:ijms221810032. [PMID: 34576197 PMCID: PMC8467468 DOI: 10.3390/ijms221810032] [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: 07/30/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 11/26/2022] Open
Abstract
Gephyrin has long been thought of as a master regulator for inhibitory synapses, acting as a scaffold to organize γ-aminobutyric acid type A receptors (GABAARs) at the post-synaptic density. Accordingly, gephyrin immunostaining has been used as an indicator of inhibitory synapses; despite this, the pan-synaptic localization of gephyrin to specific classes of inhibitory synapses has not been demonstrated. Genetically encoded fibronectin intrabodies generated with mRNA display (FingRs) against gephyrin (Gephyrin.FingR) reliably label endogenous gephyrin, and can be tagged with fluorophores for comprehensive synaptic quantitation and monitoring. Here we investigated input- and target-specific localization of gephyrin at a defined class of inhibitory synapse, using Gephyrin.FingR proteins tagged with EGFP in brain tissue from transgenic mice. Parvalbumin-expressing (PV) neuron presynaptic boutons labeled using Cre- dependent synaptophysin-tdTomato were aligned with postsynaptic Gephyrin.FingR puncta. We discovered that more than one-third of PV boutons adjacent to neocortical pyramidal (Pyr) cell somas lack postsynaptic gephyrin labeling. This finding was confirmed using correlative fluorescence and electron microscopy. Our findings suggest some inhibitory synapses may lack gephyrin. Gephyrin-lacking synapses may play an important role in dynamically regulating cell activity under different physiological conditions.
Collapse
Affiliation(s)
- Dika A. Kuljis
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Kristina D. Micheva
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; (K.D.M.); (D.V.M.)
| | - Ajit Ray
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Waja Wegner
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37075 Göttingen, Germany; (W.W.); (K.I.W.)
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Ryan Bowman
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Daniel V. Madison
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; (K.D.M.); (D.V.M.)
| | - Katrin I. Willig
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37075 Göttingen, Germany; (W.W.); (K.I.W.)
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Alison L. Barth
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
- Correspondence: ; Tel.: +1-412-268-1198
| |
Collapse
|
26
|
Wang L, Challis C, Li S, Fowlkes CC, Kumar SR, Yuan PQ, Taché YF. Multicolor sparse viral labeling and 3D digital tracing of enteric plexus in mouse proximal colon using a novel adeno-associated virus capsid. Neurogastroenterol Motil 2021; 33:e14014. [PMID: 33094876 PMCID: PMC8568587 DOI: 10.1111/nmo.14014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 08/15/2020] [Accepted: 09/22/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intravenous administration of adeno-associated virus (AAV) can be used as a noninvasive approach to trace neuronal morphology and links. AAV-PHP.S is a variant of AAV9 that effectively transduces the peripheral nervous system. The objective was to label randomly and sparsely enteric plexus in the mouse colon using AAV-PHP.S with a tunable two-component multicolor vector system and digitally trace individual neurons and nerve fibers within microcircuits in three dimensions (3D). METHODS A vector system including a tetracycline inducer with a tet-responsive element driving three separate fluorophores was packaged in the AAV-PHP.S capsid. The vectors were injected retro-orbitally in mice, and the colon was harvested 3 weeks after. Confocal microscopic images of enteric plexus were digitally segmented and traced in 3D using Neurolucida 360, neuTube, or Imaris software. KEY RESULTS The transduction of multicolor AAV vectors induced random sparse spectral labeling of soma and neurites primarily in the myenteric plexus of the proximal colon, while neurons in the submucosal plexus were occasionally transduced. Digital tracing in 3D showed various types of wiring, including multiple conjunctions of one neuron with other neurons, neurites en route, and endings; clusters of neurons in close apposition between each other; axon-axon parallel conjunctions; and intraganglionic nerve endings consisting of multiple nerve endings and passing fibers. Most of digitally traced neuronal somas were of small or medium in size. CONCLUSIONS & INFERENCES The multicolor AAV-PHP.S-packaged vectors enabled random sparse spectral labeling and revealed complexities of enteric microcircuit in the mouse proximal colon. The techniques can facilitate digital modeling of enteric micro-circuitry.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Medicine, Taman Manoukisan Digestive Diseases Division, UCLA, Los Angeles, CA, USA,Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Collin Challis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Songlin Li
- Department of Medicine, Taman Manoukisan Digestive Diseases Division, UCLA, Los Angeles, CA, USA
| | | | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Pu-Qing Yuan
- Department of Medicine, Taman Manoukisan Digestive Diseases Division, UCLA, Los Angeles, CA, USA
| | - Yvette F. Taché
- Department of Medicine, Taman Manoukisan Digestive Diseases Division, UCLA, Los Angeles, CA, USA,Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
27
|
Universal autofocus for quantitative volumetric microscopy of whole mouse brains. Nat Methods 2021; 18:953-958. [PMID: 34312564 DOI: 10.1038/s41592-021-01208-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 06/14/2021] [Indexed: 11/08/2022]
Abstract
Unbiased quantitative analysis of macroscopic biological samples demands fast imaging systems capable of maintaining high resolution across large volumes. Here we introduce RAPID (rapid autofocusing via pupil-split image phase detection), a real-time autofocus method applicable in every widefield-based microscope. RAPID-enabled light-sheet microscopy reliably reconstructs intact, cleared mouse brains with subcellular resolution, and allowed us to characterize the three-dimensional (3D) spatial clustering of somatostatin-positive neurons in the whole encephalon, including densely labeled areas. Furthermore, it enabled 3D morphological analysis of microglia across the entire brain. Beyond light-sheet microscopy, we demonstrate that RAPID maintains high image quality in various settings, from in vivo fluorescence imaging to 3D tracking of fast-moving organisms. RAPID thus provides a flexible autofocus solution that is suitable for traditional automated microscopy tasks as well as for quantitative analysis of large biological specimens.
Collapse
|
28
|
Cai A, Zheng N, Thompson GJ, Wu Y, Nie B, Lin K, Su P, Wu J, Manyande A, Zhu L, Wang J, Xu F. Longitudinal neural connection detection using a ferritin-encoding adeno-associated virus vector and in vivo MRI method. Hum Brain Mapp 2021; 42:5010-5022. [PMID: 34288264 PMCID: PMC8449107 DOI: 10.1002/hbm.25596] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/20/2021] [Accepted: 07/06/2021] [Indexed: 01/17/2023] Open
Abstract
The investigation of neural circuits is important for interpreting both healthy brain function and psychiatric disorders. Currently, the architecture of neural circuits is always investigated with fluorescent protein encoding neurotropic virus and ex vivo fluorescent imaging technology. However, it is difficult to obtain a whole‐brain neural circuit connection in living animals, due to the limited fluorescent imaging depth. Herein, the noninvasive, whole‐brain imaging technique of MRI and the hypotoxicity virus vector AAV (adeno‐associated virus) were combined to investigate the whole‐brain neural circuits in vivo. AAV2‐retro are an artificially‐evolved virus vector that permits access to the terminal of neurons and retrograde transport to their cell bodies. By expressing the ferritin protein which could accumulate iron ions and influence the MRI contrast, the neurotropic virus can cause MRI signal changes in the infected regions. For mice injected with the ferritin‐encoding virus vector (rAAV2‐retro‐CAG‐Ferritin) in the caudate putamen (CPu), several regions showed significant changes in MRI contrasts, such as PFC (prefrontal cortex), HIP (hippocampus), Ins (insular cortex) and BLA (basolateral amygdala). The expression of ferritin in those regions was also verified with ex vivo fluorescence imaging. In addition, we demonstrated that changes in T2 relaxation time could be used to identify the spread area of the virus in the brain over time. Thus, the neural connections could be longitudinally detected with the in vivo MRI method. This novel technique could be utilized to observe the viral infection process and detect the neural circuits in a living animal.
