1
|
Jing Y, Zheng W, Zhou Z, Yao H, Zhang W, Wu Y, Guo Z, Huang C, Wang X. Recent research advances of c-fos in regulating cell senescence. Arch Biochem Biophys 2025; 769:110423. [PMID: 40246221 DOI: 10.1016/j.abb.2025.110423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/25/2024] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
c-fos is an immediate early gene (IEG) that forms a heterodimeric activator protein-1 (AP-1) complex with c-Jun. Following stimulation by a variety of factors, it changes the expression of genes and participates in cellular growth, proliferation, differentiation, and apoptosis. Previous studies have reported that c-fos is linked to cellular senescence and is involved in aging-related signaling pathways or damage repair processes. However, there are limited studies related to this topic. This review summarizes the findings of the connection between c-fos and cellular senescence, including the regulatory role of c-fos in the senescence of stem cells and various kinds of somatic cells. In addition, we discussed the involvement of c-fos in the cellular senescence process and related signaling pathways, as well as the importance of regulating DNA damage repair. The current studies have demonstrated that c-fos has important roles in inhibiting stem cell senescence. They can pave the way for a more thorough examination of the aging process and the regeneration of stem cells and provide new therapeutic strategies for aging-related diseases.
Collapse
Affiliation(s)
- Yuxin Jing
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Wei Zheng
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Zhou Zhou
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haiyang Yao
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenchuan Zhang
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yilun Wu
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zimo Guo
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenxuan Huang
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xianli Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
2
|
Arai M, Osanai H, Snell CC, Kitamura T, Ogawa SK. Combinative protein expression of immediate early genes c-Fos, Arc, and Npas4 along aversive- and reward-related neural networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.21.649441. [PMID: 40475597 PMCID: PMC12139746 DOI: 10.1101/2025.04.21.649441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2025]
Abstract
Expression of immediate early genes (IEGs) is critical for memory formation and has been widely used to identify the neural substrate of memory traces, termed memory engram cells. Functions of IEGs have been known to be different depending on their types. However, there is limited knowledge about the extent to which different types of IEGs are selectively or concurrently involved in the formation of memory engram. To address this question, we investigated the combinative expression of c-Fos, Arc, and Npas4 proteins using immunohistochemistry following aversive and rewarding experiences across subregions in the prefrontal cortex (PFC), basolateral amygdala (BLA), hippocampal dentate gyrus (DG), and retrosplenial cortex (RSC). Using an automated cell detection algorithm, we found that expression patterns of c-Fos, Npas4, and Arc varied across different brain areas, with a higher increase of IEG expressing cells in the PFC and posterior BLA than in the DG. The combinative expression patterns, along with their learning-induced changes, also differed across brain areas; the co-expression of IEGs increased in the PFC and BLA following learning whereas the increase was less pronounced in the DG and RSC. Furthermore, we demonstrate that different area-to-area functional connectivity networks were extracted by different IEGs. These findings provide insights into how different IEGs and their combinations identify engram cells, which will contribute to a deeper understanding of the functional significance of IEG-tagged memory engram cells.
Collapse
Affiliation(s)
- Mary Arai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hisayuki Osanai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Chris C. Snell
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sachie K. Ogawa
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
3
|
Usategui-Martín R, Esteban-López V, Chantre-Fortes E, Sánchez-Martín M, Riancho JA, López DE, González-Sarmiento R. The p.R321C mutation in the p62 protein is associated with abnormalities in the central nervous system. Sci Rep 2025; 15:16929. [PMID: 40374720 PMCID: PMC12081919 DOI: 10.1038/s41598-025-00764-2] [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: 11/18/2024] [Accepted: 04/30/2025] [Indexed: 05/17/2025] Open
Abstract
SQSTM1/p62 has an essential role in autophagy, a catabolic pathway that is vital for maintaining cell homeostasis. p62 alterations have been observed in multiple pathological conditions, including neurodegenerative diseases and bone metabolism alterations. The p.R321C p62 protein mutation has been described in patients with amyotrophic lateral sclerosis, frontotemporal lobar degeneration, and Paget's disease of bone. In vitro studies associated the p62-321C variant with a blockade of autophagy and with the activation of the NF-kB pathway. We aimed to provide a deeper understating of the pathophysiological consequences of the p.R321C p62 mutation using a humanized mouse model. Micro-computed tomography, immunohistochemistry, and western blot analysis studied the functional consequences of the p. R321C p62 mutation. Statistical analyses were performed using SPSS software. The results showed that the p62-321C mice developed seizures after tactile-vestibular stimulation, probably associated with a blockage of the autophagy and NF-kB activation. Changes in expression of cFos and p62 were found in the amygdala, hypothalamic nuclei, and hippocampi nuclei. In addition, numerous degenerating motor neurons were observed in the spinal cord of the p62-321C mice. We report that the blockage of the autophagy, caused by p.R321C p62 mutation, is associated with abnormalities in the central nervous system, mainly seizures after tactile-vestibular stimulation and degeneration of the motor neurons of the spinal cord but not with bone abnormalities in a humanized mouse model.
Collapse
Affiliation(s)
- Ricardo Usategui-Martín
- Molecular Medicine Unit, Department of Medicine, Faculty of Medicine, University of Salamanca, Campus Miguel Unamuno, 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.
| | - Vega Esteban-López
- Institute of Neuroscience of Castilla y León (INCYL), University of Salamanca, 37007, Salamanca, Spain
| | - Estefanía Chantre-Fortes
- Institute of Neuroscience of Castilla y León (INCYL), University of Salamanca, 37007, Salamanca, Spain
| | - Manuel Sánchez-Martín
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
- Transgenic Facility, University of Salamanca, 37007, Salamanca, Spain
| | - José A Riancho
- Department of Medicine and Psychiatry, Faculty of Medicine, University of Cantabria, IDIVAL, 39011, Santander, Spain
- Internal Medicine Department, Marqués de Valdecilla University Hospital, 39008, Santander, Spain
| | - Dolores E López
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
- Institute of Neuroscience of Castilla y León (INCYL), University of Salamanca, 37007, Salamanca, Spain
- Department of Cell Biology and Pathology, Faculty of Medicine, University of Salamanca, 37007, Salamanca, Spain
| | - Rogelio González-Sarmiento
- Molecular Medicine Unit, Department of Medicine, Faculty of Medicine, University of Salamanca, Campus Miguel Unamuno, 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007, Salamanca, Spain.
| |
Collapse
|
4
|
Hanchate NK. Single-cell genomics meets systems neuroscience: Insights from mapping the brain circuitry of stress. J Neuroendocrinol 2025; 37:e70005. [PMID: 39956535 PMCID: PMC12045673 DOI: 10.1111/jne.70005] [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: 05/08/2024] [Revised: 12/26/2024] [Accepted: 02/03/2025] [Indexed: 02/18/2025]
Abstract
Responses to external and internal dangers is essential for survival and homeostatic regulation. Hypothalamic corticotropin-releasing hormone neurons (CRHNs) play a pivotal role in regulating neuroendocrine responses to fear and stress. In recent years, the application of neurogenetic tools, such as fiber photometry, chemogenetics and optogenetics, have provided new insights into the dynamic neuronal responses of CRHNs during stressful events, offering new perspectives into their functional significance in mediating neurobehavioural responses to stress. Transsynaptic viral tracers have facilitated the comprehensive mapping of neuronal inputs to CRHNs. Furthermore, the development and application of innovative single-cell genomic tools combined with viral tracing have begun to pave the way for a deeper understanding of the transcriptional profiles of neural circuit components, enabling molecular-anatomical circuit mapping. Here, I will discuss how these systems neuroscience approaches and novel single-cell genomic methods are advancing the molecular and functional mapping of stress neurocircuits, their associated challenges and future directions.
Collapse
Affiliation(s)
- Naresh K. Hanchate
- Genetics & Genomic Medicine DepartmentUCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
| |
Collapse
|
5
|
Norman AO, Farooq N, Sahni A, Tapia K, Breiner D, Razak KA, Ethell IM. Differential effects of sound repetition rate on auditory cortex development and behavior in fragile X syndrome mouse model. Exp Neurol 2025; 387:115184. [PMID: 39961384 DOI: 10.1016/j.expneurol.2025.115184] [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: 10/29/2024] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
Fragile X syndrome (FXS) is a leading genetic form of autism and intellectual disability that is associated with a loss-of-function mutation in the Fragile X messenger ribonucleoprotein 1 (Fmr1) gene. The Fmr1 knockout (KO) mouse model displays many aspects of FXS-related phenotypes and is used to study FXS pathophysiology. Sensory manipulations, such as sound exposure, are considered as a non-invasive approach to alleviate FXS phenotypes. However, it is unclear what specific sound attributes may have beneficial effects. In this study, we examined the effects of sound repetition rate on auditory cortex development and FXS-associated behaviors in a mouse model of FXS. KO and wild-type (WT) male littermates were exposed to 14 kHz pure tone trains with 1 Hz or 5 Hz repetition rates during postnatal day (P)9-P21 developmental period. We analyzed the effects of developmental sound exposure on PV cell development, cortical activity and exploratory behaviors in sound-exposed WT and KO mice. We found that parvalbumin (PV) cell density was lower in the auditory cortex (AuC) of KO compared to WT mice raised in sound-attenuated environment, but was increased following the exposure to both 1 Hz and 5 Hz sound trains. However, PV protein levels were upregulated only in AuC of 5 Hz rate exposed KO mice. Interestingly, analysis of baseline cortical activity using electroencephalography (EEG) recordings showed that sound attenuation or exposure to sound trains with 5 Hz, but not 1 Hz, repetition rates corrected enhanced resting state gamma power in AuC of KO mice to WT levels. In addition, sound attenuation and exposure to 5 Hz showed some beneficial effects on the synchronization to frequency-modulated chirp in the frontal cortex (FC) of both WT and KO mice. Analysis of event-related potentials (ERP) in response to broadband sound showed increased ongoing responses and decreased habituation to noise stimuli in the AuC and FC of naive KO mice. While sound-attenuation and exposure to 5 Hz showed no significant effects on the power of onset and ongoing responses, exposure to 1 Hz further enhanced ongoing responses and decreased habituation to sound in both WT and KO mice. Finally, developmental exposure to sound trains with 5 Hz, but not 1 Hz, repetition rates normalized exploratory behaviors and improved social novelty preference but not hyperactivity in KO mice. Summarizing, our results show that developmental exposure of mice to sound trains with 5 Hz, but not 1 Hz, repetition rate had beneficial effects on PV cell development, overall cortical activity and behaviors in KO mice. While sound attenuation alone normalized some EEG phenotypes, it did not improve PV development or behaviors. These findings may have a significant impact on developing new approaches to alleviate FXS phenotypes and open possibilities for a combination of sound exposure with drug treatment which may offer highly novel therapeutic approaches.
Collapse
Affiliation(s)
- A O Norman
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - N Farooq
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - A Sahni
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - K Tapia
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - D Breiner
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - K A Razak
- Graduate Neuroscience Program, University of California Riverside, Riverside, CA, USA; Department of Psychology, University of California Riverside, Riverside, CA, USA
| | - I M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Graduate Neuroscience Program, University of California Riverside, Riverside, CA, USA.
| |
Collapse
|
6
|
Singh A, Singh L, Dalal D. Neuroprotective potential of hispidulin and diosmin: a review of molecular mechanisms. Metab Brain Dis 2025; 40:188. [PMID: 40257619 DOI: 10.1007/s11011-025-01615-9] [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: 07/01/2024] [Accepted: 04/11/2025] [Indexed: 04/22/2025]
Abstract
Flavonoids are an important class of natural products, particularly, belong to a class of plant secondary metabolites having a polyphenolic structure. They are widely found in fruits, vegetables, and certain beverages. Hispidulin and diosmin are naturally occurring flavonoids recognized for their potential health benefits, such as antioxidant, anti-inflammatory, and neuroprotective properties. Hispidulin is present in several plants, including Arnica montana, Salvia officinalis (sage), and Eupatorium arnottianum. Diosmin is mainly extracted from citrus fruits like lemons and oranges and can also be synthesized from hesperidin, another flavonoid found in citrus fruits. Neurodegenerative diseases are characterized by complex signaling pathways that contribute to neuronal deterioration. The JAK/STAT pathway is involved in inflammatory responses, while the NF-κB/NLRP3 pathway is associated with metabolic stress and inflammation, both facilitating neurodegeneration. Conversely, the AMPK/pGSK3β pathway is crucial for neuroprotection, regulating cellular responses to oxidative stress and promoting neuronal survival. Additionally, the BACE/Aβ pathway exacerbates neuronal damage by triggering inflammatory and oxidative stress responses, highlighting critical targets for therapeutic strategies. Hispidulin and diosmin have emerged as promising agents in the modulation of mediators involved in neuroinflammation and neurodegenerative diseases. Oxidative stress and inflammatory pathways, including those driven by Aβ/BACE1 and JAK/STAT signaling, are central to neuronal damage and disease progression. Recent studies highlight that hispidulin and diosmin exhibit notable neuroprotective effects by targeting these mediators. Hispidulin has been shown to impact key inflammatory cytokines and adhesion molecules, while diosmin influences proinflammatory cytokine production and inflammasome activation. Both compounds offer potential therapeutic benefits by modulating crucial mediators linked to neuroinflammation and neurodegeneration. This review article is designed to explore the intricate mechanistic interplay underlying the neuroprotective effects of hispidulin and diosmin.
