1
|
Fu K, Yang J. Protocol for whole-tissue immunolabeling, optical clearing, and lightsheet imaging of c-Fos protein expression in unsectioned mouse brains. STAR Protoc 2025; 6:103868. [PMID: 40455684 PMCID: PMC12163136 DOI: 10.1016/j.xpro.2025.103868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/28/2025] [Accepted: 05/15/2025] [Indexed: 06/16/2025] Open
Abstract
The c-Fos protein has been broadly utilized as a marker of neuronal activity, and conventional immunohistochemistry to determine its expression relies on tissue sections. Here, we present a protocol to visualize the endogenous c-Fos protein in intact, unsectioned mouse brains responding to specific stimuli based on the immunolabeling-enabled three-dimensional imaging of solvent-cleared organs (iDISCO) method. We describe steps for tissue harvesting, fixation, decolorization, and permeabilization followed by whole-tissue anti-c-Fos immunolabeling. We then detail procedures for tissue embedding and optical clearing for imaging by lightsheet microscopy. For complete details on the use and execution of this protocol, please refer to Chen et al.1.
Collapse
Affiliation(s)
- Koukou Fu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Center for Life Sciences, IDG/McGovern Institute for Brain Research, Peking University Third Hospital Cancer Center, Peking University, Beijing 100871, China
| | - Jing Yang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Center for Life Sciences, IDG/McGovern Institute for Brain Research, Peking University Third Hospital Cancer Center, Peking University, Beijing 100871, China; Peking Union Medical College Hospital, Beijing 100730, China.
| |
Collapse
|
2
|
Borghi SM, Carvalho TT, Bertozzi MM, Bernardy CCF, Zarpelon AC, Pinho-Ribeiro FA, Calixto-Campos C, Fattori V, Alves-Filho JC, Cunha TM, Cunha FQ, Casagrande R, Verri WA. Role of the interleukin-33 (IL-33)/suppressor of tumorigenicity 2 (ST2) signaling in superoxide anion-triggered inflammation and pain behavior in mice. Chem Biol Interact 2025; 413:111476. [PMID: 40097042 DOI: 10.1016/j.cbi.2025.111476] [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: 03/23/2024] [Revised: 02/26/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Reactive oxygen species such as superoxide anion have varied roles in inflammation and pain, which can be mimicked by potassium superoxide (KO2), the superoxide anion donor. Interleukin (IL)-33 has pleiotropic functions by activating its receptor suppression of tumorigenicity 2 (ST2). However, the role of IL-33/ST2 signaling in inflammatory pain initiated by reactive oxygen species (ROS) such as superoxide anion has not been investigated, which was the aim of the present study. IL-33 levels were assessed by enzyme-linked immunosorbent assay (ELISA). Mechanical and thermal hyperalgesia and overt pain were evaluated by electronic von Frey, hot plate, and abdominal writhing/paw flinching/licking, respectively. Edema and leukocyte recruitment (myeloperoxidase assay and total/differential cell count), antioxidant capacity, superoxide anion production and lipid peroxidation were assessed. Paw skin and spinal cord messenger ribonucleic acid (mRNA) expression of pro-inflammatory mediators and glial markers in the spinal cord were evaluated. Immunofluorescence was used to detect spinal glial and neuronal c-Fos activation. KO2 injection triggered IL-33 production in the paw skin and spinal cord of mice, induced hyperalgesia, edema, neutrophil recruitment to the paw tissue, overt pain-like behavior, and leukocyte recruitment to the peritoneum that were reduced in ST2 deficient mice. In the paw skin and spinal cord, KO2 triggered IL-33/ST2-dependent oxidative stress, and mRNA expression of inflammatory molecules, which were reduced by ST2 deficiency. KO2 induced spinal cord glial (at mRNA/protein levels) and neuronal activation in IL-33/ST2-dependent manner. IL-33/ST2 signaling mediates, at least in part, superoxide anion-induced inflammatory pain by modulating local and spinal inflammatory events.
Collapse
Affiliation(s)
- Sergio M Borghi
- Department of Immunology, Parasitology and General Pathology, Center for Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Thacyana T Carvalho
- Department of Immunology, Parasitology and General Pathology, Center for Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Mariana M Bertozzi
- Department of Immunology, Parasitology and General Pathology, Center for Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Cátia C F Bernardy
- Department of Nursing, Health Sciences Center, University Hospital, State University of Londrina, Londrina, Paraná, Brazil
| | - Ana C Zarpelon
- Department of Immunology, Parasitology and General Pathology, Center for Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Felipe A Pinho-Ribeiro
- Department of Immunology, Parasitology and General Pathology, Center for Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil; Division of Dermatology, Department of Medicine, Washington University School of Medicine in St. Louis, Saint Louis, MO, USA
| | - Cássia Calixto-Campos
- Department of Immunology, Parasitology and General Pathology, Center for Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Victor Fattori
- Department of Immunology, Parasitology and General Pathology, Center for Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - José C Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, Ribeirão Preto, São Paulo, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, Ribeirão Preto, São Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, Ribeirão Preto, São Paulo, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina, 86038-440, PR, Brazil
| | - Waldiceu A Verri
- Department of Immunology, Parasitology and General Pathology, Center for Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil.
| |
Collapse
|
3
|
Nguyen TXD, Chen KT, Liu HL, Kuo CW, Peng CW, Chang MY, Hsieh TH. Temporal interference stimulation over the motor cortex enhances cortical excitability in rats. Sci Rep 2025; 15:16933. [PMID: 40374770 PMCID: PMC12081686 DOI: 10.1038/s41598-025-01008-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 05/02/2025] [Indexed: 05/18/2025] Open
Abstract
Temporal Interference Stimulation (TIS) represents a novel non-invasive brain stimulation technique that deeply targets specific brain regions using the differential beat frequency of two high-frequency stimulation pairs. This study investigated the neuromodulatory effects of TIS at different beat frequencies on cortical excitability in the rat motor cortex. Rats were randomly assigned into four groups, receiving TIS at alpha (10 Hz), beta (20 Hz), gamma (70 Hz), or sham frequencies targeting the motor cortex for 20 min under anesthesia. Cortical excitability and inhibition were evaluated by measuring motor-evoked potentials (MEPs), input-output (I/O) curves, and long-interval intracortical inhibition (LICI) before and after TIS. Additionally, immunohistochemistry was performed for neural biomarkers c-Fos and glutamic acid decarboxylase (GAD-65) to confirm targeted neural activation following TIS. We also examined glial fibrillary acidic protein (GFAP)-positive cells in the stimulated region to assess astrocyte responses associated with TIS. Alpha and gamma TIS significantly increased MEP amplitudes compared to sham stimulation. The analysis of I/O curves revealed a significant enhancement in the area under the curve (AUC) post-stimulation in the alpha and gamma TIS groups. Notably, only gamma TIS significantly reduced intracortical inhibition, indicated by an increased LICI ratio post-stimulation. Immunohistochemical analysis demonstrated a significant 35% increase in c-Fos-positive cells in the stimulated motor cortex regions after TIS compared to sham, whereas no significant changes in GAD-65-positive cells or GFAP expression were observed. These findings indicate that a single session of alpha or gamma TIS effectively modulates cortical excitability, highlighting its potential for targeted neuromodulation applications.
Collapse
Affiliation(s)
- Thi Xuan Dieu Nguyen
- School of Physical Therapy and Graduate Institute of Rehabilitation Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ko-Ting Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital Linkou, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital Linkou, Taoyuan, Taiwan
- School of medicine, Chang Gung university, Taoyuan, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan
| | - Chi-Wei Kuo
- School of Physical Therapy and Graduate Institute of Rehabilitation Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Wei Peng
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Ming-Yuan Chang
- Division of Neurosurgery, Department of Surgery, Min-Sheng General Hospital, Taoyuan, Taiwan
| | - Tsung-Hsun Hsieh
- School of Physical Therapy and Graduate Institute of Rehabilitation Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Neuroscience Research Center, Chang Gung Memorial Hospital Linkou, Taoyuan, Taiwan.
- Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
4
|
Yi MH, Liu Y, Liu YU, Lee J, Hanumaihgari P, Parusel S, Bosco DB, Wang L, Zheng J, Shi W, Eauchai L, Chompoopong S, Hunt CL, Wu LJ. Optogenetic activation of cortical microglia promotes neuronal activity and pain hypersensitivity. Cell Rep 2025; 44:115717. [PMID: 40381194 DOI: 10.1016/j.celrep.2025.115717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 02/10/2025] [Accepted: 04/28/2025] [Indexed: 05/20/2025] Open
Abstract
Chronic pain following peripheral nerve injury is accompanied by increased neuronal activity in the somatosensory cortex. However, whether and how cortical microglia contribute to these changes is less understood. To this end, we applied an optogenetic strategy to specifically target cortical microglia and investigate their function in behavioral pain sensitization. We found that optogenetic activation of microglia in the primary somatosensory cortex (S1) via red-activated channelrhodopsin (ReaChR) triggered pain hypersensitivity and affective-motivational responses in mice. Remarkably, S1-targeted optogenetic stimulation increased microglial landscape changes and ATP release. In addition, optogenetic stimulation altered the microglial proteomic profile, upregulated neuronal c-Fos expression, and enhanced neuronal Ca2+ signaling in the S1. Our results provide mechanistic evidence linking cortical microglia with neuronal hyperactivity and chronic pain behaviors.
Collapse
Affiliation(s)
- Min-Hee Yi
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun, Jeollanam-do 58128, Republic of Korea; Institute for Biomedical Science (IBS) of Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do 58128, Republic of Korea; BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun, Jeollanam-do 58128, Republic of Korea
| | - Yi Liu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA; Department of Anesthesiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yong U Liu
- Department of Anesthesiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jinkyung Lee
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun, Jeollanam-do 58128, Republic of Korea
| | - Priyanka Hanumaihgari
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Neuroscience, Johns Hopkins Medical Institute, Baltimore, MD 21205, USA
| | | | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Wu Shi
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Lattawat Eauchai
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Supin Chompoopong
- Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Christine L Hunt
- Department of Pain Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|
5
|
Simon RC, Fleming WT, Briones BA, Trzeciak M, Senthilkumar P, Ishii KK, Hjort MM, Martin MM, Hashikawa K, Sanders AD, Golden SA, Stuber GD. Opioid-driven disruption of the septum reveals a role for neurotensin-expressing neurons in withdrawal. Neuron 2025:S0896-6273(25)00307-1. [PMID: 40378834 DOI: 10.1016/j.neuron.2025.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 03/14/2025] [Accepted: 04/25/2025] [Indexed: 05/19/2025]
Abstract
Opioid withdrawal is an intensively aversive experience and often drives relapse. The lateral septum (LS) is a forebrain structure that is important in aversion processing and has been linked to substance use disorders, but which LS cell types contribute to the maladaptive state of withdrawal is unknown. We used single-nucleus RNA sequencing to interrogate cell-type-specific gene expression changes induced by chronic morphine exposure and discovered that morphine globally disrupts LS cell types, but neurotensin-expressing neurons (LS-Nts) are selectively activated by naloxone. Using two-photon calcium imaging and ex vivo electrophysiology, we next demonstrate that LS-Nts neurons receive elevated glutamatergic drive in morphine-dependent mice and remain hyperactivated during withdrawal. Finally, we show that manipulating LS-Nts neurons during opioid withdrawal regulates pain coping and sociability. Together, these results suggest that LS-Nts neurons are a key neural substrate involved in opioid withdrawal and establish the LS as a crucial regulator of adaptive behaviors.
Collapse
Affiliation(s)
- Rhiana C Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Weston T Fleming
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Brandy A Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Marta Trzeciak
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Pranav Senthilkumar
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA
| | - Kentaro K Ishii
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Madelyn M Hjort
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Madison M Martin
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Koichi Hashikawa
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Andrea D Sanders
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA
| | - Sam A Golden
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Neurobiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
6
|
Bao ST, Rao F, Yin C, Niu Y, Cao JL, Xiao C, Zhou C. Excitatory projections from the nucleus reuniens to the medial prefrontal cortex modulate pain and depression-like behaviors in mice. PLoS Biol 2025; 23:e3003170. [PMID: 40392890 PMCID: PMC12091829 DOI: 10.1371/journal.pbio.3003170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/17/2025] [Indexed: 05/22/2025] Open
Abstract
The medial prefrontal cortex (mPFC) is implicated in emotional processing, cognition, and pain sensation, moreover, its circuitry undergoes neuroplastic changes in chronic pain. Although the nucleus reuniens (RE) of the thalamus provides significant glutamatergic inputs to the mPFC, it remains unclear whether this projection contributes to plasticity changes in the mPFC and pain-related behaviors in chronic pain. Using fiber photometry, we demonstrated that RE neurons responded to pain stimulation and emotional changes. Optogenetic activation of RE neurons and their projections to the mPFC (RE-mPFC projection) elicits hyperalgesia and depression-like behaviors in naïve mice. In a neuropathic pain mouse model, RE neurons were hyperactive, and the RE-mPFC projection was enhanced with a marked preference for the part innervating GABAergic circuits in the mPFC to that controlling mPFC neurons projecting to the ventrolateral periaqueductal gray (vlPAG). Expectedly, optogenetic inhibition of RE neurons and the RE-mPFC projection ameliorated pain-like and depression-like behaviors in neuropathic pain mice. Additionally, chemogenetic inhibition of RE-mPFC neurons conferred analgesia in neuropathic pain mice exposed to both acute and chronic morphine. Our findings highlight the significant role of the RE-mPFC pathway in neuropathic pain comorbid with depression, suggesting its potential as a target for treatment of neuropathic pain.