Collapse
Affiliation(s)
- Aoling Cai
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | - Ning Zheng
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | | | - Yang Wu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Binbin Nie
- Key Laboratory of Nuclear Radiation and Nuclear Energy Technology, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Kunzhang Lin
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Peng Su
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Jinfeng Wu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, UK
| | - LingQiang Zhu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China.,Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, 2nd Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Fuqiang Xu
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China.,Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Center for Excellence in Brain Science and Intelligent Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
29
|
Yamamori T. Functional visualization and manipulation in the marmoset brain using viral vectors. Curr Opin Pharmacol 2021; 60:11-16. [PMID: 34280704 DOI: 10.1016/j.coph.2021.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/30/2021] [Accepted: 06/14/2021] [Indexed: 02/08/2023]
Abstract
The common marmoset, a New World monkey, has a primate-specific cortex with approximately 40 Brodmann areas. Genetically encoded calcium indicator (GECI) techniques have been applied to study the functional organization of the marmoset cortex. The success of GCaMP (a green fluorescent of GECI) imaging and other advances, including optogenetic approaches, provide an interesting and exciting opportunity to study the primate brain at the molecular and cellular levels, leading to an understanding of primate neural circuits. These approaches will help advance our knowledge on cognition in primates, including humans, and therapy for human neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Tetsuo Yamamori
- Center for Brain Science, Laboratory for Molecular Analysis of Higher Brain Function, RIKEN, 2-1 Hirosawa, Wako, 351-0198, Japan.
| |
Collapse
|
30
|
Jia F, Li L, Liu H, Lv P, Shi X, Wu Y, Ling C, Xu F. Development of a rabies virus-based retrograde tracer with high trans-monosynaptic efficiency by reshuffling glycoprotein. Mol Brain 2021; 14:109. [PMID: 34238335 PMCID: PMC8265122 DOI: 10.1186/s13041-021-00821-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/04/2021] [Indexed: 11/12/2022] Open
Abstract
Rabies virus (RV) is the most widely used vector for mapping neural circuits. Previous studies have shown that the RV glycoprotein can be a target to improve the retrograde transsynaptic tracing efficiency. However, the current versions still label only a small portion of all presynaptic neurons. Here, we reshuffled the oG sequence, a chimeric glycoprotein, with positive codon pair bias score (CPBS) based on bioinformatic analysis of mouse codon pair bias, generating ooG, a further optimized glycoprotein. Our experimental data reveal that the ooG has a higher expression level than the oG in vivo, which significantly increases the tracing efficiency by up to 12.6 and 62.1-fold compared to oG and B19G, respectively. The new tool can be used for labeling neural circuits Therefore, the approach reported here provides a convenient, efficient and universal strategy to improve protein expression for various application scenarios such as trans-synaptic tracing efficiency, cell engineering, and vaccine and oncolytic virus designs.
Collapse
Affiliation(s)
- Fan 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 (BCBDI), Translational Research Center for the Nervous System (TRCNS), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- 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, Guangdong Provincial Medical Products Administration, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Li Li
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems,, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Haizhou Liu
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Pei Lv
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems,, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiangwei Shi
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems,, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems,, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Chen Ling
- Division of Molecular and Cellular Therapy, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fuqiang Xu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Translational Research Center for the Nervous System (TRCNS), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- 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, Guangdong Provincial Medical Products Administration, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems,, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China.
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
31
|
Yook JS, Kim J, Kim J. Convergence Circuit Mapping: Genetic Approaches From Structure to Function. Front Syst Neurosci 2021; 15:688673. [PMID: 34234652 PMCID: PMC8255632 DOI: 10.3389/fnsys.2021.688673] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/28/2021] [Indexed: 12/22/2022] Open
Abstract
Understanding the complex neural circuits that underpin brain function and behavior has been a long-standing goal of neuroscience. Yet this is no small feat considering the interconnectedness of neurons and other cell types, both within and across brain regions. In this review, we describe recent advances in mouse molecular genetic engineering that can be used to integrate information on brain activity and structure at regional, cellular, and subcellular levels. The convergence of structural inputs can be mapped throughout the brain in a cell type-specific manner by antero- and retrograde viral systems expressing various fluorescent proteins and genetic switches. Furthermore, neural activity can be manipulated using opto- and chemo-genetic tools to interrogate the functional significance of this input convergence. Monitoring neuronal activity is obtained with precise spatiotemporal resolution using genetically encoded sensors for calcium changes and specific neurotransmitters. Combining these genetically engineered mapping tools is a compelling approach for unraveling the structural and functional brain architecture of complex behaviors and malfunctioned states of neurological disorders.
Collapse
Affiliation(s)
- Jang Soo Yook
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Jihyun Kim
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, South Korea.,Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, South Korea
| | - Jinhyun Kim
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, South Korea.,Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, South Korea
| |
Collapse
|
32
|
Faulkner RL, Wall NR, Callaway EM, Cline HT. Application of Recombinant Rabies Virus to Xenopus Tadpole Brain. eNeuro 2021; 8:ENEURO.0477-20.2021. [PMID: 34099488 PMCID: PMC8260272 DOI: 10.1523/eneuro.0477-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/13/2021] [Accepted: 05/31/2021] [Indexed: 12/25/2022] Open
Abstract
The Xenopus laevis experimental system has provided significant insight into the development and plasticity of neural circuits. Xenopus neuroscience research would be enhanced by additional tools to study neural circuit structure and function. Rabies viruses are powerful tools to label and manipulate neural circuits and have been widely used to study mesoscale connectomics. Whether rabies virus can be used to transduce neurons and express transgenes in Xenopus has not been systematically investigated. Glycoprotein-deleted rabies virus transduces neurons at the axon terminal and retrogradely labels their cell bodies. We show that glycoprotein-deleted rabies virus infects local and projection neurons in the Xenopus tadpole when directly injected into brain tissue. Pseudotyping glycoprotein-deleted rabies with EnvA restricts infection to cells with exogenous expression of the EnvA receptor, TVA. EnvA pseudotyped virus specifically infects tadpole neurons with promoter-driven expression of TVA, demonstrating its utility to label targeted neuronal populations. Neuronal cell types are defined by a combination of features including anatomical location, expression of genetic markers, axon projection sites, morphology, and physiological properties. We show that driving TVA expression in one hemisphere and injecting EnvA pseudotyped virus into the contralateral hemisphere, retrogradely labels neurons defined by cell body location and axon projection site. Using this approach, rabies can be used to identify cell types in Xenopus brain and simultaneously to express transgenes which enable monitoring or manipulation of neuronal activity. This makes rabies a valuable tool to study the structure and function of neural circuits in Xenopus.Significance StatementStudies in Xenopus have contributed a great deal to our understanding of brain circuit development and plasticity, regeneration, and hormonal regulation of behavior and metamorphosis. Here, we show that recombinant rabies virus transduces neurons in the Xenopus tadpole, enlarging the toolbox that can be applied to studying Xenopus brain. Rabies can be used for retrograde labeling and expression of a broad range of transgenes including fluorescent proteins for anatomical tracing and studying neuronal morphology, voltage or calcium indicators to visualize neuronal activity, and photo- or chemosensitive channels to control neuronal activity. The versatility of these tools enables diverse experiments to analyze and manipulate Xenopus brain structure and function, including mesoscale connectivity.