Collapse
Affiliation(s)
- Anish Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Diksha Dalal
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
7
|
Dogra D, Phan VA, Zhang S, Gavrilovici C, DiMarzo N, Narang A, Ibhazehiebo K, Kurrasch DM. Modulation of NMDA receptor signaling and zinc chelation prevent seizure-like events in a zebrafish model of SLC13A5 epilepsy. PLoS Biol 2025; 23:e3002499. [PMID: 40208862 PMCID: PMC12047791 DOI: 10.1371/journal.pbio.3002499] [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: 01/03/2024] [Revised: 05/02/2025] [Accepted: 02/26/2025] [Indexed: 04/12/2025] Open
Abstract
SLC13A5 encodes a citrate transporter highly expressed in the brain and is important for regulating intra- and extracellular citrate levels. Mutations in this gene cause rare infantile epilepsy characterized by lifelong seizures, developmental delays, behavioral deficits, poor motor progression, and language impairments. SLC13A5 individuals respond poorly to treatment options; yet drug discovery programs are limited due to a paucity of animal models that phenocopy human symptoms. Here, we used CRISPR/Cas9 to create loss-of-function mutations in slc13a5a and slc13a5b, the zebrafish paralogs to human SLC13A5. slc13a5 mutant larvae showed cognitive dysfunction and sleep disturbances, consistent with SLC13A5 individuals. These mutants also exhibited fewer neurons and a concomitant increase in apoptosis across the optic tectum, a region important for sensory processing. Further, slc13a5 mutants displayed hallmark features of epilepsy, including an imbalance in glutamatergic and GABAergic excitatory-inhibitory gene expression, increased fosab expression, disrupted neurometabolism, and neuronal hyperexcitation as measured in vivo by extracellular field recordings and live calcium imaging. Mechanistically, we tested the involvement of NMDA signaling and zinc chelation in slc13a5 mutant epilepsy-like phenotypes. Slc13a5 protein co-localizes with excitatory NMDA receptors in wild-type zebrafish and NMDA receptor expression is upregulated in the brain of slc13a5 mutant larvae. Additionally, low levels of zinc are found in the plasma membrane of slc13a5 mutants. NMDA receptor suppression and ZnCl2 treatment in slc13a5 mutant larvae rescued neurometabolic and hyperexcitable calcium events, as well as behavioral defects. These data provide empirical evidence in support of the hypothesis that excess extracellular citrate over-chelates the zinc ions needed to regulate NMDA receptor function, leading to sustained channel opening and an exaggerated excitatory response that manifests as seizures. These data show the utility of slc13a5 mutant zebrafish for studying SLC13A5 epilepsy and open new avenues for drug discovery.
Collapse
Affiliation(s)
- Deepika Dogra
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Van Anh Phan
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sinan Zhang
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Cezar Gavrilovici
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Departments of Pediatrics, Clinical Neurosciences, Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nadia DiMarzo
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ankita Narang
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Kingsley Ibhazehiebo
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Deborah M. Kurrasch
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
8
|
Sarafinovska S, Koester SK, Fang LZ, Thorpe JW, Chaturvedi SM, Ji J, Jones EF, Selmanovic D, Kornbluth DJ, Barrett MR, Rurak GM, Maloney SE, Creed MC, Mitra RD, Dougherty JD. Single-Cell Resolution of Individual Variation in Hypothalamic Neurons Allows Targeted Manipulation Affecting Social Motivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642464. [PMID: 40161710 PMCID: PMC11952468 DOI: 10.1101/2025.03.10.642464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Despite decades of research, connecting molecular and cellular phenotypes to complex behavioral traits remains an elusive goal1. Social motivation exhibits individual trait variation2, which we hypothesize is mediated by molecular and cellular variability across hypothalamic neurons. To test this, we generated single-nucleus RNA-sequencing profiles3,4 of >120,000 neurons from tuberal hypothalamus and adjacent thalamus in 36 mice, balanced across sex and autism-associated mutation5, with all mice assessed for social motivation2. First, we show that molecular activation patterns predict behavior across individuals: specifically, activation of paraventricular Agtr1a+ (angiotensin receptor 1a) neurons predicted reduced social behavior. Subsequent inhibition of AGTR1A with telmisartan-an FDA-approved antihypertensive6-improved social orienting. Second, we show natural variation in neuronal proportions-likely arising from stochastic developmental events7-is sufficient to shape adult behavior even among genetically-identical individuals: we identified multiple neuronal populations whose relative abundance predicted social reward-seeking behavior. Chemogenetic inhibition of one such population, Nxph4+ neurons of the postero-lateral hypothalamus8, suppressed multiple aspects of social motivation. This work establishes proof-of-principle for an approach where single-cell genomics precisely maps neural substrates governing behavior. This approach revealed that stochastic variations in neuronal architecture deterministically influence social motivation, and enabled identification of therapeutically-actionable targets with immediate translational potential for disorders with social deficits.
Collapse
Affiliation(s)
- S Sarafinovska
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - S K Koester
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - L Z Fang
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA
| | - J W Thorpe
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - S M Chaturvedi
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - J Ji
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - E F Jones
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - D Selmanovic
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - D J Kornbluth
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - M R Barrett
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110-1093, USA
| | - G M Rurak
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - S E Maloney
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110-1093, USA
| | - M C Creed
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA
| | - R D Mitra
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - J D Dougherty
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110-1093, USA
| |
Collapse
|
9
|
Osanai H, Arai M, Kitamura T, Ogawa SK. Automated detection of c-Fos-expressing neurons using inhomogeneous background subtraction in fluorescent images. Neurobiol Learn Mem 2025; 218:108035. [PMID: 39986434 DOI: 10.1016/j.nlm.2025.108035] [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: 11/11/2024] [Revised: 02/04/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Although many methods for automated fluorescent-labeled cell detection have been proposed, not all of them assume a highly inhomogeneous background arising from complex biological structures. Here, we propose an automated cell detection algorithm that accounts for and subtracts the inhomogeneous background by avoiding high-intensity pixels in the blur filtering calculation. Cells were detected by intensity thresholding in the background-subtracted image, and the algorithm's performance was tested on NeuN- and c-Fos-stained images in the mouse prefrontal cortex and hippocampal dentate gyrus. In addition, applications in c-Fos positive cell counting and the quantification for the expression level in double-labeled cells were demonstrated. Our method of automated detection after background assumption (ADABA) offers the advantage of high-throughput and unbiased analysis in regions with complex biological structures that produce inhomogeneous background.
Collapse
Affiliation(s)
- Hisayuki Osanai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Mary Arai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Sachie K Ogawa
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
10
|
Marin-Rodero M, Cintado E, Walker AJ, Jayewickreme T, Pinho-Ribeiro FA, Richardson Q, Jackson R, Chiu IM, Benoist C, Stevens B, Trejo JL, Mathis D. The meninges host a distinct compartment of regulatory T cells that preserves brain homeostasis. Sci Immunol 2025; 10:eadu2910. [PMID: 39873623 PMCID: PMC11924117 DOI: 10.1126/sciimmunol.adu2910] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025]
Abstract
Our understanding of the meningeal immune system has recently burgeoned, particularly regarding how innate and adaptive effector cells are mobilized to meet brain challenges. However, information on how meningeal immunocytes guard brain homeostasis in healthy individuals remains limited. This study highlights the heterogeneous, polyfunctional regulatory T cell (Treg) compartment in the meninges. A Treg subtype specialized in controlling interferon-γ (IFN-γ) responses and another dedicated to regulating follicular B cell responses were substantial components of this compartment. Accordingly, punctual Treg ablation rapidly unleashed IFN-γ production by meningeal lymphocytes, unlocked access to the brain parenchyma, and altered meningeal B cell profiles. Distally, the hippocampus assumed a reactive state, with morphological and transcriptional changes in multiple glial cell types. Within the dentate gyrus, neural stem cells underwent more death and were blocked from further differentiation, which coincided with impairments in short-term spatial-reference memory. Thus, meningeal Tregs are a multifaceted safeguard of brain homeostasis at steady state.
Collapse
Affiliation(s)
| | - Elisa Cintado
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas; Madrid, Spain
| | - Alec J. Walker
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | | | | | | | - Ruaidhrí Jackson
- Department of Immunology, Harvard Medical School; Boston, MA, USA
| | - Isaac M. Chiu
- Department of Immunology, Harvard Medical School; Boston, MA, USA
| | | | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital; Boston, MA, USA
| | - José Luís Trejo
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas; Madrid, Spain
| | - Diane Mathis
- Department of Immunology, Harvard Medical School; Boston, MA, USA
| |
Collapse
|
11
|
Bonauto SM, Patel KA, Honeycutt JA. 22 and 50 kHz rat ultrasonic vocalization playback reveals sex differences in behavior and cFos in brain regions associated with affective processing. Behav Brain Res 2025; 478:115326. [PMID: 39521142 PMCID: PMC11610767 DOI: 10.1016/j.bbr.2024.115326] [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: 08/23/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Adult rats communicate using ultrasonic vocalization (USV) frequencies indicating negative (22 kHz) or positive (50 kHz) affective states. Playback of USVs can serve as an ethologically translational method to study affective processing in response to socially communicated states. However, few studies have examined behavioral and neural effects of USV playback in both male and female rats. Here, adult male and female Sprague-Dawley rats experienced a 20-min open field test (OFT) with either silence, 22 kHz, or 50 kHz recorded USV playback. Center exploration and locomotor activity were analyzed to characterize sex differences in playback effects. Results suggest that females display greater sensitivity to frequency-specific effects of USV playback in this paradigm compared to males. 50 kHz USV playback evoked an immediate increase in center exploration and locomotor activity in females, indicating the appetitive nature of 50 kHz USVs. Initially, 22 kHz playback inhibited center exploration in the OFT compared to 50 kHz. However, females exhibited a switch in behavioral strategy in response to 22 kHz following prolonged playback. Following OFT, neural activity was quantified via the immediate early gene cFos. Results from cFos quantification showed sex- and region-specific differences in neural recruitment in areas of the brain associated with affective processing, including the prefrontal cortex, amygdala, bed nucleus of the stria terminalis, and nucleus accumbens. Taken together, this work provides a normative baseline for understanding how sex influences behavioral and neural responses to USV playback, which can be leveraged to study anxiety, communication, and affect in an ethologically relevant assay.
