1
|
Neřoldová M, Stuchlík A. Chemogenetic Tools and their Use in Studies of Neuropsychiatric Disorders. Physiol Res 2024; 73:S449-S470. [PMID: 38957949 PMCID: PMC11412350 DOI: 10.33549/physiolres.935401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Chemogenetics is a newly developed set of tools that allow for selective manipulation of cell activity. They consist of a receptor mutated irresponsive to endogenous ligands and a synthetic ligand that does not interact with the wild-type receptors. Many different types of these receptors and their respective ligands for inhibiting or excitating neuronal subpopulations were designed in the past few decades. It has been mainly the G-protein coupled receptors (GPCRs) selectively responding to clozapine-N-oxide (CNO), namely Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), that have been employed in research. Chemogenetics offers great possibilities since the activity of the receptors is reversible, inducible on demand by the ligand, and non-invasive. Also, specific groups or types of neurons can be selectively manipulated thanks to the delivery by viral vectors. The effect of the chemogenetic receptors on neurons lasts longer, and even chronic activation can be achieved. That can be useful for behavioral testing. The great advantage of chemogenetic tools is especially apparent in research on brain diseases since they can manipulate whole neuronal circuits and connections between different brain areas. Many psychiatric or other brain diseases revolve around the dysfunction of specific brain networks. Therefore, chemogenetics presents a powerful tool for investigating the underlying mechanisms causing the disease and revealing the link between the circuit dysfunction and the behavioral or cognitive symptoms observed in patients. It could also contribute to the development of more effective treatments.
Collapse
Affiliation(s)
- M Neřoldová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic. E-mail:
| | | |
Collapse
|
2
|
Zhu J, Yang Y, Du L, Yang W, Yang Y, Yi T, Maoying Q, Chu Y, Wang Y, Mi W. A regulatory role of the medial septum in the chloroquine-induced acute itch through local GABAergic system and GABAergic pathway to the anterior cingulate cortex. Biochem Biophys Res Commun 2024; 721:150145. [PMID: 38795633 DOI: 10.1016/j.bbrc.2024.150145] [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: 03/11/2024] [Revised: 05/04/2024] [Accepted: 05/18/2024] [Indexed: 05/28/2024]
Abstract
Itch, a common somatic sensation, serves as a crucial protective system. Recent studies have unraveled the neural mechanisms of itch at peripheral, spinal cord as well as cerebral levels. However, a comprehensive understanding of the central mechanism governing itch transmission and regulation remains elusive. Here, we report the role of the medial septum (MS), an integral component of the basal forebrain, in modulating the acute itch processing. The increases in c-Fos+ neurons and calcium signals within the MS during acute itch processing were observed. Pharmacogenetic activation manipulation of global MS neurons suppressed the scratching behaviors induced by chloroquine or compound 48/80. Microinjection of GABA into the MS or pharmacogenetic inhibition of non-GABAergic neurons markedly suppressed chloroquine-induced scratching behaviors. Pharmacogenetic activation of the MS-ACC GABAergic pathway attenuated chloroquine-induced acute itch. Hence, our findings reveal that MS has a regulatory role in the chloroquine-induced acute itch through local increased GABA to inhibit non-GABAergic neurons and the activation of MS-ACC GABAergic pathway.
Collapse
Affiliation(s)
- Jianyu Zhu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yayue Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lixia Du
- Department of Biochemistry, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yachen Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ting Yi
- Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangdong, 510006, China
| | - Qiliang Maoying
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuxia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Wenli Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Griffin H, Hanson J, Phelan KD, Baldini G. MC4R Localizes at Excitatory Postsynaptic and Peri-Postsynaptic Sites of Hypothalamic Neurons in Primary Culture. Cells 2024; 13:1235. [PMID: 39120267 PMCID: PMC11311852 DOI: 10.3390/cells13151235] [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: 04/28/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Abstract
The melanocortin-4 receptor (MC4R) is a G protein-coupled receptor (GPCR) that is expressed in several brain locations encompassing the hypothalamus and the brainstem, where the receptor controls several body functions, including metabolism. In a well-defined pathway to decrease appetite, hypothalamic proopiomelanocortin (POMC) neurons localized in the arcuate nucleus (Arc) project to MC4R neurons in the paraventricular nuclei (PVN) to release the natural MC4R agonist α-melanocyte-stimulating hormone (α-MSH). Arc neurons also project excitatory glutamatergic fibers to the MC4R neurons in the PVN for a fast synaptic transmission to regulate a satiety pathway potentiated by α-MSH. By using super-resolution microscopy, we found that in hypothalamic neurons in a primary culture, postsynaptic density protein 95 (PSD95) colocalizes with GluN1, a subunit of the ionotropic N-methyl-D-aspartate receptor (NMDAR). Thus, hypothalamic neurons form excitatory postsynaptic specializations. To study the MC4R distribution at these sites, tagged HA-MC4R under the synapsin promoter was expressed in neurons by adeno-associated virus (AAV) gene transduction. HA-MC4R immunofluorescence peaked at the center and in proximity to the PSD95- and NMDAR-expressing sites. These data provide morphological evidence that MC4R localizes together with glutamate receptors at postsynaptic and peri-postsynaptic sites.
Collapse
Affiliation(s)
- Haven Griffin
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| | - Jude Hanson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| | - Kevin D. Phelan
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| |
Collapse
|
4
|
Kojima L, Seiriki K, Rokujo H, Nakazawa T, Kasai A, Hashimoto H. Optimization of AAV vectors for transactivator-regulated enhanced gene expression within targeted neuronal populations. iScience 2024; 27:109878. [PMID: 38799556 PMCID: PMC11126825 DOI: 10.1016/j.isci.2024.109878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 03/03/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Adeno-associated virus (AAV) vectors are potential tools for cell-type-selective gene delivery to the central nervous system. Although cell-type-specific enhancers and promoters have been identified for AAV systems, there is limited information regarding the effects of AAV genomic components on the selectivity and efficiency of gene expression. Here, we offer an alternative strategy to provide specific and efficient gene delivery to a targeted neuronal population by optimizing recombinant AAV genomic components, named TAREGET (TransActivator-Regulated Enhanced Gene Expression within Targeted neuronal populations). We established this strategy in oxytocinergic neurons and showed that the TAREGET enabled sufficient gene expression to label long-projecting axons in wild-type mice. Its application to other cell types, including serotonergic and dopaminergic neurons, was also demonstrated. These results demonstrate that optimization of AAV expression cassettes can improve the specificity and efficiency of cell-type-specific gene expression and that TAREGET can renew previously established cell-type-specific promoters with improved performance.
Collapse
Affiliation(s)
- Leo Kojima
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kaoru Seiriki
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroki Rokujo
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takanobu Nakazawa
- Department of Bioscience, Tokyo University of Agriculture, Setagaya-ku, Tokyo 156-8502, Japan
| | - Atsushi Kasai
- Systems Neuropharmacology, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka 565-0871, Japan
- Institute for Datability Science, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Molecular Pharmaceutical Sciences, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
5
|
Chen H, Xie Y, Zhang M, Huang J, Jiang W, Zhang R, Li C, Du X, Chen H, Nie Q, Liang S, Tan Q, Yang J, Jin M, Huang S, Kuang L, Su N, Qi H, Luo X, Xu X, Deng C, Chen L, Luo F. An Hsp70 promoter-based mouse for heat shock-induced gene modulation. J Mol Med (Berl) 2024; 102:693-707. [PMID: 38492027 DOI: 10.1007/s00109-024-02433-9] [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: 01/04/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Physical therapy is extensively employed in clinical settings. Nevertheless, the absence of suitable animal models has resulted in an incomplete understanding of the in vivo mechanisms and cellular distribution that respond to physical stimuli. The objective of this research was to create a mouse model capable of indicating the cells affected by physical stimuli. In this study, we successfully established a mouse line based on the heat shock protein 70 (Hsp70) promoter, wherein the expression of CreERT2 can be induced by physical stimuli. Following stimulation of the mouse tail, ear, or cultured calvarias with heat shock (generated by heating, ultrasound, or laser), a distinct Cre-mediated excision was observed in cells stimulated by these physical factors with minimal occurrence of leaky reporter expression. The application of heat shock to Hsp70-CreERT2; FGFR2-P253R double transgenic mice or Hsp70-CreERT2 mice infected with AAV-BMP4 at calvarias induced the activation of Cre-dependent mutant FGFR2-P253R or BMP4 respectively, thereby facilitating the premature closure of cranial sutures or the repair of calvarial defects. This novel mouse line holds significant potential for investigating the underlying mechanisms of physical therapy, tissue repair and regeneration, lineage tracing, and targeted modulation of gene expression of cells in local tissue stimulated by physical factor at the interested time points. KEY MESSAGES: In the study, an Hsp70-CreERT2 transgenic mouse was generated for heat shock-induced gene modulation. Heat shock, ultrasound, and laser stimulation effectively activated Cre expression in Hsp70-CreERT2; reporter mice, which leads to deletion of floxed DNA sequence in the tail, ear, and cultured calvaria tissues of mice. Local laser stimuli on cultured calvarias effectively induce Fgfr2-P253R expression in Hsp70-mTmG-Fgfr2-P253R mice and result in accelerated premature closure of cranial suture. Heat shock activated AAV9-FLEX-BMP4 expression and subsequently promoted the repair of calvarial defect of Hsp70-CreERT2; Rosa26-mTmG mice.
Collapse
Affiliation(s)
- Hangang Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yangli Xie
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Mei Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Junlan Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Wanling Jiang
- Department of Chinese Medicine Rehabilitation, Chongqing Emergency Medical Center, Chongqing University Central Hospital), Chongqing, 400042, China
| | - Ruobin Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Can Li
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiaolan Du
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Hua Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Qiang Nie
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Sen Liang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Qiaoyan Tan
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Jing Yang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Min Jin
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Shuo Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Liang Kuang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Nan Su
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Huabing Qi
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Xiaoqing Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiaoling Xu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chuxia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lin Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
| | - Fengtao Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
6
|
Stranford DM, Simons LM, Berman KE, Cheng L, DiBiase BN, Hung ME, Lucks JB, Hultquist JF, Leonard JN. Genetically encoding multiple functionalities into extracellular vesicles for the targeted delivery of biologics to T cells. Nat Biomed Eng 2024; 8:397-414. [PMID: 38012307 PMCID: PMC11088532 DOI: 10.1038/s41551-023-01142-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 10/20/2023] [Indexed: 11/29/2023]
Abstract
The genetic modification of T cells has advanced cellular immunotherapies, yet the delivery of biologics specifically to T cells remains challenging. Here we report a suite of methods for the genetic engineering of cells to produce extracellular vesicles (EVs)-which naturally encapsulate and transfer proteins and nucleic acids between cells-for the targeted delivery of biologics to T cells without the need for chemical modifications. Specifically, the engineered cells secreted EVs that actively loaded protein cargo via a protein tag and that displayed high-affinity T-cell-targeting domains and fusogenic glycoproteins. We validated the methods by engineering EVs that delivered Cas9-single-guide-RNA complexes to ablate the gene encoding the C-X-C chemokine co-receptor type 4 in primary human CD4+ T cells. The strategy is amenable to the targeted delivery of biologics to other cell types.
Collapse
Affiliation(s)
- Devin M Stranford
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
| | - Lacy M Simons
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL, USA
| | - Katherine E Berman
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
- Interdisciplinary Biological Sciences Training Program, Northwestern University, Evanston, IL, USA
| | - Luyi Cheng
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
- Interdisciplinary Biological Sciences Training Program, Northwestern University, Evanston, IL, USA
| | - Beth N DiBiase
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
| | - Michelle E Hung
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
- Interdisciplinary Biological Sciences Training Program, Northwestern University, Evanston, IL, USA
| | - Julius B Lucks
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
- Interdisciplinary Biological Sciences Training Program, Northwestern University, Evanston, IL, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Judd F Hultquist
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL, USA
| | - Joshua N Leonard
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA.
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA.