Collapse
Affiliation(s)
- Shu-Ting Bao
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Fang Rao
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Cui Yin
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Yong Niu
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Department of Anesthesia, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Cheng Xiao
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Chunyi Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
7
|
Wu KY, Tsao CH, Su NC, Deng SM, Huang GJ. Stk24 deficiency causes disrupted hippocampal neurogenesis and anxiety-like behavior in mice. Commun Biol 2025; 8:663. [PMID: 40281197 PMCID: PMC12032016 DOI: 10.1038/s42003-025-08035-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Protein kinases regulate protein activity through phosphorylation, and many have been reported to participate in brain development. Among them, serine/threonine-protein kinase 24 (STK24) is believed to influence apoptosis, spinal synaptogenesis, and neuronal migration. Despite its recognized roles, the functions of STK24 in the brain remains insufficiently explored. Here, we present an in vivo study of brain-specific Stk24 conditional knockout mice. We investigate the impact of Stk24 deletion through histological analysis, behavior assays, and the molecular changes. In our results, Stk24 deletion disrupts the hippocampal formation during development and decreased subsequent adult hippocampal neurogenesis whilst neuronal morphology is relatively unaffected. Additionally, Stk24-deficient mice exhibit anxiety-like behavior and altered stress responses, featuring increased hippocampal neuronal activity, dysregulated HPA axis reactivity, and modified expression patterns of glucocorticoid receptor signaling-related genes. In conclusion, our findings highlight the involvement of Stk24 in brain development, adult hippocampal neurogenesis, as well as anxiety and stress responses.
Collapse
Affiliation(s)
- Kuan-Yu Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Chi-Hui Tsao
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Nicole Ching Su
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Shin-Meng Deng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Guo-Jen Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan.
| |
Collapse
|
8
|
Li X, Gao J, Liu X, Guo J, Liu Y, Cui P, Yan D, Fei T, Chen M, Gao Y. Modulation of neural activity and gene expression by arecoline. Front Integr Neurosci 2025; 19:1545260. [PMID: 40271198 PMCID: PMC12014716 DOI: 10.3389/fnint.2025.1545260] [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: 12/14/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Arecoline, a biologically active alkaloid extracted from the areca nut, serves as the primary psychoactive ingredient in betel quid, one of the most widely consumed psychoactive substances worldwide. Despite its extensive use, the central nervous system (CNS) effects of arecoline remain inadequately understood. This study aims to investigate the central actions of arecoline through a comprehensive, multi-dimensional approach that integrates behavioral assays, neuroimaging techniques, calcium signaling analysis, and transcriptomic profiling. Our findings demonstrate dose-dependent addictive properties of arecoline, alongside distinct behavioral alterations that highlight its potential for addiction. Neuroimaging and calcium signaling data revealed region-specific alterations in neural activity, particularly in areas associated with learning, memory, and reward processing. Furthermore, transcriptomic analysis identified significant changes in gene expression, particularly in pathways related to synaptic plasticity, calcium signaling, and metal ion transport. These results provide valuable insights into the addictive potential of arecoline and its underlying neurobiological mechanisms, offering crucial information for understanding its broader impact on CNS function. The study's findings hold significant implications for informing public health strategies aimed at addressing arecoline misuse and its potential role in addiction-related disorders.
Collapse
Affiliation(s)
- Xiaonan Li
- Shanghai New Tobacco Products Research Institute Co., Ltd., Shanghai, China
| | - Jie Gao
- Shanghai New Tobacco Products Research Institute Co., Ltd., Shanghai, China
| | - Xiaomin Liu
- Shanghai New Tobacco Products Research Institute Co., Ltd., Shanghai, China
| | - Jianfeng Guo
- Shanghai New Tobacco Products Research Institute Co., Ltd., Shanghai, China
| | - Yifan Liu
- Shanghai New Tobacco Products Research Institute Co., Ltd., Shanghai, China
| | - Peicai Cui
- Shanghai New Tobacco Products Research Institute Co., Ltd., Shanghai, China
| | - Dawei Yan
- Shanghai New Tobacco Products Research Institute Co., Ltd., Shanghai, China
| | - Ting Fei
- Shanghai New Tobacco Products Research Institute Co., Ltd., Shanghai, China
| | | | - Yihan Gao
- Shanghai New Tobacco Products Research Institute Co., Ltd., Shanghai, China
| |
Collapse
|
9
|
Tucker A, Baltazar A, Eisdorfer JT, Thackray JK, Vo K, Thomas H, Tandon A, Moses J, Singletary B, Gillespie T, Smith A, Pauken A, Nadella S, Pitonak M, Letchuman S, Jang J, Totty M, Jalufka FL, Aceves M, Adler AF, Maren S, Blackmon H, McCreedy DA, Abraira V, Dulin JN. Functional synaptic connectivity of engrafted spinal cord neurons with locomotor circuitry in the injured spinal cord. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.05.644402. [PMID: 40236108 PMCID: PMC11996546 DOI: 10.1101/2025.04.05.644402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Spinal cord injury (SCI) results in significant neurological deficits, with no currently available curative therapies. Neural progenitor cell (NPC) transplantation has emerged as a promising approach for neural repair, as graft-derived neurons (GDNs) can integrate into the host spinal cord and support axon regeneration. However, the mechanisms underlying functional recovery remain poorly understood. In this study, we investigate the synaptic integration of NPC-derived neurons into locomotor circuits, the projection patterns of distinct neuronal subtypes, and their potential to modulate motor circuit activity. Using transsynaptic tracing in a mouse thoracic contusion SCI model, we found that NPC-derived neurons form synaptic connections with host locomotor circuits, albeit at low frequencies. Furthermore, we mapped the axon projections of V0C and V2a interneurons, revealing distinct termination patterns within host spinal cord laminae. To assess functional integration, we employed chemogenetic activation of GDNs, which induced muscle activity in a subset of transplanted animals. However, NPC transplantation alone did not significantly improve locomotor recovery, highlighting a key challenge in the field. Our findings suggest that while GDNs can integrate into host circuits and modulate motor activity, synaptic connectivity remains a limiting factor in functional recovery. Future studies should focus on enhancing graft-host connectivity and optimizing transplantation strategies to maximize therapeutic benefits for SCI.
Collapse
|
10
|
Araj SK, Brzezik J, Mądra-Gackowska K, Szeleszczuk Ł. Overview of Epitalon-Highly Bioactive Pineal Tetrapeptide with Promising Properties. Int J Mol Sci 2025; 26:2691. [PMID: 40141333 PMCID: PMC11943447 DOI: 10.3390/ijms26062691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/15/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
Epitalon, also known as Epithalon or Epithalone, is a tetrapeptide, Ala-Glu-Asp-Gly (AEDG), which was synthesized based on the amino acids composition of Epithalamin, a bovine pineal gland extract, prior to its discovery in pineal gland polypeptide complex solution. During the last 25 years, this compound has been extensively studied using in vitro, in vivo, and in silico methods. The results of these studies indicate significant geroprotective and neuroendocrine effects of Epitalone, resulting from its antioxidant, neuro-protective, and antimutagenic effects, originating from both specific and nonspecific mechanisms. Although it has been demonstrated that Epitalon exerts, among other effects, a direct influence on melatonin synthesis, alters the mRNA levels of interleukin-2, modulates the mitogenic activity of murine thymocytes, and enhances the activity of various enzymes, including AChE, BuChE, and telomerase, it remains uncertain whether these are the sole mechanisms of action of this compound. Moreover, despite the considerable volume of research on the biological and pharmacodynamic characteristics of Epitalon, the quantity of physico-chemical and structural investigations of this peptide remains quite limited. This review aims to conclude the most important findings from such studies, thus presenting the current state of knowledge on Epitalon.
Collapse
Affiliation(s)
- Szymon Kamil Araj
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-093 Warsaw, Poland; (S.K.A.); (J.B.)
| | - Jakub Brzezik
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-093 Warsaw, Poland; (S.K.A.); (J.B.)
| | - Katarzyna Mądra-Gackowska
- Department of Geriatrics, Faculty of Health Sciences, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Skłodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland;
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-093 Warsaw, Poland; (S.K.A.); (J.B.)
| |
Collapse
|
11
|
YAVAS E, FANSELOW MS. Defensive behaviors and c-fos expression in the midbrain. Integr Zool 2025; 20:394-406. [PMID: 39218997 PMCID: PMC11871047 DOI: 10.1111/1749-4877.12892] [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] [Indexed: 09/04/2024]
Abstract
Pavlovian fear conditioning serves as a valuable method for investigating species-specific defensive reactions (SSDRs) such as freezing and flight responses. The present study examines the role of white noise under different experimental conditions. Given that white noise has been shown to elicit both conditional (associative) and unconditional (nonassociative) defensive responses, we compared the response to noise following three separate training conditions: shock-only, white noise paired with shock, and context-only. Results showed that baseline freezing level significantly changed across groups: Both the shock-only group and the white noise paired with shock group froze more than the context-only group on the test day. White noise evoked differential freezing between groups on day 2: The shock-only group froze more than the context-only group although both groups were never exposed to white noise during training. Further, an activity burst triggered by white noise was similar for the shock-only and white noise paired with shock groups during testing, although shock-only group was never exposed to white noise stimuli during training. This aligned with c-fos data, indicating similar c-fos activity levels across different periaqueductal gray (PAG) regions for both shock-only and white noise paired with shock groups. However, the driving force behind c-fos activation-whether freezing, activity burst, or a combination of both-remains uncertain, warranting further analysis to explore specific correlations between SSDRs and c-fos activity within the PAG and related brain areas.
Collapse
Affiliation(s)
- Ersin YAVAS
- Department of PsychologyBartın UniversityBartınTurkey
| | - Michael S. FANSELOW
- Staglin Center for Brain and Behavioral Health, Department of Psychology, UCLALos AngelesCaliforniaUSA
- Department of Psychiatry and Biobehavioral SciencesUCLALos AngelesCaliforniaUSA
| |
Collapse
|
12
|
Okuma R, Kobayashi S, Kobayashi S, Arai Y, Matsumoto N, Motoyoshi M, Kobayashi M, Fujita S. The cortical areas processing periodontal ligament nociception in mice. J Oral Biosci 2025; 67:100597. [PMID: 39667668 DOI: 10.1016/j.job.2024.100597] [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: 10/03/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
OBJECTIVES Toothaches are often poorly localized. Although periodontal pain is better localized, it can spread to other areas. Ultimately, the cerebral cortex processes nociception, with somatotopic organization possibly playing a role in localizing the origin. However, the exact cortical area in the periodontal ligament (PDL) remains unclear. METHODS This study examined cortical responses to electrical stimulation of the molar PDL in anesthetized male mice using in vivo optical imaging with a voltage-sensitive dye, autofluorescent flavin fluorescence, and immunohistochemistry for c-Fos protein expression. RESULTS On optical imaging, cortical responses to the stimulation of the ipsilateral and contralateral PDL of the upper and lower teeth were observed in the primary somatosensory cortex (S1) and area from the insular cortex (IC) to the ventral edge of the secondary somatosensory cortex (S2), defined as the area caudal to the middle cerebral artery (C-area). Responses in S1 were faint and unstable, but were consistent in the C-area. The initial response locations were similar regardless of which PDL was stimulated, and the activated areas in the C-area almost overlapped. Three-dimensional construction of c-Fos-immunopositive cells responding to upper or lower PDL stimulation revealed bilateral distribution in the cingulate gyrus, secondary auditory cortex, temporal association cortex, ectorhinal cortex, and IC, but not in the S1 and S2. CONCLUSION These results suggest that the somatotopic organization of the S1, S2, and IC cannot explain the localization of PDL nociception. The predominance of responses in the contralateral IC may provide clues for identifying the laterality.
Collapse
Affiliation(s)
- Risako Okuma
- Department of Orthodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Department of Biology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Department of Oral Structural and Functional Biology, Nihon University Graduate School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Shutaro Kobayashi
- Department of Biology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Oral and Maxillofacial Surgery, Kameda General Hospital, 929 Higashi-cho, Kamogawa City, Chiba 296-8602, Japan
| | - Satomi Kobayashi
- Department of Biology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Yoshinori Arai
- Department of Oral and Maxillofacial Radiology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Division of Advanced Dental Treatment, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Naoyuki Matsumoto
- Department of Pathology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama City, Kanagawa 230-8501, Japan
| | - Mitsuru Motoyoshi
- Department of Orthodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Division of Clinical Research, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Masayuki Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Satoshi Fujita
- Department of Biology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan.
| |
Collapse
|
13
|
Fang LP, Lin CH, Medlej Y, Zhao R, Chang HF, Guo Q, Wu Z, Su Y, Zhao N, Gobbo D, Wyatt A, Wahl V, Fiore F, Tu SM, Boehm U, Huang W, Bian S, Agarwal A, Lauterbach MA, Yi C, Niu J, Scheller A, Kirchhoff F, Bai X. Oligodendrocyte precursor cells facilitate neuronal lysosome release. Nat Commun 2025; 16:1175. [PMID: 39885146 PMCID: PMC11782495 DOI: 10.1038/s41467-025-56484-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/17/2025] [Indexed: 02/01/2025] Open
Abstract
Oligodendrocyte precursor cells (OPCs) shape brain function through many non-canonical regulatory mechanisms beyond myelination. Here we show that OPCs form contacts with their processes on neuronal somata in a neuronal activity-dependent manner. These contacts facilitate exocytosis of neuronal lysosomes. A reduction in the number or branching of OPCs reduces these contacts, which is associated with lysosome accumulation and altered metabolism in neurons and more senescent neurons with age. A similar reduction in OPC branching and neuronal lysosome accumulation is seen in an early-stage mouse model of Alzheimer's disease. Our findings have implications for the prevention of age-related pathologies and the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Li-Pao Fang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 211198, Nanjing, China
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
| | - Ching-Hsin Lin
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Cellular Neurophysiology, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Yasser Medlej
- Molecular Imaging, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Renping Zhao
- Biophysics, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Hsin-Fang Chang
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Cellular Neurophysiology, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Qilin Guo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
| | - Zhonghao Wu
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, 518107, Shenzhen, China
| | - Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, 518107, Shenzhen, China
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, 400038, Chongqing, China
| | - Na Zhao
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- Institute of Anatomy and Cell Biology, University of Saarland, 66421, Homburg, Germany
| | - Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
| | - Amanda Wyatt
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Vanessa Wahl
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Frederic Fiore
- 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
| | - Szu-Min Tu
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Cellular Neurophysiology, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Ulrich Boehm
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Wenhui Huang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
| | - Shan Bian
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - 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
| | | | - Chenju Yi
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, 400038, Chongqing, China
| | - Jianqin Niu
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, 518107, Shenzhen, China
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany.