Collapse
Affiliation(s)
- Sydney M Bonauto
- Research in Affective and Translational Neuroscience Laboratory, Department of Psychology and Program in Neuroscience, Bowdoin College, Brunswick, ME 04011 USA, USA
| | - Kaya A Patel
- Research in Affective and Translational Neuroscience Laboratory, Department of Psychology and Program in Neuroscience, Bowdoin College, Brunswick, ME 04011 USA, USA
| | - Jennifer A Honeycutt
- Research in Affective and Translational Neuroscience Laboratory, Department of Psychology and Program in Neuroscience, Bowdoin College, Brunswick, ME 04011 USA, USA.
| |
Collapse
|
12
|
Aboharb F, Davoudian PA, Shao LX, Liao C, Rzepka GN, Wojtasiewicz C, Indajang J, Dibbs M, Rondeau J, Sherwood AM, Kaye AP, Kwan AC. Classification of psychedelics and psychoactive drugs based on brain-wide imaging of cellular c-Fos expression. Nat Commun 2025; 16:1590. [PMID: 39939591 PMCID: PMC11822132 DOI: 10.1038/s41467-025-56850-6] [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: 04/19/2024] [Accepted: 01/31/2025] [Indexed: 02/14/2025] Open
Abstract
Psilocybin, ketamine, and MDMA are psychoactive compounds that exert behavioral effects with distinguishable but also overlapping features. The growing interest in using these compounds as therapeutics necessitates preclinical assays that can accurately screen psychedelics and related analogs. We posit that a promising approach may be to measure drug action on markers of neural plasticity in native brain tissues. We therefore developed a pipeline for drug classification using light sheet fluorescence microscopy of immediate early gene expression at cellular resolution followed by machine learning. We tested male and female mice with a panel of drugs, including psilocybin, ketamine, 5-MeO-DMT, 6-fluoro-DET, MDMA, acute fluoxetine, chronic fluoxetine, and vehicle. In one-versus-rest classification, the exact drug was identified with 67% accuracy, significantly above the chance level of 12.5%. In one-versus-one classifications, psilocybin was discriminated from 5-MeO-DMT, ketamine, MDMA, or acute fluoxetine with >95% accuracy. We used Shapley additive explanation to pinpoint the brain regions driving the machine learning predictions. Our results suggest a unique approach for characterizing and validating psychoactive drugs with psychedelic properties.
Collapse
Affiliation(s)
- Farid Aboharb
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Weill Cornell Medicine/Rockefeller/Sloan-Kettering Tri-Institutional MD/PhD Program, New York, NY, USA
| | - Pasha A Davoudian
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT, USA
| | - Ling-Xiao Shao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Clara Liao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Gillian N Rzepka
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Jonathan Indajang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Mark Dibbs
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Jocelyne Rondeau
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | | | - Alfred P Kaye
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Clinical Neurosciences Division, VA National Center for PTSD, West Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Alex C Kwan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Duan X, Zhang C, Wu Y, Ju J, Xu Z, Li X, Liu Y, Ohdah S, Constantin OM, Pan Y, Lu Z, Wang C, Chen X, Gee CE, Nagel G, Hou ST, Gao S, Song K. Suppression of epileptic seizures by transcranial activation of K +-selective channelrhodopsin. Nat Commun 2025; 16:559. [PMID: 39789018 PMCID: PMC11718177 DOI: 10.1038/s41467-025-55818-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Optogenetics is a valuable tool for studying the mechanisms of neurological diseases and is now being developed for therapeutic applications. In rodents and macaques, improved channelrhodopsins have been applied to achieve transcranial optogenetic stimulation. While transcranial photoexcitation of neurons has been achieved, noninvasive optogenetic inhibition for treating hyperexcitability-induced neurological disorders has remained elusive. There is a critical need for effective inhibitory optogenetic tools that are highly light-sensitive and capable of suppressing neuronal activity in deep brain tissue. In this study, we developed a highly sensitive moderately K+-selective channelrhodopsin (HcKCR1-hs) by molecular engineering of the recently discovered Hyphochytrium catenoides kalium (potassium) channelrhodopsin 1. Transcranial activation of HcKCR1-hs significantly prolongs the time to the first seizure, increases survival, and decreases seizure activity in several status epilepticus mouse models. Our approach for transcranial optogenetic inhibition of neural hyperactivity may be adapted for cell type-specific neuromodulation in both basic and preclinical settings.
Collapse
Affiliation(s)
- Xiaodong Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Chong Zhang
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany
| | - Yujie Wu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jun Ju
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhe Xu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Xuanyi Li
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yao Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Schugofa Ohdah
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Oana M Constantin
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Yifan Pan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhonghua Lu
- Research Center for Primate Neuromodulation and Neuroimaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cheng Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaojing Chen
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Christine E Gee
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Georg Nagel
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany
| | - Sheng-Tao Hou
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Shiqiang Gao
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany.
| | - Kun Song
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
14
|
Li Z, Lu W, Yang L, Lai N, Wang Y, Chen Z. Decade of TRAP progress: Insights and future prospects for advancing functional network research in epilepsy. Prog Neurobiol 2025; 244:102707. [PMID: 39725016 DOI: 10.1016/j.pneurobio.2024.102707] [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: 08/01/2024] [Revised: 11/30/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Targeted Recombination in Active Populations (TRAP) represents an effective and extensively applied technique that has earned significant utilization in neuroscience over the past decade, primarily for identifying and modulating functionally activated neuronal ensembles associated with diverse behaviors. As epilepsy is a neurological disorder characterized by pathological hyper-excitatory networks, TRAP has already been widely applied in epilepsy research. However, the deployment of TRAP in this field remains underexplored, and there is significant potential for further application and development in epilepsy-related investigations. In this review, we embark on a concise examination of the mechanisms behind several TRAP tools, introduce the current applications of TRAP in epilepsy research, and collate the key advantages as well as limitations of TRAP. Furthermore, we sketch out perspectives on potential applications of TRAP in future epilepsy research, grounded in the present landscape and challenges of the field, as well as the ways TRAP has been embraced in other neuroscience domains.
Collapse
Affiliation(s)
- Zhisheng Li
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wangjialu Lu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Yang
- key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Nanxi Lai
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China; key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China; key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
15
|
Szczepankiewicz AA, Parobczak K, Zaręba-Kozioł M, Ruszczycki B, Bijata M, Trzaskoma P, Hajnowski G, Holm-Kaczmarek D, Włodarczyk J, Sas-Nowosielska H, Wilczyński GM, Rędowicz MJ, Magalska A. Neuronal activation affects the organization and protein composition of the nuclear speckles. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119829. [PMID: 39197592 DOI: 10.1016/j.bbamcr.2024.119829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Nuclear speckles, also known as interchromatin granule clusters (IGCs), are subnuclear domains highly enriched in proteins involved in transcription and mRNA metabolism and, until recently, have been regarded primarily as their storage and modification hubs. However, several recent studies on non-neuronal cell types indicate that nuclear speckles may directly contribute to gene expression as some of the active genes have been shown to associate with these structures. Neuronal activity is one of the key transcriptional regulators and may lead to the rearrangement of some nuclear bodies. Notably, the impact of neuronal activation on IGC/nuclear speckles organization and function remains unexplored. To address this research gap, we examined whether and how neuronal stimulation affects the organization of these bodies in granular neurons from the rat hippocampal formation. Our findings demonstrate that neuronal stimulation induces morphological and proteomic remodelling of the nuclear speckles under both in vitro and in vivo conditions. Importantly, these changes are not associated with cellular stress or cell death but are dependent on transcription and splicing.
Collapse
Affiliation(s)
- Andrzej Antoni Szczepankiewicz
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Kamil Parobczak
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Monika Zaręba-Kozioł
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Błażej Ruszczycki
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; AGH University of Krakow, Faculty of Physics and Applied Computer Science, Department of Medical Physics and Biophysics, al. A. Mickiewicza 30, 30-059 Krakow, Poland
| | - Monika Bijata
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Paweł Trzaskoma
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Grzegorz Hajnowski
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Dagmara Holm-Kaczmarek
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Jakub Włodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Hanna Sas-Nowosielska
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Grzegorz Marek Wilczyński
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Adriana Magalska
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| |
Collapse
|
16
|
Osanai H, Arai M, Kitamura T, Ogawa SK. Automated cell detection for immediate early gene-expressing neurons using inhomogeneous background subtraction in fluorescent images. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622525. [PMID: 39574706 PMCID: PMC11580981 DOI: 10.1101/2024.11.07.622525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Although many methods for automated fluorescent-labeled cell detection have been proposed, not all of them assume a highly inhomogeneous background arising from complex biological structures. Here, we propose an automated cell detection algorithm that accounts for and subtracts the inhomogeneous background by avoiding high-intensity pixels in the blur filtering calculation. Cells were detected by intensity thresholding in the background-subtracted image, and the algorithm's performance was tested on NeuN- and c-Fos-stained images in the mouse prefrontal cortex and hippocampal dentate gyrus. In addition, applications in c-Fos positive cell counting and the quantification for the expression level in double-labeled cells were demonstrated. Our method of automated detection after background assumption (ADABA) offers the advantage of high-throughput and unbiased analysis in regions with complex biological structures that produce inhomogeneous background. Highlights - We proposed a method to assume and subtract inhomogeneous background pattern. (79/85) - Cells were automatically detected in the background-subtracted image. (71/85) - The automated detection results corresponded with the manual detection. (73/85) - Detection of IEG positive cells and overlapping with neural marker were demonstrated. (85/85).
Collapse
|
17
|
Shiromani PJ. Searching for sleep in all the right places: My career in sleep research. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2024; 5:zpae055. [PMID: 39211349 PMCID: PMC11359167 DOI: 10.1093/sleepadvances/zpae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Indexed: 09/04/2024]
Abstract
My research has always focused on sleep, whether monitoring neural activity (microwires, c-Fos, calcium imaging), triggering it with optogenetics or pharmacologically (anandamide, cholinergic agonists), or measuring levels of endogenous sleep agents such as adenosine. A recurring theme of my research is to use new tools to find the sweet spot in the brain where the signal to sleep begins. My goal is to identify the circuit, determine whether it degrades with age or disease, and repair the circuit when it fails. I am deeply grateful to my mentors for introducing me to the science of sleep, to my students and colleagues for helping me in my quest, and to the NIH and VA Research for supporting the research. Because of the collective efforts of sleep researchers, the public is more aware of the importance of sleep to a healthy lifestyle.
Collapse
Affiliation(s)
- Priyattam J Shiromani
- Laboratory of Sleep Medicine and Chronobiology, Ralph H. Johnson Veterans Healthcare System, Charleston, SC, USA
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
18
|
Barbosa-Méndez S, Salazar-Juárez A. Mirtazapine decreased cocaine-induced c-fos expression and dopamine release in rats. Front Psychiatry 2024; 15:1428730. [PMID: 39188520 PMCID: PMC11346032 DOI: 10.3389/fpsyt.2024.1428730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Introduction Chronic cocaine exposure induces an increase in dopamine release and an increase in the expression of the Fos protein in the rat striatum. It has been suggested that both are necessary for the expression of cocaine-induced alterations in behavior and neural circuitry. Mirtazapine dosing attenuated the cocaine-induced psychomotor and reinforcer effects. Methods The study evaluates the effect of chronic dosing of mirtazapine on cocaine-induced extracellular dopamine levels and Fos protein expression in rats. Male Wistar rats received cocaine (10 mg/Kg; i.p.) during the induction and expression of locomotor sensitization. The mirtazapine (30 mg/Kg; MIR), was administered 30 minutes before cocaine during the cocaine withdrawal. After each treatment, the locomotor activity was recorded for 30 minutes. Animals were sacrificed after treatment administration. Dopamine levels were determined by high-performance liquid chromatographic (HPLC) in the ventral striatum, the prefrontal cortex (PFC), and the ventral tegmental area (VTA) in animals treated with mirtazapine and cocaine. The quantification of c-fos immunoreactive cells was carried out by stereology analysis. Results Mirtazapine generated a decrease in cocaine-induced locomotor activity. In addition, mirtazapine decreased the amount of cocaine-induced dopamine and the number of cells immunoreactive to the Fos protein in the striatum, PFC, and VTA. Discussion These data suggest that mirtazapine could prevent the consolidation of changes in behavior and the cocaine-induced reorganization of neuronal circuits. It would explain the mirtazapine-induced effects on cocaine behavioral sensitization. Thus, these data together could support its possible use for the treatment of patients with cocaine use disorder.
