1
|
Song Y, Fothergill LJ, Lee KS, Liu BY, Koo A, Perelis M, Diwakarla S, Callaghan B, Huang J, Wykosky J, Furness JB, Yeo GW. Stratification of enterochromaffin cells by single-cell expression analysis. eLife 2025; 12:RP90596. [PMID: 40184163 PMCID: PMC11970908 DOI: 10.7554/elife.90596] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Dynamic interactions between gut mucosal cells and the external environment are essential to maintain gut homeostasis. Enterochromaffin (EC) cells transduce both chemical and mechanical signals and produce 5-hydroxytryptamine to mediate disparate physiological responses. However, the molecular and cellular basis for functional diversity of ECs remains to be adequately defined. Here, we integrated single-cell transcriptomics with spatial image analysis to identify 14 EC clusters that are topographically organized along the gut. Subtypes predicted to be sensitive to the chemical environment and mechanical forces were identified that express distinct transcription factors and hormones. A Piezo2+ population in the distal colon was endowed with a distinctive neuronal signature. Using a combination of genetic, chemogenetic, and pharmacological approaches, we demonstrated Piezo2+ ECs are required for normal colon motility. Our study constructs a molecular map for ECs and offers a framework for deconvoluting EC cells with pleiotropic functions.
Collapse
Affiliation(s)
- Yan Song
- Department of Cellular and Molecular Medicine, University of California San DiegoLa JollaUnited States
- Stem Cell Program, University of California San DiegoLa JollaUnited States
- Institute for Genomic Medicine, University of California San DiegoLa JollaUnited States
| | - Linda J Fothergill
- Department of Anatomy & Physiology, University of MelbourneParkvilleAustralia
- Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
| | - Kari S Lee
- Department of Cellular and Molecular Medicine, University of California San DiegoLa JollaUnited States
- Stem Cell Program, University of California San DiegoLa JollaUnited States
- Institute for Genomic Medicine, University of California San DiegoLa JollaUnited States
| | - Brandon Y Liu
- Department of Cellular and Molecular Medicine, University of California San DiegoLa JollaUnited States
- Stem Cell Program, University of California San DiegoLa JollaUnited States
- Institute for Genomic Medicine, University of California San DiegoLa JollaUnited States
| | - Ada Koo
- Department of Anatomy & Physiology, University of MelbourneParkvilleAustralia
| | - Mark Perelis
- Department of Cellular and Molecular Medicine, University of California San DiegoLa JollaUnited States
- Stem Cell Program, University of California San DiegoLa JollaUnited States
- Institute for Genomic Medicine, University of California San DiegoLa JollaUnited States
| | - Shanti Diwakarla
- Department of Anatomy & Physiology, University of MelbourneParkvilleAustralia
| | - Brid Callaghan
- Department of Anatomy & Physiology, University of MelbourneParkvilleAustralia
| | - Jie Huang
- Takeda PharmaceuticalsSan DiegoUnited States
| | | | - John B Furness
- Department of Anatomy & Physiology, University of MelbourneParkvilleAustralia
- Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California San DiegoLa JollaUnited States
- Stem Cell Program, University of California San DiegoLa JollaUnited States
- Institute for Genomic Medicine, University of California San DiegoLa JollaUnited States
| |
Collapse
|
2
|
Giovanniello JR, Paredes N, Wiener A, Ramírez-Armenta K, Oragwam C, Uwadia HO, Yu AL, Lim K, Pimenta JS, Vilchez GE, Nnamdi G, Wang A, Sehgal M, Reis FM, Sias AC, Silva AJ, Adhikari A, Malvaez M, Wassum KM. A dual-pathway architecture for stress to disrupt agency and promote habit. Nature 2025; 640:722-731. [PMID: 39972126 PMCID: PMC12011321 DOI: 10.1038/s41586-024-08580-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/27/2024] [Indexed: 02/21/2025]
Abstract
Chronic stress can change how we learn and, thus, how we make decisions1-5. Here we investigated the neuronal circuit mechanisms that enable this. Using a multifaceted systems neuroscience approach in male and female mice, we reveal a dual-pathway, amygdala-striatal neuronal circuit architecture by which a recent history of chronic stress disrupts the action-outcome learning underlying adaptive agency and promotes the formation of inflexible habits. We found that the projection from the basolateral amygdala to the dorsomedial striatum is activated by rewarding events to support the action-outcome learning needed for flexible, goal-directed decision-making. Chronic stress attenuates this to disrupt action-outcome learning and, therefore, agency. Conversely, the projection from the central amygdala to the dorsomedial striatum mediates habit formation. Following stress, this pathway is progressively recruited to learning to promote the premature formation of inflexible habits. Thus, stress exerts opposing effects on two amygdala-striatal pathways to disrupt agency and promote habit. These data provide neuronal circuit insights into how chronic stress shapes learning and decision-making, and help understanding of how stress can lead to the disrupted decision-making and pathological habits that characterize substance use disorders and mental health conditions.
Collapse
Affiliation(s)
| | - Natalie Paredes
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anna Wiener
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Chukwuebuka Oragwam
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hanniel O Uwadia
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Abigail L Yu
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kayla Lim
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jenna S Pimenta
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gabriela E Vilchez
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gift Nnamdi
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alicia Wang
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Megha Sehgal
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Fernando McV Reis
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ana C Sias
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alcino J Silva
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Avishek Adhikari
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Melissa Malvaez
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kate M Wassum
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA.
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA.
- Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Keith RE, Shen Y, Janzen-Meza JA, Abramovitz J, Antonello PC, Hameed A, Mohana Krishnan B, Antoine MW. Perirhinal cortex abnormalities impair hippocampal plasticity and learning in Scn2a, Fmr1, and Cdkl5 autism mouse models. SCIENCE ADVANCES 2025; 11:eadt0780. [PMID: 40053578 PMCID: PMC11887805 DOI: 10.1126/sciadv.adt0780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
Learning and memory deficits, including spatial navigation difficulties, are common in autism spectrum disorder (ASD). Several ASD mouse models (Scn2a+/-, Fmr1-/-, Cdkl5-/-) exhibit impaired spatial learning, with these deficits often attributed to hippocampal dysfunction. However, we identify the perirhinal cortex (PRC) as a critical driver of these deficits. Cortical-wide Scn2a reduction in excitatory neurons replicated the spatial learning and long-term potentiation (LTP) impairments-a cellular correlate of learning-seen in Scn2a+/- mice, while hippocampal-wide reduction did not. PRC-specific viral-mediated Scn2a reduction in excitatory neurons decreased release probability, which consequently disrupted synaptic transmission and LTP in the hippocampus, as well as spatial learning. As PRC activity was reduced, chemogenetic activation of the PRC reversed these deficits in Scn2a+/- mice and rescued spatial learning and LTP impairments in Fmr1 and Cdkl5 knockout mice. Thus, in several genetic models of ASD, PRC abnormalities may disrupt hippocampal function to impair learning and memory.
Collapse
Affiliation(s)
- Rachel E. Keith
- Section on Neural Circuits, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Yiming Shen
- Section on Neural Circuits, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | | | - Joseph Abramovitz
- Section on Neural Circuits, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Priscila C. Antonello
- Section on Neural Circuits, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | | | - Baskar Mohana Krishnan
- Section on Neural Circuits, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Michelle W. Antoine
- Section on Neural Circuits, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Castillon C, Otsuka S, Armstrong J, Contractor A. Subregional activity in the dentate gyrus is amplified during elevated cognitive demands. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.31.621367. [PMID: 39554113 PMCID: PMC11565968 DOI: 10.1101/2024.10.31.621367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Neural activity in the dentate gyrus (DG) is required for the detection and discrimination of novelty, context and patterns, amongst other cognitive processes. Prior work has demonstrated that there are differences in the activation of granule neurons in the supra and infrapyramidal blades of the DG during a range of hippocampal dependent tasks. Here we used an automated touch screen pattern separation task combined to temporally controlled tagging of active neurons to determine how performance in a cognitively demanding task affected patterns of neural activity in the DG. We found an increase in the blade-biased activity of suprapyramidal mature granule cells (mGCs) during the performance of a high cognitive demand segment of the task, with a further characteristic distribution of active neurons along the apex to blade, and hilar to molecular layer axes. Chemogenetic inhibition of adult-born granule cells (abDGCs) beyond a critical window of their maturation significantly impaired performance of mice during high-demand conditions but not when cognitive demand was low. abDGC inhibition also elevated the total activity of mGCs and disturbed the patterned distribution of active mGCs even in mice that eventually succeeded in the task. Conversely chemogenetic inhibition of mGCs reduced success in the high cognitive demand portion of this task and decreased the global number of active GCs without affecting the patterned distribution of active cells. These findings demonstrate how a high cognitive demand pattern separation task preferentially activates mGCs in subregions of the DG and are consistent with a modulatory role for abDGCs on the dentate circuit which in part governs the spatially organized patterns of activity of mGCs.
Collapse
|
5
|
Kim JH, Hwang SY, Lee HL, Yoon SL, Ha Y, Lee HY, Ryu S. Effects of chemogenetic virus injection and clozapine administration in spinal cord injury. Neurotherapeutics 2025; 22:e00547. [PMID: 39955176 PMCID: PMC12014403 DOI: 10.1016/j.neurot.2025.e00547] [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: 09/12/2024] [Revised: 12/03/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025] Open
Abstract
Neuromodulation therapy using chemogenetic stimulation has shown potential in enhancing motor recovery and neuroregeneration following spinal cord injury (SCI). These therapeutic benefits are hypothesized to result from the promotion of neuroplasticity, particularly when administered during the acute phase of injury. In this study, we investigated the effects of chemogenetic stimulation using Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) in conjunction with clozapine, a ligand for receptor activation. DREADDs enable targeted, reversible neuromodulation, facilitating the histological characterization of engineered neurons. We utilized these receptors to modulate G-protein-coupled receptor (GPCR) signaling pathways, leading to the activation or inhibition of intracellular signaling. The objective was to determine whether the administration of DREADDs and clozapine (0.1 mg/kg) could enhance motor function and neuronal recovery, particularly when applied during the acute phase of SCI. Weekly behavioral assessments demonstrated significant improvements in motor skills and neuronal regeneration in treated animals compared to controls, with the most pronounced effects observed when stimulation was initiated early after injury. These enhancements in neuroplasticity were reflected in improved ladder rung test scores and Basso, Beattie, and Bresnahan (BBB) scale results in DREADDs-treated rats. Histological analyses, including immunohistochemistry (IHC) staining, Western blotting, and quantitative reverse transcription PCR (qRT-PCR), confirmed that the treatment group exhibited a higher density of neurons, increased signaling protein expression, and reduced inflammatory markers. These findings suggest that chemogenetic stimulation, particularly when administered during the acute phase, effectively promotes neuroregeneration and motor recovery. Future research should focus on assessing the long-term safety and efficacy of chemogenetic virus injection and clozapine administration, with an emphasis on the timing of intervention.
Collapse
Affiliation(s)
- Ji Hyeon Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea; Life Science Cluster, Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Sae Yeon Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Sol Lip Yoon
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea; POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Hye Yeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea.
| | - Seungjun Ryu
- Life Science Cluster, Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea; Department of Neurosurgery, School of Medicine, Eulji University, Daejeon, Republic of Korea.
| |
Collapse
|
6
|
Vargas-Ortiz J, Lin L, Martinez VK, Liu RJ, Babij R, Duan ZRS, Wacks S, Sun L, Wang A, Khan S, Soto-Vargas JL, De Marco García NV, Che A. Translaminar synchronous neuronal activity is required for columnar synaptic strengthening in the mouse neocortex. Nat Commun 2025; 16:1296. [PMID: 39900899 PMCID: PMC11791040 DOI: 10.1038/s41467-024-55783-w] [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: 02/28/2024] [Accepted: 12/24/2024] [Indexed: 02/05/2025] Open
Abstract
Synchronous neuronal activity is a hallmark of the developing mouse primary somatosensory cortex. While the patterns of synchronous neuronal activity in cortical layer 2/3 have been well described, the source of the robust layer 2/3 activity is still unknown. Using a novel microprism preparation and in vivo 2-photon imaging in neonatal mice, we show that synchronous neuronal activity is organized in barrel columns across layers. Monosynaptic rabies tracing and slice electrophysiology experiments reveal that layer 2/3 pyramidal neurons receive significant layer 5 inputs during the first postnatal week, and silencing layer 5 synaptic outputs results in a significant reduction in spontaneous activity, abnormal sensory-evoked activity and disrupted layer 4-layer 2/3 connectivity. Our results demonstrate that translaminar layer 5-layer 2/3 connectivity plays an important role in synchronizing the developing barrel column to ensure the strengthening of layer 4-layer 2/3 connections, supporting the formation of the canonical cortical organization in barrel cortex.