- Interdisciplinary Biological Sciences Training Program, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
- Member, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
7
|
Bendixen SM, Jakobsgaard PR, Hansen D, Hejn KH, Terkelsen MK, Bjerre FA, Thulesen AP, Eriksen NG, Hallenborg P, Geng Y, Dam TV, Larsen FT, Wernberg CW, Vijayathurai J, Scott EAH, Marcher AB, Detlefsen S, Grøntved L, Dimke H, Berdeaux R, de Aguiar Vallim TQ, Olinga P, Lauridsen MM, Krag A, Blagoev B, Ravnskjaer K. Single cell-resolved study of advanced murine MASH reveals a homeostatic pericyte signaling module. J Hepatol 2024; 80:467-481. [PMID: 37972658 DOI: 10.1016/j.jhep.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 10/06/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatohepatitis (MASH) is linked to insulin resistance and type 2 diabetes and marked by hepatic inflammation, microvascular dysfunction, and fibrosis, impairing liver function and aggravating metabolic derangements. The liver homeostatic interactions disrupted in MASH are still poorly understood. We aimed to elucidate the plasticity and changing interactions of non-parenchymal cells associated with advanced MASH. METHODS We characterized a diet-induced mouse model of advanced MASH at single-cell resolution and validated findings by assaying chromatin accessibility, bioimaging murine and human livers, and via functional experiments in vivo and in vitro. RESULTS The fibrogenic activation of hepatic stellate cells (HSCs) led to deterioration of a signaling module consisting of the bile acid receptor NR1H4/FXR and HSC-specific GS-protein-coupled receptors (GSPCRs) capable of preserving stellate cell quiescence. Accompanying HSC activation, we further observed the attenuation of HSC Gdf2 expression, and a MASH-associated expansion of a CD207-positive macrophage population likely derived from both incoming monocytes and Kupffer cells. CONCLUSION We conclude that HSC-expressed NR1H4 and GSPCRs of the healthy liver integrate postprandial cues, which sustain HSC quiescence and, through paracrine signals, overall sinusoidal health. Hence HSC activation in MASH not only drives fibrogenesis but may desensitize the hepatic sinusoid to liver homeostatic signals. IMPACT AND IMPLICATIONS Homeostatic interactions between hepatic cell types and their deterioration in metabolic dysfunction-associated steatohepatitis are poorly characterized. In our current single cell-resolved study of advanced murine metabolic dysfunction-associated steatohepatitis, we identified a quiescence-associated hepatic stellate cell-signaling module with potential to preserve normal sinusoid function. As expression levels of its constituents are conserved in the human liver, stimulation of the identified signaling module is a promising therapeutic strategy to restore sinusoid function in chronic liver disease.
Collapse
Affiliation(s)
- Sofie M Bendixen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Peter R Jakobsgaard
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Daniel Hansen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Kamilla H Hejn
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Mike K Terkelsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Frederik A Bjerre
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Annemette P Thulesen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Niels G Eriksen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Philip Hallenborg
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Yana Geng
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, the Netherlands
| | - Trine V Dam
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Frederik T Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Charlotte W Wernberg
- Department of Gastroenterology and Hepatology, Odense University Hospital, Denmark; Department of Gastroenterology and Hepatology, University Hospital of South Denmark Esbjerg, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Janusa Vijayathurai
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Emma A H Scott
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Ann-Britt Marcher
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Sönke Detlefsen
- Department of Pathology, Odense University Hospital, Denmark; Department of Clinical Research, University of Southern Denmark, Denmark
| | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Henrik Dimke
- Department of Molecular Medicine, University of Southern Denmark, Denmark; Department of Nephrology, Odense University Hospital, Denmark
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, McGovern Medical School, UT Health Houston, USA
| | - Thomas Q de Aguiar Vallim
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, the Netherlands
| | - Mette M Lauridsen
- Department of Gastroenterology and Hepatology, University Hospital of South Denmark Esbjerg, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Aleksander Krag
- Department of Gastroenterology and Hepatology, Odense University Hospital, Denmark; Department of Clinical Research, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark
| | - Kim Ravnskjaer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark; Center for Functional Genomics and Tissue Plasticity, University of Southern Denmark, Denmark.
| |
Collapse
|
8
|
Yu L, Marchisio MA. Scaffold RNA engineering in type V CRISPR-Cas systems: a potent way to enhance gene expression in the yeast Saccharomyces cerevisiae. Nucleic Acids Res 2024; 52:1483-1497. [PMID: 38142459 PMCID: PMC10853767 DOI: 10.1093/nar/gkad1216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 12/26/2023] Open
Abstract
New, orthogonal transcription factors in eukaryotic cells have been realized by engineering nuclease-deficient CRISPR-associated proteins and/or their guide RNAs. In this work, we present a new kind of orthogonal transcriptional activators, in Saccharomyces cerevisiae, made by turning type V CRISPR RNA into a scaffold RNA (ScRNA) able to recruit a variable number of VP64 activation domains. The activator arises from the complex between the synthetic ScRNA and DNase-deficient type V Cas proteins: dCas12e and denAsCas12a. The transcription activation achieved via the newly engineered dCas:ScRNA system is up to 4.7-fold higher than that obtained with the direct fusion of VP64 to Cas proteins. The new transcription factors have been proven to be functional in circuits such as Boolean gates, converters, multiplex-gene and metabolic-pathway activation. Our results extend the CRISPR-Cas-based technology with a new effective tool that only demands RNA engineering and improves the current design of transcription factors based on type V Cas proteins.
Collapse
Affiliation(s)
- Lifang Yu
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, 300072 Tianjin, China
| | - Mario Andrea Marchisio
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, 300072 Tianjin, China
| |
Collapse
|
9
|
Jin L, Sullivan HA, Zhu M, Lavin TK, Matsuyama M, Fu X, Lea NE, Xu R, Hou Y, Rutigliani L, Pruner M, Babcock KR, Ip JPK, Hu M, Daigle TL, Zeng H, Sur M, Feng G, Wickersham IR. Long-term labeling and imaging of synaptically connected neuronal networks in vivo using double-deletion-mutant rabies viruses. Nat Neurosci 2024; 27:373-383. [PMID: 38212587 PMCID: PMC10849964 DOI: 10.1038/s41593-023-01545-8] [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: 12/16/2021] [Accepted: 12/05/2023] [Indexed: 01/13/2024]
Abstract
Rabies-virus-based monosynaptic tracing is a widely used technique for mapping neural circuitry, but its cytotoxicity has confined it primarily to anatomical applications. Here we present a second-generation system for labeling direct inputs to targeted neuronal populations with minimal toxicity, using double-deletion-mutant rabies viruses. Viral spread requires expression of both deleted viral genes in trans in postsynaptic source cells. Suppressing this expression with doxycycline following an initial period of viral replication reduces toxicity to postsynaptic cells. Longitudinal two-photon imaging in vivo indicated that over 90% of both presynaptic and source cells survived for the full 12-week course of imaging. Ex vivo whole-cell recordings at 5 weeks postinfection showed that the second-generation system perturbs input and source cells much less than the first-generation system. Finally, two-photon calcium imaging of labeled networks of visual cortex neurons showed that their visual response properties appeared normal for 10 weeks, the longest we followed them.
Collapse
Affiliation(s)
- Lei Jin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Lingang Laboratory, Shanghai, China
| | - Heather A Sullivan
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mulangma Zhu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Thomas K Lavin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Makoto Matsuyama
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Fu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicholas E Lea
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ran Xu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - YuanYuan Hou
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Luca Rutigliani
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Maxwell Pruner
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kelsey R Babcock
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacque Pak Kan Ip
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ming Hu
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ian R Wickersham
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
10
|
Park MJ, Lee J, Bagon BB, Matienzo ME, Lim S, Kim K, Lee CM, Wu J, Kim DI. N G ,N G -Dimethylarginine Dimethylaminohydrolase 1 Expression Is Dispensable for Cold- or Diet-Induced Thermogenesis. Adv Biol (Weinh) 2024; 8:e2300192. [PMID: 38164809 DOI: 10.1002/adbi.202300192] [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: 05/25/2023] [Revised: 12/18/2023] [Indexed: 01/03/2024]
Abstract
The strategy to activate thermogenic adipocytes has therapeutic potential to overcome obesity as they dissipate surplus energy as heat through various mechanisms. NG,NG-dimethylarginine dimethylaminohydrolases (DDAHs) are enzymes involved in the nitric oxide-protein kinase G signaling axis which increases thermogenic gene expression. However, the role of DDAHs in thermogenic adipocytes has not been elucidated. The adipocyte-specific Ddah1 knockout mice are generated by crossing Ddah1fl/fl mice with adiponectin Cre recombinase mice. Adipocyte-specific DDAH1 overexpressing mice are generated using adeno-associated virus-double-floxed inverse open reading frame (AAV-DIO) system. These mice are analyzed under basal, cold exposure, or high-fat diet (HFD) conditions. Primary inguinal white adipose tissue cells from adipocyte-specific Ddah1 knockout mice expressed comparable amounts of Ucp1 mRNA. Adipocyte-specific DDAH1 overexpressing mice do not exhibit enhanced activation of thermogenic adipocytes. In addition, when these mice are exposed to cold environment or fed an HFD, their body temperature/weight and thermogenesis-related gene and protein expressions are unchanged. These findings indicate that DDAH1 does not play a role in either cold- or diet-induced thermogenesis. Therefore, adipocyte targeting DDAH1 gene therapy for the treatment of obesity is unlikely to be effective.
Collapse
Affiliation(s)
- Min-Jung Park
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Junhyeong Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Bernadette B Bagon
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Merc Emil Matienzo
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Sangyi Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Keon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Chang-Min Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Jun Wu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Dong-Il Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| |
Collapse
|
11
|
Sayar-Atasoy N, Yavuz Y, Laule C, Dong C, Kim H, Rysted J, Flippo K, Davis D, Aklan I, Yilmaz B, Tian L, Atasoy D. Opioidergic signaling contributes to food-mediated suppression of AgRP neurons. Cell Rep 2024; 43:113630. [PMID: 38165803 PMCID: PMC10865729 DOI: 10.1016/j.celrep.2023.113630] [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: 10/11/2022] [Revised: 11/22/2023] [Accepted: 12/13/2023] [Indexed: 01/04/2024] Open
Abstract
Opioids are generally known to promote hedonic food consumption. Although much of the existing evidence is primarily based on studies of the mesolimbic pathway, endogenous opioids and their receptors are widely expressed in hypothalamic appetite circuits as well; however, their role in homeostatic feeding remains unclear. Using a fluorescent opioid sensor, deltaLight, here we report that mediobasal hypothalamic opioid levels increase by feeding, which directly and indirectly inhibits agouti-related protein (AgRP)-expressing neurons through the μ-opioid receptor (MOR). AgRP-specific MOR expression increases by energy surfeit and contributes to opioid-induced suppression of appetite. Conversely, its antagonists diminish suppression of AgRP neuron activity by food and satiety hormones. Mice with AgRP neuron-specific ablation of MOR expression have increased fat preference without increased motivation. These results suggest that post-ingestion release of endogenous opioids contributes to AgRP neuron inhibition to shape food choice through MOR signaling.
Collapse
Affiliation(s)
- Nilufer Sayar-Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Yavuz Yavuz
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Department of Physiology, School of Medicine, Yeditepe University, Istanbul 34755, Turkey
| | - Connor Laule
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Chunyang Dong
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Hyojin Kim
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Jacob Rysted
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kyle Flippo
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Debbie Davis
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Iltan Aklan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Bayram Yilmaz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul 34755, Turkey
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center (FOEDRC), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
12
|
Sayar-Atasoy N, Aklan I, Yavuz Y, Laule C, Kim H, Rysted J, Alp MI, Davis D, Yilmaz B, Atasoy D. AgRP neurons encode circadian feeding time. Nat Neurosci 2024; 27:102-115. [PMID: 37957320 DOI: 10.1038/s41593-023-01482-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/05/2023] [Indexed: 11/15/2023]
Abstract
Food intake follows a predictable daily pattern and synchronizes metabolic rhythms. Neurons expressing agouti-related protein (AgRP) read out physiological energetic state and elicit feeding, but the regulation of these neurons across daily timescales is poorly understood. Using a combination of neuron dynamics measurements and timed optogenetic activation in mice, we show that daily AgRP-neuron activity was not fully consistent with existing models of homeostatic regulation. Instead of operating as a 'deprivation counter', AgRP-neuron activity primarily followed the circadian rest-activity cycle through a process that required an intact suprachiasmatic nucleus and synchronization by light. Imposing novel feeding patterns through time-restricted food access or periodic AgRP-neuron stimulation was sufficient to resynchronize the daily AgRP-neuron activity rhythm and drive anticipatory-like behavior through a process that required DMHPDYN neurons. These results indicate that AgRP neurons integrate time-of-day information of past feeding experience with current metabolic needs to predict circadian feeding time.