Collapse
|
19
|
Mojahed N, Adjei M, Qasem E, Aaflaq S, Adu T, Jacobs JT, Richardson BD, Nordman JC. Acute social defeat during adolescence promotes long-lasting aggression through activation of the medial amygdala. Front Neurosci 2024; 18:1433993. [PMID: 39050664 PMCID: PMC11266103 DOI: 10.3389/fnins.2024.1433993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
Traumatic stress, particularly during critical developmental periods such as adolescence, has been strongly linked to an increased propensity and severity of aggression. Existing literature underscores that being a victim of abuse can exacerbate aggressive behaviors, with the amygdala playing a pivotal role in mediating these effects. Historically, animal models have demonstrated that traumatic stressors can increase attack behavior, implicating various amygdala nuclei. Building on this foundation, our previous work has highlighted how traumatic stress invokes long-lasting aggression via an excitatory pathway within the posterior ventral medial amygdala (MeApv). In the current study, we sought to further delineate this mechanism by examining the effects of acute social defeat during adolescence on aggressive behaviors and neural activation in mice. Using a common social defeat paradigm, we first established that acute social defeat during late adolescence indeed promotes long-lasting aggression, measured as attack behavior 7 days after the defeat session. Immunolabeling with c-Fos demonstrated that acute social defeat activates the MeApv and ventrolateral aspect of the ventromedial hypothalamus (VmHvl), consistent with our previous studies that used foot shock as an acute stressor. Finally, chemogenetically inhibiting excitatory MeApv neurons during social defeat significantly mitigated the aggression increase without affecting non-aggressive social behavior. These results strongly suggest that the MeApv plays a critical role in the onset of aggression following traumatic social experience, and offer the MeA as a potential target for therapeutic interventions.
Collapse
Affiliation(s)
- Nooshin Mojahed
- Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Magdalene Adjei
- Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Elana Qasem
- Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Sophia Aaflaq
- Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Temitope Adu
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Jessica T. Jacobs
- Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Ben D. Richardson
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Jacob C. Nordman
- Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| |
Collapse
|
20
|
Dong P, Bakhurin K, Li Y, Mikati MA, Cui J, Grill WM, Yin HH, Yang H. Attenuating midline thalamus bursting to mitigate absence epilepsy. Proc Natl Acad Sci U S A 2024; 121:e2403763121. [PMID: 38968111 PMCID: PMC11252967 DOI: 10.1073/pnas.2403763121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/31/2024] [Indexed: 07/07/2024] Open
Abstract
Advancing the mechanistic understanding of absence epilepsy is crucial for developing new therapeutics, especially for patients unresponsive to current treatments. Utilizing a recently developed mouse model of absence epilepsy carrying the BK gain-of-function channelopathy D434G, here we report that attenuating the burst firing of midline thalamus (MLT) neurons effectively prevents absence seizures. We found that enhanced BK channel activity in the BK-D434G MLT neurons promotes synchronized bursting during the ictal phase of absence seizures. Modulating MLT neurons through pharmacological reagents, optogenetic stimulation, or deep brain stimulation effectively attenuates burst firing, leading to reduced absence seizure frequency and increased vigilance. Additionally, enhancing vigilance by amphetamine, a stimulant medication, or physical perturbation also effectively suppresses MLT bursting and prevents absence seizures. These findings suggest that the MLT is a promising target for clinical interventions. Our diverse approaches offer valuable insights for developing next generation therapeutics to treat absence epilepsy.
Collapse
Affiliation(s)
- Ping Dong
- Department of Biochemistry, Duke University Medical Center, Durham, NC27710
| | | | - Yuhui Li
- Department of Biomedical Engineering, Duke University, Durham, NC27708
| | - Mohamad A. Mikati
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
- Department of Pediatrics, Duke University Medical Center, Durham, NC27710
| | - Jianmin Cui
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO63130
| | - Warren M. Grill
- Department of Biomedical Engineering, Duke University, Durham, NC27708
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
- Department of Neurosurgery, Duke University Medical Center, Durham, NC27710
| | - Henry H. Yin
- Department of Psychology and Neuroscience, Duke University, Durham, NC27708
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical Center, Durham, NC27710
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
| |
Collapse
|
21
|
Ben Musa R, Cornelius-Green J, Zhang H, Li DP, Kline DD, Hasser EM, Cummings KJ. Orexin Facilitates the Peripheral Chemoreflex via Corticotropin-Releasing Hormone Neurons Projecting to the Nucleus of the Solitary Tract. J Neurosci 2024; 44:e2383232024. [PMID: 38789262 PMCID: PMC11223477 DOI: 10.1523/jneurosci.2383-23.2024] [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: 01/02/2024] [Revised: 04/14/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
We previously showed that orexin neurons are activated by hypoxia and facilitate the peripheral chemoreflex (PCR)-mediated hypoxic ventilatory response (HVR), mostly by promoting the respiratory frequency response. Orexin neurons project to the nucleus of the solitary tract (nTS) and the paraventricular nucleus of the hypothalamus (PVN). The PVN contributes significantly to the PCR and contains nTS-projecting corticotropin-releasing hormone (CRH) neurons. We hypothesized that in male rats, orexin neurons contribute to the PCR by activating nTS-projecting CRH neurons. We used neuronal tract tracing and immunohistochemistry (IHC) to quantify the degree that hypoxia activates PVN-projecting orexin neurons. We coupled this with orexin receptor (OxR) blockade with suvorexant (Suvo, 20 mg/kg, i.p.) to assess the degree that orexin facilitates the hypoxia-induced activation of CRH neurons in the PVN, including those projecting to the nTS. In separate groups of rats, we measured the PCR following systemic orexin 1 receptor (Ox1R) blockade (SB-334867; 1 mg/kg) and specific Ox1R knockdown in PVN. OxR blockade with Suvo reduced the number of nTS and PVN neurons activated by hypoxia, including those CRH neurons projecting to nTS. Hypoxia increased the number of activated PVN-projecting orexin neurons but had no effect on the number of activated nTS-projecting orexin neurons. Global Ox1R blockade and partial Ox1R knockdown in the PVN significantly reduced the PCR. Ox1R knockdown also reduced the number of activated PVN neurons and the number of activated tyrosine hydroxylase neurons in the nTS. Our findings suggest orexin facilitates the PCR via nTS-projecting CRH neurons expressing Ox1R.
Collapse
Affiliation(s)
- Ruwaida Ben Musa
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Jennifer Cornelius-Green
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Hua Zhang
- Department of Medicine, Center for Precision Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65212
| | - De-Pei Li
- Department of Medicine, Center for Precision Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65212
| | - David D Kline
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Eileen M Hasser
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Kevin J Cummings
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| |
Collapse
|
22
|
Marin-Rodero M, Reyes EC, Walker AJ, Jayewickreme T, Pinho-Ribeiro FA, Richardson Q, Jackson R, Chiu IM, Benoist C, Stevens B, Trejo JL, Mathis D. The meninges host a unique compartment of regulatory T cells that bulwarks adult hippocampal neurogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599387. [PMID: 38948783 PMCID: PMC11212874 DOI: 10.1101/2024.06.17.599387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Our knowledge about the meningeal immune system has recently burgeoned, particularly our understanding of how innate and adaptive effector cells are mobilized to meet brain challenges. However, information on how meningeal immunocytes guard brain homeostasis in healthy individuals remains sparse. This study highlights the heterogeneous and polyfunctional regulatory-T (Treg) cell compartment in the meninges. A Treg subtype specialized in controlling Th1-cell responses and another known to control responses in B-cell follicles were substantial components of this compartment, foretelling that punctual Treg-cell ablation rapidly unleashed interferon-gamma production by meningeal lymphocytes, unlocked their access to the brain parenchyma, and altered meningeal B-cell profiles. Distally, the hippocampus assumed a reactive state, with morphological and transcriptional changes in multiple glial-cell types; within the dentate gyrus, neural stem cells showed exacerbated death and desisted from further differentiation, associated with inhibition of spatial-reference memory. Thus, meningeal Treg cells are a multifaceted bulwark to brain homeostasis at steady-state. One sentence summary A distinct population of regulatory T cells in the murine meninges safeguards homeostasis by keeping local interferon-γ-producing lymphocytes in check, thereby preventing their invasion of the parenchyma, activation of hippocampal glial cells, death of neural stem cells, and memory decay.
Collapse
|
23
|
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
|
24
|
Mori D, Inami C, Ikeda R, Sawahata M, Urata S, Yamaguchi ST, Kobayashi Y, Fujita K, Arioka Y, Okumura H, Kushima I, Kodama A, Suzuki T, Hirao T, Yoshimi A, Sobue A, Ito T, Noda Y, Mizoguchi H, Nagai T, Kaibuchi K, Okabe S, Nishiguchi K, Kume K, Yamada K, Ozaki N. Mice with deficiency in Pcdh15, a gene associated with bipolar disorders, exhibit significantly elevated diurnal amplitudes of locomotion and body temperature. Transl Psychiatry 2024; 14:216. [PMID: 38806495 PMCID: PMC11133426 DOI: 10.1038/s41398-024-02952-6] [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: 12/29/2023] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 05/30/2024] Open
Abstract
Genetic factors significantly affect the pathogenesis of psychiatric disorders. However, the specific pathogenic mechanisms underlying these effects are not fully understood. Recent extensive genomic studies have implicated the protocadherin-related 15 (PCDH15) gene in the onset of psychiatric disorders, such as bipolar disorder (BD). To further investigate the pathogenesis of these psychiatric disorders, we developed a mouse model lacking Pcdh15. Notably, although PCDH15 is primarily identified as the causative gene of Usher syndrome, which presents with visual and auditory impairments, our mice with Pcdh15 homozygous deletion (Pcdh15-null) did not exhibit observable structural abnormalities in either the retina or the inner ear. The Pcdh15-null mice showed very high levels of spontaneous motor activity which was too disturbed to perform standard behavioral testing. However, the Pcdh15 heterozygous deletion mice (Pcdh15-het) exhibited enhanced spontaneous locomotor activity, reduced prepulse inhibition, and diminished cliff avoidance behavior. These observations agreed with the symptoms observed in patients with various psychiatric disorders and several mouse models of psychiatric diseases. Specifically, the hyperactivity may mirror the manic episodes in BD. To obtain a more physiological, long-term quantification of the hyperactive phenotype, we implanted nano tag® sensor chips in the animals, to enable the continuous monitoring of both activity and body temperature. During the light-off period, Pcdh15-null exhibited elevated activity and body temperature compared with wild-type (WT) mice. However, we observed a decreased body temperature during the light-on period. Comprehensive brain activity was visualized using c-Fos mapping, which was assessed during the activity and temperature peak and trough. There was a stark contrast between the distribution of c-Fos expression in Pcdh15-null and WT brains during both the light-on and light-off periods. These results provide valuable insights into the neural basis of the behavioral and thermal characteristics of Pcdh15-deletion mice. Therefore, Pcdh15-deletion mice can be a novel model for BD with mania and other psychiatric disorders, with a strong genetic component that satisfies both construct and surface validity.
Collapse
Affiliation(s)
- Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan.