Collapse
Affiliation(s)
- John Vargas-Ortiz
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Lin Lin
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Vena K Martinez
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Rong-Jian Liu
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Rachel Babij
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Zhe Ran S Duan
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Sam Wacks
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Liyuan Sun
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Amanda Wang
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Sajida Khan
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | | | - Natalia V De Marco García
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Alicia Che
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Wu Tsai Institute, Yale University, New Haven, CT, USA.
| |
Collapse
|
7
|
Baldo BA. Opioid-induced respiratory depression: clinical aspects and pathophysiology of the respiratory network effects. Am J Physiol Lung Cell Mol Physiol 2025; 328:L267-L289. [PMID: 39726397 DOI: 10.1152/ajplung.00314.2024] [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: 10/22/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Important insights and consensus remain lacking for risk prediction of opioid-induced respiratory depression (OIRD), reversal of respiratory depression (RD), the pathophysiology of OIRD, and which sites make the most significant contribution to its induction. The ventilatory response to inhaled carbon dioxide is the most sensitive biomarker of OIRD. To accurately predict respiratory depression (RD), a multivariant RD prospective trial using continuous capnography and oximetry examining five independent variables, age ≥60, sex, opioid naivety, sleep disorders, and chronic heart failure (PRODIGY trial), were undertaken. Intermittent oximetry alone substantially underestimates the incidence of RD. Naloxone, with an elimination half-life of ∼33 min (cf. morphine 2-3 h; fentanyl and congeners only 5-15 min), has limitations for the rescue of patients with severe OIRD. Buprenorphine is potentially valuable in patients being treated long term since its high µ-receptor (MOR) affinity makes it difficult for an opioid of lower affinity (e.g., fentanyl) to displace it from the receptor. In the last decade, synthetic opioids, for example, fentanyl, its potent analogs such as carfentanil, and the benzimidazole derivative nitazene "superagonists" have contributed to the exponential growth in opioid deaths due to RD. The MOR, encoded by gene Oprm1, is widely expressed in the central and peripheral nervous systems, including centers that modulate breathing. Opioids bind to the receptors, but consensus is lacking on which site(s) makes the most significant contribution to the induction of OIRD. Both the preBötzinger complex (preBötC), the inspiratory rhythm generator, and the Kölliker-Fuse nucleus (KFN), the respiratory modulator, contribute to RD, but receptor binding is not restricted to a single site. Breathing is composed of three phases, inspiration, postinspiration, and active expiration, each generated by distinct rhythm-generating networks: the preBötC, the postinspiratory complex (PiCo), and the lateral parafacial nucleus (pFL), respectively. Somatostatin-expressing mouse cells involved in breathing regulation are not involved in opioid-induced RD.
Collapse
Affiliation(s)
- Brian A Baldo
- Kolling Institute of Medical Research, Royal North Shore Hospital of Sydney, Sydney, New South Wales, Australia
- Department of Medicine, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
8
|
Malvaez M, Liang A, Hall BS, Giovanniello JR, Paredes N, Gonzalez JY, Blair GJ, Sias AC, Murphy MD, Guo W, Wang A, Singh M, Griffin NK, Bridges SP, Wiener A, Pimenta JS, Holley SM, Cepeda C, Levine MS, Blair HT, Wikenheiser AM, Wassum KM. Striatal cell-type specific stability and reorganization underlying agency and habit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.26.634924. [PMID: 39896502 PMCID: PMC11785256 DOI: 10.1101/2025.01.26.634924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Adaptive decision making requires agency, knowledge that actions produce particular outcomes. For well-practiced routines, agency is relinquished in favor of habit. Here, we asked how dorsomedial striatum D1+ and D2/A2A+ neurons contribute to agency and habit. We imaged calcium activity of these neurons as mice learned to lever press with agency and formed habits with overtraining. Whereas many D1+ neurons stably encoded actions throughout learning and developed encoding of reward outcomes, A2A+ neurons reorganized their encoding of actions from initial action-outcome learning to habit formation. Chemogenetic manipulations indicated that both D1+ and A2A+ neurons support action-outcome learning, but only D1+ neurons enable the use of such agency for adaptive, goal-directed decision making. These data reveal coordinated dorsomedial striatum D1+ and A2A+ function for the development of agency, cell-type specific stability and reorganization underlying agency and habit, and important insights into the neuronal circuits of how we learn and decide.
Collapse
Affiliation(s)
| | - Alvina Liang
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | - Baila S Hall
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | | | | | | | | | - Ana C Sias
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | | | - Wanyi Guo
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | - Alicia Wang
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | - Malika Singh
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | | | | | - Anna Wiener
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | | | - Sandra M Holley
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Carlos Cepeda
- Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Michael S Levine
- Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - H Tad Blair
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
- Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Andrew M Wikenheiser
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
- Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Kate M Wassum
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
- Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
9
|
Song Y, Fothergill LJ, Lee KS, Liu BY, Koo A, Perelis M, Diwakarla S, Callaghan B, Huang J, Wykosky J, Furness JB, Yeo GW. Stratification of enterochromaffin cells by single-cell expression analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.08.24.554649. [PMID: 37662229 PMCID: PMC10473706 DOI: 10.1101/2023.08.24.554649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Dynamic interactions between gut mucosal cells and the external environment are essential to maintain gut homeostasis. Enterochromaffin (EC) cells transduce both chemical and mechanical signals and produce 5-hydroxytryptamine (5-HT) to mediate disparate physiological responses. However, the molecular and cellular basis for functional diversity of ECs remains to be adequately defined. Here, we integrated single-cell transcriptomics with spatial image analysis to identify fourteen EC clusters that are topographically organized along the gut. Subtypes predicted to be sensitive to the chemical environment and mechanical forces were identified that express distinct transcription factors and hormones. A Piezo2 + population in the distal colon was endowed with a distinctive neuronal signature. Using a combination of genetic, chemogenetic and pharmacological approaches, we demonstrated Piezo2 + ECs are required for normal colon motility. Our study constructs a molecular map for ECs and offers a framework for deconvoluting EC cells with pleiotropic functions.
Collapse
Affiliation(s)
- Yan Song
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Stem Cell Program, University of California San Diego, La Jolla, CA 92093, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, United States
| | - Linda J. Fothergill
- Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia
| | - Kari S. Lee
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Stem Cell Program, University of California San Diego, La Jolla, CA 92093, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, United States
| | - Brandon Y. Liu
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Stem Cell Program, University of California San Diego, La Jolla, CA 92093, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, United States
| | - Ada Koo
- Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Mark Perelis
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Stem Cell Program, University of California San Diego, La Jolla, CA 92093, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, United States
| | - Shanti Diwakarla
- Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brid Callaghan
- Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jie Huang
- Takeda Pharmaceuticals, San Diego, CA 92121, United States
| | - Jill Wykosky
- Takeda Pharmaceuticals, San Diego, CA 92121, United States
| | - John B. Furness
- Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia
| | - Gene W. Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Stem Cell Program, University of California San Diego, La Jolla, CA 92093, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, United States
| |
Collapse
|
10
|
Zhi W, Li Y, Wang L, Hu X. Advancing Neuroscience and Therapy: Insights into Genetic and Non-Genetic Neuromodulation Approaches. Cells 2025; 14:122. [PMID: 39851550 PMCID: PMC11763439 DOI: 10.3390/cells14020122] [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: 12/12/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Neuromodulation stands as a cutting-edge approach in the fields of neuroscience and therapeutic intervention typically involving the regulation of neural activity through physical and chemical stimuli. The purpose of this review is to provide an overview and evaluation of different neuromodulation techniques, anticipating a clearer understanding of the future developmental trajectories and the challenges faced within the domain of neuromodulation that can be achieved. This review categorizes neuromodulation techniques into genetic neuromodulation methods (including optogenetics, chemogenetics, sonogenetics, and magnetogenetics) and non-genetic neuromodulation methods (including deep brain stimulation, transcranial magnetic stimulation, transcranial direct current stimulation, transcranial ultrasound stimulation, photobiomodulation therapy, infrared neuromodulation, electromagnetic stimulation, sensory stimulation therapy, and multi-physical-factor stimulation techniques). By systematically evaluating the principles, mechanisms, advantages, limitations, and efficacy in modulating neuronal activity and the potential applications in interventions of neurological disorders of these neuromodulation techniques, a comprehensive picture is gradually emerging regarding the advantages and challenges of neuromodulation techniques, their developmental trajectory, and their potential clinical applications. This review highlights significant advancements in applying these techniques to treat neurological and psychiatric disorders. Genetic methods, such as sonogenetics and magnetogenetics, have demonstrated high specificity and temporal precision in targeting neuronal populations, while non-genetic methods, such as transcranial magnetic stimulation and photobiomodulation therapy, offer noninvasive and versatile clinical intervention options. The transformative potential of these neuromodulation techniques in neuroscience research and clinical practice is underscored, emphasizing the need for integration and innovation in technologies, the optimization of delivery methods, the improvement of mediums, and the evaluation of toxicity to fully harness their therapeutic potential.
Collapse
Affiliation(s)
- Weijia Zhi
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| | - Ying Li
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Lifeng Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| | - Xiangjun Hu
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| |
Collapse
|
11
|
Kizeev G, Witteveen I, Balmer T. Balance Performance in Aged Mice is Dependent on Unipolar Brush Cells. CEREBELLUM (LONDON, ENGLAND) 2024; 24:16. [PMID: 39699796 DOI: 10.1007/s12311-024-01767-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
The vestibular processing regions of the cerebellum integrate vestibular information with other sensory modalities and motor signals to regulate balance, gaze stability, and spatial orientation. A class of excitatory glutamatergic interneurons known as unipolar brush cells (UBCs) are highly concentrated within the granule cell layer of these regions. UBCs receive vestibular signals directly from primary vestibular afferents and indirectly from mossy fibers. Each UBC excites numerous granule cells and could contribute to computations necessary for balance-related motor function. Prior research has implicated UBCs in motor function, but their influence on balance performance remains unclear, especially in aged mice that have age-related impairment. Here we tested whether UBCs contribute to motor coordination and balance by disrupting their activity with chemogenetics in aged and young mice. Age-related balance deficits were apparent in mice > 6 months old. Disrupting the activity of a subpopulation of UBCs caused aged mice to fall off a balance beam more frequently and altered swimming behaviors that are sensitive to vestibular dysfunction. These effects were not seen in young (7-week-old) mice. Thus, disrupting the activity of UBCs impairs mice with age-related balance issues and suggest that UBCs are essential for balance and vestibular function in aged mice.
Collapse
Affiliation(s)
- Gabrielle Kizeev
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Isabelle Witteveen
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Timothy Balmer
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
12
|
Chen S, Cheng N, Chen X, Wang C. Integration and competition between space and time in the hippocampus. Neuron 2024; 112:3651-3664.e8. [PMID: 39241779 DOI: 10.1016/j.neuron.2024.08.007] [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: 03/12/2024] [Revised: 07/11/2024] [Accepted: 08/09/2024] [Indexed: 09/09/2024]
Abstract
Episodic memory is organized in both spatial and temporal contexts. The hippocampus is crucial for episodic memory and has been demonstrated to encode spatial and temporal information. However, how the representations of space and time interact in the hippocampal memory system is still unclear. Here, we recorded the activity of hippocampal CA1 neurons in mice in a variety of one-dimensional navigation tasks while systematically varying the speed of the animals. For all tasks, we found neurons simultaneously represented space and elapsed time. There was a negative correlation between the preferred space and lap duration, e.g., the preferred spatial position shifted more toward the origin when the lap duration became longer. A similar relationship between the preferred time and traveled distance was also observed. The results strongly suggest a competitive and integrated representation of space-time by single hippocampal neurons, which may provide the neural basis for spatiotemporal contexts.