Collapse
Affiliation(s)
- Nilufer Sayar-Atasoy
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Iltan Aklan
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Yavuz Yavuz
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Connor Laule
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Hyojin Kim
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Jacob Rysted
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Muhammed Ikbal Alp
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Debbie Davis
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Bayram Yilmaz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
13
|
Zhang XJ, Han BB, Shao ZY, Yan R, Gao J, Liu T, Jin ZY, Lai W, Xu ZM, Wang CH, Zhang F, Gu C, Wang Y, Wang H, Walsh CP, Guo F, Xu GL, Du YR. Auto-suppression of Tet dioxygenases protects the mouse oocyte genome from oxidative demethylation. Nat Struct Mol Biol 2024; 31:42-53. [PMID: 38177668 DOI: 10.1038/s41594-023-01125-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 09/14/2023] [Indexed: 01/06/2024]
Abstract
DNA cytosine methylation plays a vital role in repressing retrotransposons, and such derepression is linked with developmental failure, tumorigenesis and aging. DNA methylation patterns are formed by precisely regulated actions of DNA methylation writers (DNA methyltransferases) and erasers (TET, ten-eleven translocation dioxygenases). However, the mechanisms underlying target-specific oxidation of 5mC by TET dioxygenases remain largely unexplored. Here we show that a large low-complexity domain (LCD), located in the catalytic part of Tet enzymes, negatively regulates the dioxygenase activity. Recombinant Tet3 lacking LCD is shown to be hyperactive in converting 5mC into oxidized species in vitro. Endogenous expression of the hyperactive Tet3 mutant in mouse oocytes results in genome-wide 5mC oxidation. Notably, the occurrence of aberrant 5mC oxidation correlates with a consequent loss of the repressive histone mark H3K9me3 at ERVK retrotransposons. The erosion of both 5mC and H3K9me3 causes ERVK derepression along with upregulation of their neighboring genes, potentially leading to the impairment of oocyte development. These findings suggest that Tet dioxygenases use an intrinsic auto-regulatory mechanism to tightly regulate their enzymatic activity, thus achieving spatiotemporal specificity of methylome reprogramming, and highlight the importance of methylome integrity for development.
Collapse
Affiliation(s)
- Xiao-Jie Zhang
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bin-Bin Han
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhen-Yu Shao
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Yan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Juan Gao
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ting Liu
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Zi-Yang Jin
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Weiyi Lai
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Zhi-Mei Xu
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chao-Han Wang
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fengjuan Zhang
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chan Gu
- Changping Laboratory, Beijing, China
| | - Yin Wang
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Chinese Academy of Medical Sciences (RU069) and Zhongshan-Xuhui Hospital, Medical College of Fudan University, Shanghai, China
| | - Hailin Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Colum P Walsh
- Genomic Medicine Research Group, Biomedical Sciences, Ulster University, Coleraine, UK
- Department of Cell Biology, Institute for Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Fan Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Guo-Liang Xu
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Chinese Academy of Medical Sciences (RU069) and Zhongshan-Xuhui Hospital, Medical College of Fudan University, Shanghai, China.
| | - Ya-Rui Du
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
14
|
Singh O, Ogden SB, Varshney S, Shankar K, Gupta D, Paul S, Osborne-Lawrence S, Richard CP, Metzger NP, Lawrence C, Leon Mercado L, Zigman JM. Ghrelin-responsive mediobasal hypothalamic neurons mediate exercise-associated food intake and exercise endurance. JCI Insight 2023; 8:e172549. [PMID: 37962950 PMCID: PMC10807726 DOI: 10.1172/jci.insight.172549] [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: 05/24/2023] [Accepted: 11/08/2023] [Indexed: 11/16/2023] Open
Abstract
Previous studies have implicated the orexigenic hormone ghrelin as a mediator of exercise endurance and the feeding response postexercise. Specifically, plasma ghrelin levels nearly double in mice when they are subjected to an hour-long bout of high-intensity interval exercise (HIIE) using treadmills. Also, growth hormone secretagogue receptor-null (GHSR-null) mice exhibit decreased food intake following HIIE and diminished running distance (time until exhaustion) during a longer, stepwise exercise endurance protocol. To investigate whether ghrelin-responsive mediobasal hypothalamus (MBH) neurons mediate these effects, we stereotaxically delivered the inhibitory designer receptor exclusively activated by designer drugs virus AAV2-hSyn-DIO-hM4(Gi)-mCherry to the MBH of Ghsr-IRES-Cre mice, which express Cre recombinase directed by the Ghsr promoter. We found that chemogenetic inhibition of GHSR-expressing MBH neurons (upon delivery of clozapine-N-oxide) 1) suppressed food intake following HIIE, 2) reduced maximum running distance and raised blood glucose and blood lactate levels during an exercise endurance protocol, 3) reduced food intake following ghrelin administration, and 4) did not affect glucose tolerance. Further, HIIE increased MBH Ghsr expression. These results indicate that activation of ghrelin-responsive MBH neurons is required for the normal feeding response to HIIE and the usual amount of running exhibited during an exercise endurance protocol.
Collapse
Affiliation(s)
- Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine
| | - Sean B. Ogden
- Center for Hypothalamic Research, Department of Internal Medicine
| | - Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine
| | - Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine
| | - Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine
| | - Subhojit Paul
- Center for Hypothalamic Research, Department of Internal Medicine
| | | | | | | | - Connor Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine
| | | | - Jeffrey M. Zigman
- Center for Hypothalamic Research, Department of Internal Medicine
- Division of Endocrinology & Metabolism, Department of Internal Medicine; and
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
15
|
Middleton SJ, Hu H, Perez-Sanchez J, Zuberi S, McGrath Williams J, Weir GA, Bennett DL. GluCl.Cre ON enables selective inhibition of molecularly defined pain circuits. Pain 2023; 164:2780-2791. [PMID: 37366588 PMCID: PMC10652717 DOI: 10.1097/j.pain.0000000000002976] [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: 10/03/2022] [Revised: 02/17/2023] [Accepted: 05/08/2023] [Indexed: 06/28/2023]
Abstract
ABSTRACT Insight into nociceptive circuits will ultimately build our understanding of pain processing and aid the development of analgesic strategies. Neural circuit analysis has been advanced greatly by the development of optogenetic and chemogenetic tools, which have allowed function to be ascribed to discrete neuronal populations. Neurons of the dorsal root ganglion, which include nociceptors, have proved challenging targets for chemogenetic manipulation given specific confounds with commonly used DREADD technology. We have developed a cre/lox dependant version of the engineered glutamate-gated chloride channel (GluCl) to restrict and direct its expression to molecularly defined neuronal populations. We have generated GluCl.Cre ON that selectively renders neurons expressing cre-recombinase susceptible to agonist-induced silencing. We have functionally validated our tool in multiple systems in vitro, and subsequently generated viral vectors and tested its applicability in vivo. Using Nav1.8 Cre mice to restrict AAV-GluCl.Cre ON to nociceptors, we demonstrate effective silencing of electrical activity in vivo and concomitant hyposensitivity to noxious thermal and noxious mechanical pain, whereas light touch and motor function remained intact. We also demonstrated that our strategy can effectively silence inflammatory-like pain in a chemical pain model. Collectively, we have generated a novel tool that can be used to selectively silence defined neuronal circuits in vitro and in vivo. We believe that this addition to the chemogenetic tool box will facilitate further understanding of pain circuits and guide future therapeutic development.
Collapse
Affiliation(s)
- Steven J. Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Huimin Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Jimena Perez-Sanchez
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Sana Zuberi
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | | | - Greg A. Weir
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
16
|
Bagon BB, Lee J, Matienzo ME, Lim S, Park JI, Kang S, Kim K, Lee CM, Moon C, Kim DI, Park MJ. AAV-mediated skeletal muscle specific irisin expression does not contribute to weight loss in mice. Biochem Biophys Res Commun 2023; 682:111-117. [PMID: 37806248 DOI: 10.1016/j.bbrc.2023.10.004] [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: 09/25/2023] [Revised: 09/27/2023] [Accepted: 10/01/2023] [Indexed: 10/10/2023]
Abstract
Obesity, a chronic disease, significantly increases the risk of various diseases, including diabetes, cardiovascular diseases, and cancers. Exercise is crucial for weight management not only through energy expenditure by muscle activity but also through stimulating the secretion of myokines, which affect various tissues. Irisin, derived from the proteolytic processing of fibronectin type III domain-containing protein 5 (Fndc5), is a well-studied myokine with beneficial effects on metabolism. This study explored the feasibility of adeno-associated virus (AAV)-mediated Fndc5 gene therapy to treat obesity in a mouse model using the AAV-DIO system to express Fndc5 specifically in skeletal muscle, and investigated its anti-obesity effect. Although Fndc5 was specifically expressed in the muscle, no significant impact on body weight under normal chow or high-fat diets was observed, and no change in thermogenic gene expression in inguinal white adipose tissue was detected. Notably, Fndc5 transduction did affect bone metabolism, consistent with previous reports. These findings suggest that AAV-mediated Fndc5 gene therapy may not be an efficient strategy for obesity, contrary to our expectations. Further research is needed to elucidate the complex mechanisms involved in irisin's role in obesity and related disorders.
Collapse
Affiliation(s)
- Bernadette B Bagon
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Junhyeong Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Merc Emil Matienzo
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Sangyi Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Jae-Il Park
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, 61751, South Korea
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Keon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea; Department of Veterinary Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Chang-Min Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea; Department of Veterinary Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Changjong Moon
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea; Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Dong-Il Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea.
| | - Min-Jung Park
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea.
| |
Collapse
|
17
|
Mirabella PN, Fenselau H. Advanced neurobiological tools to interrogate metabolism. Nat Rev Endocrinol 2023; 19:639-654. [PMID: 37674015 DOI: 10.1038/s41574-023-00885-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 09/08/2023]
Abstract
Engineered neurobiological tools for the manipulation of cellular activity, such as chemogenetics and optogenetics, have become a cornerstone of modern neuroscience research. These tools are invaluable for the interrogation of the central control of metabolism as they provide a direct means to establish a causal relationship between brain activity and biological processes at the cellular, tissue and organismal levels. The utility of these methods has grown substantially due to advances in cellular-targeting strategies, alongside improvements in the resolution and potency of such tools. Furthermore, the potential to recapitulate endogenous cellular signalling has been enriched by insights into the molecular signatures and activity dynamics of discrete brain cell types. However, each modulatory tool has a specific set of advantages and limitations; therefore, tool selection and suitability are of paramount importance to optimally interrogate the cellular and circuit-based underpinnings of metabolic outcomes within the organism. Here, we describe the key principles and uses of engineered neurobiological tools. We also highlight inspiring applications and outline critical considerations to be made when using these tools within the field of metabolism research. We contend that the appropriate application of these biotechnological advances will enable the delineation of the central circuitry regulating systemic metabolism with unprecedented potential.
Collapse
Affiliation(s)
- Paul Nicholas Mirabella
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Cologne, Germany.
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
18
|
Zhang J, Wang L, Yang Y, Wang S, Huang C, Yang L, Li B, Wang L, Wang H, Hao S. Dissection of the bed nucleus of the stria terminalis neuronal subtypes in feeding regulation. Physiol Behav 2023; 271:114333. [PMID: 37595819 DOI: 10.1016/j.physbeh.2023.114333] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/07/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
The bed nucleus of the stria terminalis (BNST) plays an important role in feeding regulation through projections to other brain areas. However, whether functional distinctions exist within different BNST cells is not clear. Here, we found optogenetic activation of LH-projecting BNST neurons induced aversion and significantly reduced consumption of normal chow but not high-fat diets (HFD). In contrast, photoactivation of vlPAG-projecting BNST neurons induced place preference and promoted HFD intake, without affecting normal chow consumption. Moreover, optogenetic silencing of LH-projecting BNST neurons reduced the consumption of normal chow in fasted mice, while photoinhibition of vlPAG-projecting BNST neurons decreased the consumption of HFD in both fed and fasted mice. We then labeled the LH- and vlPAG-projecting BNST neurons using retroAAV-GFP and retroAAV-mCherry, respectively, and found these two populations of neurons have different anatomical distribution and electrophysiological properties. Taken together, we identified vlPAG-projecting and LH-projecting BNST neurons are two distinct populations of cells with significant differences in functional and anatomic characteristics.
Collapse
Affiliation(s)
- Jiaozhen Zhang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China; Institute of Brain Science and Department of Physiology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Liangliang Wang
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China; Department of Psychiatry, Sir Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, China
| | - Yiwen Yang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Siyu Wang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Changgang Huang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Li Yang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Baoming Li
- Institute of Brain Science and Department of Physiology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Lang Wang
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China; Department of Psychiatry, Sir Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, China.
| | - Hao Wang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China.
| | - Sijia Hao
- Institute of Brain Science and Department of Physiology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China.
| |
Collapse
|
19
|
Bakoyiannis I, Ducourneau EG, Parkes SL, Ferreira G. Pathway specific interventions reveal the multiple roles of ventral hippocampus projections in cognitive functions. Rev Neurosci 2023; 34:825-838. [PMID: 37192533 DOI: 10.1515/revneuro-2023-0009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/28/2023] [Indexed: 05/18/2023]
Abstract
Since the 1950s study of Scoville and Milner on the case H.M., the hippocampus has attracted neuroscientists' attention. The hippocampus has been traditionally divided into dorsal and ventral parts, each of which projects to different brain structures and mediates various functions. Despite a predominant interest in its dorsal part in animal models, especially regarding episodic-like and spatial cognition, recent data highlight the role of the ventral hippocampus (vHPC), as the main hippocampal output, in cognitive processes. Here, we review recent studies conducted in rodents that have used advanced in vivo functional techniques to specifically monitor and manipulate vHPC efferent pathways and delineate the roles of these specific projections in learning and memory processes. Results highlight that vHPC projections to basal amygdala are implicated in emotional memory, to nucleus accumbens in social memory and instrumental actions and to prefrontal cortex in all the above as well as in object-based memory. Some of these hippocampal projections also modulate feeding and anxiety-like behaviours providing further evidence that the "one pathway-one function" view is outdated and future directions are proposed to better understand the role of hippocampal pathways and shed further light on its connectivity and function.