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
| | - Chihiro Inami
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Ryosuke Ikeda
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahito Sawahata
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shinji Urata
- Department of Otolaryngology, Graduate School of Medicine, The University of Tokyo, Tokyo Pref., Japan
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, Tokyo Pref., Japan
| | - Sho T Yamaguchi
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | | | - Kosuke Fujita
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuko Arioka
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Hiroki Okumura
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Medical Genomics Center, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Akiko Kodama
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toshiaki Suzuki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Hirao
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Yoshimi
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty of Pharmacy, Nagoya, Aichi, Japan
| | - Akira Sobue
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Takahiro Ito
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty of Pharmacy, Nagoya, Aichi, Japan
| | - Yukikiro Noda
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty of Pharmacy, Nagoya, Aichi, Japan
| | - Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
- Division of Behavioral Neuropharmacology, International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, Tokyo Pref., Japan
| | - Koji Nishiguchi
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kazuhiko Kume
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Norio Ozaki
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Aichi, Japan
| |
Collapse
|
25
|
Vaughan AJ, McMeekin LJ, Hine K, Stubbs IW, Codadu NK, Cockell S, Hill JT, Cowell R, Trevelyan AJ, Parrish RR. RNA Sequencing Demonstrates Ex Vivo Neocortical Transcriptomic Changes Induced by Epileptiform Activity in Male and Female Mice. eNeuro 2024; 11:ENEURO.0520-23.2024. [PMID: 38664009 PMCID: PMC11129778 DOI: 10.1523/eneuro.0520-23.2024] [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: 11/29/2023] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
Seizures are generally associated with epilepsy but may also be a symptom of many other neurological conditions. A hallmark of a seizure is the intensity of the local neuronal activation, which can drive large-scale gene transcription changes. Such changes in the transcriptional profile likely alter neuronal function, thereby contributing to the pathological process. Therefore, there is a strong clinical imperative to characterize how gene expression is changed by seizure activity. To this end, we developed a simplified ex vivo technique for studying seizure-induced transcriptional changes. We compared the RNA sequencing profile in mouse neocortical tissue with up to 3 h of epileptiform activity induced by 4-aminopyridine (4AP) relative to control brain slices not exposed to the drug. We identified over 100 genes with significantly altered expression after 4AP treatment, including multiple genes involved in MAPK, TNF, and neuroinflammatory signaling pathways, all of which have been linked to epilepsy previously. Notably, the patterns in male and female brain slices were almost identical. Various immediate early genes were among those showing the largest upregulation. The set of down-regulated genes included ones that might be expected either to increase or to decrease neuronal excitability. In summary, we found the seizure-induced transcriptional profile complex, but the changes aligned well with an analysis of published epilepsy-associated genes. We discuss how simple models may provide new angles for investigating seizure-induced transcriptional changes.
Collapse
Affiliation(s)
- Alec J Vaughan
- Department of Cell Biology and Physiology, Brigham Young University, Provo, Utah 84602
| | - Laura J McMeekin
- Department of Neurology, University of Alabama, Birmingham, Birmingham, Alabama 35233
| | - Kutter Hine
- Department of Cell Biology and Physiology, Brigham Young University, Provo, Utah 84602
| | - Isaac W Stubbs
- Department of Cell Biology and Physiology, Brigham Young University, Provo, Utah 84602
| | - Neela K Codadu
- Newcastle University Biosciences Institute, Medical School, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Simon Cockell
- School of Biomedical, Nutritional and Sports Science, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Jonathon T Hill
- Department of Cell Biology and Physiology, Brigham Young University, Provo, Utah 84602
| | - Rita Cowell
- Department of Neurology, University of Alabama, Birmingham, Birmingham, Alabama 35233
| | - Andrew J Trevelyan
- Newcastle University Biosciences Institute, Medical School, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - R Ryley Parrish
- Department of Cell Biology and Physiology, Brigham Young University, Provo, Utah 84602
| |
Collapse
|
26
|
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
|
27
|
Tagawa N, Mori K, Koebis M, Aiba A, Iino Y, Tsuneoka Y, Funato H. Activation of lateral preoptic neurons is associated with nest-building in male mice. Sci Rep 2024; 14:8346. [PMID: 38594484 PMCID: PMC11004109 DOI: 10.1038/s41598-024-59061-z] [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: 12/26/2023] [Accepted: 04/06/2024] [Indexed: 04/11/2024] Open
Abstract
Nest-building behavior is a widely observed innate behavior. A nest provides animals with a secure environment for parenting, sleep, feeding, reproduction, and temperature maintenance. Since animal infants spend their time in a nest, nest-building behavior has been generally studied as parental behaviors, and the medial preoptic area (MPOA) neurons are known to be involved in parental nest-building. However, nest-building of singly housed male mice has been less examined. Here we show that male mice spent longer time in nest-building at the early to middle dark phase and at the end of the dark phase. These two periods are followed by sleep-rich periods. When a nest was removed and fresh nest material was introduced, both male and female mice built nests at Zeitgeber time (ZT) 6, but not at ZT12. Using Fos-immunostaining combined with double in situ hybridization of Vgat and Vglut2, we found that Vgat- and Vglut2-positive cells of the lateral preoptic area (LPOA) were the only hypothalamic neuron population that exhibited a greater number of activated cells in response to fresh nest material at ZT6, compared to being naturally awake at ZT12. Fos-positive LPOA neurons were negative for estrogen receptor 1 (Esr1). Both Vgat-positive and Vglut2-positive neurons in both the LPOA and MPOA were activated at pup retrieval by male mice. Our findings suggest the possibility that GABAergic and glutamatergic neurons in the LPOA are associated with nest-building behavior in male mice.
Collapse
Affiliation(s)
- Natsuki Tagawa
- Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, 143-8540, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Keita Mori
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Michinori Koebis
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Atsu Aiba
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-Ku, Tokyo, 113-0033, Japan
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Yuichi Iino
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, 143-8540, Japan.
| | - Hiromasa Funato
- Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, 143-8540, Japan.
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
28
|
Fang W, Yin B, Fang Z, Tian M, Ke L, Ma X, Di Q. Heat stroke-induced cerebral cortex nerve injury by mitochondrial dysfunction: A comprehensive multi-omics profiling analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 919:170869. [PMID: 38342446 DOI: 10.1016/j.scitotenv.2024.170869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/20/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
In recent years, global warming has led to frequent instances of extremely high temperatures during summer, arousing significant concern about the adverse effects of high temperature. Among these, heat stroke is the most serious, which has detrimental effects on the all organs of human body, especially on brain. However, the comprehensive pathogenesis leading to brain damage remains unclear. In this study, we constructed a mouse model of heat stroke and conducted multi-omics profiling to identify relevant pathogenesis induced by heat stroke. The mice were placed in a constant temperature chamber at 42 °C with a humidity of 50 %, and the criteria for success in modeling were that the rectal temperature reached 42 °C and that the mice were trembling. Then the mice were immediately taken out for further experiments. Firstly, we conducted cFos protein localization and identified the cerebral cortex, especially the anterior cingulate cortex as the region exhibiting the most pronounced damage. Secondly, we performed metabolomics, transcriptomics, and proteomics analysis on cerebral cortex. This multi-omics investigation unveiled noteworthy alterations in proteins and metabolites within pathways associated with neurotransmitter systems, heatstroke-induced mitochondrial dysfunction, encompassing histidine and pentose phosphate metabolic pathways, as well as oxidative stress. In addition, the cerebral cortex exhibited pronounced Reactive Oxygen Species (ROS) production, alongside significant downregulation of the mitochondrial outer membrane protein Tomm40 and mitochondrial permeability transition pore, implicating cerebral cortex mitochondrial dysfunction as the primary instigator of neural impairment. This study marks a significant milestone as the first to employ multi-omics analysis in exploring the molecular mechanisms underlying heat stroke-induced damage in cerebral cortex neurons. It comprehensively identifies all potentially impacted pathways by heat stroke, laying a solid foundation for ensuing research endeavors. Consequently, this study introduces a fresh angle to clinical approaches in heatstroke prevention and treatment, as well as establishes an innovative groundwork for shaping future-forward environmental policies.
Collapse
Affiliation(s)
- Wen Fang
- Division of Sports Science& Physical Education, Tsinghua University, Beijing, China; Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Bo Yin
- School of Medicine, Tsinghua University, Beijing, China
| | - Zijian Fang
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | - Mengyi Tian
- School of Medicine, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Limei Ke
- School of Medicine, Tsinghua University, Beijing, China
| | - Xindong Ma
- Division of Sports Science& Physical Education, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| | - Qian Di
- Vanke School of Public Health, Tsinghua University, Beijing, China; Institute for Healthy China, Tsinghua University, Beijing, China.
| |
Collapse
|
29
|
Sakurai K. Rethinking c-Fos for understanding drug action in the brain. J Biochem 2024; 175:377-381. [PMID: 38153290 DOI: 10.1093/jb/mvad110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 12/29/2023] Open
Abstract
Understanding the mechanisms of drug action in the brain, from the genetic to the neural circuit level, is crucial for the development of new agents that act upon the central nervous system. Determining the brain regions and neurons affected by a drug is essential for revealing its mechanism of action in the brain. c-Fos, a marker of neuronal activation, has been widely used to detect neurons activated by stimuli with high spatial resolution. In this review, the use of c-Fos for the visualization and manipulation of activated neurons is introduced. I also explain that a higher temporal resolution can be achieved by changing the staining method for visualization of c-Fos. Moreover, a new method that allows labeling and manipulating commonly activated neurons using two different stimuli is proposed.
Collapse
Affiliation(s)
- Katsuyasu Sakurai
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
30
|
Milczarek MM, Perry JC, Amin E, Haniffa S, Hathaway T, Vann SD. Impairments in the early consolidation of spatial memories via group II mGluR agonism in the mammillary bodies. Sci Rep 2024; 14:5977. [PMID: 38472268 PMCID: PMC10933409 DOI: 10.1038/s41598-024-56015-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
mGluR2 receptors are widely expressed in limbic brain regions associated with memory, including the hippocampal formation, retrosplenial and frontal cortices, as well as subcortical regions including the mammillary bodies. mGluR2/3 agonists have been proposed as potential therapeutics for neurological and psychiatric disorders, however, there is still little known about the role of these receptors in cognitive processes, including memory consolidation. To address this, we assessed the effect of the mGluR2/3 agonist, eglumetad, on spatial memory consolidation in both mice and rats. Using the novel place preference paradigm, we found that post-sample injections of eglumetad impaired subsequent spatial discrimination when tested 6 h later. Using the immediate early gene c-fos as a marker of neural activity, we showed that eglumetad injections reduced activity in a network of limbic brain regions including the hippocampus and mammillary bodies. To determine whether the systemic effects could be replicated with more targeted manipulations, we performed post-sample infusions of the mGluR2/3 agonist 2R,4R-APDC into the mammillary bodies. This impaired novelty discrimination on a place preference task and an object-in-place task, again highlighting the role of mGluR2/3 transmission in memory consolidation and demonstrating the crucial involvement of the mammillary bodies in post-encoding processing of spatial information.
Collapse
Affiliation(s)
- Michal M Milczarek
- School of Psychology & Neuroscience and Mental Health Innovation Institute, Cardiff University, 70 Park Place, Cardiff, CF10 3AT, UK
| | - James C Perry
- School of Psychology & Neuroscience and Mental Health Innovation Institute, Cardiff University, 70 Park Place, Cardiff, CF10 3AT, UK
| | - Eman Amin
- School of Psychology & Neuroscience and Mental Health Innovation Institute, Cardiff University, 70 Park Place, Cardiff, CF10 3AT, UK
| | - Salma Haniffa
- School of Psychology & Neuroscience and Mental Health Innovation Institute, Cardiff University, 70 Park Place, Cardiff, CF10 3AT, UK
| | - Thomas Hathaway
- School of Psychology & Neuroscience and Mental Health Innovation Institute, Cardiff University, 70 Park Place, Cardiff, CF10 3AT, UK
| | - Seralynne D Vann
- School of Psychology & Neuroscience and Mental Health Innovation Institute, Cardiff University, 70 Park Place, Cardiff, CF10 3AT, UK.
| |
Collapse
|
31
|
Speidell A, Agbey C, Mocchetti I. Accelerated neurodegeneration of basal forebrain cholinergic neurons in HIV-1 gp120 transgenic mice: Critical role of the p75 neurotrophin receptor. Brain Behav Immun 2024; 117:347-355. [PMID: 38266662 PMCID: PMC10935610 DOI: 10.1016/j.bbi.2024.01.215] [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: 09/08/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 01/26/2024] Open
Abstract
Human Immunodeficiency Virus-1 (HIV) infection of the brain induces HIV-associated neurocognitive disorders (HAND). The set of molecular events employed by HIV to drive cognitive impairments in people living with HIV are diverse and remain not completely understood. We have shown that the HIV envelope protein gp120 promotes loss of synapses and decreases performance on cognitive tasks through the p75 neurotrophin receptor (p75NTR). This receptor is abundant on cholinergic neurons of the basal forebrain and contributes to cognitive impairment in various neurological disorders. In this study, we examined cholinergic neurons of gp120 transgenic (gp120tg) mice for signs of degeneration. We observed that the number of choline acetyltransferase-expressing cells is decreased in old (12-14-month-old) gp120tg mice when compared to age matched wild type. In the same animals, we observed an increase in the levels of pro-nerve growth factor, a ligand of p75NTR, as well as a disruption of consolidation of extinction of conditioned fear, a behavior regulated by cholinergic neurons of the basal forebrain. Both biochemical and behavioral outcomes of gp120tg mice were rescued by the deletion of the p75NTR gene, strongly supporting the role that this receptor plays in the neurotoxic effects of gp120. These data indicate that future p75NTR-directed pharmacotherapies could provide an adjunct therapy against synaptic simplification caused by HIV.