Collapse
Affiliation(s)
- Regina L Faulkner
- Neuroscience Department and The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla CA
| | | | | | - Hollis T Cline
- Neuroscience Department and The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla CA
| |
Collapse
|
33
|
Stepankova K, Jendelova P, Machova Urdzikova L. Planet of the AAVs: The Spinal Cord Injury Episode. Biomedicines 2021; 9:613. [PMID: 34071245 PMCID: PMC8228984 DOI: 10.3390/biomedicines9060613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
The spinal cord injury (SCI) is a medical and life-disrupting condition with devastating consequences for the physical, social, and professional welfare of patients, and there is no adequate treatment for it. At the same time, gene therapy has been studied as a promising approach for the treatment of neurological and neurodegenerative disorders by delivering remedial genes to the central nervous system (CNS), of which the spinal cord is a part. For gene therapy, multiple vectors have been introduced, including integrating lentiviral vectors and non-integrating adeno-associated virus (AAV) vectors. AAV vectors are a promising system for transgene delivery into the CNS due to their safety profile as well as long-term gene expression. Gene therapy mediated by AAV vectors shows potential for treating SCI by delivering certain genetic information to specific cell types. This review has focused on a potential treatment of SCI by gene therapy using AAV vectors.
Collapse
Affiliation(s)
- Katerina Stepankova
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská 1083, 14200 Prague, Czech Republic;
- Department of Neuroscience, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Pavla Jendelova
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská 1083, 14200 Prague, Czech Republic;
- Department of Neuroscience, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Lucia Machova Urdzikova
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská 1083, 14200 Prague, Czech Republic;
- Department of Neuroscience, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| |
Collapse
|
34
|
Morimoto MM, Uchishiba E, Saleem AB. Organization of feedback projections to mouse primary visual cortex. iScience 2021; 24:102450. [PMID: 34113813 PMCID: PMC8169797 DOI: 10.1016/j.isci.2021.102450] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 02/01/2021] [Accepted: 04/14/2021] [Indexed: 11/17/2022] Open
Abstract
Top-down, context-dependent modulation of visual processing has been a topic of wide interest, including in mouse primary visual cortex (V1). However, the organization of feedback projections to V1 is relatively unknown. Here, we investigated inputs to mouse V1 by injecting retrograde tracers. We developed a software pipeline that maps labeled cell bodies to corresponding brain areas in the Allen Reference Atlas. We identified more than 24 brain areas that provide inputs to V1 and quantified the relative strength of their projections. We also assessed the organization of the projections, based on either the organization of cell bodies in the source area (topography) or the distribution of projections across V1 (bias). Projections from most higher visual and some nonvisual areas to V1 showed both topography and bias. Such organization of feedback projections to V1 suggests that parts of the visual field are differentially modulated by context, which can be ethologically relevant for a navigating animal.
Collapse
Affiliation(s)
- Mai M. Morimoto
- UCL Institute of Behavioural Neuroscience, Department of Experimental Psychology, University College London, London, WC1H 0AP, UK
| | - Emi Uchishiba
- UCL Institute of Behavioural Neuroscience, Department of Experimental Psychology, University College London, London, WC1H 0AP, UK
| | - Aman B. Saleem
- UCL Institute of Behavioural Neuroscience, Department of Experimental Psychology, University College London, London, WC1H 0AP, UK
| |
Collapse
|
35
|
Yang H, Xiong F, Song YG, Jiang HF, Qin HB, Zhou J, Lu S, Grieco SF, Xu X, Zeng WB, Zhao F, Luo MH. HSV-1 H129-Derived Anterograde Neural Circuit Tracers: Improvements, Production, and Applications. Neurosci Bull 2021; 37:701-719. [PMID: 33367996 PMCID: PMC8099975 DOI: 10.1007/s12264-020-00614-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 07/26/2020] [Indexed: 10/22/2022] Open
Abstract
Anterograde viral tracers are powerful and essential tools for dissecting the output targets of a brain region of interest. They have been developed from herpes simplex virus 1 (HSV-1) strain H129 (H129), and have been successfully applied to map diverse neural circuits. Initially, the anterograde polysynaptic tracer H129-G4 was used by many groups. We then developed the first monosynaptic tracer, H129-dTK-tdT, which was highly successful, yet improvements are needed. Now, by inserting another tdTomato expression cassette into the H129-dTK-tdT genome, we have created H129-dTK-T2, an updated version of H129-dTK-tdT that has improved labeling intensity. To help scientists produce and apply our H129-derived viral tracers, here we provide the protocol describing our detailed and standardized procedures. Commonly-encountered technical problems and their solutions are also discussed in detail. Broadly, the dissemination of this protocol will greatly support scientists to apply these viral tracers on a large scale.
Collapse
Affiliation(s)
- Hong Yang
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Feng Xiong
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi-Ge Song
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hai-Fei Jiang
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hai-Bin Qin
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Zhou
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Sha Lu
- Shanghai Genechem Co. Ltd., Shanghai, 201203, China
| | - Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Wen-Bo Zeng
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Fei Zhao
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| | - Min-Hua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
36
|
Hayashi T, Hou Y, Glasser MF, Autio JA, Knoblauch K, Inoue-Murayama M, Coalson T, Yacoub E, Smith S, Kennedy H, Van Essen DC. The nonhuman primate neuroimaging and neuroanatomy project. Neuroimage 2021; 229:117726. [PMID: 33484849 PMCID: PMC8079967 DOI: 10.1016/j.neuroimage.2021.117726] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/13/2020] [Accepted: 01/02/2021] [Indexed: 11/29/2022] Open
Abstract
Multi-modal neuroimaging projects such as the Human Connectome Project (HCP) and UK Biobank are advancing our understanding of human brain architecture, function, connectivity, and their variability across individuals using high-quality non-invasive data from many subjects. Such efforts depend upon the accuracy of non-invasive brain imaging measures. However, 'ground truth' validation of connectivity using invasive tracers is not feasible in humans. Studies using nonhuman primates (NHPs) enable comparisons between invasive and non-invasive measures, including exploration of how "functional connectivity" from fMRI and "tractographic connectivity" from diffusion MRI compare with long-distance connections measured using tract tracing. Our NonHuman Primate Neuroimaging & Neuroanatomy Project (NHP_NNP) is an international effort (6 laboratories in 5 countries) to: (i) acquire and analyze high-quality multi-modal brain imaging data of macaque and marmoset monkeys using protocols and methods adapted from the HCP; (ii) acquire quantitative invasive tract-tracing data for cortical and subcortical projections to cortical areas; and (iii) map the distributions of different brain cell types with immunocytochemical stains to better define brain areal boundaries. We are acquiring high-resolution structural, functional, and diffusion MRI data together with behavioral measures from over 100 individual macaques and marmosets in order to generate non-invasive measures of brain architecture such as myelin and cortical thickness maps, as well as functional and diffusion tractography-based connectomes. We are using classical and next-generation anatomical tracers to generate quantitative connectivity maps based on brain-wide counting of labeled cortical and subcortical neurons, providing ground truth measures of connectivity. Advanced statistical modeling techniques address the consistency of both kinds of data across individuals, allowing comparison of tracer-based and non-invasive MRI-based connectivity measures. We aim to develop improved cortical and subcortical areal atlases by combining histological and imaging methods. Finally, we are collecting genetic and sociality-associated behavioral data in all animals in an effort to understand how genetic variation shapes the connectome and behavior.