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany.
| | - Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany.
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 211198, Nanjing, China.
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany.
| |
Collapse
|
14
|
Gutierrez-Castellanos N, Husain BFA, Dias IC, Nomoto K, Duarte MA, Ferreira L, Lacoste B, Lima SQ. A hypothalamic node for the cyclical control of female sexual rejection. Neuron 2025; 113:277-290.e8. [PMID: 39591968 DOI: 10.1016/j.neuron.2024.10.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 08/05/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024]
Abstract
Internal state-dependent behavioral flexibility, such as the ability to switch between rejecting and accepting sexual advances based on a female's reproductive capacity, is crucial for maintaining meaningful social interactions. While the role of the ventrolateral ventromedial hypothalamus (VMHvl) in sexual acceptance is well established, the neural mechanisms underlying sexual rejection remain unexplored. In this study, we identify progesterone receptor-expressing neurons in the anterior VMHvl (aVMHvlPR+) as key regulators of cyclical female sexual rejection behavior. In vivo recordings reveal that these neurons are active during sexual rejection but inactive during sexual acceptance. Slice electrophysiology demonstrates that aVMHvlPR+ neurons receive a reduced excitatory-to-inhibitory synaptic input balance in receptive females. Furthermore, activating and inhibiting aVMHvlPR+ neurons increases rejection in receptive females and reduces rejection in non-receptive females, respectively. Thus, aVMHvlPR+ neurons constitute a critical neural substrate controlling female sexual behavior, providing an additional barrier to mating when fertilization is not possible.
Collapse
Affiliation(s)
| | | | - Inês C Dias
- Neuroethology Laboratory, Champalimaud Research, Champalimaud Foundation, Lisboa 1400-038, Portugal
| | - Kensaku Nomoto
- Neuroethology Laboratory, Champalimaud Research, Champalimaud Foundation, Lisboa 1400-038, Portugal
| | - Margarida A Duarte
- Neuroethology Laboratory, Champalimaud Research, Champalimaud Foundation, Lisboa 1400-038, Portugal
| | - Liliana Ferreira
- Neuroethology Laboratory, Champalimaud Research, Champalimaud Foundation, Lisboa 1400-038, Portugal
| | - Bertrand Lacoste
- Neuroethology Laboratory, Champalimaud Research, Champalimaud Foundation, Lisboa 1400-038, Portugal
| | - Susana Q Lima
- Neuroethology Laboratory, Champalimaud Research, Champalimaud Foundation, Lisboa 1400-038, Portugal.
| |
Collapse
|
15
|
Konat GW. Neuroplasticity elicited by peripheral immune challenge with a viral mimetic. Brain Res 2025; 1846:149239. [PMID: 39284559 DOI: 10.1016/j.brainres.2024.149239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Peripheral viral infections are well known to profoundly alter brain function; however detailed mechanisms of this immune-to-brain communication have not been deciphered. This review focuses on studies of cerebral effects of peripheral viral challenge employing intraperitoneal injection of a viral mimetic, polyinosinic-polycytidylic acid (PIC). In this paradigm, PIC challenge induces the acute phase response (APR) characterized by a transient surge of circulating inflammatory factors, primarily IFNβ, IL-6 and CXCL10. The blood-borne factors, in turn, elicit the generation of CXCL10 by hippocampal neurons. Neurons also express the cognate receptor of CXCL10, i.e., CXCR3 implicating the existence of autocrine/paracrine signaling. The CXCL10/CXCR3 axis mediates the ensuing neuroplastic changes manifested as neuronal hyperexcitability, seizure hypersusceptibility, and sickness behavior. Electrophysiological studies revealed that the neuroplastic changes entail the potentiation of excitatory synapses likely at both pre- and postsynaptic loci. Excitatory synaptic transmission is further augmented by PIC challenge-induced elevation of extracellular glutamate that is mediated by astrocytes. In addition, the hyperexcitability of neuronal circuits might involve the repression of inhibitory signaling. Accordingly, CXCL10 released by neurons activates microglia whose processes invade perisomatic inhibitory synapses, resulting in a partial detachment of the presynaptic terminals, and thus, de-inhibition. This process might be facilitated by the cerebral complement system, which is also upregulated and activated by PIC challenge. Moreover, CXCL10 stimulates the expression of neuronal c-fos protein, another index of hyperexcitability. The reviewed studies form a foundation for full elucidation of the fascinating intersection between peripheral viral infections and neuroplasticity. Because the activation of such pathways may constitute a serious comorbidity factor for neuropathological conditions, this research would advance the development of preventive strategies.
Collapse
Affiliation(s)
- Gregory W Konat
- Department of Biochemistry and Molecular Medicine, Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
| |
Collapse
|
16
|
Raun N, Jones SG, Kerr O, Keung C, Butler EF, Alka K, Krupski JD, Reid-Taylor RA, Ibrahim V, Williams M, Top D, Kramer JM. Trithorax regulates long-term memory in Drosophila through epigenetic maintenance of mushroom body metabolic state and translation capacity. PLoS Biol 2025; 23:e3003004. [PMID: 39869640 PMCID: PMC11835295 DOI: 10.1371/journal.pbio.3003004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/18/2025] [Accepted: 01/06/2025] [Indexed: 01/29/2025] Open
Abstract
The role of epigenetics and chromatin in the maintenance of postmitotic neuronal cell identities is not well understood. Here, we show that the histone methyltransferase Trithorax (Trx) is required in postmitotic memory neurons of the Drosophila mushroom body (MB) to enable their capacity for long-term memory (LTM), but not short-term memory (STM). Using MB-specific RNA-, ChIP-, and ATAC-sequencing, we find that Trx maintains homeostatic expression of several non-canonical MB-enriched transcripts, including the orphan nuclear receptor Hr51, and the metabolic enzyme lactate dehydrogenase (Ldh). Through these key targets, Trx facilitates a metabolic state characterized by high lactate levels in MBγ neurons. This metabolic state supports a high capacity for protein translation, a process that is essential for LTM, but not STM. These data suggest that Trx, a classic regulator of cell type specification during development, has additional functions in maintaining underappreciated aspects of postmitotic neuron identity, such as metabolic state. Our work supports a body of evidence suggesting that a high capacity for energy metabolism is an essential cell identity characteristic for neurons that mediate LTM.
Collapse
Affiliation(s)
- Nicholas Raun
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Spencer G. Jones
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Olivia Kerr
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Crystal Keung
- Department of Physiology and Pharmacology, University of Western Ontario, London, Canada
| | - Emily F. Butler
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Kumari Alka
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Jonathan D. Krupski
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Robert A. Reid-Taylor
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Veyan Ibrahim
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - MacKayla Williams
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Deniz Top
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
- Department of Cell Biology, University of Alberta, Edmonton, Canada
| | - Jamie M. Kramer
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Canada
| |
Collapse
|
17
|
Zhou Y, Wang JL, Qiu L, Torpey J, Wixson JG, Lyon M, Chen X. NMDA Receptors Control Activity Hierarchy in Neural Network: Loss of Control in Hierarchy Leads to Learning Impairments, Dissociation, and Psychosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.06.523038. [PMID: 36712055 PMCID: PMC9881912 DOI: 10.1101/2023.01.06.523038] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
While it is known that associative memory is preferentially encoded by memory-eligible "primed" neurons, in vivo neural activity hierarchy has not been quantified and little is known about how such a hierarchy is established. Leveraging in vivo calcium imaging of hippocampal neurons on freely behaving mice, we developed the first method to quantify real-time neural activity hierarchy in the CA1 region. Neurons at the top of activity hierarchy are identified as primed neurons. In cilia knockout mice that exhibit severe learning deficits, the percentage of primed neurons is drastically reduced. We developed a simplified neural network model that incorporates simulations of linear and non-linear weighted components, modeling the synaptic ionic conductance of AMPA and NMDA receptors, respectively. We found that moderate non-linear to linear conductance ratios naturally leads a small fraction of neurons to be primed in the simulated neural network. Removal of the non-linear component eliminates the existing activity hierarchy and reinstate it to the network stochastically primes a new pool of neurons. Blockade of NMDA receptors by ketamine not only decreases general neuronal activity causing learning impairments, but also disrupts neural activity hierarchy. Additionally, ketamine-induced super-synchronized slow oscillation during anesthesia can be simulated if the non-linear NMDAR component is removed to flatten activity hierarchy. Together, this study develops a unique method to measure neural activity hierarchy and identifies NMDA receptors as a key factor that controls the hierarchy. It presents the first evidence suggesting that hierarchy disruption by NMDAR blockade causes dissociation and psychosis.
Collapse
|
18
|
Stanisavljević Ilić A, Filipović D. Mapping of c-Fos Expression in Rat Brain Sub/Regions Following Chronic Social Isolation: Effective Treatments of Olanzapine, Clozapine or Fluoxetine. Pharmaceuticals (Basel) 2024; 17:1527. [PMID: 39598437 PMCID: PMC11597560 DOI: 10.3390/ph17111527] [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: 10/15/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
The c-Fos as a marker of cell activation is used to identify brain regions involved in stimuli processing. This review summarizes a pattern of c-Fos immunoreactivity and the overlapping brain sub/regions which may provide hints for the identification of neural circuits that underlie depressive- and anxiety-like behaviors of adult male rats following three and six weeks of chronic social isolation (CSIS), relative to controls, as well as the antipsychotic-like effects of olanzapine (Olz), and clozapine (Clz), and the antidepressant-like effect of fluoxetine (Flx) in CSIS relative to CSIS alone. Additionally, drug-treated controls relative to control rats were also characterized. The overlapping rat brain sub/regions with increased expression of c-Fos immunoreactivity following three or six weeks of CSIS were the retrosplenial granular cortex, c subregion, retrosplenial dysgranular cortex, dorsal dentate gyrus, paraventricular nucleus of the thalamus (posterior part, PVP), lateral/basolateral (LA/BL) complex of the amygdala, caudate putamen, and nucleus accumbens shell. Increased activity of the nucleus accumbens core following exposure of CSIS rats either to Olz, Clz, and Flx treatments was found, whereas these treatments in controls activated the LA/BL complex of the amygdala and PVP. We also outline sub/regions that might represent potential neuroanatomical targets for the aforementioned antipsychotics or antidepressant treatments.
Collapse
Affiliation(s)
| | - Dragana Filipović
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
19
|
Mitchell JR, Vincelette L, Tuberman S, Sheppard V, Bergeron E, Calitri R, Clark R, Cody C, Kannan A, Keith J, Parakoyi A, Pikus M, Vance V, Ziane L, Brenhouse H, Laine MA, Shansky RM. Behavioral and neural correlates of diverse conditioned fear responses in male and female rats. Neurobiol Stress 2024; 33:100675. [PMID: 39391589 PMCID: PMC11465128 DOI: 10.1016/j.ynstr.2024.100675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Pavlovian fear conditioning is a widely used tool that models associative learning in rodents. For decades the field has used predominantly male rodents and focused on a sole conditioned fear response: freezing. However, recent work from our lab and others has identified darting as a female-biased conditioned response, characterized by an escape-like movement across a fear conditioning chamber. It is also accompanied by a behavioral phenotype: Darters reliably show decreased freezing compared to Non-darters and males and reach higher velocities in response to the foot shock ("shock response"). However, the relationship between shock response and conditioned darting is not known. This study investigated if this link is due to differences in general processing of aversive stimuli between Darters, Non-darters and males. Across a variety of modalities, including corticosterone measures, the acoustic startle test, and sensitivity to thermal pain, Darters were found not to be more reactive or sensitive to aversive stimuli, and, in some cases, they appear less reactive to Non-darters and males. Analyses of cFos activity in regions involved in pain and fear processing following fear conditioning identified discrete patterns of expression among Darters, Non-darters, and males exposed to low and high intensity foot shocks. The results from these studies further our understanding of the differences between Darters, Non-darters and males and highlight the importance of studying individual differences in fear conditioning as indicators of fear state.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rose Clark
- Northeastern University, Boston, MA, USA
| | | | | | - Jack Keith
- Northeastern University, Boston, MA, USA
| | | | | | | | | | | | - Mikaela A. Laine
- Northeastern University, Boston, MA, USA
- Smith College, Northampton, MA, USA
| | | |
Collapse
|
20
|
Kim P, Garner N, Tatkovic A, Parsons R, Chunduri P, Vukovic J, Piper M, Pfeffer M, Weiergräber M, Oster H, Rawashdeh O. Melatonin's role in the timing of sleep onset is conserved in nocturnal mice. NPJ BIOLOGICAL TIMING AND SLEEP 2024; 1:13. [PMID: 39493889 PMCID: PMC11530376 DOI: 10.1038/s44323-024-00013-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024]
Abstract
Melatonin supplementation strengthens non-restorative sleep rhythms and its temporal alignment in both humans and night-active rodents. Of note, although the sleep cycle is reversed in day-active and night-active (nocturnal) mammals, both, produce melatonin at night under the control of the circadian clock. The effects of exogenous melatonin on sleep and sleepiness are relatively clear, but its endogenous role in sleep, particularly, in timing sleep onset (SO), remains poorly understood. We show in nocturnal mice that the increases in mid-nighttime sleep episodes, and the mid-nighttime decline in activity, are coupled to nighttime melatonin signaling. Furthermore, we show that endogenous melatonin modulates SO by reducing the threshold for wake-to-sleep transitioning. Such link between melatonin and SO timing may explain phenomena such as increased sleep propensity in circadian rhythm sleep disorders and chronic insomnia in patients with severely reduced nocturnal melatonin levels. Our findings demonstrate that melatonin's role in sleep is evolutionarily conserved, effectively challenging the argument that melatonin cannot play a major role in sleep regulation in nocturnal mammals, where the main activity phase coincides with high melatonin levels.