Collapse
Affiliation(s)
- Shijie Chen
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ning Cheng
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaojing Chen
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Cheng Wang
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
13
|
Ben-Shaanan TL, Knöpper K, Duan L, Liu R, Taglinao H, Xu Y, An J, Plikus MV, Cyster JG. Dermal TRPV1 innervations engage a macrophage- and fibroblast-containing pathway to activate hair growth in mice. Dev Cell 2024; 59:2818-2833.e7. [PMID: 38851191 PMCID: PMC11537826 DOI: 10.1016/j.devcel.2024.05.019] [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: 10/03/2023] [Revised: 02/25/2024] [Accepted: 05/15/2024] [Indexed: 06/10/2024]
Abstract
Pain, detected by nociceptors, is an integral part of injury, yet whether and how it can impact tissue physiology and recovery remain understudied. Here, we applied chemogenetics in mice to locally activate dermal TRPV1 innervations in naive skin and found that it triggered new regenerative cycling by dormant hair follicles (HFs). This was preceded by rapid apoptosis of dermal macrophages, mediated by the neuropeptide calcitonin gene-related peptide (CGRP). TRPV1 activation also triggered a macrophage-dependent induction of osteopontin (Spp1)-expressing dermal fibroblasts. The neuropeptide CGRP and the extracellular matrix protein Spp1 were required for the nociceptor-triggered hair growth. Finally, we showed that epidermal abrasion injury induced Spp1-expressing dermal fibroblasts and hair growth via a TRPV1 neuron and CGRP-dependent mechanism. Collectively, these data demonstrated a role for TRPV1 nociceptors in orchestrating a macrophage and fibroblast-supported mechanism to promote hair growth and enabling the efficient restoration of this mechano- and thermo-protective barrier after wounding.
Collapse
Affiliation(s)
- Tamar L Ben-Shaanan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Konrad Knöpper
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lihui Duan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ruiqi Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Hanna Taglinao
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
14
|
Graf J, Samiee A, Flossmann T, Holthoff K, Kirmse K. Chemogenetic silencing reveals presynaptic G i/o protein-mediated inhibition of developing hippocampal synchrony in vivo. iScience 2024; 27:110997. [PMID: 39429781 PMCID: PMC11489827 DOI: 10.1016/j.isci.2024.110997] [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: 03/28/2024] [Revised: 07/29/2024] [Accepted: 09/17/2024] [Indexed: 10/22/2024] Open
Abstract
Recent advances in understanding how neuronal activity shapes developing brain circuits increasingly rely on Gi/o-dependent inhibitory chemogenetic tools (Gi-DREADDs). However, their mechanisms of action and efficacy in neurons with immature Gi/o signaling are elusive. Here, we express the Gi-DREADD hM4Di in glutamatergic telencephalic neurons and analyze its impact on CA1 pyramidal neurons in neonatal mice. Using acousto-optic two-photon Ca2+ imaging, we report that activation of hM4Di leads to a complete arrest of spontaneous synchrony in CA1 in vitro. We demonstrate that hM4Di does not cause somatic hyperpolarization or shunting but rather mediates presynaptic silencing of glutamatergic neurotransmission. In vivo, inhibition through hM4Di potently suppresses early sharp waves (eSPWs) and discontinuous oscillatory network activity in CA1 of head-fixed mice before eye opening. Our findings provide insights into the role of Gi/o signaling in synchronized activity in the neonatal hippocampus and bear relevance for applying chemogenetic silencing at early developmental stages.
Collapse
Affiliation(s)
- Jürgen Graf
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Arash Samiee
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| | - Tom Flossmann
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany
- Institute of Physiology I, Jena University Hospital, 07743 Jena, Germany
| | - Knut Holthoff
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Knut Kirmse
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| |
Collapse
|
15
|
Kizeev G, Witteveen I, Balmer T. Balance performance in aged mice is dependent on unipolar brush cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617602. [PMID: 39416048 PMCID: PMC11482929 DOI: 10.1101/2024.10.10.617602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The vestibular processing regions of the cerebellum integrate vestibular information with other sensory modalities and motor signals to regulate balance, gaze stability, and spatial orientation. A class of excitatory glutamatergic interneurons known as unipolar brush cells (UBCs) are highly concentrated within the granule cell layer of these regions. UBCs receive vestibular signals directly from primary vestibular afferents and indirectly from mossy fibers. Each UBC excites numerous granule cells and could contribute to computations necessary for balance-related motor function. Prior research has implicated UBCs in motor function, but their influence on balance performance remains unclear, especially in aged mice that have age-related impairment. Here we tested whether UBCs contribute to motor coordination and balance by disrupting their activity with chemogenetics in aged and young mice. Age-related balance deficits were apparent in mice > 6 months old. Disrupting the activity of a subpopulation of UBCs caused aged mice to fall off a balance beam more frequently and altered swimming behaviors that are sensitive to vestibular dysfunction. These effects were not seen in young (7-week-old) mice. Thus, disrupting the activity of UBCs impairs mice with age-related balance issues and suggest that UBCs are essential for balance and vestibular function in aged mice.
Collapse
Affiliation(s)
- Gabrielle Kizeev
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Isabelle Witteveen
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Timothy Balmer
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| |
Collapse
|
16
|
Sawyer IL, Evans MC, Mamgain A, Decourt C, Iremonger KJ, Anderson GM. Chemogenetic Activation of RFRP Neurons Reduces LH Pulse Frequency in Female but not Male Mice. J Endocr Soc 2024; 8:bvae159. [PMID: 39381686 PMCID: PMC11458915 DOI: 10.1210/jendso/bvae159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Indexed: 10/10/2024] Open
Abstract
Context The neuropeptide RFRP-3 (RFamide-related peptide-3) is thought to play a role in the negative regulation of fertility. However, the exogenous administration of RFRP-3 yields varying results depending on the dose and route of administration, sex of the subject, and many other variables. Manipulation of in vivo neuronal activity using DREADDs (designer receptor exclusively activated by designer drugs) technology enables investigation of cell type-specific neuronal activation in a manner that better reflects endogenous neuronal activity. Objective To test the effects of RFRP neuronal activation on pulsatile luteinizing hormone (LH) secretion. Methods We generated mice expressing the stimulatory hM3Dq designer receptor exclusively in RFRP cells using 2 different Cre-loxP-mediated approaches: (1) we bred mice to express hM3Dq in all Rfrp-Cre-expressing cells, including some that transiently expressed Rfrp-Cre neonatally (RFRP × hM3Dq mice), and (2) we stereotaxically injected Cre-dependent hM3Dq into the dorsomedial nucleus of RFRP-Cre mice to drive hM3Dq expression exclusively in a subpopulation of adult Rfrp-Cre neurons (RFRP-AAV-hM3Dq mice). We then investigated the effects of acute hM3Dq activation on LH pulse frequency in RFRP × hM3Dq mice, RFRP-AAV-hM3Dq mice, and their respective controls. Results In both female RFRP × hM3Dq and RFRP-AAV-hM3Dq mice, chemogenetic activation of Cre-driven hM3Dq led to a significant 35% to 50% reduction in LH pulse frequency compared with controls, while no differences in pulse amplitude or mean LH concentration were observed. In marked contrast, RFRP activation did not cause any changes to LH pulse dynamics in male mice. Conclusions These data show for the first time that activation of neurons that have expressed Rfrp, or of a subset of adult RFRP neurons, can independently suppress LH pulsatility in female, but not male mice.
Collapse
Affiliation(s)
- India L Sawyer
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| | - Maggie C Evans
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| | - Asha Mamgain
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| | - Caroline Decourt
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| | - Karl J Iremonger
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| | - Greg M Anderson
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| |
Collapse
|
17
|
Giovanniello JR, Paredes N, Wiener A, Ramírez-Armenta K, Oragwam C, Uwadia HO, Yu AL, Lim K, Pimenta JS, Vilchez GE, Nnamdi G, Wang A, Sehgal M, Reis FM, Sias AC, Silva AJ, Adhikari A, Malvaez M, Wassum KM. A dual-pathway architecture enables chronic stress to disrupt agency and promote habit formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.03.560731. [PMID: 37873076 PMCID: PMC10592885 DOI: 10.1101/2023.10.03.560731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Chronic stress can change how we learn and, thus, how we make decisions. Here we investigated the neuronal circuit mechanisms that enable this. Using a multifaceted systems neuroscience approach in male and female mice, we reveal a dual pathway, amygdala-striatal neuronal circuit architecture by which a recent history of chronic stress disrupts the action-outcome learning underlying adaptive agency and promotes the formation of inflexible habits. We found that the basolateral amygdala projection to the dorsomedial striatum is activated by rewarding events to support the action-outcome learning needed for flexible, goal-directed decision making. Chronic stress attenuates this to disrupt action-outcome learning and, therefore, agency. Conversely, the central amygdala projection to the dorsomedial striatum mediates habit formation. Following stress this pathway is progressively recruited to learning to promote the premature formation of inflexible habits. Thus, stress exerts opposing effects on two amygdala-striatal pathways to disrupt agency and promote habit. These data provide neuronal circuit insights into how chronic stress shapes learning and decision making, and help understand how stress can lead to the disrupted decision making and pathological habits that characterize substance use disorders and mental health conditions.
Collapse
Affiliation(s)
| | | | - Anna Wiener
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | | | | | | | - Abigail L Yu
- Dept. of Physiology, UCLA, Los Angeles, CA 90095
| | - Kayla Lim
- Dept. of Biological Chemistry, UCLA, Los Angeles, CA 90095
| | | | | | - Gift Nnamdi
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | - Alicia Wang
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | - Megha Sehgal
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | | | - Ana C Sias
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
| | - Alcino J Silva
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
- Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
- Integrative Center for Learning and Memory, University of California Los Angeles, Los Angeles, CA, USA
| | - Avishek Adhikari
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
- Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
- Integrative Center for Learning and Memory, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Kate M Wassum
- Dept. of Psychology, UCLA, Los Angeles, CA 90095
- Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
- Integrative Center for Learning and Memory, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
18
|
Coutinho EA, Esparza LA, Rodriguez J, Yang J, Schafer D, Kauffman AS. Targeted inhibition of kisspeptin neurons reverses hyperandrogenemia and abnormal hyperactive LH secretion in a preclinical mouse model of polycystic ovary syndrome. Hum Reprod 2024; 39:2089-2103. [PMID: 38978296 PMCID: PMC11373419 DOI: 10.1093/humrep/deae153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/11/2024] [Indexed: 07/10/2024] Open
Abstract
STUDY QUESTION Do hyperactive kisspeptin neurons contribute to abnormally high LH secretion and downstream hyperandrogenemia in polycystic ovary syndrome (PCOS)-like conditions and can inhibition of kisspeptin neurons rescue such endocrine impairments? SUMMARY ANSWER Targeted inhibition of endogenous kisspeptin neuron activity in a mouse model of PCOS reduced the abnormally hyperactive LH pulse secretion and hyperandrogenemia to healthy control levels. WHAT IS KNOWN ALREADY PCOS is a reproductive disorder characterized by hyperandrogenemia, anovulation, and/or polycystic ovaries, along with a hallmark feature of abnormal LH hyper-pulsatility, but the mechanisms underlying the endocrine impairments remain unclear. A chronic letrozole (LET; aromatase inhibitor) mouse model recapitulates PCOS phenotypes, including polycystic ovaries, anovulation, high testosterone, and hyperactive LH pulses. LET PCOS-like females also have increased hypothalamic kisspeptin neuronal activation which may drive their hyperactive LH secretion and hyperandrogenemia, but this has not been tested. STUDY DESIGN, SIZE, DURATION Transgenic KissCRE+/hM4Di female mice or littermates Cre- controls were treated with placebo, or chronic LET (50 µg/day) to induce a PCOS-like phenotype, followed by acute (once) or chronic (2 weeks) clozapine-N-oxide (CNO) exposure to chemogenetically inhibit kisspeptin cells (n = 6 to 10 mice/group). PARTICIPANTS/MATERIALS, SETTING, METHODS Key endocrine measures, including in vivo LH pulse secretion patterns and circulating testosterone levels, were assessed before and after selective kisspeptin neuron inhibition and compared between PCOS groups and healthy controls. Alterations in body weights were measured and pituitary and ovarian gene expression was determined by qRT-PCR. MAIN RESULTS AND THE ROLE OF CHANCE Acute targeted inhibition of kisspeptin neurons in PCOS mice successfully lowered the abnormally hyperactive LH pulse secretion (P < 0.05). Likewise, chronic selective suppression of kisspeptin neuron activity reversed the previously high LH and testosterone levels (P < 0.05) down to healthy control levels and rescued reproductive gene expression (P < 0. 05). LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Ovarian morphology was not assessed in this study. Additionally, mouse models can offer mechanistic insights into neuroendocrine processes in PCOS-like conditions but may not perfectly mirror PCOS in women. WIDER IMPLICATIONS OF THE FINDINGS These data support the hypothesis that overactive kisspeptin neurons can drive neuroendocrine PCOS-like impairments, and this may occur in PCOS women. Our findings complement recent clinical investigations using NKB receptor antagonists to lower LH in PCOS women and suggest that pharmacological dose-dependent modulation of kisspeptin neuron activity may be a valuable future therapeutic target to clinically treat hyperandrogenism and lower elevated LH in PCOS women. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by NIH grants R01 HD111650, R01 HD090161, R01 HD100580, P50 HD012303, R01 AG078185, and NIH R24 HD102061, and a pilot project award from the British Society for Neuroendocrinology. There are no competing interests.