Collapse
Affiliation(s)
- Andrew Speidell
- Interdisciplinary Program in Neuroscience, and Department of Neuroscience, NRB WP13, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Christy Agbey
- Interdisciplinary Program in Neuroscience, and Department of Neuroscience, NRB WP13, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Italo Mocchetti
- Interdisciplinary Program in Neuroscience, and Department of Neuroscience, NRB WP13, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
32
|
Andreyanov M, Heinrich R, Berlin S. Design of Ultrapotent Genetically Encoded Inhibitors of Kv4.2 for Gating Neural Plasticity. J Neurosci 2024; 44:e2295222023. [PMID: 38154956 PMCID: PMC10869153 DOI: 10.1523/jneurosci.2295-22.2023] [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: 12/15/2022] [Revised: 11/05/2023] [Accepted: 11/22/2023] [Indexed: 12/30/2023] Open
Abstract
The Kv4.2 potassium channel plays established roles in neuronal excitability, while also being implicated in plasticity. Current means to study the roles of Kv4.2 are limited, motivating us to design a genetically encoded membrane tethered Heteropodatoxin-2 (MetaPoda). We find that MetaPoda is an ultrapotent and selective gating-modifier of Kv4.2. We narrow its site of contact with the channel to two adjacent residues within the voltage sensitive domain (VSD) and, with docking simulations, suggest that the toxin binds the VSD from within the membrane. We also show that MetaPoda does not require an external linker of the channel for its activity. In neurons (obtained from female and male rat neonates), MetaPoda specifically, and potently, inhibits all Kv4 currents, leaving all other A-type currents unaffected. Inhibition of Kv4 in hippocampal neurons does not promote excessive excitability, as is expected from a simple potassium channel blocker. We do find that MetaPoda's prolonged expression (1 week) increases expression levels of the immediate early gene cFos and prevents potentiation. These findings argue for a major role of Kv4.2 in facilitating plasticity of hippocampal neurons. Lastly, we show that our engineering strategy is suitable for the swift engineering of another potent Kv4.2-selective membrane-tethered toxin, Phrixotoxin-1, denoted MetaPhix. Together, we provide two uniquely potent genetic tools to study Kv4.2 in neuronal excitability and plasticity.
Collapse
Affiliation(s)
- Michael Andreyanov
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa 3525433, Israel
| | - Ronit Heinrich
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa 3525433, Israel
| | - Shai Berlin
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa 3525433, Israel
| |
Collapse
|
33
|
Xing B, Barbour AJ, Vithayathil J, Li X, Dutko S, Fawcett-Patel J, Lancaster E, Talos DM, Jensen FE. Reversible synaptic adaptations in a subpopulation of murine hippocampal neurons following early-life seizures. J Clin Invest 2024; 134:e175167. [PMID: 38227384 PMCID: PMC10904056 DOI: 10.1172/jci175167] [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: 08/25/2023] [Accepted: 01/11/2024] [Indexed: 01/17/2024] Open
Abstract
Early-life seizures (ELSs) can cause permanent cognitive deficits and network hyperexcitability, but it is unclear whether ELSs induce persistent changes in specific neuronal populations and whether these changes can be targeted to mitigate network dysfunction. We used the targeted recombination of activated populations (TRAP) approach to genetically label neurons activated by kainate-induced ELSs in immature mice. The ELS-TRAPed neurons were mainly found in hippocampal CA1, remained uniquely susceptible to reactivation by later-life seizures, and displayed sustained enhancement in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-mediated (AMPAR-mediated) excitatory synaptic transmission and inward rectification. ELS-TRAPed neurons, but not non-TRAPed surrounding neurons, exhibited enduring decreases in Gria2 mRNA, responsible for encoding the GluA2 subunit of the AMPARs. This was paralleled by decreased synaptic GluA2 protein expression and heightened phosphorylated GluA2 at Ser880 in dendrites, indicative of GluA2 internalization. Consistent with increased GluA2-lacking AMPARs, ELS-TRAPed neurons showed premature silent synapse depletion, impaired long-term potentiation, and impaired long-term depression. In vivo postseizure treatment with IEM-1460, an inhibitor of GluA2-lacking AMPARs, markedly mitigated ELS-induced changes in TRAPed neurons. These findings show that enduring modifications of AMPARs occur in a subpopulation of ELS-activated neurons, contributing to synaptic dysplasticity and network hyperexcitability, but are reversible with early IEM-1460 intervention.
Collapse
|
34
|
Yelhekar TD, Meng M, Doupe J, Lin Y. All IEGs Are Not Created Equal-Molecular Sorting Within the Memory Engram. ADVANCES IN NEUROBIOLOGY 2024; 38:81-109. [PMID: 39008012 DOI: 10.1007/978-3-031-62983-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
When neurons are recruited to form the memory engram, they are driven to activate the expression of a series of immediate-early genes (IEGs). While these IEGs have been used relatively indiscriminately to identify the so-called engram neurons, recent research has demonstrated that different IEG ensembles can be physically and functionally distinct within the memory engram. This inherent heterogeneity of the memory engram is driven by the diversity in the functions and distributions of different IEGs. This process, which we call molecular sorting, is analogous to sorting the entire population of engram neurons into different sub-engrams molecularly defined by different IEGs. In this chapter, we will describe the molecular sorting process by systematically reviewing published work on engram ensemble cells defined by the following four major IEGs: Fos, Npas4, Arc, and Egr1. By comparing and contrasting these likely different components of the memory engram, we hope to gain a better understanding of the logic and significance behind the molecular sorting process for memory functions.
Collapse
Affiliation(s)
- Tushar D Yelhekar
- Department of Psychiatry, O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Meizhen Meng
- Department of Psychiatry, O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Neuroscience Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joslyn Doupe
- Neuroscience Graduate Program, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yingxi Lin
- Department of Psychiatry, O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
35
|
L’Esperance OJ, McGhee J, Davidson G, Niraula S, Smith A, Sosunov AA, Yan SS, Subramanian J. Functional Connectivity Favors Aberrant Visual Network c-Fos Expression Accompanied by Cortical Synapse Loss in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2024; 101:111-131. [PMID: 39121131 PMCID: PMC11810533 DOI: 10.3233/jad-240776] [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: 08/11/2024]
Abstract
Background While Alzheimer's disease (AD) has been extensively studied with a focus on cognitive networks, visual network dysfunction has received less attention despite compelling evidence of its significance in AD patients and mouse models. We recently reported c-Fos and synaptic dysregulation in the primary visual cortex of a pre-amyloid plaque AD-model. Objective We test whether c-Fos expression and presynaptic density/dynamics differ in cortical and subcortical visual areas in an AD-model. We also examine whether aberrant c-Fos expression is inherited through functional connectivity and shaped by light experience. Methods c-Fos+ cell density, functional connectivity, and their experience-dependent modulation were assessed for visual and whole-brain networks in both sexes of 4-6-month-old J20 (AD-model) and wildtype (WT) mice. Cortical and subcortical differences in presynaptic vulnerability in the AD-model were compared using ex vivo and in vivo imaging. Results Visual cortical, but not subcortical, networks show aberrant c-Fos expression and impaired experience-dependent modulation. The average functional connectivity of a brain region in WT mice significantly predicts aberrant c-Fos expression, which correlates with impaired experience-dependent modulation in the AD-model. We observed a subtle yet selective weakening of excitatory visual cortical synapses. The size distribution of cortical boutons in the AD-model is downscaled relative to those in WT mice, suggesting a synaptic scaling-like adaptation of bouton size. Conclusions Visual network structural and functional disruptions are biased toward cortical regions in pre-plaque J20 mice, and the cellular and synaptic dysregulation in the AD-model represents a maladaptive modification of the baseline physiology seen in WT conditions.
Collapse
Affiliation(s)
- Oliver J. L’Esperance
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Josh McGhee
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Garett Davidson
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Suraj Niraula
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Adam Smith
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Alexandre A. Sosunov
- Department of Neurosurgery, Columbia University Medical Center,630 W. 168th St. New York, NY 10032
| | - Shirley Shidu Yan
- Department of Neurosurgery, Columbia University Medical Center,630 W. 168th St. New York, NY 10032
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
36
|
Davidson CJ, Mascarin AT, Yahya MA, Rubio FJ, Gheidi A. Approaches and considerations of studying neuronal ensembles: a brief review. Front Cell Neurosci 2023; 17:1310724. [PMID: 38155864 PMCID: PMC10752959 DOI: 10.3389/fncel.2023.1310724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
First theorized by Hebb, neuronal ensembles have provided a framework for understanding how the mammalian brain operates, especially regarding learning and memory. Neuronal ensembles are discrete, sparsely distributed groups of neurons that become activated in response to a specific stimulus and are thought to provide an internal representation of the world. Beyond the study of region-wide or projection-wide activation, the study of ensembles offers increased specificity and resolution to identify and target specific memories or associations. Neuroscientists interested in the neurobiology of learning, memory, and motivated behavior have used electrophysiological-, calcium-, and protein-based proxies of neuronal activity in preclinical models to better understand the neurobiology of learned and motivated behaviors. Although these three approaches may be used to pursue the same general goal of studying neuronal ensembles, technical differences lead to inconsistencies in the output and interpretation of data. This mini-review highlights some of the methodologies used in electrophysiological-, calcium-, and protein-based studies of neuronal ensembles and discusses their strengths and weaknesses.
Collapse
Affiliation(s)
- Cameron J. Davidson
- William Beaumont School of Medicine, Oakland University, Rochester, MI, United States
| | - Alixandria T. Mascarin
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Majd A. Yahya
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - F. Javier Rubio
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Bethesda, MD, United States
| | - Ali Gheidi
- Department of Biomedical Sciences, Mercer University, Macon, GA, United States
| |
Collapse
|
37
|
Danis A, Gallagher AA, Anderson AN, Isakharov A, Beeson KA, Schnell E. Altered hippocampal activation in seizure-prone CACNA2D2 knockout mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.565511. [PMID: 37986872 PMCID: PMC10659305 DOI: 10.1101/2023.11.08.565511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
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 in hippocampal tissue from wildtype (WT) and α2δ-2 knockout (CACNA2D2 KO) mice using immunohistochemical staining and confocal microscopy. Both genotypes demonstrated similarly sparse c-fos expression 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 hour after handling-associated convulsions, KO mice had fewer c-fos-positive cells in the dentate gyrus, indicating that activity in the dentate gyrus actually decreased. However, the dentate was significantly more active in KO mice compared to WT after administration of a subthreshold pentylenetetrazole dose, consistent with increased susceptibility to proconvulsant stimuli. Other histopathological markers of temporal lobe epilepsy in these mice, including markers of neurogenesis, glial activation, and mossy fiber sprouting, were similar in WT and KO mice, apart from a small but significant increase in hilar mossy cell density, opposite to what is typically found in mice with temporal lobe epilepsy. This suggests that the differences in seizure-associated hippocampal 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 Danis
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239
- Research and Development Service, Portland VA Health Care System, Portland, OR, 97239, Portland, OR, 97239
| | - Ashlynn A. Gallagher
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239
- Research and Development Service, Portland VA Health Care System, Portland, OR, 97239, Portland, OR, 97239
| | - Ashley N. Anderson
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239
- Research and Development Service, Portland VA Health Care System, Portland, OR, 97239, Portland, OR, 97239
| | - Arielle Isakharov
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, 97239
| | - Kathleen A. Beeson
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, 97239
| | - Eric Schnell
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239
- Research and Development Service, Portland VA Health Care System, Portland, OR, 97239, Portland, OR, 97239
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, 97239
| |
Collapse
|
38
|
Li S, Xu X, Li C, Xu Z, Wu K, Ye Q, Zhang Y, Jiang X, Cang C, Tian C, Wen J. In vivo labeling and quantitative imaging of neuronal populations using MRI. Neuroimage 2023; 281:120374. [PMID: 37729795 DOI: 10.1016/j.neuroimage.2023.120374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/30/2023] [Accepted: 09/10/2023] [Indexed: 09/22/2023] Open
Abstract
The study of neural circuits, which underlies perception, cognition, emotion, and behavior, is essential for understanding the mammalian brain, a complex organ consisting of billions of neurons. To study the structure and function of the brain, in vivo neuronal labeling and imaging techniques are crucial as they provide true physiological information that ex vivo methods cannot offer. In this paper, we present a new strategy for in vivo neuronal labeling and quantification using MRI. We demonstrate the efficacy of this method by delivering the oatp1a1 gene to the target neurons using rAAV2-retro virus. OATP1A1 protein expression on the neuronal membrane increased the uptake of a specific MRI contrast agent (Gd-EOB-DTPA), leading to hyperintense signals on T1W images of labeled neuronal populations. We also used dynamic contrast enhancement-based methods to obtain quantitative information on labeled neuronal populations in vivo.