Collapse
Affiliation(s)
- Pureum Kim
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| | - Nicholas Garner
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| | - Annaleis Tatkovic
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| | - Rex Parsons
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
- Present Address: Australian Centres for Health Services Innovation and Healthcare Transformation, School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Kelvin Grove, QLD Australia
| | - Prasad Chunduri
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| | - Jana Vukovic
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
- Queensland Brain Institute, University of Queensland, Brisbane, QLD Australia
| | - Michael Piper
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
- Queensland Brain Institute, University of Queensland, Brisbane, QLD Australia
| | - Martina Pfeffer
- Institute of Anatomy 2, Faculty of Medicine, Heinrich Heine University, Düsseldorf, Germany
| | - Marco Weiergräber
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior & Metabolism, University of Lübeck, Lübeck, Germany
| | - Oliver Rawashdeh
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| |
Collapse
|
21
|
Aguilar-Delgadillo A, Cruz-Mendoza F, Luquin-de Andais teh S, Ruvalcaba-Delgadillo Y, Jáuregui-Huerta F. Stress-induced c-fos expression in the medial prefrontal cortex differentially affects the main residing cell phenotypes. Heliyon 2024; 10:e39325. [PMID: 39498004 PMCID: PMC11532284 DOI: 10.1016/j.heliyon.2024.e39325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 11/07/2024] Open
Abstract
Stress poses a challenge to the body's equilibrium and triggers a series of responses that enable organisms to adapt to stressful stimuli. The medial prefrontal cortex (mPFC), particularly in acute stress conditions, undergoes significant physiological changes to cope with the demands associated with cellular activation. The proto-oncogene c-fos and its protein product c-Fos have long been utilized to investigate the effects of external factors on the central nervous system (CNS). While c-Fos expression has traditionally been attributed to neurons, emerging evidence suggests its potential expression in glial cells. In this study, our main objective was to explore the expression of c-Fos in glial cells and examine how acute stress influences these activity patterns. We conducted our experiments on male Wistar rats, subjecting them to acute stress and sacrificing them 2 h after the stressor initiation. Using double-labelling fluorescent immunohistochemistry targeting c-Fos, along with markers such as GFAP, Iba-1, Olig2, NG2, and NeuN, we analyzed 35 μm brain slices obtained from the mPFC. Our findings compellingly demonstrate that c-Fos expression extends beyond neurons and is present in astrocytes, oligodendrocytes, microglia, and NG2 cells-the diverse population of glial cells. Moreover, we observed distinct regulation of c-Fos expression in response to stress across different subregions of the mPFC. These results emphasize the importance of considering glial cells and their perspective in studies investigating brain activity, highlighting c-Fos as a response marker in glial cells. By shedding light on the differential regulation of c-Fos expression in response to stress, our study contributes to the understanding of glial cell involvement in stress-related processes. This underscores the significance of including glial cells in investigations of brain activity and expands our knowledge of c-Fos as a potential marker for glial cell responses.
Collapse
Affiliation(s)
| | - Fernando Cruz-Mendoza
- Neurosciences Department, Health sciences center, University of Guadalajara, Guadalajara, Mexico
| | | | | | - Fernando Jáuregui-Huerta
- Neurosciences Department, Health sciences center, University of Guadalajara, Guadalajara, Mexico
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
22
|
Hill A, Amendolara AB, Small C, Guzman SC, Pfister D, McFarland K, Settelmayer M, Baker S, Donnelly S, Payne A, Sant D, Kriak J, Bills KB. Metabolic Pathophysiology of Cortical Spreading Depression: A Review. Brain Sci 2024; 14:1026. [PMID: 39452037 PMCID: PMC11505892 DOI: 10.3390/brainsci14101026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
Cortical spreading depression (CSD) is an electrophysiologic pathological state in which a wave of depolarization in the cerebral cortex is followed by the suppression of spontaneous neuronal activity. This transient spread of neuronal depolarization on the surface of the cortex is the hallmark of CSD. Numerous investigations have demonstrated that transmembrane ion transport, astrocytic ion clearing and fatigue, glucose metabolism, the presence of certain genetic markers, point mutations, and the expression of the enzyme responsible for the production of various arachidonic acid derivatives that participate in the inflammatory response, namely, cyclooxygenase (COX), all influence CSD. Here, we explore the associations between CSD occurrence in the cortex and various factors, including how CSD is related to migraines, how the glucose state affects CSD, the effect of TBI and its relationship with CSD and glucose metabolism, how different markers can be measured to determine the severity of CSD, and possible connections to oligemia, orexin, and leptin.
Collapse
|
23
|
Bi JR, Zha HW, Gao QL, Wu H, Liu ZJ, Sun D. Pleasant Odor Decreases Mouse Anxiety-like Behaviors by Regulating Hippocampal Endocannabinoid Signaling. Int J Mol Sci 2024; 25:10699. [PMID: 39409026 PMCID: PMC11477034 DOI: 10.3390/ijms251910699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Anxiety disorder is one of the most common neuropsychiatric disorders, and affects many people's daily activities. Although the pathogenesis and treatments of anxiety disorder have been studied for several decades, the underlying mechanisms remain elusive. Here, we provide evidence that olfactory stimuli with inhaled linalool or 2-phenylethanol decreased mouse anxiety-like behaviors and increased the activities of hippocampal dentate granule cells (DGCs). RNA-sequencing analysis identified retrograde endocannabinoid signaling, which is a critical pathway for mood regulation and neuron activation, is altered in the hippocampus of both linalool- and 2-phenylethanol-exposed mice. Further studies found that selective inhibition of endocannabinoid signaling by injecting rimonabant abolished the activation of DGCs and the anxiolytic effect induced by linalool or 2-phenylethanol. Together, these results uncovered a novel mechanism by which linalool or 2-phenylethanol decreases mouse anxiety-like behaviors and increases DG activity likely through activating hippocampal retrograde endocannabinoid signaling.
Collapse
Affiliation(s)
- Jia-Rui Bi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (J.-R.B.)
| | - Hai-Wei Zha
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (J.-R.B.)
| | - Qing-Lin Gao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (J.-R.B.)
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (J.-R.B.)
| | - Zhen-Jiang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (J.-R.B.)
| | - Dong Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (J.-R.B.)
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
24
|
Brandt HB, Sinning S, Hasselstrøm JB, Andersen CU. A review of possible biomarkers for opioid tolerance. Forensic Sci Int 2024; 363:112187. [PMID: 39154523 DOI: 10.1016/j.forsciint.2024.112187] [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: 03/01/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/20/2024]
Abstract
Knowledge of opioid tolerance in a deceased person is important for distinguishing between therapeutic and toxic opioid concentrations for that particular individual when interpreting postmortem toxicological results. However, no biomarkers for opioid tolerance are currently available. This review aimed to study the existing literature on mechanisms or changes in signaling pathways related to chronic opioid use, which could be relevant for further studies to identify biomarkers for opioid tolerance. We performed a systematic literature search using the PRISMA 2020 guidelines using the MeSH terms "opioid tolerance AND biomarkers" in PubMed, Embase, WebofScience, and the Cochrane library. A review of the search results yielded seven studies on animal models or humans, identifying and evaluating thirteen possible biomarkers in terms of specificity for changes induced by opioids and other aspects to be considered as potential biomarkers. We evaluated nine potential biomarkers as unlikely to be specific for opioid tolerance, and one had contradictory results in terms of upregulation or downregulation. However, methylation of the promoter region of the μ-opioid receptor gene, increased activity of soluble puromycin-sensitive aminopeptidase, altered miRNA profile, or other multiple component profiling may be interesting to study further as biomarkers for opioid tolerance in forensic postmortem cases.
Collapse
Affiliation(s)
| | | | | | - Charlotte Uggerhøj Andersen
- Department of Forensic Medicine, Aarhus University, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Denmark.
| |
Collapse
|
25
|
Robertson RV, Meylakh N, Crawford LS, Tinoco Mendoza FA, Macey PM, Macefield VG, Keay KA, Henderson LA. Differential activation of lateral parabrachial nuclei and their limbic projections during head compared with body pain: A 7-Tesla functional magnetic resonance imaging study. Neuroimage 2024; 299:120832. [PMID: 39236852 DOI: 10.1016/j.neuroimage.2024.120832] [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: 06/28/2024] [Revised: 08/16/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
Pain is a complex experience that involves sensory, emotional, and motivational components. It has been suggested that pain arising from the head and orofacial regions evokes stronger emotional responses than pain from the body. Indeed, recent work in rodents reports different patterns of activation in ascending pain pathways during noxious stimulation of the skin of the face when compared to noxious stimulation of the body. Such differences may dictate different activation patterns in higher brain regions, specifically in those areas processing the affective component of pain. We aimed to use ultra-high field functional magnetic resonance imaging (fMRI at 7-Tesla) to determine whether noxious thermal stimuli applied to the surface of the face and body evoke differential activation patterns within the ascending pain pathway in awake humans (n=16). Compared to the body, noxious heat stimulation to the face evoked more widespread signal changes in prefrontal cortical regions and numerous brainstem and subcortical limbic areas. Moreover, facial pain evoked significantly different signal changes in the lateral parabrachial nucleus, substantia nigra, paraventricular hypothalamus, and paraventricular thalamus, to those evoked by body pain. These results are consistent with recent preclinical findings of differential activation in the brainstem and subcortical limbic nuclei and associated cortices during cutaneous pain of the face when compared with the body. The findings suggest one potential mechanism by which facial pain could evoke a greater emotional impact than that evoked by body pain.
Collapse
Affiliation(s)
- Rebecca V Robertson
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Noemi Meylakh
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Lewis S Crawford
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Fernando A Tinoco Mendoza
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Paul M Macey
- UCLA School of Nursing and Brain Research Institute, University of California, Los Angeles, California, 90095, USA
| | | | - Kevin A Keay
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Luke A Henderson
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia.
| |
Collapse
|
26
|
Harriott AM, Kaya M, Ayata C. Oxytocin shortens spreading depolarization-induced periorbital allodynia. J Headache Pain 2024; 25:152. [PMID: 39289629 PMCID: PMC11406737 DOI: 10.1186/s10194-024-01855-7] [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: 07/07/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Migraine is among the most prevalent and burdensome neurological disorders in the United States based on disability-adjusted life years. Cortical spreading depolarization (SD) is the most likely electrophysiological cause of migraine aura and may be linked to trigeminal nociception. We previously demonstrated, using a minimally invasive optogenetic approach of SD induction (opto-SD), that opto-SD triggers acute periorbital mechanical allodynia that is reversed by 5HT1B/1D receptor agonists, supporting SD-induced activation of migraine-relevant trigeminal pain pathways in mice. Recent data highlight hypothalamic neural circuits in migraine, and SD may activate hypothalamic neurons. Furthermore, neuroanatomical, electrophysiological, and behavioral data suggest a homeostatic analgesic function of hypothalamic neuropeptide hormone, oxytocin. We, therefore, examined the role of hypothalamic paraventricular nucleus (PVN) and oxytocinergic (OXT) signaling in opto-SD-induced trigeminal pain behavior. METHODS We induced a single opto-SD in adult male and female Thy1-ChR2-YFP transgenic mice and quantified fos immunolabeling in the PVN and supraoptic nucleus (SON) compared with sham controls. Oxytocin expression was also measured in fos-positive neurons in the PVN. Periorbital mechanical allodynia was tested after treatment with selective OXT receptor antagonist L-368,899 (5 to 25 mg/kg i.p.) or vehicle at 1, 2, and 4 h after opto-SD or sham stimulation using von Frey monofilaments. RESULTS Opto-SD significantly increased the number of fos immunoreactive cells in the PVN and SON as compared to sham stimulation (p < 0.001, p = 0.018, respectively). A subpopulation of fos-positive neurons also stained positive for oxytocin. Opto-SD evoked periorbital mechanical allodynia 1 h after SD (p = 0.001 vs. sham), which recovered quickly within 2 h (p = 0.638). OXT receptor antagonist L-368,899 dose-dependently prolonged SD-induced periorbital allodynia (p < 0.001). L-368,899 did not affect mechanical thresholds in the absence of opto-SD. CONCLUSIONS These data support an SD-induced activation of PVN neurons and a role for endogenous OXT in alleviating acute SD-induced trigeminal allodynia by shortening its duration.