Collapse
Affiliation(s)
- Takuya Hayashi
- Laboratory for Brain Connectomics Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 MI R&D Center 3F, Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan; Department of Neurobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yujie Hou
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Matthew F Glasser
- Department of Neuroscience, Washington University Medical School, St Louis, MO USA; Department of Neuroscience and Radiology, Washington University Medical School, St Louis, MO USA
| | - Joonas A Autio
- Laboratory for Brain Connectomics Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 MI R&D Center 3F, Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Kenneth Knoblauch
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, Bron, France
| | | | - Tim Coalson
- Department of Neuroscience, Washington University Medical School, St Louis, MO USA
| | - Essa Yacoub
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, USA
| | - Stephen Smith
- Oxford Centre for Functional Magnetic Resonance Imaging of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging (WIN), Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Henry Kennedy
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, Bron, France; Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences (CAS) Key Laboratory of Primate Neurobiology, CAS, Shanghai, China
| | - David C Van Essen
- Department of Neuroscience, Washington University Medical School, St Louis, MO USA
| |
Collapse
|
37
|
Farmer AD, Strzelczyk A, Finisguerra A, Gourine AV, Gharabaghi A, Hasan A, Burger AM, Jaramillo AM, Mertens A, Majid A, Verkuil B, Badran BW, Ventura-Bort C, Gaul C, Beste C, Warren CM, Quintana DS, Hämmerer D, Freri E, Frangos E, Tobaldini E, Kaniusas E, Rosenow F, Capone F, Panetsos F, Ackland GL, Kaithwas G, O'Leary GH, Genheimer H, Jacobs HIL, Van Diest I, Schoenen J, Redgrave J, Fang J, Deuchars J, Széles JC, Thayer JF, More K, Vonck K, Steenbergen L, Vianna LC, McTeague LM, Ludwig M, Veldhuizen MG, De Couck M, Casazza M, Keute M, Bikson M, Andreatta M, D'Agostini M, Weymar M, Betts M, Prigge M, Kaess M, Roden M, Thai M, Schuster NM, Montano N, Hansen N, Kroemer NB, Rong P, Fischer R, Howland RH, Sclocco R, Sellaro R, Garcia RG, Bauer S, Gancheva S, Stavrakis S, Kampusch S, Deuchars SA, Wehner S, Laborde S, Usichenko T, Polak T, Zaehle T, Borges U, Teckentrup V, Jandackova VK, Napadow V, Koenig J. International Consensus Based Review and Recommendations for Minimum Reporting Standards in Research on Transcutaneous Vagus Nerve Stimulation (Version 2020). Front Hum Neurosci 2021; 14:568051. [PMID: 33854421 PMCID: PMC8040977 DOI: 10.3389/fnhum.2020.568051] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022] Open
Abstract
Given its non-invasive nature, there is increasing interest in the use of transcutaneous vagus nerve stimulation (tVNS) across basic, translational and clinical research. Contemporaneously, tVNS can be achieved by stimulating either the auricular branch or the cervical bundle of the vagus nerve, referred to as transcutaneous auricular vagus nerve stimulation(VNS) and transcutaneous cervical VNS, respectively. In order to advance the field in a systematic manner, studies using these technologies need to adequately report sufficient methodological detail to enable comparison of results between studies, replication of studies, as well as enhancing study participant safety. We systematically reviewed the existing tVNS literature to evaluate current reporting practices. Based on this review, and consensus among participating authors, we propose a set of minimal reporting items to guide future tVNS studies. The suggested items address specific technical aspects of the device and stimulation parameters. We also cover general recommendations including inclusion and exclusion criteria for participants, outcome parameters and the detailed reporting of side effects. Furthermore, we review strategies used to identify the optimal stimulation parameters for a given research setting and summarize ongoing developments in animal research with potential implications for the application of tVNS in humans. Finally, we discuss the potential of tVNS in future research as well as the associated challenges across several disciplines in research and clinical practice.
Collapse
Affiliation(s)
- Adam D. Farmer
- Department of Gastroenterology, University Hospitals of North Midlands NHS Trust, Stoke on Trent, United Kingdom
| | - Adam Strzelczyk
- Department of Neurology, Epilepsy Center Frankfurt Rhine-Main, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | | | - Alexander V. Gourine
- Department of Neuroscience, Physiology and Pharmacology, Centre for Cardiovascular and Metabolic Neuroscience, University College London, London, United Kingdom
| | - Alireza Gharabaghi
- Institute for Neuromodulation and Neurotechnology, University Hospital and University of Tuebingen, Tuebingen, Germany
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Augsburg, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Andreas M. Burger
- Laboratory for Biological Psychology, Faculty of Psychology and Educational Sciences, University of Leuven, Leuven, Belgium
| | | | - Ann Mertens
- Department of Neurology, Institute for Neuroscience, 4Brain, Ghent University Hospital, Gent, Belgium
| | - Arshad Majid
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Bart Verkuil
- Clinical Psychology and the Leiden Institute of Brain and Cognition, Leiden University, Leiden, Netherlands
| | - Bashar W. Badran
- Department of Psychiatry, Medical University of South Carolina, Charleston, SC, United States
| | - Carlos Ventura-Bort
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, Potsdam, Germany
| | - Charly Gaul
- Migraine and Headache Clinic Koenigstein, Königstein im Taunus, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany
| | | | - Daniel S. Quintana
- NORMENT, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Dorothea Hämmerer
- Medical Faculty, Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom
- Center for Behavioral Brain Sciences Magdeburg (CBBS), Otto-von-Guericke University, Magdeburg, Germany
| | - Elena Freri
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Eleni Frangos
- Pain and Integrative Neuroscience Branch, National Center for Complementary and Integrative Health, NIH, Bethesda, MD, United States
| | - Eleonora Tobaldini
- Department of Internal Medicine, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Eugenijus Kaniusas
- Institute of Electrodynamics, Microwave and Circuit Engineering, TU Wien, Vienna, Austria
- SzeleSTIM GmbH, Vienna, Austria
| | - Felix Rosenow
- Department of Neurology, Epilepsy Center Frankfurt Rhine-Main, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Fioravante Capone
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Fivos Panetsos
- Faculty of Biology and Faculty of Optics, Complutense University of Madrid and Institute for Health Research, San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - Gareth L. Ackland
- Translational Medicine and Therapeutics, Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Gaurav Kaithwas
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Georgia H. O'Leary
- Department of Psychiatry, Medical University of South Carolina, Charleston, SC, United States
| | - Hannah Genheimer
- Department of Biological Psychology, Clinical Psychology and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Heidi I. L. Jacobs
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, Netherlands
| | - Ilse Van Diest
- Research Group Health Psychology, Faculty of Psychology and Educational Sciences, University of Leuven, Leuven, Belgium
| | - Jean Schoenen
- Headache Research Unit, Department of Neurology-Citadelle Hospital, University of Liège, Liège, Belgium
| | - Jessica Redgrave
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Jiliang Fang
- Functional Imaging Lab, Department of Radiology, Guang An Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jim Deuchars