Collapse
Affiliation(s)
- Ioannis Bakoyiannis
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077 Bordeaux, France
| | - Eva-Gunnel Ducourneau
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077 Bordeaux, France
| | - Shauna L Parkes
- University of Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Guillaume Ferreira
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077 Bordeaux, France
| |
Collapse
|
20
|
Sayar-Atasoy N, Laule C, Aklan I, Kim H, Yavuz Y, Ates T, Coban I, Koksalar-Alkan F, Rysted J, Davis D, Singh U, Alp MI, Yilmaz B, Cui H, Atasoy D. Adrenergic modulation of melanocortin pathway by hunger signals. Nat Commun 2023; 14:6602. [PMID: 37857606 PMCID: PMC10587058 DOI: 10.1038/s41467-023-42362-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023] Open
Abstract
Norepinephrine (NE) is a well-known appetite regulator, and the nor/adrenergic system is targeted by several anti-obesity drugs. To better understand the circuitry underlying adrenergic appetite control, here we investigated the paraventricular hypothalamic nucleus (PVN), a key brain region that integrates energy signals and receives dense nor/adrenergic input, using a mouse model. We found that PVN NE level increases with signals of energy deficit and decreases with food access. This pattern is recapitulated by the innervating catecholaminergic axon terminals originating from NTSTH-neurons. Optogenetic activation of rostral-NTSTH → PVN projection elicited strong motivation to eat comparable to overnight fasting whereas its inhibition attenuated both fasting-induced & hypoglycemic feeding. We found that NTSTH-axons functionally targeted PVNMC4R-neurons by predominantly inhibiting them, in part, through α1-AR mediated potentiation of GABA release from ARCAgRP presynaptic terminals. Furthermore, glucoprivation suppressed PVNMC4R activity, which was required for hypoglycemic feeding response. These results define an ascending nor/adrenergic circuit, NTSTH → PVNMC4R, that conveys peripheral hunger signals to melanocortin pathway.
Collapse
Affiliation(s)
- Nilufer Sayar-Atasoy
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Connor Laule
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Iltan Aklan
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hyojin Kim
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yavuz Yavuz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Tayfun Ates
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ilknur Coban
- Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | | | - Jacob Rysted
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Debbie Davis
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Uday Singh
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Muhammed Ikbal Alp
- Department of Physiology, School of Medicine, Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Bayram Yilmaz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Huxing Cui
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Deniz Atasoy
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
21
|
Sharma K, Govar AA, Ghimire B, Nishimori K, Hammock E, Teruyama R. Sexually dimorphic oxytocin receptor-expressing (OXTR) neurons in the anteroventral periventricular nucleus (AVPV) in the postpartum female mouse are involved in maternal behavior. J Neuroendocrinol 2023; 35:e13337. [PMID: 37784240 DOI: 10.1111/jne.13337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/03/2023] [Accepted: 08/03/2023] [Indexed: 10/04/2023]
Abstract
Maternal care is crucial for the survival and development of offspring. Oxytocin modulates maternal behavior by binding to oxytocin receptors (OXTRs) in various parts of the brain. Previously, we showed that OXTRs are expressed in the anteroventral periventricular nucleus (AVPV) of female, but not male mice. Because the AVPV is involved in the regulation of maternal behavior and oxytocin enhances its induction, this finding leads to the hypothesis that the female specific population of OXTR neurons in the AVPV regulates maternal behavior. To address this hypothesis, OXTR-Venus reporter mice were used to assess if expression levels of OXTR in the AVPV are changed during the postpartum period. The total number of OXTR-Venus neurons was significantly greater in postpartum dams compared to virgin females. To assess efferent projections of the AVPV-OXTR neurons, a Cre-dependent fluorescent protein (tdTomato) expressing a viral vector was injected into one side of the AVPV of female OXTR-Cre mice. Fibers expressing tdTomato were found in hypothalamic areas containing oxytocin neurons (the supraoptic and paraventricular nuclei) and the midbrain areas (the ventral tegmental area and periaqueductal gray) that are involved in the regulation of maternal motivation. To assess if activity of the AVPV-OXTR neurons is involved in the regulation of maternal behaviors, a chemogenetic approach was employed. Specific inhibition of activity of AVPV-OXTR neurons completely abolished pup retrieval and nest building behaviors. Collectively, these findings demonstrate that AVPV-OXTR neurons in postpartum female mice constitute an important node in the neural circuitry that regulates maternal behavior.
Collapse
Affiliation(s)
- Kaustubh Sharma
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Armita A Govar
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bandana Ghimire
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Katsuhiko Nishimori
- Laboratory of Obesity and Internal Inflammation, Fukushima Medical University, Fukushima, Japan
| | - Elizabeth Hammock
- Department of Psychology, Florida State University, Tallahassee, Florida, USA
| | - Ryoichi Teruyama
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
22
|
de Araujo Salgado I, Li C, Burnett CJ, Rodriguez Gonzalez S, Becker JJ, Horvath A, Earnest T, Kravitz AV, Krashes MJ. Toggling between food-seeking and self-preservation behaviors via hypothalamic response networks. Neuron 2023; 111:2899-2917.e6. [PMID: 37442130 PMCID: PMC10528369 DOI: 10.1016/j.neuron.2023.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 05/02/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023]
Abstract
Motivated behaviors are often studied in isolation to assess labeled lines of neural connections underlying innate actions. However, in nature, multiple systems compete for expression of goal-directed behaviors via complex neural networks. Here, we examined flexible survival decisions in animals tasked with food seeking under predation threat. We found that predator exposure rapidly induced physiological, neuronal, and behavioral adaptations in mice highlighted by reduced food seeking and consumption contingent on current threat level. Diminishing conflict via internal state or external environment perturbations shifted feeding strategies. Predator introduction and/or selective manipulation of danger-responsive cholecystokinin (Cck) cells of the dorsal premammilary nucleus (PMd) suppressed hunger-sensitive Agouti-related peptide (AgRP) neurons, providing a mechanism for threat-evoked hypophagia. Increased caloric need enhanced food seeking under duress through AgRP pathways to the bed nucleus of the stria terminalis (BNST) and/or lateral hypothalamus (LH). Our results suggest oscillating interactions between systems underlying self-preservation and food seeking to promote optimal behavior.
Collapse
Affiliation(s)
- Isabel de Araujo Salgado
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chia Li
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - C Joseph Burnett
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shakira Rodriguez Gonzalez
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jordan J Becker
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Allison Horvath
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas Earnest
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Alexxai V Kravitz
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
23
|
Kaminosono J, Kambe Y, Tanimoto A, Kuwaki T, Yamashita A. The physiological response during optogenetic-based cardiac pacing in awake freely moving mice. Front Physiol 2023; 14:1130956. [PMID: 37736488 PMCID: PMC10509767 DOI: 10.3389/fphys.2023.1130956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 07/10/2023] [Indexed: 09/23/2023] Open
Abstract
There are several methods to control a heart rate, such as electrical stimulation and drug administration. However, these methods may be invasive or affect other organs. Recently, an optogenetic-based cardiac pacing method has enabled us to stimulate the cardiac muscle in non-contact. In many previous studies, the pacing was applied ex vivo or in anesthetized animals. Therefore, the physiologic response of animals during optogenetic pacing remains unclear. Here, we established a method of optogenetic-based cardiac pacing in awake, freely moving mice and simultaneously measured electrocardiogram, blood pressure, and respiration. As a result, light-induced myocardial contraction produces blood flow and indirectly affects the respiration rhythm. Additionally, light illumination enabled heart rate recovery in bradycardic mice. These findings may be employed for further research that relates a heartbeat state to animal behavior. Together, this method may drive the development of less invasive pacemakers without pacing leads.
Collapse
Affiliation(s)
- Jun Kaminosono
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuki Kambe
- Department of Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akihide Tanimoto
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tomoyuki Kuwaki
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akira Yamashita
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- Department of Medical Neuropharmacology, Wakayama Medical University School of Pharmaceutical Sciences, Wakayama, Japan
| |
Collapse
|
24
|
Haroun R, Gossage SJ, Luiz AP, Arcangeletti M, Sikandar S, Zhao J, Cox JJ, Wood JN. Chemogenetic Silencing of Na V1.8-Positive Sensory Neurons Reverses Chronic Neuropathic and Bone Cancer Pain in FLEx PSAM 4-GlyR Mice. eNeuro 2023; 10:ENEURO.0151-23.2023. [PMID: 37679042 PMCID: PMC10523839 DOI: 10.1523/eneuro.0151-23.2023] [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: 05/10/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Drive from peripheral neurons is essential in almost all pain states, but pharmacological silencing of these neurons to effect analgesia has proved problematic. Reversible gene therapy using long-lived chemogenetic approaches is an appealing option. We used the genetically activated chloride channel PSAM4-GlyR to examine pain pathways in mice. Using recombinant AAV9-based delivery to sensory neurons, we found a reversal of acute pain behavior and diminished neuronal activity using in vitro and in vivo GCaMP imaging on activation of PSAM4-GlyR with varenicline. A significant reduction in inflammatory heat hyperalgesia and oxaliplatin-induced cold allodynia was also observed. Importantly, there was no impairment of motor coordination, but innocuous von Frey sensation was inhibited. We generated a transgenic mouse that expresses a CAG-driven FLExed PSAM4-GlyR downstream of the Rosa26 locus that requires Cre recombinase to enable the expression of PSAM4-GlyR and tdTomato. We used NaV1.8 Cre to examine the role of predominantly nociceptive NaV1.8+ neurons in cancer-induced bone pain (CIBP) and neuropathic pain caused by chronic constriction injury (CCI). Varenicline activation of PSAM4-GlyR in NaV1.8-positive neurons reversed CCI-driven mechanical, thermal, and cold sensitivity. Additionally, varenicline treatment of mice with CIBP expressing PSAM4-GlyR in NaV1.8+ sensory neurons reversed cancer pain as assessed by weight-bearing. Moreover, when these mice were subjected to acute pain assays, an elevation in withdrawal thresholds to noxious mechanical and thermal stimuli was detected, but innocuous mechanical sensations remained unaffected. These studies confirm the utility of PSAM4-GlyR chemogenetic silencing in chronic pain states for mechanistic analysis and potential future therapeutic use.
Collapse
Affiliation(s)
- Rayan Haroun
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - Samuel J Gossage
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - Ana Paula Luiz
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - Manuel Arcangeletti
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - Shafaq Sikandar
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Jing Zhao
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - James J Cox
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - John N Wood
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
25
|
Priest MF, Freda SN, Rieth IJ, Badong D, Dumrongprechachan V, Kozorovitskiy Y. Peptidergic and functional delineation of the Edinger-Westphal nucleus. Cell Rep 2023; 42:112992. [PMID: 37594894 PMCID: PMC10512657 DOI: 10.1016/j.celrep.2023.112992] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 06/15/2023] [Accepted: 07/31/2023] [Indexed: 08/20/2023] Open
Abstract
Many neuronal populations that release fast-acting excitatory and inhibitory neurotransmitters in the brain also contain slower-acting neuropeptides. These facultative peptidergic cell types are common, but it remains uncertain whether neurons that solely release peptides exist. Our fluorescence in situ hybridization, genetically targeted electron microscopy, and electrophysiological characterization suggest that most neurons of the non-cholinergic, centrally projecting Edinger-Westphal nucleus in mice are obligately peptidergic. We further show, using anterograde projection mapping, monosynaptic retrograde tracing, angled-tip fiber photometry, and chemogenetic modulation and genetically targeted ablation in conjunction with canonical assays for anxiety, that this peptidergic population activates in response to loss of motor control and promotes anxiety responses. Together, these findings elucidate an integrative, ethologically relevant role for the Edinger-Westphal nucleus and functionally align the nucleus with the periaqueductal gray, where it resides. This work advances our understanding of peptidergic modulation of anxiety and provides a framework for future investigations of peptidergic systems.