Collapse
Affiliation(s)
- Shana Li
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Xiang Xu
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Canjun Li
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Ziyan Xu
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Ke Wu
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Qiong Ye
- Key Laboratory of High Field Magnetic Resonance Image of Anhui Province, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, PR China
| | - Yan Zhang
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Xiaohua Jiang
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Chunlei Cang
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Changlin Tian
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China; Key Laboratory of High Field Magnetic Resonance Image of Anhui Province, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, PR China.
| | - Jie Wen
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China.
| |
Collapse
|
39
|
Chen CH, Newman LN, Stark AP, Bond KE, Zhang D, Nardone S, Vanderburg CR, Nadaf NM, Yao Z, Mutume K, Flaquer I, Lowell BB, Macosko EZ, Regehr WG. A Purkinje cell to parabrachial nucleus pathway enables broad cerebellar influence over the forebrain. Nat Neurosci 2023; 26:1929-1941. [PMID: 37919612 PMCID: PMC11348979 DOI: 10.1038/s41593-023-01462-w] [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: 03/15/2023] [Accepted: 09/11/2023] [Indexed: 11/04/2023]
Abstract
In addition to its motor functions, the cerebellum is involved in emotional regulation, anxiety and affect. We found that suppressing the firing of cerebellar Purkinje cells (PCs) rapidly excites forebrain areas that contribute to such functions (including the amygdala, basal forebrain and septum), but that the classic cerebellar outputs, the deep cerebellar nuclei, do not directly project there. We show that PCs directly inhibit parabrachial nuclei (PBN) neurons that project to numerous forebrain regions. Suppressing the PC-PBN pathway influences many regions in the forebrain and is aversive. Molecular profiling shows that PCs directly inhibit numerous types of PBN neurons that control diverse behaviors that are not involved in motor control. Therefore, the PC-PBN pathway allows the cerebellum to directly regulate activity in the forebrain, and may be an important substrate for cerebellar disorders arising from damage to the posterior vermis.
Collapse
Affiliation(s)
- Christopher H Chen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Leannah N Newman
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Amanda P Stark
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Katherine E Bond
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Dawei Zhang
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Stefano Nardone
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Charles R Vanderburg
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Naeem M Nadaf
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Zhiyi Yao
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Kefiloe Mutume
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Isabella Flaquer
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Evan Z Macosko
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Saito S, Hashimoto H, Wakashin H, Ishibane M, Pae S, Saito S, Reien Y, Hirayama Y, Seo Y, Mizushima T, Anzai N. Central administered xenin induced Fos expression in nesfatin-1 neurons in rats. Brain Res Bull 2023; 204:110788. [PMID: 37844783 DOI: 10.1016/j.brainresbull.2023.110788] [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: 07/13/2023] [Revised: 10/03/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Xenin is a 25-amino acid peptide identified in human gastric mucosa, which is widely expressed in peripheral and central tissues. It is known that the central or peripheral administration of xenin decreases food intake in rodents. Nesfatin-1/NUCB2 (nesfatin-1) has been identified as an anorexic neuropeptide, it is often found co-localized with many peptides in the central nervous system. After the intracerebroventricular administration of xenin on nesfain-1-like immunoreactivity (LI) neurons, we examined its effects on food intake and water intake in rats. As a result, Fos-LI neurons were observed in the organum vasculosum of the laminae terminalis (OVLT), the median preoptic nucleus (MnPO), the subfornical organ (SFO), the supraoptic nucleus (SON), the paraventricular nucleus (PVN), the arcuate nucleus (Arc), the lateral hypothalamic area (LHA), the central amygdaloid nucleus (CAN), the dorsal raphe nucleus (DR), the locus coeruleus (LC), the area postrema (AP) and the nucleus of the solitary tract (NTS). After the administration, the number of Fos-LI neurons was significantly increased in the LC and the OVLT, the MnPO, the SFO, the SON, the PVN, the Arc, the LHA, the CAN, the DR, the AP and the NTS, compared with the control group. After the administration of xenin, we conducted double immunohistochemistry for Fos and nesfatin-1, and found that the number of nesfatin-1-LI neurons expressing Fos were significantly increased in the SON, the PVN, the Arc, the LHA, the CAN, the DR, the AP and the NTS, compared with the control group. The pretreatment of nesfatin-1 antisense significantly attenuated this xenin-induced feeding suppression, while that of nesfatin-1 missense showed no improvement. These results indicate that central administered xenin may have anorexia effects associated with activated central nesfatin-1 neurons.
Collapse
Affiliation(s)
- Shota Saito
- Department of Pharmacology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, JAPAN; Department of Rehabilitation, Dokkyo Medical University, 8880 Kitakobayashi, Mibu, Shimotsugagun, Tochigi 321-0293, Japan
| | - Hirofumi Hashimoto
- Department of Pharmacology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, JAPAN; Department of Rehabilitation, Dokkyo Medical University, 8880 Kitakobayashi, Mibu, Shimotsugagun, Tochigi 321-0293, Japan; Department of Regulatory Physiology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsugagun, Tochigi 321-0293, Japan.
| | - Hidefumi Wakashin
- Department of Regulatory Physiology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsugagun, Tochigi 321-0293, Japan
| | - Misaki Ishibane
- Department of Pharmacology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, JAPAN
| | - Sangjon Pae
- Department of Pharmacology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, JAPAN
| | - Shinpei Saito
- Department of Pharmacology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, JAPAN
| | - Yoshie Reien
- Department of Pharmacology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, JAPAN
| | - Yuri Hirayama
- Department of Pharmacology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, JAPAN
| | - Yoshiteru Seo
- Department of Regulatory Physiology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsugagun, Tochigi 321-0293, Japan; Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi 444-8787, Japan
| | - Takashi Mizushima
- Department of Rehabilitation, Dokkyo Medical University, 8880 Kitakobayashi, Mibu, Shimotsugagun, Tochigi 321-0293, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, JAPAN
| |
Collapse
|
41
|
Rijsketic DR, Casey AB, Barbosa DAN, Zhang X, Hietamies TM, Ramirez-Ovalle G, Pomrenze MB, Halpern CH, Williams LM, Malenka RC, Heifets BD. UNRAVELing the synergistic effects of psilocybin and environment on brain-wide immediate early gene expression in mice. Neuropsychopharmacology 2023; 48:1798-1807. [PMID: 37248402 PMCID: PMC10579391 DOI: 10.1038/s41386-023-01613-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/25/2023] [Accepted: 05/15/2023] [Indexed: 05/31/2023]
Abstract
The effects of context on the subjective experience of serotonergic psychedelics have not been fully examined in human neuroimaging studies, partly due to limitations of the imaging environment. Here, we administered saline or psilocybin to mice in their home cage or an enriched environment, immunofluorescently-labeled brain-wide c-Fos, and imaged iDISCO+ cleared tissue with light sheet fluorescence microscopy (LSFM) to examine the impact of environmental context on psilocybin-elicited neural activity at cellular resolution. Voxel-wise analysis of c-Fos-immunofluorescence revealed clusters of neural activity associated with main effects of context and psilocybin-treatment, which were validated with c-Fos+ cell density measurements. Psilocybin increased c-Fos expression in subregions of the neocortex, caudoputamen, central amygdala, and parasubthalamic nucleus while it decreased c-Fos in the hypothalamus, cortical amygdala, striatum, and pallidum in a predominantly context-independent manner. To gauge feasibility of future mechanistic studies on ensembles activated by psilocybin, we confirmed activity- and Cre-dependent genetic labeling in a subset of these neurons using TRAP2+/-;Ai14+ mice. Network analyses treating each psilocybin-sensitive cluster as a node indicated that psilocybin disrupted co-activity between highly correlated regions, reduced brain modularity, and dramatically attenuated intermodular co-activity. Overall, our results indicate that main effects of context and psilocybin were robust, widespread, and reorganized network architecture, whereas context×psilocybin interactions were surprisingly sparse.
Collapse
Affiliation(s)
- Daniel Ryskamp Rijsketic
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Austen B Casey
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Daniel A N Barbosa
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xue Zhang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Tuuli M Hietamies
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Grecia Ramirez-Ovalle
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Matthew B Pomrenze
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
- Nancy Pritzker Laboratory, Stanford University, Stanford, CA, 94305, USA
| | - Casey H Halpern
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Leanne M Williams
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
- Sierra-Pacific Mental Illness Research, Education, and Clinical Center (MIRECC) Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Robert C Malenka
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
- Nancy Pritzker Laboratory, Stanford University, Stanford, CA, 94305, USA
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
42
|
Manning A, Han V, Stephens A, Wang R, Bush N, Bard M, Ramirez JM, Kalume F. Elevated susceptibility to exogenous seizure triggers and impaired interneuron excitability in a mouse model of Leigh syndrome epilepsy. Neurobiol Dis 2023; 187:106288. [PMID: 37704057 PMCID: PMC10621616 DOI: 10.1016/j.nbd.2023.106288] [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: 05/28/2023] [Revised: 08/12/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023] Open
Abstract
Mutations in the NADH dehydrogenase (ubiquinone reductase) iron‑sulfur protein 4 (NDUFS4) gene, which encodes for a key structural subunit of the OXFOS complex I (CI), lead to the most common form of mitochondrial disease in children known as Leigh syndrome (LS). As in other mitochondrial diseases, epileptic seizures constitute one of the most significant clinical features of LS. These seizures are often very difficult to treat and are a sign of poor disease prognosis. Mice with whole-body Ndufs4 KO are a well-validated model of LS; they exhibit epilepsy and several other clinical features of LS. We have previously shown that mice with Ndufs4 KO in only GABAergic interneurons (Gad2-Ndufs4-KO) reproduce the severe epilepsy phenotype observed in the global KO mice. This observation indicated that these mice represent an excellent model of LS epilepsy isolated from other clinical manifestations of the disease. To further characterize this epilepsy phenotype, we investigated seizure susceptibility to selected exogenous seizure triggers in Gad2-Ndufs4-KO mice. Then, using electrophysiology, imaging, and immunohistochemistry, we studied the cellular, physiological, and neuroanatomical consequences of Ndufs4 KO in GABAergic interneurons. Homozygous KO of Ndufs4 in GABAergic interneurons leads to a prominent susceptibility to exogenous seizure triggers, impaired interneuron excitability and interneuron loss. Finally, we found that the hippocampus and cortex participate in the generation of seizure activity in Gad2-Ndufs4-KO mice. These findings further define the LS epilepsy phenotype and provide important insights into the cellular mechanisms underlying epilepsy in LS and other mitochondrial diseases.