- School of Biomedical Science, Faculty of Biological Science, University of Leeds, Leeds, United Kingdom
| | - Jozsef C. Széles
- Division for Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Julian F. Thayer
- Department of Psychological Science, University of California, Irvine, Irvine, CA, United States
| | - Kaushik More
- Institute for Cognitive Neurology and Dementia Research, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Neuromodulatory Networks, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Kristl Vonck
- Department of Neurology, Institute for Neuroscience, 4Brain, Ghent University Hospital, Gent, Belgium
| | - Laura Steenbergen
- Clinical and Cognitive Psychology and the Leiden Institute of Brain and Cognition, Leiden University, Leiden, Netherlands
| | - Lauro C. Vianna
- NeuroV̇ASQ̇ - Integrative Physiology Laboratory, Faculty of Physical Education, University of Brasilia, Brasilia, Brazil
| | - Lisa M. McTeague
- Department of Psychiatry, Medical University of South Carolina, Charleston, SC, United States
| | - Mareike Ludwig
- Department of Anatomy, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Maria G. Veldhuizen
- Mental Health and Wellbeing Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marijke De Couck
- Faculty of Health Care, University College Odisee, Aalst, Belgium
- Division of Epileptology, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Marina Casazza
- Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | - Marius Keute
- Institute for Neuromodulation and Neurotechnology, University Hospital and University of Tuebingen, Tuebingen, Germany
| | - Marom Bikson
- Department of Biomedical Engineering, City College of New York, New York, NY, United States
| | - Marta Andreatta
- Department of Biological Psychology, Clinical Psychology and Psychotherapy, University of Würzburg, Würzburg, Germany
- Department of Psychology, Education and Child Studies, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Martina D'Agostini
- Research Group Health Psychology, Faculty of Psychology and Educational Sciences, University of Leuven, Leuven, Belgium
| | - Mathias Weymar
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, Potsdam, Germany
- Faculty of Health Sciences Brandenburg, University of Potsdam, Potsdam, Germany
| | - Matthew Betts
- Department of Anatomy, Faculty of Medicine, Mersin University, Mersin, Turkey
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany
| | - Matthias Prigge
- Neuromodulatory Networks, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Michael Kaess
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- Section for Translational Psychobiology in Child and Adolescent Psychiatry, Department of Child and Adolescent Psychiatry, Centre for Psychosocial Medicine, University of Heidelberg, Heidelberg, Germany
| | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Munich, Germany
| | - Michelle Thai
- Department of Psychology, College of Liberal Arts, University of Minnesota, Minneapolis, MN, United States
| | - Nathaniel M. Schuster
- Department of Anesthesiology, Center for Pain Medicine, University of California, San Diego Health System, La Jolla, CA, United States
| | - Nicola Montano
- Department of Internal Medicine, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University of Göttingen, Göttingen, Germany
- Laboratory of Systems Neuroscience and Imaging in Psychiatry (SNIPLab), University of Göttingen, Göttingen, Germany
| | - Nils B. Kroemer
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Peijing Rong
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rico Fischer
- Department of Psychology, University of Greifswald, Greifswald, Germany
| | - Robert H. Howland
- Department of Psychiatry, University of Pittsburgh School of Medicine, UPMC Western Psychiatric Hospital, Pittsburgh, PA, United States
| | - Roberta Sclocco
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
- Department of Radiology, Logan University, Chesterfield, MO, United States
| | - Roberta Sellaro
- Cognitive Psychology Unit, Institute of Psychology, Leiden University, Leiden, Netherlands
- Leiden Institute for Brain and Cognition, Leiden, Netherlands
- Department of Developmental Psychology and Socialisation, University of Padova, Padova, Italy
| | - Ronald G. Garcia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sebastian Bauer
- Department of Neurology, Epilepsy Center Frankfurt Rhine-Main, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Sofiya Gancheva
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Stavros Stavrakis
- Faculty of Biological Science, School of Biomedical Science, University of Leeds, Leeds, United Kingdom
| | - Stefan Kampusch
- Institute of Electrodynamics, Microwave and Circuit Engineering, TU Wien, Vienna, Austria
- SzeleSTIM GmbH, Vienna, Austria
| | - Susan A. Deuchars
- School of Biomedical Science, Faculty of Biological Science, University of Leeds, Leeds, United Kingdom
| | - Sven Wehner
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Sylvain Laborde
- Department of Performance Psychology, Institute of Psychology, Deutsche Sporthochschule, Köln, Germany
| | - Taras Usichenko
- Department of Anesthesiology, University Medicine Greifswald, Greifswald, Germany
- Department of Anesthesia, McMaster University, Hamilton, ON, Canada
| | - Thomas Polak
- Laboratory of Functional Neurovascular Diagnostics, AG Early Diagnosis of Dementia, Department of Psychiatry, Psychosomatics and Psychotherapy, University Clinic Würzburg, Würzburg, Germany
| | - Tino Zaehle
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Uirassu Borges
- Department of Performance Psychology, Institute of Psychology, Deutsche Sporthochschule, Köln, Germany
- Department of Social and Health Psychology, Institute of Psychology, Deutsche Sporthochschule, Köln, Germany
| | - Vanessa Teckentrup
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Vera K. Jandackova
- Department of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
- Department of Human Movement Studies, Faculty of Education, University of Ostrava, Ostrava, Czechia
| | - Vitaly Napadow
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
- Department of Radiology, Logan University, Chesterfield, MO, United States
| | - Julian Koenig
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- Section for Experimental Child and Adolescent Psychiatry, Department of Child and Adolescent Psychiatry, Centre for Psychosocial Medicine, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
38
|
Brown BL, Zalla RM, Shepard CT, Howard RM, Kopechek JA, Magnuson DSK, Whittemore SR. Dual-Viral Transduction Utilizing Highly Efficient Retrograde Lentivirus Improves Labeling of Long Propriospinal Neurons. Front Neuroanat 2021; 15:635921. [PMID: 33828464 PMCID: PMC8019739 DOI: 10.3389/fnana.2021.635921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/12/2021] [Indexed: 11/13/2022] Open
Abstract
The nervous system coordinates pathways and circuits to process sensory information and govern motor behaviors. Mapping these pathways is important to further understand the connectivity throughout the nervous system and is vital for developing treatments for neuronal diseases and disorders. We targeted long ascending propriospinal neurons (LAPNs) in the rat spinal cord utilizing Fluoro-Ruby (FR) [10kD rhodamine dextran amine (RDA)], and two dual-viral systems. Dual-viral tracing utilizing a retrograde adeno-associated virus (retroAAV), which confers robust labeling in the brain, resulted in a small number of LAPNs being labeled, but dual-viral tracing using a highly efficient retrograde (HiRet) lentivirus provided robust labeling similar to FR. Additionally, dual-viral tracing with HiRet lentivirus and tracing with FR may preferentially label different subpopulations of LAPNs. These data demonstrate that dual-viral tracing in the spinal cord employing a HiRet lentivirus provides robust and specific labeling of LAPNs and emphasizes the need to empirically optimize viral systems to target specific neuronal population(s).