Collapse
Affiliation(s)
- Michael F Priest
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sara N Freda
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Isabelle J Rieth
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Deanna Badong
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Vasin Dumrongprechachan
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
26
|
Laing BT, Anderson MS, Bonaventura J, Jayan A, Sarsfield S, Gajendiran A, Michaelides M, Aponte Y. Anterior hypothalamic parvalbumin neurons are glutamatergic and promote escape behavior. Curr Biol 2023; 33:3215-3228.e7. [PMID: 37490921 PMCID: PMC10529150 DOI: 10.1016/j.cub.2023.06.070] [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: 09/21/2022] [Revised: 05/19/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023]
Abstract
The anterior hypothalamic area (AHA) is a critical structure for defensive responding. Here, we identified a cluster of parvalbumin-expressing neurons in the AHA (AHAPV) that are glutamatergic with fast-spiking properties and send axonal projections to the dorsal premammillary nucleus (PMD). Using in vivo functional imaging, optogenetics, and behavioral assays, we determined the role of these AHAPV neurons in regulating behaviors essential for survival. We observed that AHAPV neuronal activity significantly increases when mice are exposed to a predator, and in a real-time place preference assay, we found that AHAPV neuron photoactivation is aversive. Moreover, activation of both AHAPV neurons and the AHAPV → PMD pathway triggers escape responding during a predator-looming test. Furthermore, escape responding is impaired after AHAPV neuron ablation, and anxiety-like behavior as measured by the open field and elevated plus maze assays does not seem to be affected by AHAPV neuron ablation. Finally, whole-brain metabolic mapping using positron emission tomography combined with AHAPV neuron photoactivation revealed discrete activation of downstream areas involved in arousal, affective, and defensive behaviors including the amygdala and the substantia nigra. Our results indicate that AHAPV neurons are a functional glutamatergic circuit element mediating defensive behaviors, thus expanding the identity of genetically defined neurons orchestrating fight-or-flight responses. Together, our work will serve as a foundation for understanding neuropsychiatric disorders triggered by escape such as post-traumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Brenton T Laing
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224-6823, USA
| | - Megan S Anderson
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224-6823, USA
| | - Jordi Bonaventura
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224-6823, USA
| | - Aishwarya Jayan
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224-6823, USA
| | - Sarah Sarsfield
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224-6823, USA
| | - Anjali Gajendiran
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224-6823, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224-6823, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yeka Aponte
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224-6823, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
27
|
Douglass AM, Resch JM, Madara JC, Kucukdereli H, Yizhar O, Grama A, Yamagata M, Yang Z, Lowell BB. Neural basis for fasting activation of the hypothalamic-pituitary-adrenal axis. Nature 2023; 620:154-162. [PMID: 37495689 PMCID: PMC11168300 DOI: 10.1038/s41586-023-06358-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 06/21/2023] [Indexed: 07/28/2023]
Abstract
Fasting initiates a multitude of adaptations to allow survival. Activation of the hypothalamic-pituitary-adrenal (HPA) axis and subsequent release of glucocorticoid hormones is a key response that mobilizes fuel stores to meet energy demands1-5. Despite the importance of the HPA axis response, the neural mechanisms that drive its activation during energy deficit are unknown. Here, we show that fasting-activated hypothalamic agouti-related peptide (AgRP)-expressing neurons trigger and are essential for fasting-induced HPA axis activation. AgRP neurons do so through projections to the paraventricular hypothalamus (PVH), where, in a mechanism not previously described for AgRP neurons, they presynaptically inhibit the terminals of tonically active GABAergic afferents from the bed nucleus of the stria terminalis (BNST) that otherwise restrain activity of corticotrophin-releasing hormone (CRH)-expressing neurons. This disinhibition of PVHCrh neurons requires γ-aminobutyric acid (GABA)/GABA-B receptor signalling and potently activates the HPA axis. Notably, stimulation of the HPA axis by AgRP neurons is independent of their induction of hunger, showing that these canonical 'hunger neurons' drive many distinctly different adaptations to the fasted state. Together, our findings identify the neural basis for fasting-induced HPA axis activation and uncover a unique means by which AgRP neurons activate downstream neurons: through presynaptic inhibition of GABAergic afferents. Given the potency of this disinhibition of tonically active BNST afferents, other activators of the HPA axis, such as psychological stress, may also work by reducing BNST inhibitory tone onto PVHCrh neurons.
Collapse
Affiliation(s)
- Amelia M Douglass
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jon M Resch
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hakan Kucukdereli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ofer Yizhar
- Departments of Brain Sciences and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Abhinav Grama
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Masahito Yamagata
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Zongfang Yang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Liu YS, Wang ML, Hu NY, Li ZM, Wu JL, Li H, Li JT, Li XW, Yang JM, Gao TM, Chen YH. A comparison of the impact on neuronal transcriptome and cognition of rAAV5 transduction with three different doses in the mouse hippocampus. Front Mol Neurosci 2023; 16:1195327. [PMID: 37520430 PMCID: PMC10375024 DOI: 10.3389/fnmol.2023.1195327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/20/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Recombinant adeno-associated viruses (rAAVs) are widely used in genetic therapeutics. AAV5 has shown superior transduction efficiency, targeting neurons and glial cells in primate brains. Nonetheless, the comprehensive impact of AAV5 transduction on molecular and behavioral alterations remains unexplored. This study focuses on evaluating the effects of AAV5 transduction in the hippocampus, a critical region for memory formation and emotional processes. Methods In this experiment, fluorescence-activated cell sorting (FACS) was utilized to isolate the mCherry-labeled pyramidal neurons in the hippocampus of CaMkIIα-cre mice following three different doses rAAV5-mCherry infusion after 3 weeks, which were then subjected to RNA sequencing (RNA-seq) to assess gene expression profiles. The cytokines concentration, mRNA expression, and glial response in hippocampi were confirmed by ELASA, digital droplet PCR and immunohistochemistry respectively. Locomotion and anxiety-like behaviors were elevated by Open Field Test and Elevated Plus Maze Test, while the Y-Maze were used to assessed spatial working memory. Recognition memory and fear responses were examined by the Novel Object Recognition Test and Fear Conditioning Test, respectively. Results We found that 2.88 × 1010 v.g rAAV5 transduction significantly upregulated genes related to the immune response and apoptosis, and downregulated genes associated with mitochondrial function and synaptic plasticity in hippocampal pyramidal neurons, while did not induce neuronal loss and gliosis compared with 2.88 × 109 v.g and 2.88 × 108 v.g. Furthermore, the same doses impaired working memory and contextual fear memory, without effects on locomotion and anxiety-related behaviors. Discussion Our findings highlight the detrimental impact of high-dose administration compared to median-dose or low-dose, resulting in increased neural vulnerability and impaired memory. Therefore, when considering the expression effectiveness of exogenous genes, it is crucial to also take potential side effects into account in clinical settings. However, the precise molecular mechanisms underlying these drawbacks of high-dose rAAV5-mCherry still require further investigation in future studies.
Collapse
|
29
|
Zengel J, Wang YX, Seo JW, Ning K, Hamilton JN, Wu B, Raie M, Holbrook C, Su S, Clements DR, Pillay S, Puschnik AS, Winslow MM, Idoyaga J, Nagamine CM, Sun Y, Mahajan VB, Ferrara KW, Blau HM, Carette JE. Hardwiring tissue-specific AAV transduction in mice through engineered receptor expression. Nat Methods 2023; 20:1070-1081. [PMID: 37291262 PMCID: PMC10333121 DOI: 10.1038/s41592-023-01896-x] [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: 05/23/2022] [Accepted: 04/25/2023] [Indexed: 06/10/2023]
Abstract
The development of transgenic mouse models that express genes of interest in specific cell types has transformed our understanding of basic biology and disease. However, generating these models is time- and resource-intensive. Here we describe a model system, SELective Expression and Controlled Transduction In Vivo (SELECTIV), that enables efficient and specific expression of transgenes by coupling adeno-associated virus (AAV) vectors with Cre-inducible overexpression of the multi-serotype AAV receptor, AAVR. We demonstrate that transgenic AAVR overexpression greatly increases the efficiency of transduction of many diverse cell types, including muscle stem cells, which are normally refractory to AAV transduction. Superior specificity is achieved by combining Cre-mediated AAVR overexpression with whole-body knockout of endogenous Aavr, which is demonstrated in heart cardiomyocytes, liver hepatocytes and cholinergic neurons. The enhanced efficacy and exquisite specificity of SELECTIV has broad utility in development of new mouse model systems and expands the use of AAV for gene delivery in vivo.
Collapse
Affiliation(s)
- James Zengel
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yu Xin Wang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Center for Genetic Disorders and Aging, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jai Woong Seo
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ke Ning
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
| | - James N Hamilton
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Bo Wu
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marina Raie
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Colin Holbrook
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shiqi Su
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Derek R Clements
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Sirika Pillay
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Andreas S Puschnik
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Juliana Idoyaga
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Claude M Nagamine
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Administration, Palo Alto, CA, USA
| | - Vinit B Mahajan
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Administration, Palo Alto, CA, USA
| | - Katherine W Ferrara
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jan E Carette
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
30
|
Zhuo M, Mao J, David CJ. Protocol to package and concentrate adeno-associated virus serotype 8 for use in autochthonous mouse models of pancreatic cancer. STAR Protoc 2023; 4:102108. [PMID: 36853729 PMCID: PMC9943862 DOI: 10.1016/j.xpro.2023.102108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/23/2022] [Accepted: 01/23/2023] [Indexed: 02/12/2023] Open
Abstract
We have developed an economical and rapid protocol to package and concentrate adeno-associated virus serotype 8, allowing production of high-titer virus for use in vivo within 1 week. When combined with the CRISPR-Cas9 system, this provides a straightforward method for knockout of genes of interest in the pancreas. The method can also be used to express cDNAs in the pancreas. This method shows great potential to accelerate pancreatic cancer research in autochthonous models. For complete details on the use and execution of this protocol, please refer to Li et al. (2021).1.
Collapse
Affiliation(s)
- Meilian Zhuo
- Tsinghua University School of Medicine, Beijing, China; Peking University-Tsinghua Center for Life Sciences, Beijing, China.
| | - Jiangjiao Mao
- Tsinghua University School of Medicine, Beijing, China
| | - Charles J David
- Tsinghua University School of Medicine, Beijing, China; Peking University-Tsinghua Center for Life Sciences, Beijing, China.
| |
Collapse
|
31
|
Salimi-Nezhad N, Missault S, Notario-Reinoso A, Hassani A, Amiri M, Keliris GA. The impact of selective and non-selective medial septum stimulation on hippocampal neuronal oscillations: A study based on modeling and experiments. Neurobiol Dis 2023; 180:106052. [PMID: 36822547 DOI: 10.1016/j.nbd.2023.106052] [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: 11/18/2022] [Revised: 02/09/2023] [Accepted: 02/19/2023] [Indexed: 02/23/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with a rising socioeconomic impact on societies. The hippocampus (HPC), which plays an important role in AD, is affected in the early stages. The medial septum (MS) in the forebrain provides major cholinergic input to the HPC and has been shown to play a significant role in generating oscillations in hippocampal neurons. Cholinergic neurons in the basal forebrain are particularly vulnerable to neurodegeneration in AD. To better understand the role of MS neurons including the cholinergic, glutamatergic, and GABAergic subpopulations in generating the well-known brain rhythms in HPC including delta, theta, slow gamma, and fast gamma oscillations, we designed a detailed computational model of the septohippocampal pathway. We validated the results of our model, using electrophysiological recordings in HPC with and without stimulation of the cholinergic neurons in MS using designer receptors exclusively activated by designer drugs (DREADDs) in healthy male ChAT-cre rats. Then, we eliminated 75% of the MS cholinergic neurons in the model to simulate degeneration in AD. A series of selective and non-selective stimulations of the remaining MS neurons were performed to understand the dynamics of oscillation regulation in the HPC during the degenerated state. In this way, appropriate stimulation strategies able to normalize the aberrant oscillations are proposed. We found that selectively stimulating the remaining healthy cholinergic neurons was sufficient for network recovery and compare this to stimulating other subpopulations and a non-selective stimulation of all MS neurons. Our data provide valuable information for the development of new therapeutic strategies in AD and a tool to test and predict the outcome of potential theranostic manipulations.