Collapse
Affiliation(s)
- Andrea M Harriott
- Neurovascular Research Unit, Department of Neurology, Massachusetts General Hospital, 149 13th Street, Charlestown, Boston MA, 02129, USA.
| | - Melih Kaya
- Neurovascular Research Unit, Department of Neurology, Massachusetts General Hospital, 149 13th Street, Charlestown, Boston MA, 02129, USA
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Neurology, Massachusetts General Hospital, 149 13th Street, Charlestown, Boston MA, 02129, USA
| |
Collapse
|
27
|
Akpınar G, Ketenci S, Sarıdoğan GE, Aydın B, Tekin N, Cabadak H, Zafer Gören M. The epigenetic changes are affected by sex and valproic acid treatment in a rat model of post-traumatic stress disorder. Neurosci Lett 2024; 839:137957. [PMID: 39218294 DOI: 10.1016/j.neulet.2024.137957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/17/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Post-traumatic stress disorder (PTSD) presents distinct sex-specific differences in both symptom expression and treatment outcomes, with the underlying biological mechanisms still remain unclear. Epigenetic modifications, particularly histone acetylation, have been increasingly recognized as critical factors in the pathophysiology of PTSD. Valproic acid (VPA), a potent histone deacetylase (HDAC) inhibitor, has shown promise in modulating epigenetic responses and improving therapeutic outcomes is PTSD, though its effect may differ between sexes. This study aimed to explore the sex-specific epigenetic changes in response to trauma and the impact of VPA treatment in a rat model of PTSD induced by predator scent stress. Sprague-Dawley rats of both sexes were randomly assigned to stressed and non-stressed groups and treated with either VPA (100 mg/kg) or vehicle. Anxiety levels were assessed using the elevated plus maze, followed by analysis of histone H3 and H4 acetylation, HDAC activity, and c-fos expression in the hippocampus. Our findings revealed that traumatic stress led to increased freezing time and anxiety levels, with more pronounced effects observed in females. Additionally, we have identified sex-specific differences in hippocampal epigenetic modifications; stressed females exhibited higher H3 acetylation, and VPA-treated stressed males showed increased H4 acetylation. These results highlight the importance of considering sex differences in the epigenetic mechanism underlying PTSD and suggest that personalized therapeutic approaches may be necessary to address these complexities.
Collapse
Affiliation(s)
- Gökçe Akpınar
- Department of Medical Pharmacology, Marmara University, School of Medicine, Istanbul, Turkey
| | - Sema Ketenci
- Department of Medical Pharmacology, Marmara University, School of Medicine, Istanbul, Turkey
| | - Gökçe E Sarıdoğan
- Department of Medical Pharmacology, Marmara University, School of Medicine, Istanbul, Turkey
| | - Banu Aydın
- Department of Biophysics, Marmara University, School of Medicine, Istanbul, Turkey
| | - Nurdan Tekin
- Department of Medical Pharmacology, University of Health Sciences, Hamidiye Faculty of Medicine, Istanbul, Turkey
| | - Hülya Cabadak
- Department of Biophysics, Marmara University, School of Medicine, Istanbul, Turkey
| | - M Zafer Gören
- Department of Medical Pharmacology, Marmara University, School of Medicine, Istanbul, Turkey.
| |
Collapse
|
28
|
Fernández-Peña C, Pace RL, Fernando LM, Pittman BG, Schwarz LA. Adrenergic C1 neurons enhance anxiety via projections to PAG. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612440. [PMID: 39314285 PMCID: PMC11419123 DOI: 10.1101/2024.09.11.612440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Anxiety is an emotional state precipitated by the anticipation of real or potential threats. Anxiety disorders are the most prevalent psychiatric illnesses globally and increase the risk of developing comorbid conditions that negatively impact the brain and body. The etiology of anxiety disorders remains unresolved, limiting improvement of therapeutic strategies to alleviate anxiety-related symptoms with increased specificity and efficacy. Here, we applied novel intersectional tools to identify a discrete population of brainstem adrenergic neurons, named C1 cells, that promote aversion and anxiety-related behaviors via projections to the periaqueductal gray matter (PAG). While C1 cells have traditionally been implicated in modulation of autonomic processes, rabies tracing revealed that they receive input from brain areas with diverse functions. Calcium-based in vivo imaging showed that activation of C1 cells enhances excitatory responses in vlPAG, activity that is exacerbated in times of heightened stress. Furthermore, inhibition of C1 cells impedes the development of anxiety-like behaviors in response to stressful situations. Overall, these findings suggest that C1 neurons are positioned to integrate complex information from the brain and periphery for the promotion of anxiety-like behaviors.
Collapse
Affiliation(s)
- Carlos Fernández-Peña
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
| | - Rachel L. Pace
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
| | - Lourds M. Fernando
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
| | - Brittany G. Pittman
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
| | - Lindsay A. Schwarz
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
| |
Collapse
|
29
|
Borland JM. The effects of different types of social interactions on the electrophysiology of neurons in the nucleus accumbens in rodents. Neurosci Biobehav Rev 2024; 164:105809. [PMID: 39004323 PMCID: PMC11771367 DOI: 10.1016/j.neubiorev.2024.105809] [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: 04/23/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
BORLAND, J.M., The effects of different types of social interactions on the electrophysiology of neurons in the nucleus accumbens in rodents, NEUROSCI BIOBEH REV 21(1) XXX-XXX, 2024.-Sociality shapes an organisms' life. The nucleus accumbens is a critical brain region for mental health. In the following review, the effects of different types of social interactions on the physiology of neurons in the nucleus accumbens is synthesized. More specifically, the effects of sex behavior, aggression, social defeat, pair-bonding, play behavior, affiliative interactions, parental behaviors, the isolation from social interactions and maternal separation on measures of excitatory synaptic transmission, intracellular signaling and factors of transcription and translation in neurons in the nucleus accumbens in rodent models are reviewed. Similarities and differences in effects depending on the type of social interaction is then discussed. This review improves the understanding of the molecular and synaptic mechanisms of sociality.
Collapse
|
30
|
Shu Q, Zhou J, Zhang B, Zhang F, Zhou X, Wu Y, Chang H, Hu L, Cai R, Yu Q. Electroacupuncture alleviates myocardial ischemia-reperfusion injury by inhibiting hypothalamic paraventricular nucleus neurons projecting to the rostral ventrolateral medulla. Eur J Neurosci 2024; 60:4861-4876. [PMID: 39054660 DOI: 10.1111/ejn.16480] [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: 10/20/2023] [Revised: 05/20/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024]
Abstract
Accumulating evidence suggests that electroacupuncture (EA) has obvious therapeutic effects and unique advantages in alleviating myocardial ischemia-reperfusion injury (MIRI), while the underlying neuromolecular mechanisms of EA intervention for MIRI have not been fully elucidated. The aim of the study is to investigate the role of the neural pathway of hypothalamic paraventricular nucleus (PVN) neurons projecting to the rostral ventrolateral medulla (RVLM) in the alleviation of MIRI rats by EA preconditioning. MIRI models were established by ligating the left anterior descending coronary artery for 30 min followed by reperfusion for 2 h. Electrocardiogram recording, chemogenetics, enzyme-linked immunosorbent assay, multichannel physiology recording and haematoxylin-eosin and immunofluorescence staining methods were conducted to demonstrate that the firing frequencies of neurons in the PVN and the expression of c-Fos decreased by EA pretreatment. Meanwhile, EA preconditioning significantly reduced the levels of creatine kinase isoenzymes (CK-MB), cardiac troponin I (cTnI) and lactic dehydrogenase (LDH). Virus tracing showed a projection connection between PVN and RVLM. The inhibition of the PVN-RVLM neural pathway could replicate the protective effect of EA pretreatment on MIRI rats. However, the activation of the pathway weakened the effect of EA preconditioning. EA pretreatment alleviated MIRI by regulating PVN neurons projecting to RVLM. This work provides novel evidence of EA pretreatment for alleviating MIRI.
Collapse
Affiliation(s)
- Qi Shu
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jie Zhou
- Medical College of Acu-Moxi, Anhui University of Chinese Medicine, Hefei, China
| | - Bin Zhang
- Medical College of Acu-Moxi, Anhui University of Chinese Medicine, Hefei, China
| | - Fan Zhang
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xiang Zhou
- Medical College of Acu-Moxi, Anhui University of Chinese Medicine, Hefei, China
| | - Yan Wu
- Medical College of Acu-Moxi, Anhui University of Chinese Medicine, Hefei, China
| | - Huimin Chang
- Medical College of Acu-Moxi, Anhui University of Chinese Medicine, Hefei, China
| | - Ling Hu
- Medical College of Acu-Moxi, Anhui University of Chinese Medicine, Hefei, China
- Institute of Acupuncture and Moxibustion Meridian, Anhui University of Chinese Medicine, Hefei, China
| | - Ronglin Cai
- Institute of Acupuncture and Moxibustion Meridian, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Meridian Viscera Correlationship, Anhui University of Chinese Medicine, Hefei, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute for Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Qing Yu
- Institute of Acupuncture and Moxibustion Meridian, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Meridian Viscera Correlationship, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
31
|
Sanguino-Gómez J, Huijgens S, den Hartog M, Schenk IJM, Kluck W, Versluis TD, Krugers HJ. Neural correlates of learning and memory are altered by early-life stress. Neurobiol Learn Mem 2024; 213:107952. [PMID: 38906243 DOI: 10.1016/j.nlm.2024.107952] [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: 01/11/2024] [Revised: 04/12/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024]
Abstract
The ability to learn and remember, which is fundamental for behavioral adaptation, is susceptible to stressful experiences during the early postnatal period, such as abnormal levels of maternal care. The exact mechanisms underlying these effects still remain elusive. This study examined whether early life stress (ELS) alters memory and brain activation patterns in male mice. Therefore, we examined the expression of the immediate early genes (IEGs) c-Fos and Arc in the dentate gyrus (DG) and basolateral amygdala (BLA) after training and memory retrieval in a fear conditioning task. Furthermore, we examined the potential of RU38486 (RU486), a glucocorticoid receptor antagonist, to mitigate ELS-induced memory deficits by blocking stress signalling during adolescence. Arc::dVenus reporter mice, which allow investigating experience-dependent expression of the immediate early gene Arc also at more remote time points, were exposed to ELS by housing dams and offspring with limited bedding and nesting material (LBN) between postnatal days (PND) 2-9 and trained in a fear conditioning task at adult age. We found that ELS reduced both fear acquisition and contextual memory retrieval. RU486 did not prevent these effects. ELS reduced the number of Arc::dVenus+ cells in DG and BLA after training, while the number of c-Fos+ cells were left unaffected. After memory retrieval, ELS decreased c-Fos+ cells in the ventral DG and BLA. ELS also altered the colocalization of c-Fos+ cells with Arc::dVenus+ cells in the ventral DG, possibly indicating impaired engram allocation in the ventral DG after memory retrieval. In conclusion, this study shows that ELS alters neuronal activation patterns after fear acquisition and retrieval, which may provide mechanistic insights into enduring impact of ELS on the processing of fear memories, possibly via changes in cell (co-) activation and engram cell allocation.
Collapse
Affiliation(s)
| | - Stefan Huijgens
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Maxine den Hartog
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Inim J M Schenk
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Wenya Kluck
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Tamara D Versluis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
32
|
Mitchell JR, Vincelette L, Tuberman S, Sheppard V, Bergeron E, Calitri R, Clark R, Cody C, Kannan A, Keith J, Parakoyi A, Pikus M, Vance V, Ziane L, Brenhouse H, Laine MA, Shansky RM. Behavioral and neural correlates of diverse conditioned fear responses in male and female rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608817. [PMID: 39229164 PMCID: PMC11370446 DOI: 10.1101/2024.08.20.608817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Pavlovian fear conditioning is a widely used tool that models associative learning in rodents. For decades the field has used predominantly male rodents and focused on a sole conditioned fear response: freezing. However, recent work from our lab and others has identified darting as a female-biased conditioned response, characterized by an escape-like movement across a fear conditioning chamber. It is also accompanied by a behavioral phenotype: Darters reliably show decreased freezing compared to Non-darters and males and reach higher velocities in response to the foot shock ("shock response"). However, the relationship between shock response and conditioned darting is not known. This study investigated if this link is due to differences in general processing of aversive stimuli between Darters, Non-darters and males. Across a variety of modalities, including corticosterone measures, the acoustic startle test, and sensitivity to thermal pain, Darters were found not to be more reactive or sensitive to aversive stimuli, and, in some cases, they appear less reactive to Non-darters and males. Analyses of cFos activity in regions involved in pain and fear processing following fear conditioning identified discrete patterns of expression among Darters, Non-darters, and males exposed to low and high intensity foot shocks. The results from these studies further our understanding of the differences between Darters, Non-darters and males and highlight the importance of studying individual differences in fear conditioning as indicators of fear state.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mikaela A. Laine
- Northeastern University, Boston, MA
- Smith College, Northampton, MA
| | | |
Collapse
|
33
|
Roh SH, Mendez-Vazquez H, Sathler MF, Doolittle MJ, Zaytseva A, Brown H, Sainsbury M, Kim S. Prenatal exposure to valproic acid reduces synaptic δ-catenin levels and disrupts ultrasonic vocalization in neonates. Neuropharmacology 2024; 253:109963. [PMID: 38657945 PMCID: PMC11127754 DOI: 10.1016/j.neuropharm.2024.109963] [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: 12/14/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Valproic acid (VPA) is an effective and commonly prescribed drug for epilepsy and bipolar disorder. However, children born from mothers treated with VPA during pregnancy exhibit an increased incidence of autism spectrum disorder (ASD). Although VPA may impair brain development at the cellular level, the mechanism of VPA-induced ASD has not been completely addressed. A previous study has found that VPA treatment strongly reduces δ-catenin mRNA levels in cultured human neurons. δ-catenin is important for the control of glutamatergic synapses and is strongly associated with ASD. VPA inhibits dendritic morphogenesis in developing neurons, an effect that is also found in neurons lacking δ-catenin expression. We thus hypothesize that prenatal exposure to VPA significantly reduces δ-catenin levels in the brain, which impairs glutamatergic synapses to cause ASD. Here, we found that prenatal exposure to VPA markedly reduced δ-catenin levels in the brain of mouse pups. VPA treatment also impaired dendritic branching in developing mouse cortical neurons, which was partially reversed by elevating δ-catenin expression. Prenatal VPA exposure significantly reduced synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor levels and postsynaptic density 95 (PSD95) in the brain of mouse pups, indicating dysfunctions in glutamatergic synaptic transmission. VPA exposure also significantly altered ultrasonic vocalization (USV) in newly born pups when they were isolated from their nest. Moreover, VPA-exposed pups show impaired hypothalamic response to isolation, which is required to produce animals' USVs following isolation from the nest. Therefore, these results suggest that VPA-induced ASD pathology can be mediated by the loss of δ-catenin functions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Morgan Sainsbury
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Seonil Kim
- Department of Biomedical Sciences, USA; Molecular, Cellular and Integrative Neurosciences Program, USA.