Collapse
Affiliation(s)
- Brandon L Brown
- Interdisciplinary Program in Translational Neuroscience, University of Louisville, Louisville, KY, United States.,Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, United States.,Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, United States
| | - Rachel M Zalla
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, United States.,Department of Bioengineering, J.B. Speed School of Engineering, University of Louisville, Louisville, KY, United States
| | - Courtney T Shepard
- Interdisciplinary Program in Translational Neuroscience, University of Louisville, Louisville, KY, United States.,Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, United States.,Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, United States
| | - Russell M Howard
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, United States.,Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, KY, United States
| | - Jonathan A Kopechek
- Department of Bioengineering, J.B. Speed School of Engineering, University of Louisville, Louisville, KY, United States
| | - David S K Magnuson
- Interdisciplinary Program in Translational Neuroscience, University of Louisville, Louisville, KY, United States.,Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, United States.,Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, United States.,Department of Bioengineering, J.B. Speed School of Engineering, University of Louisville, Louisville, KY, United States.,Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, KY, United States
| | - Scott R Whittemore
- Interdisciplinary Program in Translational Neuroscience, University of Louisville, Louisville, KY, United States.,Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, United States.,Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, United States.,Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, KY, United States
| |
Collapse
|
39
|
Chadney OMT, Blankvoort S, Grimstvedt JS, Utz A, Kentros CG. Multiplexing viral approaches to the study of the neuronal circuits. J Neurosci Methods 2021; 357:109142. [PMID: 33753126 DOI: 10.1016/j.jneumeth.2021.109142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 02/27/2021] [Accepted: 03/10/2021] [Indexed: 12/16/2022]
Abstract
Neural circuits are composed of multitudes of elaborately interconnected cell types. Understanding neural circuit function requires not only cell-specific knowledge of connectivity, but the ability to record and manipulate distinct cell types independently. Recent advances in viral vectors promise the requisite specificity to perform true "circuit-breaking" experiments. However, such new avenues of multiplexed, cell-specific investigation raise new technical issues: one must ensure that both the viral vectors and their transgene payloads do not overlap with each other in both an anatomical and a functional sense. This review describes benefits and issues regarding the use of viral vectors to analyse the function of neural circuits and provides a resource for the design and implementation of such multiplexing experiments.
Collapse
Affiliation(s)
- Oscar M T Chadney
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway.
| | - Stefan Blankvoort
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway
| | - Joachim S Grimstvedt
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway
| | - Annika Utz
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway
| | - Clifford G Kentros
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway.
| |
Collapse
|
40
|
Cassel JC, Pereira de Vasconcelos A. Routes of the thalamus through the history of neuroanatomy. Neurosci Biobehav Rev 2021; 125:442-465. [PMID: 33676963 DOI: 10.1016/j.neubiorev.2021.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/19/2021] [Accepted: 03/01/2021] [Indexed: 12/24/2022]
Abstract
The most distant roots of neuroanatomy trace back to antiquity, with the first human dissections, but no document which would identify the thalamus as a brain structure has reached us. Claudius Galenus (Galen) gave to the thalamus the name 'thalamus nervorum opticorum', but later on, other names were used (e.g., anchae, or buttocks-like). In 1543, Andreas Vesalius provided the first quality illustrations of the thalamus. During the 19th century, tissue staining techniques and ablative studies contributed to the breakdown of the thalamus into subregions and nuclei. The next step was taken using radiomarkers to identify connections in the absence of lesions. Anterograde and retrograde tracing methods arose in the late 1960s, supporting extension, revision, or confirmation of previously established knowledge. The use of the first viral tracers introduced a new methodological breakthrough in the mid-1970s. Another important step was supported by advances in neuroimaging of the thalamus in the 21th century. The current review follows the history of the thalamus through these technical revolutions from Antiquity to the present day.
Collapse
Affiliation(s)
- Jean-Christophe Cassel
- Laboratoire de Neurosciences Cognitives et Adaptatives, Université de Strasbourg, F-67000 Strasbourg, France; LNCA, UMR 7364 - CNRS, F-67000 Strasbourg, France.
| | - Anne Pereira de Vasconcelos
- Laboratoire de Neurosciences Cognitives et Adaptatives, Université de Strasbourg, F-67000 Strasbourg, France; LNCA, UMR 7364 - CNRS, F-67000 Strasbourg, France
| |
Collapse
|
41
|
Nonhuman Primate Optogenetics: Current Status and Future Prospects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:345-358. [PMID: 33398825 DOI: 10.1007/978-981-15-8763-4_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonhuman primates (NHPs) have widely and crucially been utilized as model animals for understanding various higher brain functions and neurological disorders since their behavioral actions mimic both normal and disease states in humans. To know about how such behaviors emerge from the functions and dysfunctions of complex neural networks, it is essential to define the role of a particular pathway or neuron-type constituting these networks. Optogenetics is a potential technique that enables analyses of network functions. However, because of the large size of the NHP brain and the difficulty in creating genetically modified animal models, this technique is currently still hard to apply effectively and efficiently to NHP neuroscience. In this article, we focus on the issues that should be overcome for the development of NHP optogenetics, with special reference to the gene introduction strategy. We review the recent breakthroughs that have been made in NHP optogenetics to address these issues and discuss future prospects regarding more effective and efficient approaches to successful optogenetic manipulation in NHPs.
Collapse
|
42
|
Vezoli J, Magrou L, Goebel R, Wang XJ, Knoblauch K, Vinck M, Kennedy H. Cortical hierarchy, dual counterstream architecture and the importance of top-down generative networks. Neuroimage 2021; 225:117479. [PMID: 33099005 PMCID: PMC8244994 DOI: 10.1016/j.neuroimage.2020.117479] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/29/2020] [Accepted: 10/15/2020] [Indexed: 12/18/2022] Open
Abstract
Hierarchy is a major organizational principle of the cortex and underscores modern computational theories of cortical function. The local microcircuit amplifies long-distance inter-areal input, which show distance-dependent changes in their laminar profiles. Statistical modeling of these changes in laminar profiles demonstrates that inputs from multiple hierarchical levels to their target areas show remarkable consistency, allowing the construction of a cortical hierarchy based on a principle of hierarchical distance. The statistical modeling that is applied to structure can also be applied to laminar differences in the oscillatory coherence between areas thereby determining a functional hierarchy of the cortex. Close examination of the anatomy of inter-areal connectivity reveals a dual counterstream architecture with well-defined distance-dependent feedback and feedforward pathways in both the supra- and infragranular layers, suggesting a multiplicity of feedback pathways with well-defined functional properties. These findings are consistent with feedback connections providing a generative network involved in a wide range of cognitive functions. A dynamical model constrained by connectivity data sheds insight into the experimentally observed signatures of frequency-dependent Granger causality for feedforward versus feedback signaling. Concerted experiments capitalizing on recent technical advances and combining tract-tracing, high-resolution fMRI, optogenetics and mathematical modeling hold the promise of a much improved understanding of lamina-constrained mechanisms of neural computation and cognition. However, because inter-areal interactions involve cortical layers that have been the target of important evolutionary changes in the primate lineage, these investigations will need to include human and non-human primate comparisons.
Collapse
Affiliation(s)
- Julien Vezoli
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt, Germany
| | - Loïc Magrou
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Rainer Goebel
- Faculty of Psychology and Neuroscience, Department of Cognitive Neuroscience, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| | - Xiao-Jing Wang
- Center for Neural Science, New York University (NYU), New York, NY 10003, USA
| | - Kenneth Knoblauch
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Martin Vinck
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt, Germany.
| | - Henry Kennedy
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France; Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences (CAS) Key Laboratory of Primate Neurobiology, CAS, Shanghai 200031, China.
| |
Collapse
|
43
|
In vivo cell tracking with viral vector mediated genetic labeling. J Neurosci Methods 2020; 350:109021. [PMID: 33316318 DOI: 10.1016/j.jneumeth.2020.109021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022]
Abstract
Cell tracking is a useful technique to monitor specific cell populations for their morphology, development, proliferation, migration, interaction, function, and other properties, both in vitro and in vivo. Using different materials and methodologies to label the target cells directly or indirectly, the dynamic biological processes in living organisms can be visualized with appropriate detection techniques. Viruses, with the unique ability to deliver exogenous genes into host cells, have been used as vectors to mediate gene transfer. Genetic labeling of target cells by viral vectors endows the cells to express reporter genes with high efficiency and specificity. In conjunction with corresponding imaging techniques, cells labeled with different genetic reporters mediated by different viral vectors can be monitored across spatial and temporal scales to fulfill various purposes and address different questions. In the present review, we introduce the basic principle of viral vectors in cell tracking and highlight the examples of cell tracking in various research areas.