Collapse
Affiliation(s)
- Eulalia A Coutinho
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Lourdes A Esparza
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Julian Rodriguez
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jason Yang
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Danielle Schafer
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Alexander S Kauffman
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
19
|
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
|
20
|
Kabanova A, Fedorov L, Eschenko O. The Projection-Specific Noradrenergic Modulation of Perseverative Spatial Behavior in Adult Male Rats. eNeuro 2024; 11:ENEURO.0063-24.2024. [PMID: 39160074 PMCID: PMC11334950 DOI: 10.1523/eneuro.0063-24.2024] [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: 02/13/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 08/21/2024] Open
Abstract
Adaptive behavior relies on efficient cognitive control. The anterior cingulate cortex (ACC) is a key node within the executive prefrontal network. The reciprocal connectivity between the locus ceruleus (LC) and ACC is thought to support behavioral reorganization triggered by the detection of an unexpected change. We transduced LC neurons with either excitatory or inhibitory chemogenetic receptors in adult male rats and trained rats on a spatial task. Subsequently, we altered LC activity and confronted rats with an unexpected change of reward locations. In a new spatial context, rats with decreased noradrenaline (NA) in the ACC entered unbaited maze arms more persistently which was indicative of perseveration. In contrast, the suppression of the global NA transmission reduced perseveration. Neither chemogenetic manipulation nor inactivation of the ACC by muscimol affected the rate of learning, possibly due to partial virus transduction of the LC neurons and/or the compensatory engagement of other prefrontal regions. Importantly, we observed behavioral deficits in rats with LC damage caused by virus injection. The latter finding highlights the importance of careful histological assessment of virus-transduced brain tissue as inadvertent damage of the targeted cell population due to virus neurotoxicity or other factors might cause unwanted side effects. Although the specific role of ACC in the flexibility of spatial behavior has not been convincingly demonstrated, our results support the beneficial role of noradrenergic transmission for an optimal function of the ACC. Overall, our findings suggest the LC exerts the projection-specific modulation of neural circuits mediating the flexibility of spatial behavior.
Collapse
Affiliation(s)
- Anna Kabanova
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, 72076 Tübingen, Germany
| | - Leonid Fedorov
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, 72076 Tübingen, Germany
| | - Oxana Eschenko
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, 72076 Tübingen, Germany
| |
Collapse
|
21
|
Frechou MA, Martin SS, McDermott KD, Huaman EA, Gökhan Ş, Tomé WA, Coen-Cagli R, Gonçalves JT. Adult neurogenesis improves spatial information encoding in the mouse hippocampus. Nat Commun 2024; 15:6410. [PMID: 39080283 PMCID: PMC11289285 DOI: 10.1038/s41467-024-50699-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: 12/15/2022] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Adult neurogenesis is a unique form of neuronal plasticity in which newly generated neurons are integrated into the adult dentate gyrus in a process that is modulated by environmental stimuli. Adult-born neurons can contribute to spatial memory, but it is unknown whether they alter neural representations of space in the hippocampus. Using in vivo two-photon calcium imaging, we find that male and female mice previously housed in an enriched environment, which triggers an increase in neurogenesis, have increased spatial information encoding in the dentate gyrus. Ablating adult neurogenesis blocks the effect of enrichment and lowers spatial information, as does the chemogenetic silencing of adult-born neurons. Both ablating neurogenesis and silencing adult-born neurons decreases the calcium activity of dentate gyrus neurons, resulting in a decreased amplitude of place-specific responses. These findings are in contrast with previous studies that suggested a predominantly inhibitory action for adult-born neurons. We propose that adult neurogenesis improves representations of space by increasing the gain of dentate gyrus neurons and thereby improving their ability to tune to spatial features. This mechanism may mediate the beneficial effects of environmental enrichment on spatial learning and memory.
Collapse
Affiliation(s)
- M Agustina Frechou
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA
| | - Sunaina S Martin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Psychology, University of California San Diego, La Jolla, CA, USA
| | - Kelsey D McDermott
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Evan A Huaman
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Şölen Gökhan
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wolfgang A Tomé
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ruben Coen-Cagli
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - J Tiago Gonçalves
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
22
|
Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, Chen Z, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, Rich LM, Barilli A, Sabat M, Debley JS, Wu C, Myers R, Howitt MR, von Moltke J. Tuft cell-derived acetylcholine promotes epithelial chloride secretion and intestinal helminth clearance. Immunity 2024; 57:1243-1259.e8. [PMID: 38744291 PMCID: PMC11168877 DOI: 10.1016/j.immuni.2024.03.023] [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: 04/05/2023] [Revised: 02/05/2024] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
Epithelial cells secrete chloride to regulate water release at mucosal barriers, supporting both homeostatic hydration and the "weep" response that is critical for type 2 immune defense against parasitic worms (helminths). Epithelial tuft cells in the small intestine sense helminths and release cytokines and lipids to activate type 2 immune cells, but whether they regulate epithelial secretion is unknown. Here, we found that tuft cell activation rapidly induced epithelial chloride secretion in the small intestine. This response required tuft cell sensory functions and tuft cell-derived acetylcholine (ACh), which acted directly on neighboring epithelial cells to stimulate chloride secretion, independent of neurons. Maximal tuft cell-induced chloride secretion coincided with immune restriction of helminths, and clearance was delayed in mice lacking tuft cell-derived ACh, despite normal type 2 inflammation. Thus, we have uncovered an epithelium-intrinsic response unit that uses ACh to couple tuft cell sensing to the secretory defenses of neighboring epithelial cells.
Collapse
Affiliation(s)
- Tyler E Billipp
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lily M Webeck
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Derek B Sargent
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Matthew B Gologorsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Margaret M McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Darshan N Kasal
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - John W McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Kaitlyn A Barrow
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | - Lucille M Rich
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Mark Sabat
- Takeda Pharmaceuticals, San Diego, CA, USA
| | - Jason S Debley
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Michael R Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
23
|
Engström Ruud L, Font-Gironès F, Zajdel J, Kern L, Teixidor-Deulofeu J, Mannerås-Holm L, Carreras A, Becattini B, Björefeldt A, Hanse E, Fenselau H, Solinas G, Brüning JC, Wunderlich TF, Bäckhed F, Ruud J. Activation of GFRAL + neurons induces hypothermia and glucoregulatory responses associated with nausea and torpor. Cell Rep 2024; 43:113960. [PMID: 38507407 DOI: 10.1016/j.celrep.2024.113960] [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: 09/21/2023] [Revised: 01/11/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
GFRAL-expressing neurons actuate aversion and nausea, are targets for obesity treatment, and may mediate metformin effects by long-term GDF15-GFRAL agonism. Whether GFRAL+ neurons acutely regulate glucose and energy homeostasis is, however, underexplored. Here, we report that cell-specific activation of GFRAL+ neurons using a variety of techniques causes a torpor-like state, including hypothermia, the release of stress hormones, a shift from glucose to lipid oxidation, and impaired insulin sensitivity, glucose tolerance, and skeletal muscle glucose uptake but augmented glucose uptake in visceral fat. Metabolomic analysis of blood and transcriptomics of muscle and fat indicate alterations in ketogenesis, insulin signaling, adipose tissue differentiation and mitogenesis, and energy fluxes. Our findings indicate that acute GFRAL+ neuron activation induces endocrine and gluco- and thermoregulatory responses associated with nausea and torpor. While chronic activation of GFRAL signaling promotes weight loss in obesity, these results show that acute activation of GFRAL+ neurons causes hypothermia and hyperglycemia.
Collapse
Affiliation(s)
- Linda Engström Ruud
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ferran Font-Gironès
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joanna Zajdel
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lara Kern
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Júlia Teixidor-Deulofeu
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise Mannerås-Holm
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Alba Carreras
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Barbara Becattini
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Andreas Björefeldt
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eric Hanse
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Giovanni Solinas
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | | | - Fredrik Bäckhed
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Ruud
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
24
|
Privitera M, von Ziegler LM, Floriou-Servou A, Duss SN, Zhang R, Waag R, Leimbacher S, Sturman O, Roessler FK, Heylen A, Vermeiren Y, Van Dam D, De Deyn PP, Germain PL, Bohacek J. Noradrenaline release from the locus coeruleus shapes stress-induced hippocampal gene expression. eLife 2024; 12:RP88559. [PMID: 38477670 DOI: 10.7554/elife.88559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024] Open
Abstract
Exposure to an acute stressor triggers a complex cascade of neurochemical events in the brain. However, deciphering their individual impact on stress-induced molecular changes remains a major challenge. Here, we combine RNA sequencing with selective pharmacological, chemogenetic, and optogenetic manipulations to isolate the contribution of the locus coeruleus-noradrenaline (LC-NA) system to the acute stress response in mice. We reveal that NA release during stress exposure regulates a large and reproducible set of genes in the dorsal and ventral hippocampus via β-adrenergic receptors. For a smaller subset of these genes, we show that NA release triggered by LC stimulation is sufficient to mimic the stress-induced transcriptional response. We observe these effects in both sexes, and independent of the pattern and frequency of LC activation. Using a retrograde optogenetic approach, we demonstrate that hippocampus-projecting LC neurons directly regulate hippocampal gene expression. Overall, a highly selective set of astrocyte-enriched genes emerges as key targets of LC-NA activation, most prominently several subunits of protein phosphatase 1 (Ppp1r3c, Ppp1r3d, Ppp1r3g) and type II iodothyronine deiodinase (Dio2). These results highlight the importance of astrocytic energy metabolism and thyroid hormone signaling in LC-mediated hippocampal function and offer new molecular targets for understanding how NA impacts brain function in health and disease.
Collapse
Affiliation(s)
- Mattia Privitera
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Lukas M von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Amalia Floriou-Servou
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Sian N Duss
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Runzhong Zhang
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Rebecca Waag
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Sebastian Leimbacher
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Oliver Sturman
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Fabienne K Roessler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Annelies Heylen
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Yannick Vermeiren
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Division of Human Nutrition and Health, Chair Group of Nutritional Biology, Wageningen University & Research (WUR), Wageningen, Netherlands
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen (UMCG), Groningen, Netherlands
| | - Peter P De Deyn
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen (UMCG), Groningen, Netherlands
- Department of Neurology, Memory Clinic of Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Pierre-Luc Germain
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
- Computational Neurogenomics, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
- Laboratory of Statistical Bioinformatics, University of Zürich, Zürich, Switzerland
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| |
Collapse
|
25
|
Jensen VN, Huffman EE, Jalufka FL, Pritchard AL, Baumgartner S, Walling I, C. Gibbs H, McCreedy DA, Alilain WJ, Crone SA. V2a neurons restore diaphragm function in mice following spinal cord injury. Proc Natl Acad Sci U S A 2024; 121:e2313594121. [PMID: 38442182 PMCID: PMC10945804 DOI: 10.1073/pnas.2313594121] [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: 08/07/2023] [Accepted: 01/15/2024] [Indexed: 03/07/2024] Open
Abstract
The specific roles that different types of neurons play in recovery from injury is poorly understood. Here, we show that increasing the excitability of ipsilaterally projecting, excitatory V2a neurons using designer receptors exclusively activated by designer drugs (DREADDs) restores rhythmic bursting activity to a previously paralyzed diaphragm within hours, days, or weeks following a C2 hemisection injury. Further, decreasing the excitability of V2a neurons impairs tonic diaphragm activity after injury as well as activation of inspiratory activity by chemosensory stimulation, but does not impact breathing at rest in healthy animals. By examining the patterns of muscle activity produced by modulating the excitability of V2a neurons, we provide evidence that V2a neurons supply tonic drive to phrenic circuits rather than increase rhythmic inspiratory drive at the level of the brainstem. Our results demonstrate that the V2a class of neurons contribute to recovery of respiratory function following injury. We propose that altering V2a excitability is a potential strategy to prevent respiratory motor failure and promote recovery of breathing following spinal cord injury.