| |
Collapse
|
34
|
Kopsidas CA, Lowe CC, McDaniel DP, Zhou X, Feng Y. Sustained generation of neurons destined for neocortex with oxidative metabolic upregulation upon filamin abrogation. iScience 2024; 27:110199. [PMID: 38989458 PMCID: PMC11233971 DOI: 10.1016/j.isci.2024.110199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 04/01/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Neurons in the neocortex are generated during embryonic development. While the adult ventricular-subventricular zone (V-SVZ) contains cells with neural stem/progenitors' characteristics, it remains unclear whether it has the capacity of producing neocortical neurons. Here, we show that generating neurons with transcriptomic resemblance to upper layer neocortical neurons continues in the V-SVZ of mouse models of a human condition known as periventricular heterotopia by abrogating Flna and Flnb. We found such surplus neurogenesis was associated with V-SVZ's upregulation of oxidative phosphorylation, mitochondrial biogenesis, and vascular abundance. Additionally, spatial transcriptomics analyses showed V-SVZ's neurogenic activation was coupled with transcriptional enrichment of genes in diverse pathways for energy metabolism, angiogenesis, cell signaling, synaptic transmission, and turnovers of nucleic acids and proteins in upper cortical layers. These findings support the potential of generating neocortical neurons in adulthood through boosting brain-wide vascular circulation, aerobic adenosine triphosphate synthesis, metabolic turnover, and neuronal activity.
Collapse
Affiliation(s)
- Caroline A. Kopsidas
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Clara C. Lowe
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Dennis P. McDaniel
- Biomedical Instrumentation Center, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Xiaoming Zhou
- Department of Medicine, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Yuanyi Feng
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
35
|
Cruz-Ochoa NA, Motta-Teixeira LC, Cruz-Ochoa PF, Lopez-Paredes S, Ochoa-Amaya JE, Takada SH, Xavier GF, Nogueira MI. Post-weaning social isolation modifies neonatal anoxia-induced changes in energy metabolism and growth of rats. Int J Dev Neurosci 2024; 84:293-304. [PMID: 38530155 DOI: 10.1002/jdn.10327] [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: 08/24/2023] [Revised: 02/08/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
Neonatal oxygen deficiency in rats may disturb growth and long-term metabolic homeostasis. In order to facilitate metabolic evaluation, the subjects are usually housed individually. However, social isolation associated with individually housed conditions alters animal behavior, which may influence the experimental results. This study investigated the effects of social isolation on neonatal anoxia-induced changes in growth and energy metabolism. Male and female Wistar rats were exposed, on postnatal day 2 (P2), to either 25-min of anoxia or control treatment. From P27 onward, part of the subjects of each group was isolated in standard cages, and the remaining subjects were housed in groups. At P34 or P95, the subjects were fasted for 18 h, refeed for 1 h, and then perfused 30 min later. Glycemia, leptin, insulin, and morphology of the pancreas were evaluated at both ages. For subjects perfused at P95, body weight and food intake were recorded up to P90, and the brain was collected for Fos and NeuN immunohistochemistry. Results showed that male rats exposed to neonatal anoxia and social isolation exhibited increased body weight gain despite the lack of changes in food intake. In addition, social isolation (1) decreased post-fasting weight loss and post-fasting food intake and (2) increased glycemia, insulin, and leptin levels of male and female rats exposed to anoxia and control treatments, both at P35 and P95. Furthermore, although at P35, anoxia increased insulin levels of males, it decreased the area of the β-positive cells in the pancreas of females. At P95, anoxia increased post-prandial weight loss of males, post-fasting food intake, insulin, and leptin, and decreased Fos expression in the arcuate nucleus (ARC) of males and females. Hyperphagia was associated with possible resistance to leptin and insulin, suspected by the high circulating levels of these hormones and poor neuronal activation of ARC. This study demonstrated that continuous social isolation from weaning modifies, in a differentiated way, the long-term energy metabolism and growth of male and female Wistar rats exposed to neonatal anoxia or even control treatments. Therefore, social isolation should be considered as a factor that negatively influences experimental results and the outcomes of the neonatal injury. These results should also be taken into account in clinical procedures, since the used model simulates the preterm babies' conditions and some therapeutic approaches require isolation.
Collapse
Affiliation(s)
- Natalia Andrea Cruz-Ochoa
- Neurosciences Laboratory, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Lívia Clemente Motta-Teixeira
- Neurobiology Laboratory, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Pablo Felipe Cruz-Ochoa
- Laboratory of Wildlife Comparative Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Santiago Lopez-Paredes
- Research Group of Pathology of Domestic and Wild Animals. Facultad de Ciencias Agropecuarias y Recursos Naturales, Universidad de los Llanos, Villavicencio, Colombia
| | - Julieta Esperanza Ochoa-Amaya
- Research Group of Pathology of Domestic and Wild Animals. Facultad de Ciencias Agropecuarias y Recursos Naturales, Universidad de los Llanos, Villavicencio, Colombia
| | - Silvia Honda Takada
- Laboratory of Neurogenetics. Center for Mathematics, Computing and Cognition, Federal University of ABC, São Bernardo do Campo, São Paolo, Brazil
| | - Gilberto Fernando Xavier
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Maria Inês Nogueira
- Neurosciences Laboratory, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
Glavonic E, Dragic M, Mitic M, Aleksic M, Lukic I, Ivkovic S, Adzic M. Ketamine's Amelioration of Fear Extinction in Adolescent Male Mice Is Associated with the Activation of the Hippocampal Akt-mTOR-GluA1 Pathway. Pharmaceuticals (Basel) 2024; 17:669. [PMID: 38931336 PMCID: PMC11206546 DOI: 10.3390/ph17060669] [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: 03/01/2024] [Revised: 03/29/2024] [Accepted: 05/14/2024] [Indexed: 06/28/2024] Open
Abstract
Fear-related disorders, including post-traumatic stress disorder (PTSD), and anxiety disorders are pervasive psychiatric conditions marked by persistent fear, stemming from its dysregulated acquisition and extinction. The primary treatment for these disorders, exposure therapy (ET), relies heavily on fear extinction (FE) principles. Adolescence, a vulnerable period for developing psychiatric disorders, is characterized by neurobiological changes in the fear circuitry, leading to impaired FE and increased susceptibility to relapse following ET. Ketamine, known for relieving anxiety and reducing PTSD symptoms, influences fear-related learning processes and synaptic plasticity across the fear circuitry. Our study aimed to investigate the effects of ketamine (10 mg/kg) on FE in adolescent male C57 BL/6 mice at the behavioral and molecular levels. We analyzed the protein and gene expression of synaptic plasticity markers in the hippocampus (HPC) and prefrontal cortex (PFC) and sought to identify neural correlates associated with ketamine's effects on adolescent extinction learning. Ketamine ameliorated FE in the adolescent males, likely affecting the consolidation and/or recall of extinction memory. Ketamine also increased the Akt and mTOR activity and the GluA1 and GluN2A levels in the HPC and upregulated BDNF exon IV mRNA expression in the HPC and PFC of the fear-extinguished mice. Furthermore, ketamine increased the c-Fos expression in specific brain regions, including the ventral HPC (vHPC) and the left infralimbic ventromedial PFC (IL vmPFC). Providing a comprehensive exploration of ketamine's mechanisms in adolescent FE, our study suggests that ketamine's effects on FE in adolescent males are associated with the activation of hippocampal Akt-mTOR-GluA1 signaling, with the vHPC and the left IL vmPFC as the proposed neural correlates.
Collapse
Affiliation(s)
- Emilija Glavonic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| | - Milorad Dragic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11158 Belgrade, Serbia
| | - Milos Mitic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| | - Minja Aleksic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| | - Iva Lukic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| | - Sanja Ivkovic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| |
Collapse
|
37
|
Harari R, Chatterjee I, Getselter D, Elliott E. Psilocybin induces acute anxiety and changes in amygdalar phosphopeptides independently from the 5-HT2A receptor. iScience 2024; 27:109686. [PMID: 38660396 PMCID: PMC11039401 DOI: 10.1016/j.isci.2024.109686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/29/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024] Open
Abstract
Psilocybin, and its metabolite psilocin, induces psychedelic effects through activation of the 5-HT2A receptor. Psilocybin has been proposed as a treatment for depression and anxiety but sometimes induces anxiety in humans. An understanding of mechanisms underlying the anxiety response will help to better develop therapeutic prospects of psychedelics. In the current study, psilocybin induced an acute increase in anxiety in behavioral paradigms in mice. Importantly, pharmacological blocking of the 5-HT2A receptor attenuates psilocybin-induced head twitch response, a behavioral proxy for the psychedelic response, but does not rescue psilocybin's effect on anxiety-related behavior. Phosphopeptide analysis in the amygdala uncovered signal transduction pathways that are dependent or independent of the 5-HT2A receptor. Furthermore, presynaptic proteins are specifically involved in psilocybin-induced acute anxiety. These insights into how psilocybin may induce short-term anxiety are important for understanding how psilocybin may best be used in the clinical framework.
Collapse
Affiliation(s)
- Ram Harari
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 13215, Israel
| | - Ipsita Chatterjee
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 13215, Israel
- Department of Neuroscience, Yale University, New Haven, CT 06520, USA
| | - Dmitriy Getselter
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 13215, Israel
| | - Evan Elliott
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 13215, Israel
| |
Collapse
|
38
|
Wood CP, Alvarez C, DiPatrizio NV. Cholinergic Neurotransmission Controls Orexigenic Endocannabinoid Signaling in the Gut in Diet-Induced Obesity. J Neurosci 2024; 44:e0813232024. [PMID: 38594069 PMCID: PMC11097264 DOI: 10.1523/jneurosci.0813-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 03/12/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
The brain bidirectionally communicates with the gut to control food intake and energy balance, which becomes dysregulated in obesity. For example, endocannabinoid (eCB) signaling in the small-intestinal (SI) epithelium is upregulated in diet-induced obese (DIO) mice and promotes overeating by a mechanism that includes inhibiting gut-brain satiation signaling. Upstream neural and molecular mechanism(s) involved in overproduction of orexigenic gut eCBs in DIO, however, are unknown. We tested the hypothesis that overactive parasympathetic signaling at the muscarinic acetylcholine receptors (mAChRs) in the SI increases biosynthesis of the eCB, 2-arachidonoyl-sn-glycerol (2-AG), which drives hyperphagia via local CB1Rs in DIO. Male mice were maintained on a high-fat/high-sucrose Western-style diet for 60 d, then administered several mAChR antagonists 30 min prior to tissue harvest or a food intake test. Levels of 2-AG and the activity of its metabolic enzymes in the SI were quantitated. DIO mice, when compared to those fed a low-fat/no-sucrose diet, displayed increased expression of cFos protein in the dorsal motor nucleus of the vagus, which suggests an increased activity of efferent cholinergic neurotransmission. These mice exhibited elevated levels of 2-AG biosynthesis in the SI, that was reduced to control levels by mAChR antagonists. Moreover, the peripherally restricted mAChR antagonist, methylhomatropine bromide, and the peripherally restricted CB1R antagonist, AM6545, reduced food intake in DIO mice for up to 24 h but had no effect in mice conditionally deficient in SI CB1Rs. These results suggest that hyperactivity at mAChRs in the periphery increases formation of 2-AG in the SI and activates local CB1Rs, which drives hyperphagia in DIO.