Collapse
|
44
|
Cheffer A, Flitsch LJ, Krutenko T, Röderer P, Sokhranyaeva L, Iefremova V, Hajo M, Peitz M, Schwarz MK, Brüstle O. Human stem cell-based models for studying autism spectrum disorder-related neuronal dysfunction. Mol Autism 2020; 11:99. [PMID: 33308283 PMCID: PMC7733257 DOI: 10.1186/s13229-020-00383-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
The controlled differentiation of pluripotent stem cells (PSCs) into neurons and glia offers a unique opportunity to study early stages of human central nervous system development under controlled conditions in vitro. With the advent of cell reprogramming and the possibility to generate induced pluripotent stem cells (iPSCs) from any individual in a scalable manner, these studies can be extended to a disease- and patient-specific level. Autism spectrum disorder (ASD) is considered a neurodevelopmental disorder, with substantial evidence pointing to early alterations in neurogenesis and network formation as key pathogenic drivers. For that reason, ASD represents an ideal candidate for stem cell-based disease modeling. Here, we provide a concise review on recent advances in the field of human iPSC-based modeling of syndromic and non-syndromic forms of ASD, with a particular focus on studies addressing neuronal dysfunction and altered connectivity. We further discuss recent efforts to translate stem cell-based disease modeling to 3D via brain organoid and cell transplantation approaches, which enable the investigation of disease mechanisms in a tissue-like context. Finally, we describe advanced tools facilitating the assessment of altered neuronal function, comment on the relevance of iPSC-based models for the assessment of pharmaceutical therapies and outline potential future routes in stem cell-based ASD research.
Collapse
Affiliation(s)
- Arquimedes Cheffer
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Lea Jessica Flitsch
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Tamara Krutenko
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Pascal Röderer
- Life & Brain GmbH, Platform Cellomics, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Liubov Sokhranyaeva
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Vira Iefremova
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Mohamad Hajo
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
- Life & Brain GmbH, Platform Cellomics, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
- Cell Programming Core Facility, University of Bonn Medical Faculty, Bonn, Germany
| | - Martin Karl Schwarz
- Life & Brain GmbH, Platform Cellomics, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany.
| |
Collapse
|
45
|
Albaugh DL, Smith Y, Galvan A. Comparative analyses of transgene expression patterns after intra-striatal injections of rAAV2-retro in rats and rhesus monkeys: A light and electron microscopic study. Eur J Neurosci 2020; 52:4824-4839. [PMID: 33113247 PMCID: PMC7902345 DOI: 10.1111/ejn.15027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/30/2020] [Accepted: 10/21/2020] [Indexed: 12/18/2022]
Abstract
Retrogradely-transducing viral vectors are versatile tools for anatomical and functional interrogations of neural circuits. These vectors can be applied in nonhuman primates (NHPs), powerful model species for neuroscientific studies with limited genetic tractability, but limited data are available regarding the tropism and transgene expression patterns of such viruses after injections in NHP brains. Consequently, NHP researchers must often rely on related data available from other species for experimental planning. To evaluate the suitability of rAAV2-retro in the NHP basal ganglia, we studied the transgene expression patterns at the light and electron microscope level after injections of rAAV2-retro vector encoding the opsin Jaws conjugated to a green fluorescent protein (GFP) in the putamen of rhesus macaques. For inter-species comparison, we injected the same vector in the rat dorsal striatum. In both species, GFP expression was observed in numerous cortical and subcortical regions with known striatal projections. However, important inter-species differences in pathway transduction were seen, including labeling of the intralaminar thalamostriatal projection in rats, but not monkeys. Electron microscopic ultrastructural observations within the basal ganglia revealed GFP labeling in both postsynaptic dendrites and presynaptic axonal terminals; the latter likely derived from anterograde transgene transport in neurons that project to the striatum, and from collaterals of these neurons. Our results suggest that certain neural pathways may be refractory to transduction by retrograde vectors in a species-specific manner, highlighting the need for caution when determining the suitability of a retrograde vector for NHP studies based solely on rodent data.
Collapse
Affiliation(s)
- Daniel L Albaugh
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Udall Center of Excellence for Parkinson's Disease Research, Atlanta, GA, USA
| | - Yoland Smith
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Udall Center of Excellence for Parkinson's Disease Research, Atlanta, GA, USA
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Adriana Galvan
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Udall Center of Excellence for Parkinson's Disease Research, Atlanta, GA, USA
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
46
|
Luchicchi A, Pattij T, Viaña JNM, de Kloet S, Marchant N. Tracing goes viral: Viruses that introduce expression of fluorescent proteins in chemically-specific neurons. J Neurosci Methods 2020; 348:109004. [PMID: 33242528 DOI: 10.1016/j.jneumeth.2020.109004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 12/26/2022]
Abstract
Over the last century, there has been great progress in understanding how the brain works. In particular, the last two decades have been crucial in gaining more awareness over the complex functioning of neurotransmitter systems. The use of viral vectors in neuroscience has been pivotal for such development. Exploiting the properties of viral particles, modifying them according to the research needs, and making them target chemically-specific neurons, techniques such as optogenetics and chemogenetics have been developed, which could lead to a giant step toward gene therapy for brain disorders. In this review, we aim to provide an overview of some of the most widely used viral techniques in neuroscience. We will discuss advantages and disadvantages of these methods. In particular, attention is dedicated to the pivotal role played by the introduction of adeno-associated virus and the retrograde tracer canine-associated-2 Cre virus in order to achieve optimal visualization, and interrogation, of chemically-specific neuronal populations and their projections.
Collapse
Affiliation(s)
- Antonio Luchicchi
- Department of Anatomy and Neurosciences, Amsterdam UMC, VU University Medical Center, de Boelelaan 1108, 1081HZ, Amsterdam, the Netherlands.
| | - Tommy Pattij
- Department of Anatomy and Neurosciences, Amsterdam UMC, VU University Medical Center, de Boelelaan 1108, 1081HZ, Amsterdam, the Netherlands
| | - John Noel M Viaña
- Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, de Boelelaan 1085, 1081HZ, Amsterdam, the Netherlands; Australian National Centre for the Public Awareness of Science, ANU College of Science, The Australian National University, Linnaeus Way, Acton, ACT 2601, Australia
| | - Sybren de Kloet
- Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, de Boelelaan 1085, 1081HZ, Amsterdam, the Netherlands
| | - Nathan Marchant
- Department of Anatomy and Neurosciences, Amsterdam UMC, VU University Medical Center, de Boelelaan 1108, 1081HZ, Amsterdam, the Netherlands
| |
Collapse
|
47
|
Lunde A, Glover JC. A versatile toolbox for semi-automatic cell-by-cell object-based colocalization analysis. Sci Rep 2020; 10:19027. [PMID: 33149236 PMCID: PMC7643144 DOI: 10.1038/s41598-020-75835-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/14/2020] [Indexed: 11/09/2022] Open
Abstract
Differential fluorescence labeling and multi-fluorescence imaging followed by colocalization analysis is commonly used to investigate cellular heterogeneity in situ. This is particularly important when investigating the biology of tissues with diverse cell types. Object-based colocalization analysis (OBCA) tools can employ automatic approaches, which are sensitive to errors in cell segmentation, or manual approaches, which can be impractical and tedious. Here, we present a novel set of tools for OBCA using a semi-automatic approach, consisting of two ImageJ plugins, a Microsoft Excel macro, and a MATLAB script. One ImageJ plugin enables customizable processing of multichannel 3D images for enhanced visualization of features relevant to OBCA, and another enables semi-automatic colocalization quantification. The Excel macro and the MATLAB script enable data organization and 3D visualization of object data across image series. The tools are well suited for experiments involving complex and large image data sets, and can be used in combination or as individual components, allowing flexible, efficient and accurate OBCA. Here we demonstrate their utility in immunohistochemical analyses of the developing central nervous system, which is characterized by complexity in the number and distribution of cell types, and by high cell packing densities, which can both create challenging situations for OBCA.