Collapse
Affiliation(s)
- Victoria N. Jensen
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH45219
| | - Emily E. Huffman
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY40536
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY40536
| | - Frank L. Jalufka
- Department of Biology, Texas A&M University, College Station, TX77843
| | - Anna L. Pritchard
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
| | - Sarah Baumgartner
- Division of Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Ian Walling
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH45219
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH45267
| | - Holly C. Gibbs
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
- Microscopy and Imaging Center, Texas A&M University, College Station, TX77843
| | - Dylan A. McCreedy
- Department of Biology, Texas A&M University, College Station, TX77843
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX77843
| | - Warren J. Alilain
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY40536
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY40536
| | - Steven A. Crone
- Division of Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH45267
| |
Collapse
|
26
|
Paniccia JE, Vollmer KM, Green LM, Grant RI, Winston KT, Buchmaier S, Westphal AM, Clarke RE, Doncheck EM, Bordieanu B, Manusky LM, Martino MR, Ward AL, Rinker JA, McGinty JF, Scofield MD, Otis JM. Restoration of a paraventricular thalamo-accumbal behavioral suppression circuit prevents reinstatement of heroin seeking. Neuron 2024; 112:772-785.e9. [PMID: 38141605 PMCID: PMC10939883 DOI: 10.1016/j.neuron.2023.11.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 10/17/2023] [Accepted: 11/29/2023] [Indexed: 12/25/2023]
Abstract
Lack of behavioral suppression typifies substance use disorders, yet the neural circuit underpinnings of drug-induced behavioral disinhibition remain unclear. Here, we employ deep-brain two-photon calcium imaging in heroin self-administering mice, longitudinally tracking adaptations within a paraventricular thalamus to nucleus accumbens behavioral inhibition circuit from the onset of heroin use to reinstatement. We find that select thalamo-accumbal neuronal ensembles become profoundly hypoactive across the development of heroin seeking and use. Electrophysiological experiments further reveal persistent adaptations at thalamo-accumbal parvalbumin interneuronal synapses, whereas functional rescue of these synapses prevents multiple triggers from initiating reinstatement of heroin seeking. Finally, we find an enrichment of μ-opioid receptors in output- and cell-type-specific paraventricular thalamic neurons, which provide a mechanism for heroin-induced synaptic plasticity and behavioral disinhibition. These findings reveal key circuit adaptations that underlie behavioral disinhibition in opioid dependence and further suggest that recovery of this system would reduce relapse susceptibility.
Collapse
Affiliation(s)
- Jacqueline E Paniccia
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kelsey M Vollmer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Lisa M Green
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Roger I Grant
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kion T Winston
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sophie Buchmaier
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Annaka M Westphal
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Rachel E Clarke
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Elizabeth M Doncheck
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bogdan Bordieanu
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Logan M Manusky
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael R Martino
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Amy L Ward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jennifer A Rinker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jacqueline F McGinty
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - James M Otis
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Ralph Johnson Veterans Administration, Charleston, SC 29425, USA.
| |
Collapse
|
27
|
Pan S, A.C. Souza L, Worker CJ, Reyes Mendez ME, Gayban AJB, Cooper SG, Sanchez Solano A, Bergman RN, Stefanovski D, Morton GJ, Schwartz MW, Feng Earley Y. (Pro)renin receptor signaling in hypothalamic tyrosine hydroxylase neurons is required for obesity-associated glucose metabolic impairment. JCI Insight 2024; 9:e174294. [PMID: 38349753 PMCID: PMC11063935 DOI: 10.1172/jci.insight.174294] [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: 09/01/2023] [Accepted: 02/08/2024] [Indexed: 03/06/2024] Open
Abstract
Glucose homeostasis is achieved via complex interactions between the endocrine pancreas and other peripheral tissues and glucoregulatory neurocircuits in the brain that remain incompletely defined. Within the brain, neurons in the hypothalamus appear to play a particularly important role. Consistent with this notion, we report evidence that (pro)renin receptor (PRR) signaling within a subset of tyrosine hydroxylase (TH) neurons located in the hypothalamic paraventricular nucleus (PVNTH neurons) is a physiological determinant of the defended blood glucose level. Specifically, we demonstrate that PRR deletion from PVNTH neurons restores normal glucose homeostasis in mice with diet-induced obesity (DIO). Conversely, chemogenetic inhibition of PVNTH neurons mimics the deleterious effect of DIO on glucose. Combined with our finding that PRR activation inhibits PVNTH neurons, these findings suggest that, in mice, (a) PVNTH neurons play a physiological role in glucose homeostasis, (b) PRR activation impairs glucose homeostasis by inhibiting these neurons, and (c) this mechanism plays a causal role in obesity-associated metabolic impairment.
Collapse
Affiliation(s)
- Shiyue Pan
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Lucas A.C. Souza
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Caleb J. Worker
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Miriam E. Reyes Mendez
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Ariana Julia B. Gayban
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Silvana G. Cooper
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Alfredo Sanchez Solano
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Darko Stefanovski
- New Bolton Center, School of Veterinary Medicine, University of Pennsylvania Philadelphia, Pennsylvania, USA
| | - Gregory J. Morton
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, Washington, USA
| | - Michael W. Schwartz
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, Washington, USA
| | - Yumei Feng Earley
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| |
Collapse
|
28
|
Bachor TP, Hwang E, Yulyaningsih E, Attal K, Mifsud F, Pham V, Vagena E, Huarcaya R, Valdearcos M, Vaisse C, Williams KW, Emmerson PJ, Xu AW. Identification of AgRP cells in the murine hindbrain that drive feeding. Mol Metab 2024; 80:101886. [PMID: 38246589 PMCID: PMC10844855 DOI: 10.1016/j.molmet.2024.101886] [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: 11/10/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
OBJECTIVE The central melanocortin system is essential for the regulation of food intake and body weight. Agouti-related protein (AgRP) is the sole orexigenic component of the central melanocortin system and is conserved across mammalian species. AgRP is currently known to be expressed exclusively in the mediobasal hypothalamus, and hypothalamic AgRP-expressing neurons are essential for feeding. Here we characterized a previously unknown population of AgRP cells in the mouse hindbrain. METHODS Expression of AgRP in the hindbrain was investigated using gene expression analysis, single-cell RNA sequencing, immunofluorescent analysis and multiple transgenic mice with reporter expressions. Activation of AgRP neurons was achieved by Designer Receptors Exclusively Activated by Designer Drugs (DREADD) and by transcranial focal photo-stimulation using a step-function opsin with ultra-high light sensitivity (SOUL). RESULTS AgRP expressing cells were present in the area postrema (AP) and the adjacent subpostrema area (SubP) and commissural nucleus of the solitary tract (cNTS) of the mouse hindbrain (termed AgRPHind herein). AgRPHind cells consisted of locally projecting neurons as well as tanycyte-like cells. Food deprivation stimulated hindbrain Agrp expression as well as neuronal activity of subsets of AgRPHind cells. In adult mice that lacked hypothalamic AgRP neurons, chemogenetic activation of AgRP neurons resulted in hyperphagia and weight gain. In addition, transcranial focal photo-stimulation of hindbrain AgRP cells increased food intake in adult mice with or without hypothalamic AgRP neurons. CONCLUSIONS Our study indicates that the central melanocortin system in the hindbrain possesses an orexigenic component, and that AgRPHind neurons stimulate feeding independently of hypothalamic AgRP neurons.
Collapse
Affiliation(s)
- Tomas P Bachor
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Eunsang Hwang
- Center for Hypothalamic Research, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Ernie Yulyaningsih
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Kush Attal
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Francois Mifsud
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Viana Pham
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Eirini Vagena
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Renzo Huarcaya
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Martin Valdearcos
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Christian Vaisse
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Kevin W Williams
- Center for Hypothalamic Research, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Paul J Emmerson
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly & Company, Indianapolis, IN, USA
| | - Allison W Xu
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA.
| |
Collapse
|
29
|
Huang JL, Pourhosseinzadeh MS, Lee S, Krämer N, Guillen JV, Cinque NH, Aniceto P, Momen AT, Koike S, Huising MO. Paracrine signalling by pancreatic δ cells determines the glycaemic set point in mice. Nat Metab 2024; 6:61-77. [PMID: 38195859 PMCID: PMC10919447 DOI: 10.1038/s42255-023-00944-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/09/2023] [Indexed: 01/11/2024]
Abstract
While pancreatic β and α cells are considered the main drivers of blood glucose homeostasis through insulin and glucagon secretion, the contribution of δ cells and somatostatin (SST) secretion to glucose homeostasis remains unresolved. Here we provide a quantitative assessment of the physiological contribution of δ cells to the glycaemic set point in mice. Employing three orthogonal mouse models to remove SST signalling within the pancreas or transplanted islets, we demonstrate that ablating δ cells or SST leads to a sustained decrease in the glycaemic set point. This reduction coincides with a decreased glucose threshold for insulin response from β cells, leading to increased insulin secretion to the same glucose challenge. Our data demonstrate that β cells are sufficient to maintain stable glycaemia and reveal that the physiological role of δ cells is to provide tonic feedback inhibition that reduces the β cell glucose threshold and consequently lowers the glycaemic set point in vivo.
Collapse
Affiliation(s)
- Jessica L Huang
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Mohammad S Pourhosseinzadeh
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Sharon Lee
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Niels Krämer
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
- Department of Animal Ecology and Physiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Jaresley V Guillen
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Naomi H Cinque
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Paola Aniceto
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Ariana T Momen
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Shinichiro Koike
- Department of Nutrition, University of California, Davis, CA, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA.
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
30
|
Dheer A, Bosco DB, Zheng J, Wang L, Zhao S, Haruwaka K, Yi MH, Barath A, Tian DS, Wu LJ. Chemogenetic approaches reveal dual functions of microglia in seizures. Brain Behav Immun 2024; 115:406-418. [PMID: 37926132 PMCID: PMC10841657 DOI: 10.1016/j.bbi.2023.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/14/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023] Open
Abstract
Microglia are key players in maintaining brain homeostasis and exhibit phenotypic alterations in response to epileptic stimuli. However, it is still relatively unknown if these alterations are pro- or anti-epileptic. To unravel this dilemma, we employed chemogenetic manipulation of microglia using the artificial Gi-Dreadd receptor within a kainic acid (KA) induced murine seizure model. Our results indicate that acute Gi-Dreadd activation with Clozapine-N-Oxide can reduce seizure severity. Additionally, we observed increased interaction between microglia and neuronal soma, which correlated with reduced neuronal hyperactivity. Interestingly, prolonged activation of microglial Gi-Dreadds by repeated doses of CNO over 3 days, arrested microglia in a less active, homeostatic-like state, which associated with increased neuronal loss after KA induced seizures. RNAseq analysis revealed that prolonged activation of Gi-Dreadd interferes with interferon β signaling and microglia proliferation. Thus, our findings highlight the importance of microglial Gi signaling not only during status epilepticus (SE) but also within later seizure induced pathology.
Collapse
Affiliation(s)
- Aastha Dheer
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | | | - Min-Hee Yi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Abhijeet Barath
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
31
|
He Y, Liu T, He Q, Ke W, Li X, Du J, Deng S, Shu Z, Wu J, Yang B, Wang Y, Mao Y, Rao Y, Shu Y, Peng B. Microglia facilitate and stabilize the response to general anesthesia via modulating the neuronal network in a brain region-specific manner. eLife 2023; 12:RP92252. [PMID: 38131301 PMCID: PMC10746144 DOI: 10.7554/elife.92252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
General anesthesia leads to a loss of consciousness and an unrousable state in patients. Although general anesthetics are widely used in clinical practice, their underlying mechanisms remain elusive. The potential involvement of nonneuronal cells is unknown. Microglia are important immune cells in the central nervous system (CNS) that play critical roles in CNS function and dysfunction. We unintentionally observed delayed anesthesia induction and early anesthesia emergence in microglia-depleted mice. We found that microglial depletion differentially regulates neuronal activities by suppressing the neuronal network of anesthesia-activated brain regions and activating emergence-activated brain regions. Thus, microglia facilitate and stabilize the anesthesia status. This influence is not mediated by dendritic spine plasticity. Instead, it relies on the activation of microglial P2Y12 and subsequent calcium influx, which facilitates the general anesthesia response. Together, we elucidate the regulatory role of microglia in general anesthesia, extending our knowledge of how nonneuronal cells modulate neuronal activities.