Collapse
Affiliation(s)
- Nima Salimi-Nezhad
- Medical Biology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Anaïs Notario-Reinoso
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium; Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Atefe Hassani
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahmood Amiri
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Georgios A Keliris
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium; Institute of Computer Science, Foundation for Research and Technology Hellas, Heraklion, Crete, Greece.
| |
Collapse
|
32
|
Jin L, Matsuyama M, Sullivan HA, Zhu M, Lavin TK, Hou Y, Lea NE, Pruner MT, Dam Ferdínez ML, Wickersham IR. "Self-inactivating" rabies viruses are susceptible to loss of their intended attenuating modification. Proc Natl Acad Sci U S A 2023; 120:e2023481120. [PMID: 37053554 PMCID: PMC9963760 DOI: 10.1073/pnas.2023481120] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 11/29/2022] [Indexed: 02/09/2023] Open
Abstract
Monosynaptic tracing using rabies virus is an important technique in neuroscience, allowing brain-wide labeling of neurons directly presynaptic to a targeted neuronal population. A 2017 article reported the development of a noncytotoxic version-a major advance-based on attenuating the rabies virus by the addition of a destabilization domain to the C terminus of a viral protein. However, this modification did not appear to hinder the ability of the virus to spread between neurons. We analyzed two viruses provided by the authors and show here that both were mutants that had lost the intended modification, explaining the paper's paradoxical results. We then made a virus that actually did have the intended modification in at least the majority of virions and found that it did not spread efficiently under the conditions described in the original paper, namely, without an exogenous protease being expressed in order to remove the destabilization domain. We found that it did spread when the protease was supplied, although this also appeared to result in the deaths of most source cells by 3 wk postinjection. We conclude that the new approach is not robust but that it could become a viable technique given further optimization and validation.
Collapse
Affiliation(s)
- Lei Jin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Makoto Matsuyama
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Heather A. Sullivan
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Mulangma Zhu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Thomas K. Lavin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - YuanYuan Hou
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Nicholas E. Lea
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Maxwell T. Pruner
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - María Lucía Dam Ferdínez
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Ian R. Wickersham
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
33
|
The deep cerebellar nuclei to striatum disynaptic connection contributes to skilled forelimb movement. Cell Rep 2023; 42:112000. [PMID: 36656714 DOI: 10.1016/j.celrep.2023.112000] [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: 06/17/2022] [Revised: 12/20/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
Cerebellar-thalamo-striatal synaptic communication has been implicated in a wide range of behaviors, including goal-directed actions, and is altered in cerebellar dystonia. However, its detailed connectivity through the thalamus and its contribution to the execution of forelimb movements is unclear. Here, we use trans-synaptic and retrograde tracing, ex vivo slice recordings, and optogenetic inhibitions during the execution of unidirectional or sequential joystick displacements to demonstrate that the deep cerebellar nuclei (DCN) influence the dorsal striatum with a very high probability. We show that this mainly occurs through the centrolateral (CL), parafascicular (PF), and ventrolateral (VL) nuclei of the thalamus, observing that the DCN→VL and DCN→CL pathways contribute to the execution of unidirectional forelimb displacements while the DCN→PF and DCN→thalamo→striatal pathways contribute to the appropriate execution of forelimb reaching and sequential displacements. These findings highlight specific contributions of the different cerebellar-thalamo-striatal paths to the control of skilled forelimb movement.
Collapse
|
34
|
Okamoto K, Kamikubo Y, Yamauchi K, Okamoto S, Takahashi M, Ishida Y, Koike M, Ikegaya Y, Sakurai T, Hioki H. Specific AAV2/PHP.eB-mediated gene transduction of CA2 pyramidal cells via injection into the lateral ventricle. Sci Rep 2023; 13:323. [PMID: 36609635 PMCID: PMC9822962 DOI: 10.1038/s41598-022-27372-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023] Open
Abstract
Given its limited accessibility, the CA2 area has been less investigated compared to other subregions of the hippocampus. While the development of transgenic mice expressing Cre recombinase in the CA2 has revealed unique features of this area, the use of mouse lines has several limitations, such as lack of specificity. Therefore, a specific gene delivery system is required. Here, we confirmed that the AAV-PHP.eB capsid preferably infected CA2 pyramidal cells following retro-orbital injection and demonstrated that the specificity was substantially higher after injection into the lateral ventricle. In addition, a tropism for the CA2 area was observed in organotypic slice cultures. Combined injection into the lateral ventricle and stereotaxic injection into the CA2 area specifically introduced the transgene into CA2 pyramidal cells, enabling us to perform targeted patch-clamp recordings and optogenetic manipulation. These results suggest that AAV-PHP.eB is a versatile tool for specific gene transduction in CA2 pyramidal cells.
Collapse
Affiliation(s)
- Kazuki Okamoto
- grid.258269.20000 0004 1762 2738Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Juntendo Advanced Research Institute for Health Science, Juntendo University, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Yuji Kamikubo
- grid.258269.20000 0004 1762 2738Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Kenta Yamauchi
- grid.258269.20000 0004 1762 2738Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Juntendo Advanced Research Institute for Health Science, Juntendo University, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Shinichiro Okamoto
- grid.258269.20000 0004 1762 2738Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Juntendo Advanced Research Institute for Health Science, Juntendo University, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Megumu Takahashi
- grid.258269.20000 0004 1762 2738Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258799.80000 0004 0372 2033Department of Neuroscience, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto 606-8501 Japan ,grid.54432.340000 0001 0860 6072Research Fellow of Japan Society for the Promotion of Science (JSPS), Chiyoda-ku, Tokyo, 102-0083 Japan
| | - Yoko Ishida
- grid.258269.20000 0004 1762 2738Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Juntendo Advanced Research Institute for Health Science, Juntendo University, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Masato Koike
- grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Juntendo Advanced Research Institute for Health Science, Juntendo University, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Yuji Ikegaya
- grid.26999.3d0000 0001 2151 536XLaboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo‐ku, Tokyo, 113‐0033 Japan ,grid.28312.3a0000 0001 0590 0962Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita, Osaka 565-0871 Japan ,grid.26999.3d0000 0001 2151 536XInstitute for AI and Beyond, The University of Tokyo, Bunkyo‐ku, Tokyo, 113‐0033 Japan
| | - Takashi Sakurai
- grid.258269.20000 0004 1762 2738Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Hiroyuki Hioki
- Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421, Japan. .,Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421, Japan. .,Department of Multi-Scale Brain Structure Imaging, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
35
|
Swanson JL, Ortiz-Guzman J, Srivastava S, Chin PS, Dooling SW, Hanson Moss E, Kochukov MY, Hunt PJ, Patel JM, Pekarek BT, Tong Q, Arenkiel BR. Activation of basal forebrain-to-lateral habenula circuitry drives reflexive aversion and suppresses feeding behavior. Sci Rep 2022; 12:22044. [PMID: 36543829 PMCID: PMC9772215 DOI: 10.1038/s41598-022-26306-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Environmental cues and internal states such as mood, reward, or aversion directly influence feeding behaviors beyond homeostatic necessity. The hypothalamus has been extensively investigated for its role in homeostatic feeding. However, many of the neural circuits that drive more complex, non-homeostatic feeding that integrate valence and sensory cues (such as taste and smell) remain unknown. Here, we describe a basal forebrain (BF)-to-lateral habenula (LHb) circuit that directly modulates non-homeostatic feeding behavior. Using viral-mediated circuit mapping, we identified a population of glutamatergic neurons within the BF that project to the LHb, which responds to diverse sensory cues, including aversive and food-related odors. Optogenetic activation of BF-to-LHb circuitry drives robust, reflexive-like aversion. Furthermore, activation of this circuitry suppresses the drive to eat in a fasted state. Together, these data reveal a role of basal forebrain glutamatergic neurons in modulating LHb-associated aversion and feeding behaviors by sensing environmental cues.
Collapse
Affiliation(s)
- Jessica L Swanson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Joshua Ortiz-Guzman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Snigdha Srivastava
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Pey-Shyuan Chin
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Sean W Dooling
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Elizabeth Hanson Moss
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Mikhail Y Kochukov
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Patrick J Hunt
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Jay M Patel
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Brandon T Pekarek
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Qingchun Tong
- Center for Metabolic and Degenerative Disease, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
36
|
Lewis JE, Woodward OR, Nuzzaci D, Smith CA, Adriaenssens AE, Billing L, Brighton C, Phillips BU, Tadross JA, Kinston SJ, Ciabatti E, Göttgens B, Tripodi M, Hornigold D, Baker D, Gribble FM, Reimann F. Relaxin/insulin-like family peptide receptor 4 (Rxfp4) expressing hypothalamic neurons modulate food intake and preference in mice. Mol Metab 2022; 66:101604. [PMID: 36184065 PMCID: PMC9579047 DOI: 10.1016/j.molmet.2022.101604] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Insulin-like peptide 5 (INSL5) signalling, through its cognate receptor relaxin/insulin-like family peptide receptor 4 (RXFP4), has been reported to be orexigenic, and the high fat diet (HFD) preference observed in wildtype mice is altered in Rxfp4 knock-out mice. In this study, we used a new Rxfp4-Cre mouse model to investigate the mechanisms underlying these observations. METHODS We generated transgenic Rxfp4-Cre mice and investigated central expression of Rxfp4 by RT-qPCR, RNAscope and intraparenchymal infusion of INSL5. Rxfp4-expressing cells were chemogenetically manipulated in global Cre-reporter mice using designer receptors exclusively activated by designer drugs (DREADDs) or after stereotactic injection of a Cre-dependent AAV-DIO-Dq-DREADD targeting a population located in the ventromedial hypothalamus (RXFP4VMH). Food intake and feeding motivation were assessed in the presence and absence of a DREADD agonist. Rxfp4-expressing cells in the hypothalamus were characterised by single-cell RNA-sequencing (scRNAseq) and the connectivity of RXFP4VMH cells was investigated using viral tracing. RESULTS Rxfp4-Cre mice displayed Cre-reporter expression in the hypothalamus. Active expression of Rxfp4 in the adult mouse brain was confirmed by RT-qPCR and RNAscope. Functional receptor expression was supported by cyclic AMP-responses to INSL5 application in ex vivo brain slices and increased HFD and highly palatable liquid meal (HPM), but not chow, intake after intra-VMH INSL5 infusion. scRNAseq of hypothalamic RXFP4 neurons defined a cluster expressing VMH markers, alongside known appetite-modulating neuropeptide receptors (Mc4r, Cckar and Nmur2). Viral tracing demonstrated RXFP4VMH neural projections to nuclei implicated in hedonic feeding behaviour. Whole body chemogenetic inhibition (Di-DREADD) of Rxfp4-expressing cells, mimicking physiological INSL5-RXFP4 Gi-signalling, increased intake of the HFD and HPM, but not chow, whilst activation (Dq-DREADD), either at whole body level or specifically within the VMH, reduced HFD and HPM intake and motivation to work for the HPM. CONCLUSION These findings identify RXFP4VMH neurons as regulators of food intake and preference, and hypothalamic RXFP4 signalling as a target for feeding behaviour manipulation.
Collapse
Affiliation(s)
- Jo E Lewis
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Orla Rm Woodward
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Danaé Nuzzaci
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Christopher A Smith
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Alice E Adriaenssens
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Lawrence Billing
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Cheryl Brighton
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Benjamin U Phillips
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - John A Tadross
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK; Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Sarah J Kinston
- Department of Haematology, Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Ernesto Ciabatti
- Department of Haematology, Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Berthold Göttgens
- Department of Haematology, Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Marco Tripodi
- MRC Laboratory of Molecular Biology, Neurobiology Division, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - David Hornigold
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca Ltd, Cambridge, UK
| | - David Baker
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca Ltd, Cambridge, UK
| | - Fiona M Gribble
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Frank Reimann
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
37
|
Saunders A, Huang KW, Vondrak C, Hughes C, Smolyar K, Sen H, Philson AC, Nemesh J, Wysoker A, Kashin S, Sabatini BL, McCarroll SA. Ascertaining cells' synaptic connections and RNA expression simultaneously with barcoded rabies virus libraries. Nat Commun 2022; 13:6993. [PMID: 36384944 PMCID: PMC9668842 DOI: 10.1038/s41467-022-34334-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/21/2022] [Indexed: 11/17/2022] Open
Abstract
Brain function depends on synaptic connections between specific neuron types, yet systematic descriptions of synaptic networks and their molecular properties are not readily available. Here, we introduce SBARRO (Synaptic Barcode Analysis by Retrograde Rabies ReadOut), a method that uses single-cell RNA sequencing to reveal directional, monosynaptic relationships based on the paths of a barcoded rabies virus from its "starter" postsynaptic cell to that cell's presynaptic partners. Thousands of these partner relationships can be ascertained in a single experiment, alongside genome-wide RNAs. We use SBARRO to describe synaptic networks formed by diverse mouse brain cell types in vitro, finding that different cell types have presynaptic networks with differences in average size and cell type composition. Patterns of RNA expression suggest that functioning synapses are critical for rabies virus uptake. By tracking individual rabies clones across cells, SBARRO offers new opportunities to map the synaptic organization of neural circuits.