Collapse
Affiliation(s)
- Courtney P Wood
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California 92521
- University of California Riverside Center for Cannabinoid Research, Riverside, California 92521
| | - Camila Alvarez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California 92521
- University of California Riverside Center for Cannabinoid Research, Riverside, California 92521
| | - Nicholas V DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California 92521
- University of California Riverside Center for Cannabinoid Research, Riverside, California 92521
| |
Collapse
|
39
|
Danis AB, Gallagher AA, Anderson AN, Isakharov A, Beeson KA, Schnell E. Altered Hippocampal Activation in Seizure-Prone CACNA2D2 Knock-out Mice. eNeuro 2024; 11:ENEURO.0486-23.2024. [PMID: 38749701 PMCID: PMC11097259 DOI: 10.1523/eneuro.0486-23.2024] [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: 11/21/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 05/18/2024] Open
Abstract
The voltage-gated calcium channel subunit α2δ-2 controls calcium-dependent signaling in neurons, and loss of this subunit causes epilepsy in both mice and humans. To determine whether mice without α2δ-2 demonstrate hippocampal activation or histopathological changes associated with seizure activity, we measured expression of the activity-dependent gene c-fos and various histopathological correlates of temporal lobe epilepsy (TLE) in hippocampal tissue from wild-type (WT) and α2δ-2 knock-out (CACNA2D2 KO) mice using immunohistochemical staining and confocal microscopy. Both genotypes demonstrated similarly sparse c-fos and ΔFosB expressions within the hippocampal dentate granule cell layer (GCL) at baseline, consistent with no difference in basal activity of granule cells between genotypes. Surprisingly, when mice were assayed 1 h after handling-associated convulsions, KO mice had fewer c-fos-positive cells but dramatically increased ΔFosB expression in the dentate gyrus compared with WT mice. After administration of a subthreshold pentylenetetrazol dose, however, KO mice dentate had significantly more c-fos expression compared with WT mice. Other histopathological markers of TLE in these mice, including markers of neurogenesis, glial activation, and mossy fiber sprouting, were similar between WT and KO mice, apart from a small but statistically significant increase in hilar mossy cell density, opposite to what is typically found in mice with TLE. This suggests that the differences in seizure-associated dentate gyrus function in the absence of α2δ-2 protein are likely due to altered functional properties of the network without associated structural changes in the hippocampus at the typical age of seizure onset.
Collapse
Affiliation(s)
- Alyssa B Danis
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon 97239
- Research and Development Service, Portland VA Health Care System, Portland, Oregon 97239
| | - Ashlynn A Gallagher
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon 97239
- Research and Development Service, Portland VA Health Care System, Portland, Oregon 97239
| | - Ashley N Anderson
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon 97239
- Research and Development Service, Portland VA Health Care System, Portland, Oregon 97239
| | - Arielle Isakharov
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, Oregon 97239
| | - Kathleen A Beeson
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, Oregon 97239
| | - Eric Schnell
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon 97239
- Research and Development Service, Portland VA Health Care System, Portland, Oregon 97239
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, Oregon 97239
| |
Collapse
|
40
|
Sun Y, Darmani NA. A Comparative Study of the Antiemetic Effects of α 2-Adrenergic Receptor Agonists Clonidine and Dexmedetomidine against Diverse Emetogens in the Least Shrew ( Cryptotis parva) Model of Emesis. Int J Mol Sci 2024; 25:4603. [PMID: 38731821 PMCID: PMC11083949 DOI: 10.3390/ijms25094603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/09/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
In contrast to cats and dogs, here we report that the α2-adrenergic receptor antagonist yohimbine is emetic and corresponding agonists clonidine and dexmedetomidine behave as antiemetics in the least shrew model of vomiting. Yohimbine (0, 0.5, 0.75, 1, 1.5, 2, and 3 mg/kg, i.p.) caused vomiting in shrews in a bell-shaped and dose-dependent manner, with a maximum frequency (0.85 ± 0.22) at 1 mg/kg, which was accompanied by a key central contribution as indicated by increased expression of c-fos, serotonin and substance P release in the shrew brainstem emetic nuclei. Our comparative study in shrews demonstrates that clonidine (0, 0.1, 1, 5, and 10 mg/kg, i.p.) and dexmedetomidine (0, 0.01, 0.05, and 0.1 mg/kg, i.p.) not only suppress yohimbine (1 mg/kg, i.p.)-evoked vomiting in a dose-dependent manner, but also display broad-spectrum antiemetic effects against diverse well-known emetogens, including 2-Methyl-5-HT, GR73632, McN-A-343, quinpirole, FPL64176, SR141716A, thapsigargin, rolipram, and ZD7288. The antiemetic inhibitory ID50 values of dexmedetomidine against the evoked emetogens are much lower than those of clonidine. At its antiemetic doses, clonidine decreased shrews' locomotor activity parameters (distance moved and rearing), whereas dexmedetomidine did not do so. The results suggest that dexmedetomidine represents a better candidate for antiemetic potential with advantages over clonidine.
Collapse
Affiliation(s)
| | - Nissar A. Darmani
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 East Second Street, Pomona, CA 91766, USA;
| |
Collapse
|
41
|
Maejima Y, Yokota S, Hidema S, Nishimori K, de Wet H, Shimomura K. Systemic Co-Administration of Low-Dose Oxytocin and Glucagon-Like Peptide 1 Additively Decreases Food Intake and Body Weight. Neuroendocrinology 2024; 114:639-657. [PMID: 38599201 DOI: 10.1159/000538792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/08/2024] [Indexed: 04/12/2024]
Abstract
INTRODUCTION GLP-1 receptor agonists are the number one drug prescribed for the treatment of obesity and type 2 diabetes. These drugs are not, however, without side effects, and in an effort to maximize therapeutic effect while minimizing adverse effects, gut hormone co-agonists received considerable attention as new drug targets in the fight against obesity. Numerous previous reports identified the neuropeptide oxytocin (OXT) as a promising anti-obesity drug. The aims of this study were to evaluate OXT as a possible co-agonist for GLP-1 and examine the effects of its co-administration on food intake (FI) and body weight (BW) in mice. METHODS FI and c-Fos levels were measured in the feeding centers of the brain in response to an intraperitoneal injection of saline, OXT, GLP-1, or OXT/GLP-1. The action potential frequency and cytosolic Ca2+ ([Ca2+]i) in response to OXT, GLP-1, or OXT/GLP-1 were measured in ex vivo paraventricular nucleus (PVN) neuronal cultures. Finally, FI and BW changes were compared in diet-induced obese mice treated with saline, OXT, GLP-1, or OXT/GLP-1 for 13 days. RESULTS Single injection of OXT/GLP-1 additively decreased FI and increased c-Fos expression specifically in the PVN and supraoptic nucleus. Seventy percent of GLP-1 receptor-positive neurons in the PVN also expressed OXT receptors, and OXT/GLP-1 co-administration dramatically increased firing and [Ca2+]i in the PVN OXT neurons. The chronic OXT/GLP-1 co-administration decreased BW without changing FI. CONCLUSION Chronic OXT/GLP-1 co-administration decreases BW, possibly via the activation of PVN OXT neurons. OXT might be a promising candidate as an incretin co-agonist in obesity treatment.
Collapse
Affiliation(s)
- Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
- Departments of Obesity and Inflammation Research, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford, UK
| | - Shoko Yokota
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shizu Hidema
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Katsuhiko Nishimori
- Departments of Obesity and Inflammation Research, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Heidi de Wet
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford, UK
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
- Departments of Obesity and Inflammation Research, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
42
|
Lee Y, Kim S, Cho YK, Kong C, Chang JW, Jun SB. Amygdala electrical stimulation for operant conditioning in rat navigation. Biomed Eng Lett 2024; 14:291-306. [PMID: 38374898 PMCID: PMC10874353 DOI: 10.1007/s13534-023-00336-1] [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: 09/13/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 02/21/2024] Open
Abstract
There have been several attempts to navigate the locomotion of animals by neuromodulation. The most common method is animal training with electrical brain stimulation for directional cues and rewards; the basic principle is to activate dopamine-mediated neural reward pathways such as the medial forebrain bundle (MFB) when the animal correctly follows the external commands. In this study, the amygdala, which is the brain region responsible for fear modulation, was targeted for punishment training. The brain regions of MFB, amygdala, and barrel cortex were electrically stimulated for reward, punishment, and directional cues, respectively. Electrical stimulation was applied to the amygdala of rats when they failed to follow directional commands. First, two different amygdala regions, i.e., basolateral amygdala (BLA) and central amygdala (CeA), were stimulated and compared in terms of behavior responses, success and correction rates for training, and gene expression for learning and memory. Then, the training was performed in three groups: group R (MFB stimulation for reward), group P (BLA stimulation for punishment), and group RP (both MFB and BLA stimulation for reward and punishment). In group P, after the training, RNA sequencing was conducted to detect gene expression and demonstrate the effect of punishment learning. Group P showed higher success rates than group R, and group RP exhibited the most effective locomotion control among the three groups. Gene expression results imply that BLA stimulation can be more effective as a punishment in the learning process than CeA stimulation. We developed a new method to navigate rat locomotion behaviors by applying amygdala stimulation.
Collapse
Affiliation(s)
- Youjin Lee
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760 Republic of Korea
- Graduate Program in Smart Factory, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Soonyoung Kim
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005 USA
| | - Yoon Kyung Cho
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Chanho Kong
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Jin Woo Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science and Brain Research Institute, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Sang Beom Jun
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760 Republic of Korea
- Graduate Program in Smart Factory, Ewha Womans University, Seoul, 03760 Republic of Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| |
Collapse
|
43
|
Bimbi G, Tongiorgi E. Chemical LTP induces confinement of BDNF mRNA under dendritic spines and BDNF protein accumulation inside the spines. Front Mol Neurosci 2024; 17:1348445. [PMID: 38450041 PMCID: PMC10914971 DOI: 10.3389/fnmol.2024.1348445] [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: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) plays a key role in neuronal development and synaptic plasticity. The discovery that BDNF mRNA can be transported in neuronal dendrites in an activity-dependent manner has suggested that its local translation may support synapse maturation and plasticity. However, a clear demonstration that BDNF mRNA is locally transported and translated at activated synapses in response to long-term potentiation (LTP) is still lacking. Here, we study the dynamics of BDNF mRNA dendritic trafficking following the induction of chemical LTP (cLTP). Dendritic transport of BDNF transcripts was analyzed using the MS2 system for mRNA visualization, and chimeric BDNF-GFP constructs were used to monitor protein synthesis in living neurons. We found that within 15 min from cLTP induction, most BDNF mRNA granules become stationary and transiently accumulate in the dendritic shaft at the base of the dendritic spines, while at 30 min they accumulate inside the spine, similar to the control CamkIIα mRNA which also increased inside the spines at 60 min post-cLTP. At 60 min but not at 15 min from cLTP induction, we observed an increase in BDNF protein levels within the spines. Taken together, these findings suggest that BDNF mRNA trafficking is arrested in the early phase of cLTP, providing a local source of mRNA for BDNF translation at the base of the spine followed by translocation of both the BDNF mRNA and protein within the spine head in the late phase of LTP.
Collapse
Affiliation(s)
| | - Enrico Tongiorgi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
44
|
Abdelhamid M, Jung CG, Zhou C, Inoue R, Chen Y, Sento Y, Hida H, Michikawa M. Potential Therapeutic Effects of Bifidobacterium breve MCC1274 on Alzheimer's Disease Pathologies in AppNL-G-F Mice. Nutrients 2024; 16:538. [PMID: 38398861 PMCID: PMC10893354 DOI: 10.3390/nu16040538] [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: 01/24/2024] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
We previously demonstrated that orally supplemented Bifidobacterium breve MCC1274 (B. breve MCC1274) mitigated Alzheimer's disease (AD) pathologies in both 7-month-old AppNL-G-F mice and wild-type mice; thus, B. breve MCC1274 supplementation might potentially prevent the progression of AD. However, the possibility of using this probiotic as a treatment for AD remains unclear. Thus, we investigated the potential therapeutic effects of this probiotic on AD using 17-month-old AppNL-G-F mice with memory deficits and amyloid beta saturation in the brain. B. breve MCC1274 supplementation ameliorated memory impairment via an amyloid-cascade-independent pathway. It reduced hippocampal and cortical levels of phosphorylated extracellular signal-regulated kinase and c-Jun N-terminal kinase as well as heat shock protein 90, which might have suppressed tau hyperphosphorylation and chronic stress. Moreover, B. breve MCC1274 supplementation increased hippocampal synaptic protein levels and upregulated neuronal activity. Thus, B. breve MCC1274 supplementation may alleviate cognitive dysfunction by reducing chronic stress and tau hyperphosphorylation, thereby enhancing both synaptic density and neuronal activity in 17-month-old AppNL-G-F mice. Overall, this study suggests that B. breve MCC1274 has anti-AD effects and can be used as a potential treatment for AD.
Collapse
Affiliation(s)
- Mona Abdelhamid
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
| | - Cha-Gyun Jung
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
- Department of Neurophysiology and Brain Science, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan;
| | - Chunyu Zhou
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
| | - Rieko Inoue
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
| | - Yuxin Chen
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
| | - Yoshiki Sento
- Department of Anesthesiology and Intensive Care Medicine, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan;
| | - Hideki Hida
- Department of Neurophysiology and Brain Science, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan;
| | - Makoto Michikawa
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
- Department of Geriatric Medicine School of Life, Dentistry at Niigata, Nippon Dental University, 1-8 Hamaura-cho, Chuo-ku, Niigata 951-8580, Japan
| |
Collapse
|
45
|
Xie Y, Brynildsen JK, Windisch K, Blendy JA. Neural Network Connectivity Following Opioid Dependence is Altered by a Common Genetic Variant in the µ-Opioid Receptor, OPRM1 A118G. J Neurosci 2024; 44:e1492232023. [PMID: 38124015 PMCID: PMC10866092 DOI: 10.1523/jneurosci.1492-23.2023] [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: 08/24/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023] Open
Abstract
Opioid use disorder is a chronic, relapsing disease associated with persistent changes in brain plasticity. A common single nucleotide polymorphism (SNP) in the µ-opioid receptor gene, OPRM1 A118G, is associated with altered vulnerability to opioid addiction. Reconfiguration of neuronal connectivity may explain dependence risk in individuals with this SNP. Mice with the equivalent Oprm1 variant, A112G, demonstrate sex-specific alterations in the rewarding properties of morphine and heroin. To determine whether this SNP influences network-level changes in neuronal activity, we compared FOS expression in male and female mice that were opioid-naive or opioid-dependent. Network analyses identified significant differences between the AA and GG Oprm1 genotypes. Based on several graph theory metrics, including small-world analysis and degree centrality, we show that GG females in the opioid-dependent state exhibit distinct patterns of connectivity compared to other groups of the same genotype. Using a network control theory approach, we identified key cortical brain regions that drive the transition between opioid-naive and opioid-dependent brain states; however, these regions are less influential in GG females leading to sixfold higher average minimum energy needed to transition from the acute to the dependent state. In addition, we found that the opioid-dependent brain state is significantly less stable in GG females compared to other groups. Collectively, our findings demonstrate sex- and genotype-specific modifications in local, mesoscale, and global properties of functional brain networks following opioid exposure and provide a framework for identifying genotype differences in specific brain regions that play a role in opioid dependence.