Collapse
Affiliation(s)
- Yang He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Taohui Liu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Quansheng He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Wei Ke
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Xiaoyu Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Jinjin Du
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Suixin Deng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Zhenfeng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Jialin Wu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Baozhi Yang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Yuqing Wang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
| | - Yousheng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- Co-Innovation Center of Neurodegeneration, Nantong UniversityNantongChina
| |
Collapse
|
32
|
Thompson A, Arano R, Saleem U, Preciado R, Munoz L, Nelson I, Ramos K, Kim Y, Li Y, Xu W. Brain-wide circuit-specific targeting of astrocytes. CELL REPORTS METHODS 2023; 3:100653. [PMID: 38052209 PMCID: PMC10753298 DOI: 10.1016/j.crmeth.2023.100653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 10/04/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023]
Abstract
Astrocytes are integral components of brain circuitry. They enwrap synapses, react to neuronal activity, and regulate synaptic transmission. Astrocytes are heterogeneous and exhibit distinct features and functions in different circuits. Selectively targeting the astrocytes associated with a given neuronal circuit would enable elucidation of their circuit-specific functions but has been technically challenging to date. Recently, we constructed anterograde transneuronal viral vectors based on yellow fever vaccine YFV-17D. Among them, the replication-incompetent YFVΔNS1-Cre can selectively turn on reporter genes in postsynaptic neurons if the viral gene NS1 is expressed in postsynaptic neurons. Here we show that without exogenous expression of NS1 at the postsynaptic sites, locally injected YFVΔNS1-Cre selectively turns on reporter genes in astrocytes in downstream brain regions. The targeting of astrocytes can occur across the whole brain but is specific for the neuronal circuits traced. Therefore, YFVΔNS1-Cre provides a tool for selective genetic targeting of astrocytes to reveal their circuit-specific roles.
Collapse
Affiliation(s)
- Alyssa Thompson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rachel Arano
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Uzair Saleem
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rebecca Preciado
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lizbeth Munoz
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ian Nelson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katarina Ramos
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yerim Kim
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ying Li
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Wei Xu
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
33
|
Fiore F, Alhalaseh K, Dereddi RR, Bodaleo Torres F, Çoban I, Harb A, Agarwal A. Norepinephrine regulates calcium signals and fate of oligodendrocyte precursor cells in the mouse cerebral cortex. Nat Commun 2023; 14:8122. [PMID: 38065932 PMCID: PMC10709653 DOI: 10.1038/s41467-023-43920-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Oligodendrocyte precursor cells (OPCs) generate oligodendrocytes, contributing to myelination and myelin repair. OPCs contact axons and respond to neuronal activity, but how the information relayed by the neuronal activity translates into OPC Ca2+ signals, which in turn influence their fate, remains unknown. We generated transgenic mice for concomitant monitoring of OPCs Ca2+ signals and cell fate using 2-photon microscopy in the somatosensory cortex of awake-behaving mice. Ca2+ signals in OPCs mainly occur within processes and confine to Ca2+ microdomains. A subpopulation of OPCs enhances Ca2+ transients while mice engaged in exploratory locomotion. We found that OPCs responsive to locomotion preferentially differentiate into oligodendrocytes, and locomotion-non-responsive OPCs divide. Norepinephrine mediates locomotion-evoked Ca2+ increases in OPCs by activating α1 adrenergic receptors, and chemogenetic activation of OPCs or noradrenergic neurons promotes OPC differentiation. Hence, we uncovered that for fate decisions OPCs integrate Ca2+ signals, and norepinephrine is a potent regulator of OPC fate.
Collapse
Affiliation(s)
- Frederic Fiore
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Khaleel Alhalaseh
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Ram R Dereddi
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Felipe Bodaleo Torres
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Ilknur Çoban
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Ali Harb
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Amit Agarwal
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
34
|
Klima ML, Kruger KA, Goldstein N, Pulido S, Low AYT, Assenmacher CA, Alhadeff AL, Betley JN. Anti-inflammatory effects of hunger are transmitted to the periphery via projection-specific AgRP circuits. Cell Rep 2023; 42:113338. [PMID: 37910501 DOI: 10.1016/j.celrep.2023.113338] [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: 02/14/2019] [Revised: 07/31/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023] Open
Abstract
Caloric restriction has anti-inflammatory effects. However, the coordinated physiological actions that lead to reduced inflammation in a state of caloric deficit (hunger) are largely unknown. Using a mouse model of injury-induced peripheral inflammation, we find that food deprivation reduces edema, temperature, and cytokine responses that occur after injury. The magnitude of the anti-inflammatory effect that occurs during hunger is more robust than that of non-steroidal anti-inflammatory drugs. The effects of hunger are recapitulated centrally by activity in nutrient-sensing hypothalamic agouti-related protein (AgRP)-expressing neurons. We find that AgRP neurons projecting to the paraventricular nucleus of the hypothalamus rapidly and robustly reduce inflammation and mediate the majority of hunger's anti-inflammatory effects. Intact vagal efferent signaling is required for the anti-inflammatory action of hunger, revealing a brain-to-periphery pathway for this reduction in inflammation. Taken together, these data begin to unravel a potent anti-inflammatory pathway engaged by hypothalamic AgRP neurons to reduce inflammation.
Collapse
Affiliation(s)
- Michelle L Klima
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kayla A Kruger
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nitsan Goldstein
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Santiago Pulido
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aloysius Y T Low
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charles-Antoine Assenmacher
- Comparative Pathology Core, Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Amber L Alhadeff
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA; Monell Chemical Senses Center, Philadelphia, PA 19104, USA.
| | - J Nicholas Betley
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
Broersen R, Albergaria C, Carulli D, Carey MR, Canto CB, De Zeeuw CI. Synaptic mechanisms for associative learning in the cerebellar nuclei. Nat Commun 2023; 14:7459. [PMID: 37985778 PMCID: PMC10662440 DOI: 10.1038/s41467-023-43227-w] [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: 11/17/2022] [Accepted: 11/03/2023] [Indexed: 11/22/2023] Open
Abstract
Associative learning during delay eyeblink conditioning (EBC) depends on an intact cerebellum. However, the relative contribution of changes in the cerebellar nuclei to learning remains a subject of ongoing debate. In particular, little is known about the changes in synaptic inputs to cerebellar nuclei neurons that take place during EBC and how they shape the membrane potential of these neurons. Here, we probed the ability of these inputs to support associative learning in mice, and investigated structural and cell-physiological changes within the cerebellar nuclei during learning. We find that optogenetic stimulation of mossy fiber afferents to the anterior interposed nucleus (AIP) can substitute for a conditioned stimulus and is sufficient to elicit conditioned responses (CRs) that are adaptively well-timed. Further, EBC induces structural changes in mossy fiber and inhibitory inputs, but not in climbing fiber inputs, and it leads to changes in subthreshold processing of AIP neurons that correlate with conditioned eyelid movements. The changes in synaptic and spiking activity that precede the CRs allow for a decoder to distinguish trials with a CR. Our data reveal how structural and physiological modifications of synaptic inputs to cerebellar nuclei neurons can facilitate learning.
Collapse
Affiliation(s)
- Robin Broersen
- Department of Cerebellar Coordination and Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Catarina Albergaria
- Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
- University College London, Sainsbury Wellcome Centre, London, UK
| | - Daniela Carulli
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Megan R Carey
- Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal.
| | - Cathrin B Canto
- Department of Cerebellar Coordination and Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands.
| | - Chris I De Zeeuw
- Department of Cerebellar Coordination and Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
36
|
Moghimyfiroozabad S, Paul MA, Sigoillot SM, Selimi F. Mapping and targeting of C1ql1-expressing cells in the mouse. Sci Rep 2023; 13:17563. [PMID: 37845276 PMCID: PMC10579299 DOI: 10.1038/s41598-023-42924-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/16/2023] [Indexed: 10/18/2023] Open
Abstract
The C1Q complement protein C1QL1 is highly conserved in mammals where it is expressed in various tissues including the brain. This secreted protein interacts with Brain-specific Angiogenesis Inhibitor 3, BAI3/ADGRB3, and controls synapse formation and maintenance. C1ql1 is expressed in the inferior olivary neurons that send projections to cerebellar Purkinje cells, but its expression in the rest of the brain is less documented. To map C1ql1 expression and enable the specific targeting of C1ql1-expressing cells, we generated a knockin mouse model expressing the Cre recombinase under the control of C1ql1 regulatory sequences. We characterized the capacity for Cre-driven recombination in the brain and mapped Cre expression in various neuron types using reporter mouse lines. Using an intersectional strategy with viral particle injections, we show that this mouse line can be used to target specific afferents of Purkinje cells. As C1ql1 is also expressed in other regions of the brain, as well as in other tissues such as adrenal glands and colon, our mouse model is a useful tool to target C1ql1-expressing cells in a broad variety of tissues.
Collapse
Affiliation(s)
- Shayan Moghimyfiroozabad
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Maëla A Paul
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Séverine M Sigoillot
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France.
| | - Fekrije Selimi
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France.
| |
Collapse
|
37
|
Lisgaras CP, Scharfman HE. Interictal spikes in Alzheimer's disease: Preclinical evidence for dominance of the dentate gyrus and cholinergic control by the medial septum. Neurobiol Dis 2023; 187:106294. [PMID: 37714307 PMCID: PMC10617404 DOI: 10.1016/j.nbd.2023.106294] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023] Open
Abstract
Interictal spikes (IIS) are a common type of abnormal electrical activity in Alzheimer's disease (AD) and preclinical models. The brain regions where IIS are largest are not known but are important because such data would suggest sites that contribute to IIS generation. Because hippocampus and cortex exhibit altered excitability in AD models, we asked which areas dominate the activity during IIS along the cortical-CA1-dentate gyrus (DG) dorso-ventral axis. Because medial septal (MS) cholinergic neurons are overactive when IIS typically occur, we also tested the novel hypothesis that silencing the MS cholinergic neurons selectively would reduce IIS. We used mice that simulate aspects of AD: Tg2576 mice, presenilin 2 (PS2) knockout mice and Ts65Dn mice. To selectively silence MS cholinergic neurons, Tg2576 mice were bred with choline-acetyltransferase (ChAT)-Cre mice and offspring were injected in the MS with AAV encoding inhibitory designer receptors exclusively activated by designer drugs (DREADDs). We recorded local field potentials along the cortical-CA1-DG axis using silicon probes during wakefulness, slow-wave sleep (SWS) and rapid eye movement (REM) sleep. We detected IIS in all transgenic or knockout mice but not age-matched controls. IIS were detectable throughout the cortical-CA1-DG axis and occurred primarily during REM sleep. In all 3 mouse lines, IIS amplitudes were significantly greater in the DG granule cell layer vs. CA1 pyramidal layer or overlying cortex. Current source density analysis showed robust and early current sources in the DG, and additional sources in CA1 and the cortex also. Selective chemogenetic silencing of MS cholinergic neurons significantly reduced IIS rate during REM sleep without affecting the overall duration, number of REM bouts, latency to REM sleep, or theta power during REM. Notably, two control interventions showed no effects. Consistent maximal amplitude and strong current sources of IIS in the DG suggest that the DG is remarkably active during IIS. In addition, selectively reducing MS cholinergic tone, at times when MS is hyperactive, could be a new strategy to reduce IIS in AD.