Collapse
Affiliation(s)
- Arpiar Saunders
- grid.38142.3c000000041936754XDepartment of Genetics, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.340000 0004 0546 1623Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA ,grid.5288.70000 0000 9758 5690Vollum Institute, Oregon Health & Science University, Portland, OR 97239 USA
| | - Kee Wui Huang
- grid.38142.3c000000041936754XHoward Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115 USA
| | - Cassandra Vondrak
- grid.38142.3c000000041936754XDepartment of Genetics, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.340000 0004 0546 1623Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Christina Hughes
- grid.38142.3c000000041936754XDepartment of Genetics, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.340000 0004 0546 1623Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Karina Smolyar
- grid.38142.3c000000041936754XDepartment of Genetics, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.340000 0004 0546 1623Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Harsha Sen
- grid.38142.3c000000041936754XDepartment of Genetics, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.340000 0004 0546 1623Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Adrienne C. Philson
- grid.38142.3c000000041936754XHoward Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115 USA
| | - James Nemesh
- grid.38142.3c000000041936754XDepartment of Genetics, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.340000 0004 0546 1623Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Alec Wysoker
- grid.38142.3c000000041936754XDepartment of Genetics, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.340000 0004 0546 1623Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Seva Kashin
- grid.38142.3c000000041936754XDepartment of Genetics, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.340000 0004 0546 1623Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Bernardo L. Sabatini
- grid.38142.3c000000041936754XHoward Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115 USA
| | - Steven A. McCarroll
- grid.38142.3c000000041936754XDepartment of Genetics, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.340000 0004 0546 1623Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| |
Collapse
|
38
|
Okoro SU, Goz RU, Njeri BW, Harish M, Ruff CF, Ross SE, Gerfen C, Hooks BM. Organization of Cortical and Thalamic Input to Inhibitory Neurons in Mouse Motor Cortex. J Neurosci 2022; 42:8095-8112. [PMID: 36104281 PMCID: PMC9637002 DOI: 10.1523/jneurosci.0950-22.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 11/21/2022] Open
Abstract
Intracortical inhibition in motor cortex (M1) regulates movement and motor learning. If cortical and thalamic inputs target different inhibitory cell types in different layers, then these afferents may play different roles in regulating M1 output. Using mice of both sexes, we quantified input to two main classes of M1 interneurons, parvalbumin+ (PV+) cells and somatostatin+ (SOM+) cells, using monosynaptic rabies tracing. We then compared anatomic and functional connectivity based on synaptic strength from sensory cortex and thalamus. Functionally, each input innervated M1 interneurons with a unique laminar profile. Different interneuron types were excited in a distinct, complementary manner, suggesting feedforward inhibition proceeds selectively via distinct circuits. Specifically, somatosensory cortex (S1) inputs primarily targeted PV+ neurons in upper layers (L2/3) but SOM+ neurons in middle layers (L5). Somatosensory thalamus [posterior nucleus (PO)] inputs targeted PV+ neurons in middle layers (L5). In contrast to sensory cortical areas, thalamic input to SOM+ neurons was equivalent to that of PV+ neurons. Thus, long-range excitatory inputs target inhibitory neurons in an area and a cell type-specific manner, which contrasts with input to neighboring pyramidal cells. In contrast to feedforward inhibition providing generic inhibitory tone in cortex, circuits are selectively organized to recruit inhibition matched to incoming excitatory circuits.SIGNIFICANCE STATEMENT M1 integrates sensory information and frontal cortical inputs to plan and control movements. Although inputs to excitatory cells are described, the synaptic circuits by which these inputs drive specific types of M1 interneurons are unknown. Anatomical results with rabies tracing and physiological quantification of synaptic strength shows that two main classes of inhibitory cells (PV+ and SOM+ interneurons) both receive substantial cortical and thalamic input, in contrast to interneurons in sensory areas (where thalamic input strongly prefers PV+ interneurons). Further, each input studied targets PV+ and SOM+ interneurons in a different fashion, suggesting that separate, specific circuits exist for recruitment of feedforward inhibition.
Collapse
Affiliation(s)
- Sandra U Okoro
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Roman U Goz
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Brigdet W Njeri
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Madhumita Harish
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Catherine F Ruff
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Sarah E Ross
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Charles Gerfen
- National Institute of Mental Health, Bethesda, Maryland 20892
| | - Bryan M Hooks
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
39
|
Nanobody-based RFP-dependent Cre recombinase for selective anterograde tracing in RFP-expressing transgenic animals. Commun Biol 2022; 5:979. [PMID: 36114373 PMCID: PMC9481622 DOI: 10.1038/s42003-022-03944-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 09/05/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractTransgenic animals expressing fluorescent proteins are widely used to label specific cells and proteins. By using a split Cre recombinase fused with mCherry-binding nanobodies or designed ankyrin repeat proteins, we created Cre recombinase dependent on red fluorescent protein (RFP) (Cre-DOR). Functional binding units for monomeric RFPs are different from those for polymeric RFPs. We confirmed selective target RFP-dependent gene expression in the mouse cerebral cortex using stereotaxic injection of adeno-associated virus vectors. In estrogen receptor-beta (Esr2)-mRFP1 mice and gastrin-releasing peptide receptor (Grpr)-mRFP1 rats, we confirmed that Cre-DOR can be used for selective tracing of the neural projection from RFP-expressing specific neurons. Cellular localization of RFPs affects recombination efficiency of Cre-DOR, and light and chemical-induced nuclear translocation of an RFP-fused protein can modulate Cre-DOR efficiency. Our results provide a method for manipulating gene expression in specific cells expressing RFPs and expand the repertory of nanobody-based genetic tools.
Collapse
|
40
|
Laing BT, Jayan A, Erbaugh LJ, Park AS, Wilson DJ, Aponte Y. Regulation of body weight and food intake by AGRP neurons during opioid dependence and abstinence in mice. Front Neural Circuits 2022; 16:977642. [PMID: 36110920 PMCID: PMC9468932 DOI: 10.3389/fncir.2022.977642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of body weight maintenance and opioid dependence are often treated as independent disorders. Here, we assessed the effects of both acute and long-term administration of morphine with and without chemogenetic activation of agouti-related peptide (AGRP)-expressing neurons in the arcuate nucleus (ARCAGRP neurons) to elucidate whether morphine and neuronal activation affect feeding behavior and body weight. First, we characterized interactions of opioids and energy deficit in wild-type mice. We observed that opioid administration attenuated both fasting-induced refeeding and ghrelin-stimulated feeding. Moreover, antagonism of opioid receptors blocked fasting-induced refeeding behavior. Next, we interfaced chemogenetics with opioid dependence. For chemogenetic experiments of ARCAGRP neurons, we conducted a priori behavioral qualification and post-mortem FOS immunostaining verification of arcuate activation following ARCAGRP chemogenetic activation. We administered clozapine during short-term and long-term morphine administration paradigms to determine the effects of dependence on food intake and body weight. We found that morphine occluded feeding behavior characteristic of chemogenetic activation of ARCAGRP neurons. Notably, activation of ARCAGRP neurons attenuated opioid-induced weight loss but did not evoke weight gain during opioid dependence. Consistent with these findings, we observed that morphine administration did not block fasting-induced activation of the ARC. Together, these results highlight the strength of opioidergic effects on body weight maintenance and demonstrate the utility of ARCAGRP neuron manipulations as a lever to influence energy balance throughout the development of opioid dependence.
Collapse
Affiliation(s)
- Brenton T. Laing
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Aishwarya Jayan
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Lydia J. Erbaugh
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Anika S. Park
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Danielle J. Wilson
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Yeka Aponte
- Neuronal Circuits and Behavior Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
41
|
Agnew-Svoboda W, Ubina T, Figueroa Z, Wong YC, Vizcarra EA, Roebini B, Wilson EH, Fiacco TA, Riccomagno MM. A genetic tool for the longitudinal study of a subset of post-inflammatory reactive astrocytes. CELL REPORTS METHODS 2022; 2:100276. [PMID: 36046623 PMCID: PMC9421582 DOI: 10.1016/j.crmeth.2022.100276] [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] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 06/01/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022]
Abstract
Astrocytes are vital support cells that ensure proper brain function. In brain disease, astrocytes reprogram into a reactive state that alters many of their cellular roles. A long-standing question in the field is whether downregulation of reactive astrocyte (RA) markers during resolution of inflammation is because these astrocytes revert back to a non-reactive state or die and are replaced. This has proven difficult to answer mainly because existing genetic tools cannot distinguish between healthy versus RAs. Here we describe the generation of an inducible genetic tool that can be used to specifically target and label a subset of RAs. Longitudinal analysis of an acute inflammation model using this tool revealed that the previously observed downregulation of RA markers after inflammation is likely due to changes in gene expression and not because of cell death. Our findings suggest that cellular changes associated with astrogliosis after acute inflammation are largely reversible.
Collapse
Affiliation(s)
- William Agnew-Svoboda
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Teresa Ubina
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Zoe Figueroa
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
| | - Yiu-Cheung Wong
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Edward A. Vizcarra
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| | - Bryan Roebini
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Emma H. Wilson
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| | - Todd A. Fiacco
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| | - Martin M. Riccomagno
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| |
Collapse
|
42
|
Sciolino NR, Hsiang M, Mazzone CM, Wilson LR, Plummer NW, Amin J, Smith KG, McGee CA, Fry SA, Yang CX, Powell JM, Bruchas MR, Kravitz AV, Cushman JD, Krashes MJ, Cui G, Jensen P. Natural locus coeruleus dynamics during feeding. SCIENCE ADVANCES 2022; 8:eabn9134. [PMID: 35984878 PMCID: PMC9390985 DOI: 10.1126/sciadv.abn9134] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 07/07/2022] [Indexed: 06/15/2023]
Abstract
Recent data demonstrate that noradrenergic neurons of the locus coeruleus (LC-NE) are required for fear-induced suppression of feeding, but the role of endogenous LC-NE activity in natural, homeostatic feeding remains unclear. Here, we found that LC-NE activity was suppressed during food consumption, and the magnitude of this neural response was attenuated as mice consumed more pellets throughout the session, suggesting that LC responses to food are modulated by satiety state. Visual-evoked LC-NE activity was also attenuated in sated mice, suggesting that satiety state modulates LC-NE encoding of multiple behavioral states. We also found that food intake could be attenuated by brief or longer durations of LC-NE activation. Last, we found that activation of the LC to the lateral hypothalamus pathway suppresses feeding and enhances avoidance and anxiety-like responding. Our findings suggest that LC-NE neurons modulate feeding by integrating both external cues (e.g., anxiogenic environmental cues) and internal drives (e.g., satiety).
Collapse
Affiliation(s)
- Natale R. Sciolino
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Madeline Hsiang
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Christopher M. Mazzone
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Leslie R. Wilson
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Nicholas W. Plummer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Jaisal Amin
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Kathleen G. Smith
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Christopher A. McGee
- Comparative Medicine, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Sydney A. Fry
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Cindy X. Yang
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Jeanne M. Powell
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Michael R. Bruchas
- Departments of Anesthesiology and Pharmacology, Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | | | - Jesse D. Cushman
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Michael J. Krashes
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Guohong Cui
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Patricia Jensen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| |
Collapse
|
43
|
Ansai S, Kitano J. Speciation and adaptation research meets genome editing. Philos Trans R Soc Lond B Biol Sci 2022; 377:20200516. [PMID: 35634923 PMCID: PMC9149800 DOI: 10.1098/rstb.2020.0516] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 02/07/2022] [Indexed: 07/20/2023] Open
Abstract
Understanding the genetic basis of reproductive isolation and adaptive traits in natural populations is one of the fundamental goals in evolutionary biology. Genome editing technologies based on CRISPR-Cas systems and site-specific recombinases have enabled us to modify a targeted genomic region as desired and thus to conduct functional analyses of target loci, genes and mutations even in non-conventional model organisms. Here, we review the technical properties of genome editing techniques by classifying them into the following applications: targeted gene knock-out for investigating causative gene functions, targeted gene knock-in of marker genes for visualizing expression patterns and protein functions, precise gene replacement for identifying causative alleles and mutations, and targeted chromosomal rearrangement for investigating the functional roles of chromosomal structural variations. We describe examples of their application to demonstrate functional analysis of naturally occurring genetic variations and discuss how these technologies can be applied to speciation and adaptation research. This article is part of the theme issue 'Genetic basis of adaptation and speciation: from loci to causative mutations'.