Collapse
Affiliation(s)
- Anders Lunde
- Laboratory of Neural Development and Optical Recording (NDEVOR), Division of Physiology, Department of Molecular Medicine, University of Oslo, Blindern, 1105, Oslo, Norway
| | - Joel C Glover
- Laboratory of Neural Development and Optical Recording (NDEVOR), Division of Physiology, Department of Molecular Medicine, University of Oslo, Blindern, 1105, Oslo, Norway.
| |
Collapse
|
48
|
Cushnie AK, El-Nahal HG, Bohlen MO, May PJ, Basso MA, Grimaldi P, Wang MZ, de Velasco Ezequiel MF, Sommer MA, Heilbronner SR. Using rAAV2-retro in rhesus macaques: Promise and caveats for circuit manipulation. J Neurosci Methods 2020; 345:108859. [PMID: 32668316 PMCID: PMC7539563 DOI: 10.1016/j.jneumeth.2020.108859] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/01/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recent genetic technologies such as opto- and chemogenetics allow for the manipulation of brain circuits with unprecedented precision. Most studies employing these techniques have been undertaken in rodents, but a more human-homologous model for studying the brain is the nonhuman primate (NHP). Optimizing viral delivery of transgenes encoding actuator proteins could revolutionize the way we study neuronal circuits in NHPs. NEW METHOD: rAAV2-retro, a popular new capsid variant, produces robust retrograde labeling in rodents. Whether rAAV2-retro's highly efficient retrograde transport would translate to NHPs was unknown. Here, we characterized the anatomical distribution of labeling following injections of rAAV2-retro encoding opsins or DREADDs in the cortico-basal ganglia and oculomotor circuits of rhesus macaques. RESULTS rAAV2-retro injections in striatum, frontal eye field, and superior colliculus produced local labeling at injection sites and robust retrograde labeling in many afferent regions. In every case, however, a few brain regions with well-established projections to the injected structure lacked retrogradely labeled cells. We also observed robust terminal field labeling in downstream structures. COMPARISON WITH EXISTING METHOD(S) Patterns of labeling were similar to those obtained with traditional tract-tracers, except for some afferent labeling that was noticeably absent. CONCLUSIONS rAAV2-retro promises to be useful for circuit manipulation via retrograde transduction in NHPs, but caveats were revealed by our findings. Some afferently connected regions lacked retrogradely labeled cells, showed robust axon terminal labeling, or both. This highlights the importance of anatomically characterizing rAAV2-retro's expression in target circuits in NHPs before moving to manipulation studies.
Collapse
Affiliation(s)
- Adriana K Cushnie
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| | - Hala G El-Nahal
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States
| | - Martin O Bohlen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States
| | - Paul J May
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, 39216, United States
| | - Michele A Basso
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences and Neurobiology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, Univ. of California Los Angeles, Los Angeles, CA 90095, United States
| | - Piercesare Grimaldi
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences and Neurobiology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, Univ. of California Los Angeles, Los Angeles, CA 90095, United States
| | - Maya Zhe Wang
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| | | | - Marc A Sommer
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States; Department of Neurobiology, Duke University School of Medicine, Durham, NC 27708, United States; Center for Cognitive Neuroscience, Duke University, Durham, NC 27708, United States
| | - Sarah R Heilbronner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States.
| |
Collapse
|
49
|
Functional interrogation of neural circuits with virally transmitted optogenetic tools. J Neurosci Methods 2020; 345:108905. [PMID: 32795553 DOI: 10.1016/j.jneumeth.2020.108905] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022]
Abstract
The vertebrate brain comprises a plethora of cell types connected by intertwined pathways. Optogenetics enriches the neuroscientific tool set for disentangling these neuronal circuits in a manner which exceeds the spatio-temporal precision of previously existing techniques. Technically, optogenetics can be divided in three types of optical and genetic combinations: (1) it is primarily understood as the manipulation of the activity of genetically modified cells (typically neurons) with light, i.e. optical actuators. (2) A second combination refers to visualizing the activity of genetically modified cells (again typically neurons), i.e. optical sensors. (3) A completely different interpretation of optogenetics refers to the light activated expression of a genetically induced construct. Here, we focus on the first two types of optogenetics, i.e. the optical actuators and sensors in an attempt to give an overview into the topic. We first cover methods to express opsins into neurons and introduce strategies of targeting specific neuronal populations in different animal species. We then summarize combinations of optogenetics with behavioral read out and neuronal imaging. Finally, we give an overview of the current state-of-the-art and an outlook on future perspectives.
Collapse
|
50
|
Xu X, Holmes TC, Luo MH, Beier KT, Horwitz GD, Zhao F, Zeng W, Hui M, Semler BL, Sandri-Goldin RM. Viral Vectors for Neural Circuit Mapping and Recent Advances in Trans-synaptic Anterograde Tracers. Neuron 2020; 107:1029-1047. [PMID: 32755550 DOI: 10.1016/j.neuron.2020.07.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/23/2020] [Accepted: 07/12/2020] [Indexed: 12/17/2022]
Abstract
Viral tracers are important tools for neuroanatomical mapping and genetic payload delivery. Genetically modified viruses allow for cell-type-specific targeting and overcome many limitations of non-viral tracers. Here, we summarize the viruses that have been developed for neural circuit mapping, and we provide a primer on currently applied anterograde and retrograde viral tracers with practical guidance on experimental uses. We also discuss and highlight key technical and conceptual considerations for developing new safer and more effective anterograde trans-synaptic viral vectors for neural circuit analysis in multiple species.
Collapse
Affiliation(s)
- Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697-1275, USA; Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697-4025, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697-2715, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA.
| | - Todd C Holmes
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697-4560, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Min-Hua Luo
- State Key Laboratory of Virology, Wuhan Institute of Virology, CAS Center for Excellence in Brain Science, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697-4560, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Gregory D Horwitz
- The Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA; Department of Physiology & Biophysics, University of Washington, Seattle, WA 98195, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Fei Zhao
- School of Basic Medical Sciences, Capital Medical University, Beijing 102206, China; Chinese Institute for Brain Research (CIBR), Beijing 102206, China
| | - Wenbo Zeng
- State Key Laboratory of Virology, Wuhan Institute of Virology, CAS Center for Excellence in Brain Science, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697-4560, USA
| | - Bert L Semler
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697-4025, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Rozanne M Sandri-Goldin
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697-4025, USA; The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|