Collapse
Affiliation(s)
- Christos Panagiotis Lisgaras
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, and the Neuroscience Institute New York University Langone Health, 550 First Ave., New York, NY 10016, United States of America; Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, New York State Office of Mental Health, 140 Old Orangeburg Road, Bldg. 35, Orangeburg, NY 10962, United States of America.
| | - Helen E Scharfman
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, and the Neuroscience Institute New York University Langone Health, 550 First Ave., New York, NY 10016, United States of America; Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, New York State Office of Mental Health, 140 Old Orangeburg Road, Bldg. 35, Orangeburg, NY 10962, United States of America
| |
Collapse
|
38
|
van Bruggen R, Patel ZH, Wang M, Suk TR, Rousseaux MWC, Tan Q. A Versatile Strategy for Genetic Manipulation of Cajal-Retzius Cells in the Adult Mouse Hippocampus. eNeuro 2023; 10:ENEURO.0054-23.2023. [PMID: 37775311 PMCID: PMC10585607 DOI: 10.1523/eneuro.0054-23.2023] [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: 02/14/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023] Open
Abstract
Cajal-Retzius (CR) cells are transient neurons with long-lasting effects on the architecture and circuitry of the neocortex and hippocampus. Contrary to the prevailing assumption that CR cells completely disappear in rodents shortly after birth, a substantial portion of these cells persist in the hippocampus throughout adulthood. The role of these surviving CR cells in the adult hippocampus is largely unknown, partly because of the paucity of suitable tools to dissect their functions in the adult versus the embryonic brain. Here, we show that genetic crosses of the ΔNp73-Cre mouse line, widely used to target CR cells, to reporter mice induce reporter expression not only in CR cells, but also progressively in postnatal dentate gyrus granule neurons. Such a lack of specificity may confound studies of CR cell function in the adult hippocampus. To overcome this, we devise a method that not only leverages the temporary CR cell-targeting specificity of the ΔNp73-Cre mice before the first postnatal week, but also capitalizes on the simplicity and effectiveness of freehand neonatal intracerebroventricular injection of adeno-associated virus. We achieve robust Cre-mediated recombination that remains largely restricted to hippocampal CR cells from early postnatal age to adulthood. We further demonstrate the utility of this method to manipulate neuronal activity of CR cells in the adult hippocampus. This versatile and scalable strategy will facilitate experiments of CR cell-specific gene knockdown and/or overexpression, lineage tracing, and neural activity modulation in the postnatal and adult brain.
Collapse
Affiliation(s)
- Rebekah van Bruggen
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Zain H Patel
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Mi Wang
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Terry R Suk
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Eric Poulin Center for Neuromuscular Diseases, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Maxime W C Rousseaux
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Eric Poulin Center for Neuromuscular Diseases, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Qiumin Tan
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta T6G 1C9, Canada
| |
Collapse
|
39
|
Lisgaras CP, Scharfman HE. Interictal Spikes in Alzheimer's Disease: Preclinical Evidence for Dominance of the Dentate Gyrus and Cholinergic Control by Medial Septum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.24.537999. [PMID: 37163065 PMCID: PMC10168266 DOI: 10.1101/2023.04.24.537999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
HIGHLIGHTS Interictal spikes (IIS) occur in 3 mouse lines with Alzheimer's disease featuresIIS in all 3 mouse lines were most frequent during rapid eye movement (REM) sleepThe dentate gyrus showed larger IIS and earlier current sources vs. CA1 or cortexChemogenetic silencing of medial septum (MS) cholinergic neurons reduced IIS during REMMS silencing did not change REM latency, duration, number of bouts or theta power. Interictal spikes (IIS) are a common type of abnormal electrical activity in Alzheimer's disease (AD) and preclinical models. The brain regions where IIS are largest are not known but are important because such data would suggest sites that contribute to IIS generation. Because hippocampus and cortex exhibit altered excitability in AD models, we asked which areas dominate the activity during IIS along the cortical-CA1-dentate gyrus (DG) dorso-ventral axis. Because medial septal (MS) cholinergic neurons are overactive when IIS typically occur, we also tested the novel hypothesis that silencing the MS cholinergic neurons selectively would reduce IIS.We used mice that simulate aspects of AD: Tg2576 mice, presenilin 2 (PS2) knockout mice and Ts65Dn mice. To selectively silence MS cholinergic neurons, Tg2576 mice were bred with choline-acetyltransferase (ChAT)-Cre mice and offspring were injected in the MS with AAV encoding inhibitory designer receptors exclusively activated by designer drugs (DREADDs). We recorded local field potentials along the cortical-CA1-DG axis using silicon probes during wakefulness, slow-wave sleep (SWS) and rapid eye movement (REM) sleep.We detected IIS in all transgenic or knockout mice but not age-matched controls. IIS were detectable throughout the cortical-CA1-DG axis and occurred primarily during REM sleep. In all 3 mouse lines, IIS amplitudes were significantly greater in the DG granule cell layer vs. CA1 pyramidal layer or overlying cortex. Current source density analysis showed robust and early current sources in the DG, and additional sources in CA1 and the cortex also. Selective chemogenetic silencing of MS cholinergic neurons significantly reduced IIS rate during REM sleep without affecting the overall duration, number of REM bouts, latency to REM sleep, or theta power during REM. Notably, two control interventions showed no effects.Consistent maximal amplitude and strong current sources of IIS in the DG suggest that the DG is remarkably active during IIS. In addition, selectively reducing MS cholinergic tone, at times when MS is hyperactive, could be a new strategy to reduce IIS in AD.
Collapse
|
40
|
Bohic M, Upadhyay A, Eisdorfer JT, Keating J, Simon RC, Briones BA, Azadegan C, Nacht HD, Oputa O, Martinez AM, Bethell BN, Gradwell MA, Romanienko P, Ramer MS, Stuber GD, Abraira VE. A new Hoxb8FlpO mouse line for intersectional approaches to dissect developmentally defined adult sensorimotor circuits. Front Mol Neurosci 2023; 16:1176823. [PMID: 37603775 PMCID: PMC10437123 DOI: 10.3389/fnmol.2023.1176823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/04/2023] [Indexed: 08/23/2023] Open
Abstract
Improvements in the speed and cost of expression profiling of neuronal tissues offer an unprecedented opportunity to define ever finer subgroups of neurons for functional studies. In the spinal cord, single cell RNA sequencing studies support decades of work on spinal cord lineage studies, offering a unique opportunity to probe adult function based on developmental lineage. While Cre/Flp recombinase intersectional strategies remain a powerful tool to manipulate spinal neurons, the field lacks genetic tools and strategies to restrict manipulations to the adult mouse spinal cord at the speed at which new tools develop. This study establishes a new workflow for intersectional mouse-viral strategies to dissect adult spinal function based on developmental lineages in a modular fashion. To restrict manipulations to the spinal cord, we generate a brain-sparing Hoxb8FlpO mouse line restricting Flp recombinase expression to caudal tissue. Recapitulating endogenous Hoxb8 gene expression, Flp-dependent reporter expression is present in the caudal embryo starting day 9.5. This expression restricts Flp activity in the adult to the caudal brainstem and below. Hoxb8FlpO heterozygous and homozygous mice do not develop any of the sensory or locomotor phenotypes evident in Hoxb8 heterozygous or mutant animals, suggesting normal developmental function of the Hoxb8 gene and protein in Hoxb8FlpO mice. Compared to the variability of brain recombination in available caudal Cre and Flp lines, Hoxb8FlpO activity is not present in the brain above the caudal brainstem, independent of mouse genetic background. Lastly, we combine the Hoxb8FlpO mouse line with dorsal horn developmental lineage Cre mouse lines to express GFP in developmentally determined dorsal horn populations. Using GFP-dependent Cre recombinase viruses and Cre recombinase-dependent inhibitory chemogenetics, we target developmentally defined lineages in the adult. We show how developmental knock-out versus transient adult silencing of the same ROR𝛃 lineage neurons affects adult sensorimotor behavior. In summary, this new mouse line and viral approach provides a blueprint to dissect adult somatosensory circuit function using Cre/Flp genetic tools to target spinal cord interneurons based on genetic lineage.
Collapse
Affiliation(s)
- Manon Bohic
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Aman Upadhyay
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- Neuroscience PhD Program at Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Jaclyn T. Eisdorfer
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Jessica Keating
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- School of Medicine, Oregon Health and Science University, Portland, OR, United States
- M.D./PhD Program in Neuroscience, School of Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Rhiana C. Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Brandy A. Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Chloe Azadegan
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Hannah D. Nacht
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Olisemeka Oputa
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Alana M. Martinez
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Bridget N. Bethell
- International Collaboration on Repair Discoveries and Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Mark A. Gradwell
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Peter Romanienko
- Genome Editing Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Matt S. Ramer
- International Collaboration on Repair Discoveries and Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Garret D. Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Victoria E. Abraira
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| |
Collapse
|
41
|
Kim SK, Tran LT, NamKoong C, Choi HJ, Chun HJ, Lee YH, Cheon M, Chung C, Hwang J, Lim HH, Shin DM, Choi YH, Kim KW. Mitochondria-derived peptide SHLP2 regulates energy homeostasis through the activation of hypothalamic neurons. Nat Commun 2023; 14:4321. [PMID: 37468558 PMCID: PMC10356901 DOI: 10.1038/s41467-023-40082-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
Small humanin-like peptide 2 (SHLP2) is a mitochondrial-derived peptide implicated in several biological processes such as aging and oxidative stress. However, its functional role in the regulation of energy homeostasis remains unclear, and its corresponding receptor is not identified. Hereby, we demonstrate that both systemic and intracerebroventricular (ICV) administrations of SHLP2 protected the male mice from high-fat diet (HFD)-induced obesity and improved insulin sensitivity. In addition, the activation of pro-opiomelanocortin (POMC) neurons by SHLP2 in the arcuate nucleus of the hypothalamus (ARC) is involved in the suppression of food intake and the promotion of thermogenesis. Through high-throughput structural complementation screening, we discovered that SHLP2 binds to and activates chemokine receptor 7 (CXCR7). Taken together, our study not only reveals the therapeutic potential of SHLP2 in metabolic disorders but also provides important mechanistic insights into how it exerts its effects on energy homeostasis.
Collapse
Affiliation(s)
- Seul Ki Kim
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, 03722, Korea
- Department of Applied Life Science, BK21 FOUR, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Le Trung Tran
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, 03722, Korea
- Department of Applied Life Science, BK21 FOUR, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Cherl NamKoong
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Hyung Jin Choi
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Hye Jin Chun
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Yong-Ho Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - MyungHyun Cheon
- Department of Biological Sciences, Konkuk University, Seoul, 05029, Korea
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul, 05029, Korea
| | - Junmo Hwang
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu, 41068, Korea
| | - Hyun-Ho Lim
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu, 41068, Korea
| | - Dong Min Shin
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, 03722, Korea
- Department of Applied Life Science, BK21 FOUR, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Yun-Hee Choi
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Ki Woo Kim
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, 03722, Korea.
- Department of Applied Life Science, BK21 FOUR, Yonsei University College of Dentistry, Seoul, 03722, Korea.
| |
Collapse
|
42
|
Khouma A, Moeini MM, Plamondon J, Richard D, Caron A, Michael NJ. Histaminergic regulation of food intake. Front Endocrinol (Lausanne) 2023; 14:1202089. [PMID: 37448468 PMCID: PMC10338010 DOI: 10.3389/fendo.2023.1202089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/06/2023] [Indexed: 07/15/2023] Open
Abstract
Histamine is a biogenic amine that acts as a neuromodulator within the brain. In the hypothalamus, histaminergic signaling contributes to the regulation of numerous physiological and homeostatic processes, including the regulation of energy balance. Histaminergic neurons project extensively throughout the hypothalamus and two histamine receptors (H1R, H3R) are strongly expressed in key hypothalamic nuclei known to regulate energy homeostasis, including the paraventricular (PVH), ventromedial (VMH), dorsomedial (DMH), and arcuate (ARC) nuclei. The activation of different histamine receptors is associated with differential effects on neuronal activity, mediated by their different G protein-coupling. Consequently, activation of H1R has opposing effects on food intake to that of H3R: H1R activation suppresses food intake, while H3R activation mediates an orexigenic response. The central histaminergic system has been implicated in atypical antipsychotic-induced weight gain and has been proposed as a potential therapeutic target for the treatment of obesity. It has also been demonstrated to interact with other major regulators of energy homeostasis, including the central melanocortin system and the adipose-derived hormone leptin. However, the exact mechanisms by which the histaminergic system contributes to the modification of these satiety signals remain underexplored. The present review focuses on recent advances in our understanding of the central histaminergic system's role in regulating feeding and highlights unanswered questions remaining in our knowledge of the functionality of this system.