Collapse
Affiliation(s)
- Satoshi Ansai
- Graduate School of Life Sciences, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Jun Kitano
- Ecological Genetics Laboratory, Department of Genomics and Evolutionary Biology, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 411-8540, Japan
| |
Collapse
|
44
|
Efficient spatially targeted gene editing using a near-infrared activatable protein-conjugated nanoparticle for brain applications. Nat Commun 2022; 13:4135. [PMID: 35840564 PMCID: PMC9287341 DOI: 10.1038/s41467-022-31791-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 07/05/2022] [Indexed: 12/27/2022] Open
Abstract
Spatial control of gene expression is critical to modulate cellular functions and deconstruct the function of individual genes in biological processes. Light-responsive gene-editing formulations have been recently developed; however, they have shown limited applicability in vivo due to poor tissue penetration, limited cellular transfection and the difficulty in evaluating the activity of the edited cells. Here, we report a formulation composed of upconversion nanoparticles conjugated with Cre recombinase enzyme through a photocleavable linker, and a lysosomotropic agent that facilitates endolysosomal escape. This formulation allows in vitro spatial control in gene editing after activation with near-infrared light. We further demonstrate the potential of this formulation in vivo through three different paradigms: (i) gene editing in neurogenic niches, (ii) gene editing in the ventral tegmental area to facilitate monitoring of edited cells by precise optogenetic control of reward and reinforcement, and (iii) gene editing in a localized brain region via a noninvasive administration route (i.e., intranasal). Spatial control of gene expression allows precise control over biological processes. Here, the authors develop an efficient light-responsive formulation based on upconversion nanoparticles, and demonstrate on-demand genetic manipulation in deep brain tissue.
Collapse
|
45
|
Chadwick SR, Güler AD. Local Drd1-neurons input to subgroups of arcuate AgRP/NPY-neurons. iScience 2022; 25:104605. [PMID: 35789850 PMCID: PMC9250019 DOI: 10.1016/j.isci.2022.104605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/02/2022] [Accepted: 06/08/2022] [Indexed: 11/21/2022] Open
Abstract
Obesity is a pandemic afflicting more than 300 million people worldwide, driven by consumption of calorically dense and highly rewarding foods. Dopamine (DA) signaling has been implicated in neural responses to highly palatable nutrients, but the exact mechanisms through which DA modulates homeostatic feeding circuits remains unknown. A subpopulation of arcuate (ARC) agouti-related peptide (AgRP)/neuropeptide Y (NPY) (ARCAgRP/NPY+) neurons express the D(1A) dopamine receptor (Drd1) and are stimulated by DA, suggesting one potential avenue for dopaminergic regulation of food intake. Using patch clamp electrophysiology, we evaluated the responses of ARC Drd1-expressing (ARCDrd1+) neurons to overnight fasting and leptin. Collectively, ARCDrd1+ neurons were less responsive to caloric deficit than ARCAgRP/NPY+ neurons; however, ARCDrd1+ neurons were inhibited by the satiety hormone leptin. Using Channelrhodopsin-2-Assisted Circuit Mapping, we identified novel subgroups of ARCDrd1+ neurons that inhibit or excite ARCAgRP/NPY+ neurons. These findings suggest dopamine receptive neurons have multimodal actions in food intake circuits.
Collapse
Affiliation(s)
- Sean R. Chadwick
- Program in Fundamental Neuroscience and the Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Ali D. Güler
- Program in Fundamental Neuroscience and the Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
46
|
Pouchelon G, Vergara J, McMahon J, Gorissen BL, Lin JD, Vormstein-Schneider D, Niehaus JL, Burbridge TJ, Wester JC, Sherer M, Fernandez-Otero M, Allaway KC, Pelkey K, Chittajallu R, McBain CJ, Fan M, Nasse JS, Wildenberg GA, Fishell G, Dimidschstein J. A versatile viral toolkit for functional discovery in the nervous system. CELL REPORTS METHODS 2022; 2:100225. [PMID: 35784651 PMCID: PMC9243523 DOI: 10.1016/j.crmeth.2022.100225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/21/2022] [Accepted: 05/04/2022] [Indexed: 12/24/2022]
Abstract
The ability to precisely control transgene expression is essential for basic research and clinical applications. Adeno-associated viruses (AAVs) are non-pathogenic and can be used to drive stable expression in virtually any tissue, cell type, or species, but their limited genomic payload results in a trade-off between the transgenes that can be incorporated and the complexity of the regulatory elements controlling their expression. Resolving these competing imperatives in complex experiments inevitably results in compromises. Here, we assemble an optimized viral toolkit (VTK) that addresses these limitations and allows for efficient combinatorial targeting of cell types. Moreover, their modular design explicitly enables further refinements. We achieve this in compact vectors by integrating structural improvements of AAV vectors with innovative molecular tools. We illustrate the potential of this approach through a systematic demonstration of their utility for targeting cell types and querying their biology using a wide array of genetically encoded tools.
Collapse
Affiliation(s)
- Gabrielle Pouchelon
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Josselyn Vergara
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | - Justin McMahon
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | - Bram L. Gorissen
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Jessica D. Lin
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | | | | | - Timothy J. Burbridge
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Jason C. Wester
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Mia Sherer
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Marian Fernandez-Otero
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Kathryn C. Allaway
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Kenneth Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Chris J. McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | | | | | - Gregg A. Wildenberg
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
- Biosciences Division, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Gordon Fishell
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | | |
Collapse
|
47
|
Swanson JL, Chin PS, Romero JM, Srivastava S, Ortiz-Guzman J, Hunt PJ, Arenkiel BR. Advancements in the Quest to Map, Monitor, and Manipulate Neural Circuitry. Front Neural Circuits 2022; 16:886302. [PMID: 35719420 PMCID: PMC9204427 DOI: 10.3389/fncir.2022.886302] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/27/2022] [Indexed: 01/27/2023] Open
Abstract
Neural circuits and the cells that comprise them represent the functional units of the brain. Circuits relay and process sensory information, maintain homeostasis, drive behaviors, and facilitate cognitive functions such as learning and memory. Creating a functionally-precise map of the mammalian brain requires anatomically tracing neural circuits, monitoring their activity patterns, and manipulating their activity to infer function. Advancements in cell-type-specific genetic tools allow interrogation of neural circuits with increased precision. This review provides a broad overview of recombination-based and activity-driven genetic targeting approaches, contemporary viral tracing strategies, electrophysiological recording methods, newly developed calcium, and voltage indicators, and neurotransmitter/neuropeptide biosensors currently being used to investigate circuit architecture and function. Finally, it discusses methods for acute or chronic manipulation of neural activity, including genetically-targeted cellular ablation, optogenetics, chemogenetics, and over-expression of ion channels. With this ever-evolving genetic toolbox, scientists are continuing to probe neural circuits with increasing resolution, elucidating the structure and function of the incredibly complex mammalian brain.
Collapse
Affiliation(s)
- Jessica L. Swanson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Pey-Shyuan Chin
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Juan M. Romero
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Snigdha Srivastava
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Joshua Ortiz-Guzman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Patrick J. Hunt
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Benjamin R. Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
48
|
Ledderose JMT, Benitez JA, Roberts AJ, Reed R, Bintig W, Larkum ME, Sachdev RNS, Furnari F, Eickholt BJ. The impact of phosphorylated PTEN at threonine 366 on cortical connectivity and behaviour. Brain 2022; 145:3608-3621. [PMID: 35603900 DOI: 10.1093/brain/awac188] [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: 09/29/2021] [Revised: 04/19/2022] [Accepted: 05/04/2022] [Indexed: 11/14/2022] Open
Abstract
The lipid phosphatase PTEN (phosphatase and tensin homologue on chromosome 10) is a key tumour suppressor gene and an important regulator of neuronal signalling. PTEN mutations have been identified in patients with autism spectrum disorders, characterized by macrocephaly, impaired social interactions and communication, repetitive behaviour, intellectual disability, and epilepsy. PTEN enzymatic activity is regulated by a cluster of phosphorylation sites at the C-terminus of the protein. Here, we focussed on the role of PTEN T366 phosphorylation and generated a knock-in mouse line in which Pten T366 was substituted with alanine (PtenT366A/T366A). We identify that phosphorylation of PTEN at T366 controls neuron size and connectivity of brain circuits involved in sensory processing. We show in behavioural tests that PtenT366/T366A mice exhibit cognitive deficits and selective sensory impairments, with significant differences in male individuals. We identify restricted cellular overgrowth of cortical neurons in PtenT366A/T366A brains, linked to increases in both dendritic arborization and soma size. In a combinatorial approach of anterograde and retrograde monosynaptic tracing using rabies virus, we characterize differences in connectivity to the primary somatosensory cortex of PtenT366A/T366A brains, with imbalances in long-range cortico-cortical input to neurons. We conclude that phosphorylation of PTEN at T366 controls neuron size and connectivity of brain circuits involved in sensory processing and propose that PTEN T366 signalling may account for a subset of autism-related functions of PTEN.
Collapse
Affiliation(s)
- Julia M T Ledderose
- Institute for Biochemistry, Charité Universitätsmedizin Berlin, Germany.,Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jorge A Benitez
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California, 92121, USA
| | - Amanda J Roberts
- The Scripps Research Institute, Animal Models Core, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Rachel Reed
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California, 92121, USA
| | - Willem Bintig
- Institute for Biochemistry, Charité Universitätsmedizin Berlin, Germany
| | - Matthew E Larkum
- Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany.,Neurocure Center for Excellence, Charité Universitätsmedizin Berlin, Germany
| | | | - Frank Furnari
- Ludwig Cancer Institute, San Diego, USA.,University of California San Diego, La Jolla, USA
| | - Britta J Eickholt
- Institute for Biochemistry, Charité Universitätsmedizin Berlin, Germany.,Neurocure Center for Excellence, Charité Universitätsmedizin Berlin, Germany
| |
Collapse
|
49
|
Cai X, Liu H, Feng B, Yu M, He Y, Liu H, Liang C, Yang Y, Tu L, Zhang N, Wang L, Yin N, Han J, Yan Z, Wang C, Xu P, Wu Q, Tong Q, He Y, Xu Y. A D2 to D1 shift in dopaminergic inputs to midbrain 5-HT neurons causes anorexia in mice. Nat Neurosci 2022; 25:646-658. [PMID: 35501380 PMCID: PMC9926508 DOI: 10.1038/s41593-022-01062-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/24/2022] [Indexed: 12/18/2022]
Abstract
Midbrain dopamine (DA) and serotonin (5-HT) neurons regulate motivated behaviors, including feeding, but less is known about how these circuits may interact. In this study, we found that DA neurons in the mouse ventral tegmental area bidirectionally regulate the activity of 5-HT neurons in the dorsal raphe nucleus (DRN), with weaker stimulation causing DRD2-dependent inhibition and overeating, while stronger stimulation causing DRD1-dependent activation and anorexia. Furthermore, in the activity-based anorexia (ABA) paradigm, which is a mouse model mimicking some clinical features of human anorexia nervosa (AN), we observed a DRD2 to DRD1 shift of DA neurotransmission on 5-HTDRN neurons, which causes constant activation of these neurons and contributes to AN-like behaviors. Finally, we found that systemic administration of a DRD1 antagonist can prevent anorexia and weight loss in ABA. Our results revealed regulation of feeding behavior by stimulation strength-dependent interactions between DA and 5-HT neurons, which may contribute to the pathophysiology of AN.
Collapse
Affiliation(s)
- Xing Cai
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Present address: Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,These authors contributed equally: Xing Cai, Hailan Liu
| | - Hailan Liu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,These authors contributed equally: Xing Cai, Hailan Liu
| | - Bing Feng
- Brain Glycemic and Metabolism Control Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Meng Yu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yang He
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Hesong Liu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Chen Liang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yongjie Yang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Longlong Tu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Nan Zhang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Lina Wang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Na Yin
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Junying Han
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Zili Yan
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Chunmei Wang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Pingwen Xu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Present address: Division of Endocrinology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Qi Wu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yanlin He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA. .,Brain Glycemic and Metabolism Control Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA.
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
50
|
Hui Y, Zheng X, Zhang H, Li F, Yu G, Li J, Zhang J, Gong X, Guo G. Strategies for Targeting Neural Circuits: How to Manipulate Neurons Using Virus Vehicles. Front Neural Circuits 2022; 16:882366. [PMID: 35571271 PMCID: PMC9099413 DOI: 10.3389/fncir.2022.882366] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/07/2022] [Indexed: 01/02/2023] Open
Abstract
Viral strategies are the leading methods for mapping neural circuits. Viral vehicles combined with genetic tools provide the possibility to visualize entire functional neural networks and monitor and manipulate neural circuit functions by high-resolution cell type- and projection-specific targeting. Optogenetics and chemogenetics drive brain research forward by exploring causal relationships among different brain regions. Viral strategies offer a fresh perspective for the analysis of the structure-function relationship of the neural circuitry. In this review, we summarize current and emerging viral strategies for targeting neural circuits and focus on adeno-associated virus (AAV) vectors.
Collapse
Affiliation(s)
- Yuqing Hui
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xuefeng Zheng
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Huijie Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Fang Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Guangyin Yu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Jiong Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Jifeng Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- Jifeng Zhang,
| | - Xiaobing Gong
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Xiaobing Gong,
| | - Guoqing Guo
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- *Correspondence: Guoqing Guo,
| |
Collapse
|