Collapse
Affiliation(s)
- Yihan Xie
- Department of Systems Pharmacology and Translational Therapeutics and Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, Pennsylvania
| | - Julia K Brynildsen
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia 19104, Pennsylvania
| | - Kyle Windisch
- Department of Systems Pharmacology and Translational Therapeutics and Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, Pennsylvania
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics and Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, Pennsylvania
| |
Collapse
|
46
|
Chaney R, Leger C, Wirtz J, Fontanier E, Méloux A, Quirié A, Martin A, Prigent-Tessier A, Garnier P. Cerebral Benefits Induced by Electrical Muscle Stimulation: Evidence from a Human and Rat Study. Int J Mol Sci 2024; 25:1883. [PMID: 38339161 PMCID: PMC10855504 DOI: 10.3390/ijms25031883] [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: 12/20/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Physical exercise (EX) is well established for its positive impact on brain health. However, conventional EX may not be feasible for certain individuals. In this regard, this study explores electromyostimulation (EMS) as a potential alternative for enhancing cognitive function. Conducted on both human participants and rats, the study involved two sessions of EMS applied to the quadriceps with a duration of 30 min at one-week intervals. The human subjects experienced assessments of cognition and mood, while the rats underwent histological and biochemical analyses on the prefrontal cortex, hippocampus, and quadriceps. Our findings indicated that EMS enhanced executive functions and reduced anxiety in humans. In parallel, our results from the animal studies revealed an elevation in brain-derived neurotrophic factor (BDNF), specifically in the hippocampus. Intriguingly, this increase was not associated with heightened neuronal activity or cerebral hemodynamics; instead, our data point towards a humoral interaction from muscle to brain. While no evidence of increased muscle and circulating BDNF or FNDC5/irisin pathways could be found, our data highlight lactate as a bridging signaling molecule of the muscle-brain crosstalk following EMS. In conclusion, our results suggest that EMS could be an effective alternative to conventional EX for enhancing both brain health and cognitive function.
Collapse
Affiliation(s)
- Rémi Chaney
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000 Dijon, France; (R.C.); (C.L.); (J.W.); (E.F.); (A.M.); (A.Q.); (P.G.)
| | - Clémence Leger
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000 Dijon, France; (R.C.); (C.L.); (J.W.); (E.F.); (A.M.); (A.Q.); (P.G.)
| | - Julien Wirtz
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000 Dijon, France; (R.C.); (C.L.); (J.W.); (E.F.); (A.M.); (A.Q.); (P.G.)
| | - Estelle Fontanier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000 Dijon, France; (R.C.); (C.L.); (J.W.); (E.F.); (A.M.); (A.Q.); (P.G.)
| | - Alexandre Méloux
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000 Dijon, France; (R.C.); (C.L.); (J.W.); (E.F.); (A.M.); (A.Q.); (P.G.)
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000 Dijon, France; (R.C.); (C.L.); (J.W.); (E.F.); (A.M.); (A.Q.); (P.G.)
| | - Alain Martin
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences du Sport, F-21000 Dijon, France;
| | - Anne Prigent-Tessier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000 Dijon, France; (R.C.); (C.L.); (J.W.); (E.F.); (A.M.); (A.Q.); (P.G.)
| | - Philippe Garnier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000 Dijon, France; (R.C.); (C.L.); (J.W.); (E.F.); (A.M.); (A.Q.); (P.G.)
- Département Génie Biologique, IUT, F-21000 Dijon, France
| |
Collapse
|
47
|
Zhu X, Liu S, Tian L, Li X, Yao R, Zhao Y, Gao Z, Liu XR, Liu XQ, Huo FQ, Liang L. Spinal interleukin-16 mediates inflammatory pain via promoting glial activation. Int Immunopharmacol 2024; 127:111411. [PMID: 38113689 DOI: 10.1016/j.intimp.2023.111411] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 12/21/2023]
Abstract
Proinflammatory cytokines are crucial contributors to neuroinflammation in the development of chronic pain. Here, we identified il16, which encodes interleukin-16 (IL-16), as a differentially expressed gene in spinal dorsal horn of a complete Freund's Adjuvant (CFA) inflammatory pain model in mice by RNA sequencing. We further investigated whether and how IL-16 regulates pain transmission in the spinal cord and contributes to the development of inflammatory pain hypersensitivity. RNA sequencing and bioinformatics analysis revealed elevated IL-16 transcript levels in the spinal dorsal horn after CFA injection. This increase was further confirmed by qPCR, immunofluorescence, and western blotting. Knockdown of IL-16 by intrathecal injection of IL-16 siRNA not only attenuated CFA-induced mechanical and thermal pain hypersensitivity, but also inhibited enhanced c-fos expression and glial activation in the spinal dorsal horn in male mice injected with CFA. Moreover, exogenous IL-16 induced nociceptive responses and increased c-fos expression and glial activation in spinal dorsal horn. This effect was largely impaired when CD4, the binding receptor for IL-16, was inhibited. In addition, CD4 expression was upregulated in the spinal dorsal horn after CFA injection and CD4 was present in microglia and in contact with astrocytes and activated spinal neurons. Taken together, these results suggest that enhanced IL-16-CD4 signaling triggers pain and activates microglia and astrocytes in the spinal dorsal horn, thus contributing to inflammatory pain. IL-16 may serve as a promising target for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Xuan Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Department of Anesthesiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Siyi Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Lixia Tian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Xiang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Rongrong Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Yunhan Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Zihao Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Xue-Ru Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Xin-Qi Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Fu-Quan Huo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Lingli Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
48
|
Simon RC, Fleming WT, Senthilkumar P, Briones BA, Ishii KK, Hjort MM, Martin MM, Hashikawa K, Sanders AD, Golden SA, Stuber GD. Opioid-driven disruption of the septal complex reveals a role for neurotensin-expressing neurons in withdrawal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575766. [PMID: 38293241 PMCID: PMC10827099 DOI: 10.1101/2024.01.15.575766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Because opioid withdrawal is an intensely aversive experience, persons with opioid use disorder (OUD) often relapse to avoid it. The lateral septum (LS) is a forebrain structure that is important in aversion processing, and previous studies have linked the lateral septum (LS) to substance use disorders. It is unclear, however, which precise LS cell types might contribute to the maladaptive state of withdrawal. To address this, we used single-nucleus RNA-sequencing to interrogate cell type specific gene expression changes induced by chronic morphine and withdrawal. We discovered that morphine globally disrupted the transcriptional profile of LS cell types, but Neurotensin-expressing neurons (Nts; LS-Nts neurons) were selectively activated by naloxone. Using two-photon calcium imaging and ex vivo electrophysiology, we next demonstrate that LS-Nts neurons receive enhanced glutamatergic drive in morphine-dependent mice and remain hyperactivated during opioid withdrawal. Finally, we showed that activating and silencing LS-Nts neurons during opioid withdrawal regulates pain coping behaviors and sociability. Together, these results suggest that LS-Nts neurons are a key neural substrate involved in opioid withdrawal and establish the LS as a crucial regulator of adaptive behaviors, specifically pertaining to OUD.
Collapse
Affiliation(s)
- Rhiana C. Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Weston T. Fleming
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Pranav Senthilkumar
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
| | - Brandy A. Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Kentaro K. Ishii
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Madelyn M. Hjort
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Madison M. Martin
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Koichi Hashikawa
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Andrea D. Sanders
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
| | - Sam A. Golden
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Biological Structure, University of Washington, Seattle, WA, 98195, USA
| | - Garret D. Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
- Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
49
|
Lüttig A, Perl S, Zetsche M, Richter F, Franz D, Heerdegen M, Köhling R, Richter A. Short-term stimulations of the entopeduncular nucleus induce cerebellar changes of c-Fos expression in an animal model of paroxysmal dystonia. Brain Res 2024; 1823:148672. [PMID: 37956748 DOI: 10.1016/j.brainres.2023.148672] [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: 08/25/2023] [Revised: 10/23/2023] [Accepted: 11/07/2023] [Indexed: 11/15/2023]
Abstract
Deep brain stimulation (DBS) of the globus pallidus internus (entopeduncular nucleus, EPN, in rodents) is important for the treatment of drug-refractory dystonia. The pathophysiology of this movement disorder and the mechanisms of DBS are largely unknown. Insights into the mechanisms of DBS in animal models of dystonia can be helpful for optimization of DBS and add-on therapeutics. We recently found that short-term EPN-DBS with 130 Hz (50 µA, 60 µs) for 3 h improved dystonia in dtsz hamsters and reduced spontaneous excitatory cortico-striatal activity in brain slices of this model, indicating fast effects on synaptic plasticity. Therefore, in the present study, we examined if these effects are related to changes of c-Fos, a marker of neuronal activity, in brains derived from dtsz hamsters after these short-term DBS or sham stimulations. After DBS vs. sham, c-Fos intensity was increased around the electrode, but the number of c-Fos+ cells was not altered within the whole EPN and projection areas (habenula, thalamus). DBS did not induce changes in striatal and cortical c-Fos+ cells as GABAergic (GAD67+ and parvalbumin-reactive) neurons in motor cortex and striatum. Unexpectedly, c-Fos+ cells were decreased in deep cerebellar nuclei (DCN) after DBS, suggesting that cerebellar changes may be involved in antidystonic effects already during short-term DBS. However, the present results do not exclude functional changes within the basal ganglia-thalamo-cortical network, which will be further investigated by long-term EPN stimulations. The present study indicates that the cerebellum deserves attention in ongoing examinations on the mechanisms of DBS in dystonia.
Collapse
Affiliation(s)
- Anika Lüttig
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, Leipzig University, An den Tierkliniken 15, 04103 Leipzig, Germany.
| | - Stefanie Perl
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, Leipzig University, An den Tierkliniken 15, 04103 Leipzig, Germany
| | - Maria Zetsche
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, Leipzig University, An den Tierkliniken 15, 04103 Leipzig, Germany
| | - Franziska Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, Leipzig University, An den Tierkliniken 15, 04103 Leipzig, Germany; Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Denise Franz
- Oscar Langendorff Institute of Physiology, University Rostock, Gertrudenstraße 9, 18057 Rostock, Germany
| | - Marco Heerdegen
- Oscar Langendorff Institute of Physiology, University Rostock, Gertrudenstraße 9, 18057 Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University Rostock, Gertrudenstraße 9, 18057 Rostock, Germany
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, Leipzig University, An den Tierkliniken 15, 04103 Leipzig, Germany.
| |
Collapse
|
50
|
Mitchell CS, Campbell EJ, Fisher SD, Stanton LM, Burton NJ, Pearl AJ, McNally GP, Bains JS, Füzesi T, Graham BA, Manning EE, Dayas CV. Optogenetic recruitment of hypothalamic corticotrophin-releasing-hormone (CRH) neurons reduces motivational drive. Transl Psychiatry 2024; 14:8. [PMID: 38191479 PMCID: PMC10774335 DOI: 10.1038/s41398-023-02710-0] [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: 02/07/2023] [Revised: 11/20/2023] [Accepted: 12/12/2023] [Indexed: 01/10/2024] Open
Abstract
Impaired motivational drive is a key feature of depression. Chronic stress is a known antecedent to the development of depression in humans and depressive-like states in animals. Whilst there is a clear relationship between stress and motivational drive, the mechanisms underpinning this association remain unclear. One hypothesis is that the endocrine system, via corticotropin-releasing hormone (CRH) in the paraventricular nucleus of the hypothalamus (PVN; PVNCRH), initiates a hormonal cascade resulting in glucocorticoid release, and that excessive glucocorticoids change brain circuit function to produce depression-related symptoms. Another mostly unexplored hypothesis is that the direct activity of PVNCRH neurons and their input to other stress- and reward-related brain regions drives these behaviors. To further understand the direct involvement of PVNCRH neurons in motivation, we used optogenetic stimulation to activate these neurons 1 h/day for 5 consecutive days and showed increased acute stress-related behaviors and long-lasting deficits in the motivational drive for sucrose. This was associated with increased Fos-protein expression in the lateral hypothalamus (LH). Direct stimulation of the PVNCRH inputs in the LH produced a similar pattern of effects on sucrose motivation. Together, these data suggest that PVNCRH neuronal activity may be directly responsible for changes in motivational drive and that these behavioral changes may, in part, be driven by PVNCRH synaptic projections to the LH.
Collapse
Affiliation(s)
- Caitlin S Mitchell
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Erin J Campbell
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Simon D Fisher
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Laura M Stanton
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Nicholas J Burton
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Amy J Pearl
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Gavan P McNally
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Jaideep S Bains
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tamás Füzesi
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Brett A Graham
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia
| | - Elizabeth E Manning
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia.
| | - Christopher V Dayas
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, Sydney, NSW, 2305, Australia.
| |
Collapse
|