Collapse
Affiliation(s)
- Axelle Khouma
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Moein Minbashi Moeini
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Julie Plamondon
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
| | - Denis Richard
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Medicine, Université Laval, Québec, QC, Canada
| | - Alexandre Caron
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Montreal Diabetes Research Center, Montreal, QC, Canada
| | - Natalie Jane Michael
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| |
Collapse
|
43
|
Nguyen TTT, Lu W, Zhu WS, Ansel KM, Liang HE, Weiss A. Stimulation of ectopically expressed muscarinic receptors induces IFN-γ but suppresses IL-2 production by inhibiting activation of pAKT pathways in primary T cells. Proc Natl Acad Sci U S A 2023; 120:e2300987120. [PMID: 37307442 PMCID: PMC10288620 DOI: 10.1073/pnas.2300987120] [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: 01/17/2023] [Accepted: 05/15/2023] [Indexed: 06/14/2023] Open
Abstract
T cell antigen receptor stimulation induces tyrosine phosphorylation of downstream signaling molecules and the phosphatidylinositol, Ras, MAPK, and PI3 kinase pathways, leading to T cell activation. Previously, we reported that the G-protein-coupled human muscarinic receptor could bypass tyrosine kinases to activate the phosphatidylinositol pathway and induce interleukin-2 production in Jurkat leukemic T cells. Here, we demonstrate that stimulating G-protein-coupled muscarinic receptors (M1 and synthetic hM3Dq) can activate primary mouse T cells if PLCβ1 is coexpressed. Resting peripheral hM3Dq+PLCβ1 (hM3Dq/β1) T cells did not respond to clozapine, an hM3Dq agonist, unless they were preactivated by TCR and CD28 stimulation which increased hM3Dq and PLCβ1 expression. This permitted large calcium and phosphorylated ERK responses to clozapine. Clozapine treatment induced high IFN-γ, CD69, and CD25 expression, but surprisingly did not induce substantial IL-2 in hM3Dq/β1 T cells. Importantly, costimulation of both muscarinic receptors plus the TCR even led to reduced IL-2 expression, suggesting a selective inhibitory effect of muscarinic receptor costimulation. Stimulation of muscarinic receptors induced strong nuclear translocation of NFAT and NFκB and activated AP-1. However, stimulation of hM3Dq led to reduced IL-2 mRNA stability which correlated with an effect on the IL-2 3'UTR activity. Interestingly, stimulation of hM3Dq resulted in reduced pAKT and its downstream pathway. This may explain the inhibitory impact on IL-2 production in hM3Dq/β1T cells. Moreover, an inhibitor of PI3K reduced IL-2 production in TCR-stimulated hM3Dq/β1 CD4 T cells, suggesting that activating the pAKT pathway is critical for IL-2 production in T cells.
Collapse
Affiliation(s)
- Trang T. T. Nguyen
- Rosalind Russell-Ephraim Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA 94143
| | - Wen Lu
- Rosalind Russell-Ephraim Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA 94143
| | - Wandi S. Zhu
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA94143
| | - K. Mark Ansel
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA94143
| | - Hong-Erh Liang
- Rosalind Russell-Ephraim Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA 94143
- Department of Medicine, University of California San Francisco, San Francisco, 94143
| | - Arthur Weiss
- Rosalind Russell-Ephraim Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA 94143
| |
Collapse
|
44
|
Li YD, Luo YJ, Xie L, Tart DS, Sheehy RN, Zhang L, Coleman LG, Chen X, Song J. Activation of hypothalamic-enhanced adult-born neurons restores cognitive and affective function in Alzheimer's disease. Cell Stem Cell 2023; 30:415-432.e6. [PMID: 37028406 PMCID: PMC10150940 DOI: 10.1016/j.stem.2023.02.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/29/2022] [Accepted: 02/14/2023] [Indexed: 04/09/2023]
Abstract
Patients with Alzheimer's disease (AD) exhibit progressive memory loss, depression, and anxiety, accompanied by impaired adult hippocampal neurogenesis (AHN). Whether AHN can be enhanced in impaired AD brain to restore cognitive and affective function remains elusive. Here, we report that patterned optogenetic stimulation of the hypothalamic supramammillary nucleus (SuM) enhances AHN in two distinct AD mouse models, 5×FAD and 3×Tg-AD. Strikingly, the chemogenetic activation of SuM-enhanced adult-born neurons (ABNs) rescues memory and emotion deficits in these AD mice. By contrast, SuM stimulation alone or activation of ABNs without SuM modification fails to restore behavioral deficits. Furthermore, quantitative phosphoproteomics analyses reveal activation of the canonical pathways related to synaptic plasticity and microglia phagocytosis of plaques following acute chemogenetic activation of SuM-enhanced (vs. control) ABNs. Our study establishes the activity-dependent contribution of SuM-enhanced ABNs in modulating AD-related deficits and informs signaling mechanisms mediated by the activation of SuM-enhanced ABNs.
Collapse
Affiliation(s)
- Ya-Dong Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yan-Jia Luo
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dalton S Tart
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ryan N Sheehy
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Pharmacology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Libo Zhang
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leon G Coleman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
45
|
Sutton Hickey AK, Duane SC, Mickelsen LE, Karolczak EO, Shamma AM, Skillings A, Li C, Krashes MJ. AgRP neurons coordinate the mitigation of activity-based anorexia. Mol Psychiatry 2023; 28:1622-1635. [PMID: 36577844 PMCID: PMC10782560 DOI: 10.1038/s41380-022-01932-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/29/2022]
Abstract
Anorexia nervosa (AN) is a debilitating and deadly disease characterized by low body mass index due to diminished food intake, and oftentimes concurrent hyperactivity. A high percentage of AN behavioral and metabolic phenotypes can be replicated in rodents given access to a voluntary running wheel and subject to food restriction, termed activity-based anorexia (ABA). Despite the well-documented bodyweight loss observed in AN human patients and ABA rodents, much less is understood regarding the neurobiological underpinnings of these maladaptive behaviors. Hunger-promoting hypothalamic agouti-related peptide (AgRP) neurons have been well characterized in their ability to regulate appetite, yet much less is known regarding their activity and function in the mediation of food intake during ABA. Here, feeding microstructure analysis revealed ABA mice decreased food intake due to increased interpellet interval retrieval and diminished meal number. Longitudinal activity recordings of AgRP neurons in ABA animals exhibited a maladaptive inhibitory response to food, independent of basal activity changes. We then demonstrated that ABA development or progression can be mitigated by chemogenetic AgRP activation through the reprioritization of food intake (increased meal number) over hyperactivity, but only during periods of food availability. These results elucidate a potential neural target for the amelioration of behavioral maladaptations present in AN patients.
Collapse
Affiliation(s)
- Ames K Sutton Hickey
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.
| | - Sean C Duane
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Laura E Mickelsen
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Eva O Karolczak
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Ahmed M Shamma
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Anna Skillings
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Chia Li
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
46
|
Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, Rich LM, Barilli A, Sabat M, Debley JS, Myers R, Howitt MR, von Moltke J. Tuft cell-derived acetylcholine regulates epithelial fluid secretion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.533208. [PMID: 36993541 PMCID: PMC10055254 DOI: 10.1101/2023.03.17.533208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Tuft cells are solitary chemosensory epithelial cells that can sense lumenal stimuli at mucosal barriers and secrete effector molecules to regulate the physiology and immune state of their surrounding tissue. In the small intestine, tuft cells detect parasitic worms (helminths) and microbe-derived succinate, and signal to immune cells to trigger a Type 2 immune response that leads to extensive epithelial remodeling spanning several days. Acetylcholine (ACh) from airway tuft cells has been shown to stimulate acute changes in breathing and mucocilliary clearance, but its function in the intestine is unknown. Here we show that tuft cell chemosensing in the intestine leads to release of ACh, but that this does not contribute to immune cell activation or associated tissue remodeling. Instead, tuft cell-derived ACh triggers immediate fluid secretion from neighboring epithelial cells into the intestinal lumen. This tuft cell-regulated fluid secretion is amplified during Type 2 inflammation, and helminth clearance is delayed in mice lacking tuft cell ACh. The coupling of the chemosensory function of tuft cells with fluid secretion creates an epithelium-intrinsic response unit that effects a physiological change within seconds of activation. This response mechanism is shared by tuft cells across tissues, and serves to regulate the epithelial secretion that is both a hallmark of Type 2 immunity and an essential component of homeostatic maintenance at mucosal barriers.
Collapse
Affiliation(s)
- Tyler E. Billipp
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lily M. Webeck
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Derek B. Sargent
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthew B. Gologorsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Margaret M. McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Darshan N. Kasal
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - John W. McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Kaitlyn A. Barrow
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Lucille M. Rich
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Mark Sabat
- Takeda Pharmaceuticals, San Diego, California, USA
| | - Jason S. Debley
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | | | - Michael R. Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
47
|
Banerjee T, Pati S, Tiwari P, Vaidya VA. Chronic hM3Dq-DREADD-mediated chemogenetic activation of parvalbumin-positive inhibitory interneurons in postnatal life alters anxiety and despair-like behavior in adulthood in a task- and sex-dependent manner. J Biosci 2022. [DOI: 10.1007/s12038-022-00308-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
48
|
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
|
49
|
Basilico B, Ferrucci L, Khan A, Di Angelantonio S, Ragozzino D, Reverte I. What microglia depletion approaches tell us about the role of microglia on synaptic function and behavior. Front Cell Neurosci 2022; 16:1022431. [PMID: 36406752 PMCID: PMC9673171 DOI: 10.3389/fncel.2022.1022431] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Microglia are dynamic cells, constantly surveying their surroundings and interacting with neurons and synapses. Indeed, a wealth of knowledge has revealed a critical role of microglia in modulating synaptic transmission and plasticity in the developing brain. In the past decade, novel pharmacological and genetic strategies have allowed the acute removal of microglia, opening the possibility to explore and understand the role of microglia also in the adult brain. In this review, we summarized and discussed the contribution of microglia depletion strategies to the current understanding of the role of microglia on synaptic function, learning and memory, and behavior both in physiological and pathological conditions. We first described the available microglia depletion methods highlighting their main strengths and weaknesses. We then reviewed the impact of microglia depletion on structural and functional synaptic plasticity. Next, we focused our analysis on the effects of microglia depletion on behavior, including general locomotor activity, sensory perception, motor function, sociability, learning and memory both in healthy animals and animal models of disease. Finally, we integrated the findings from the reviewed studies and discussed the emerging roles of microglia on the maintenance of synaptic function, learning, memory strength and forgetfulness, and the implications of microglia depletion in models of brain disease.
Collapse
Affiliation(s)
| | - Laura Ferrucci
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Azka Khan
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Davide Ragozzino
- Laboratory Affiliated to Institute Pasteur Italia – Fondazione Cenci Bolognetti, Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
- *Correspondence: Davide Ragozzino,
| | - Ingrid Reverte
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
- Ingrid Reverte,
| |
Collapse
|
50
|
Dickerson MT, Dadi PK, Zaborska KE, Nakhe AY, Schaub CM, Dobson JR, Wright NM, Lynch JC, Scott CF, Robinson LD, Jacobson DA. G i/o protein-coupled receptor inhibition of beta-cell electrical excitability and insulin secretion depends on Na +/K + ATPase activation. Nat Commun 2022; 13:6461. [PMID: 36309517 PMCID: PMC9617941 DOI: 10.1038/s41467-022-34166-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 10/17/2022] [Indexed: 12/25/2022] Open
Abstract
Gi/o-coupled somatostatin or α2-adrenergic receptor activation stimulated β-cell NKA activity, resulting in islet Ca2+ fluctuations. Furthermore, intra-islet paracrine activation of β-cell Gi/o-GPCRs and NKAs by δ-cell somatostatin secretion slowed Ca2+ oscillations, which decreased insulin secretion. β-cell membrane potential hyperpolarization resulting from Gi/o-GPCR activation was dependent on NKA phosphorylation by Src tyrosine kinases. Whereas, β-cell NKA function was inhibited by cAMP-dependent PKA activity. These data reveal that NKA-mediated β-cell membrane potential hyperpolarization is the primary and conserved mechanism for Gi/o-GPCR control of electrical excitability, Ca2+ handling, and insulin secretion.
Collapse
Affiliation(s)
- Matthew T Dickerson
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA
| | - Prasanna K Dadi
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA
| | - Karolina E Zaborska
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA
| | - Arya Y Nakhe
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA
| | - Charles M Schaub
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA
| | - Jordyn R Dobson
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA
| | - Nicole M Wright
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA
| | - Joshua C Lynch
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA
| | - Claire F Scott
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA
| | - Logan D Robinson
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA
| | - David A Jacobson
- Molecular Physiology and Biophysics Department, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN, USA.
| |
Collapse
|