Collapse
Affiliation(s)
- Arena Manning
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States of America; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Victor Han
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Alexa Stephens
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Rose Wang
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Nicholas Bush
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Michelle Bard
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Jan M Ramirez
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States of America; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America; Department of Neurological Surgery, University of Washington, Seattle, WA, United States of America
| | - Franck Kalume
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States of America; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America; Department of Neurological Surgery, University of Washington, Seattle, WA, United States of America.
| |
Collapse
|
43
|
Barber KR, Vizcarra VS, Zilch A, Majuta L, Diezel CC, Culver OP, Hughes BW, Taniguchi M, Streicher JM, Vanderah TW, Riegel AC. The Role of Ryanodine Receptor 2 in Drug-Associated Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560743. [PMID: 37873212 PMCID: PMC10592901 DOI: 10.1101/2023.10.03.560743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Type-2 ryanodine receptor (RyR2) ion channels facilitate the release of Ca 2+ from stores and serve an important function in neuroplasticity. The role for RyR2 in hippocampal-dependent learning and memory is well established and chronic hyperphosphorylation of RyR2 (RyR2P) is associated with pathological calcium leakage and cognitive disorders, including Alzheimer's disease. By comparison, little is known about the role of RyR2 in the ventral medial prefrontal cortex (vmPFC) circuitry important for working memory, decision making, and reward seeking. Here, we evaluated the basal expression and localization of RyR2 and RyR2P in the vmPFC. Next, we employed an operant model of sucrose, cocaine, or morphine self-administration (SA) followed by a (reward-free) recall test, to reengage vmPFC neurons and reactivate reward-seeking and re-evaluated the expression and localization of RyR2 and RyR2P in vmPFC. Under basal conditions, RyR2 was expressed in pyramidal cells but not regularly detected in PV/SST interneurons. On the contrary, RyR2P was rarely observed in PFC somata and was restricted to a different subcompartment of the same neuron - the apical dendrites of layer-5 pyramidal cells. Chronic SA of drug (cocaine or morphine) and nondrug (sucrose) rewards produced comparable increases in RyR2 protein expression. However, recalling either drug reward impaired the usual localization of RyR2P in dendrites and markedly increased its expression in somata immunoreactive for Fos, a marker of highly activated neurons. These effects could not be explained by chronic stress or drug withdrawal and instead appeared to require a recall experience associated with prior drug SA. In addition to showing the differential distribution of RyR2/RyR2P and affirming the general role of vmPFC in reward learning, this study provides information on the propensity of addictive drugs to redistribute RyR2P ion channels in a neuronal population engaged in drug-seeking. Hence, focusing on the early impact of addictive drugs on RyR2 function may serve as a promising approach to finding a treatment for substance use disorders.
Collapse
|
44
|
Matchynski JI, Cilley TS, Sadik N, Makki KM, Wu M, Manwar R, Woznicki AR, Kallakuri S, Arfken CL, Hope BT, Avanaki K, Conti AC, Perrine SA. Quantification of prefrontal cortical neuronal ensembles following conditioned fear learning in a Fos-LacZ transgenic rat with photoacoustic imaging in Vivo. PHOTOACOUSTICS 2023; 33:100551. [PMID: 38021296 PMCID: PMC10658601 DOI: 10.1016/j.pacs.2023.100551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 05/19/2023] [Accepted: 08/26/2023] [Indexed: 12/01/2023]
Abstract
Understanding the neurobiology of complex behaviors requires measurement of activity in the discrete population of active neurons, neuronal ensembles, which control the behavior. Conventional neuroimaging techniques ineffectively measure neuronal ensemble activity in the brain in vivo because they assess the average regional neuronal activity instead of the specific activity of the neuronal ensemble that mediates the behavior. Our functional molecular photoacoustic tomography (FM-PAT) system allows direct imaging of Fos-dependent neuronal ensemble activation in Fos-LacZ transgenic rats in vivo. We tested four experimental conditions and found increased FM-PAT signal in prefrontal cortical areas in rats undergoing conditioned fear or novel context exposure. A parallel immunofluorescence ex vivo study of Fos expression found similar findings. These findings demonstrate the ability of FM-PAT to measure Fos-expressing neuronal ensembles directly in vivo and support a mechanistic role for the prefrontal cortex in higher-order processing of response to specific stimuli or environmental cues.
Collapse
Affiliation(s)
- James I Matchynski
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
- Wayne State MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Timothy S Cilley
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nareen Sadik
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kassem M Makki
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | - Min Wu
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | - Rayyan Manwar
- University of Illinois at Chicago, Department of Bioengineering, Chicago, IL, USA
| | | | - Srinivasu Kallakuri
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Cynthia L Arfken
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bruce T Hope
- The National Institute on Drug Abuse (NIDA), Intramural Research Program, Baltimore, MD, USA
| | - Kamran Avanaki
- University of Illinois at Chicago, Department of Bioengineering, Chicago, IL, USA
| | - Alana C Conti
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
45
|
Li X, Li Y, Jin Y, Zhang Y, Wu J, Xu Z, Huang Y, Cai L, Gao S, Liu T, Zeng F, Wang Y, Wang W, Yuan TF, Tian H, Shu Y, Guo F, Lu W, Mao Y, Mei X, Rao Y, Peng B. Transcriptional and epigenetic decoding of the microglial aging process. NATURE AGING 2023; 3:1288-1311. [PMID: 37697166 PMCID: PMC10570141 DOI: 10.1038/s43587-023-00479-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 08/03/2023] [Indexed: 09/13/2023]
Abstract
As important immune cells, microglia undergo a series of alterations during aging that increase the susceptibility to brain dysfunctions. However, the longitudinal characteristics of microglia remain poorly understood. In this study, we mapped the transcriptional and epigenetic profiles of microglia from 3- to 24-month-old mice. We first discovered unexpected sex differences and identified age-dependent microglia (ADEM) genes during the aging process. We then compared the features of aging and reactivity in female microglia at single-cell resolution and epigenetic level. To dissect functions of aged microglia excluding the influence from other aged brain cells, we established an accelerated microglial turnover model without directly affecting other brain cells. By this model, we achieved aged-like microglia in non-aged brains and confirmed that aged-like microglia per se contribute to cognitive decline. Collectively, our work provides a comprehensive resource for decoding the aging process of microglia, shedding light on how microglia maintain brain functions.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Yuxin Li
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Yuxiao Jin
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yuheng Zhang
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Jingchuan Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen Xu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yubin Huang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lin Cai
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuai Gao
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Taohui Liu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Fanzhuo Zeng
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yafei Wang
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Wenxu Wang
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hengli Tian
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yousheng Shu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Feifan Guo
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Wei Lu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xifan Mei
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Bo Peng
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China.
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- Co-Innovation Center of Neurodegeneration, Nantong University, Nantong, China.
| |
Collapse
|
46
|
Dogra D, Meza-Santoscoy PL, Gavrilovici C, Rehak R, de la Hoz CLR, Ibhazehiebo K, Rho JM, Kurrasch DM. kcna1a mutant zebrafish model episodic ataxia type 1 (EA1) with epilepsy and show response to first-line therapy carbamazepine. Epilepsia 2023; 64:2186-2199. [PMID: 37209379 DOI: 10.1111/epi.17659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 05/18/2023] [Accepted: 05/18/2023] [Indexed: 05/22/2023]
Abstract
OBJECTIVE KCNA1 mutations are associated with a rare neurological movement disorder known as episodic ataxia type 1 (EA1), and epilepsy is a common comorbidity. Current medications provide only partial relief for ataxia and/or seizures, making new drugs needed. Here, we characterized zebrafish kcna1a-/- as a model of EA1 with epilepsy and compared the efficacy of the first-line therapy carbamazepine in kcna1a-/- zebrafish to Kcna1-/- rodents. METHODS CRISPR/Cas9 mutagenesis was used to introduce a mutation in the sixth transmembrane segment of the zebrafish Kcna1 protein. Behavioral and electrophysiological assays were performed on kcna1a-/- larvae to assess ataxia- and epilepsy-related phenotypes. Real-time quantitative polymerase chain reaction (qPCR) was conducted to measure mRNA levels of brain hyperexcitability markers in kcna1a-/- larvae, followed by bioenergetics profiling to evaluate metabolic function. Drug efficacies were tested using behavioral and electrophysiological assessments, as well as seizure frequency in kcna1a-/- zebrafish and Kcna1-/- mice, respectively. RESULTS Zebrafish kcna1a-/- larvae showed uncoordinated movements and locomotor deficits, along with scoliosis and increased mortality. The mutants also exhibited impaired startle responses when exposed to light-dark flashes and acoustic stimulation as well as hyperexcitability as measured by extracellular field recordings and upregulated fosab transcripts. Neural vglut2a and gad1b transcript levels were disrupted in kcna1a-/- larvae, indicative of a neuronal excitatory/inhibitory imbalance, as well as a significant reduction in cellular respiration in kcna1a-/- , consistent with dysregulation of neurometabolism. Notably, carbamazepine suppressed the impaired startle response and brain hyperexcitability in kcna1a-/- zebrafish but had no effect on the seizure frequency in Kcna1-/- mice, suggesting that this EA1 zebrafish model might better translate to humans than rodents. SIGNIFICANCE We conclude that zebrafish kcna1a-/- show ataxia and epilepsy-related phenotypes and are responsive to carbamazepine treatment, consistent with EA1 patients. These findings suggest that kcna1-/- zebrafish are a useful model for drug screening as well as studying the underlying disease biology.
Collapse
Affiliation(s)
- Deepika Dogra
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Paola L Meza-Santoscoy
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Cezar Gavrilovici
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Departments of Pediatrics, Clinical Neurosciences, Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Departments of Neurosciences, Pediatrics, and Pharmacology, Rady Children's Hospital San Diego, University of California San Diego, San Diego, California, USA
| | - Renata Rehak
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Cristiane L R de la Hoz
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Kingsley Ibhazehiebo
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jong M Rho
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Departments of Pediatrics, Clinical Neurosciences, Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Departments of Neurosciences, Pediatrics, and Pharmacology, Rady Children's Hospital San Diego, University of California San Diego, San Diego, California, USA
| | - Deborah M Kurrasch
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
47
|
Naik AA, Ma X, Munyeshyaka M, Leibenluft E, Li Z. A New Behavioral Paradigm for Frustrative Non-reward Reveals a Global Change in Brain Networks by Frustration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.530477. [PMID: 36909498 PMCID: PMC10002733 DOI: 10.1101/2023.02.28.530477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Background Irritability, defined as proneness to anger, can reach a pathological extent. It is a defining symptom of Disruptive Mood Dysregulation Disorder (DMDD) and one of the most common reasons youth presents for psychiatric evaluation and care. Aberrant responses to frustrative non-reward (FNR, the response to omission of expected reward) are central to the pathophysiology of irritability. FNR is a translational construct to study irritability across species. The development of preclinical FNR models would advance mechanistic studies of the important and relatively understudied clinical phenomenon of irritability. Methods We used FNR as a conceptual framework to develop a novel mouse behavioral paradigm named Alternate Poking Reward Omission (APRO). After APRO, mice were examined with a battery of behavioral tests and processed for whole brain c-Fos imaging. FNR increases locomotion and aggression in mice regardless of sex. These behavioral changes resemble the symptoms observed in youth with severe irritability. There is no change in anxiety-like, depression-like, or non-aggressive social behaviors. FNR increases c-Fos+ neurons in 13 subregions of thalamus, iso-cortex and hippocampus including the prelimbic, ACC, hippocampus, dorsal thalamus, cuneiform nucleus, pons, and pallidum areas. FNR also shifts the brain network towards a more global processing mode. Conclusion Our novel FNR paradigm produces a frustration effect and alters brain processing in ways resembling the symptoms and brain network reconfiguration observed in youth with severe irritability. The novel behavioral paradigm and identified brain regions lay the groundwork for further mechanistic studies of frustration and irritability in rodents.
Collapse
Affiliation(s)
- Aijaz Ahmad Naik
- Section on Synapse Development Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
- Center on Compulsive Behaviors, Intramural Research program, NIH, Bethesda, MD, USA
| | - Xiaoyu Ma
- Section on Synapse Development Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Maxime Munyeshyaka
- Section on Synapse Development Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Ellen Leibenluft
- Section on Mood Dysregulation and Neuroscience, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Zheng Li
- Section on Synapse Development Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
48
|
McPherson KB, Bouchet CA, Coutens B, Ingram SL. Persistent inflammation selectively activates opioid-sensitive phasic-firing neurons within the vlPAG. J Neurophysiol 2023; 129:1237-1248. [PMID: 37073984 PMCID: PMC10202481 DOI: 10.1152/jn.00016.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/06/2023] [Accepted: 04/06/2023] [Indexed: 04/20/2023] Open
Abstract
The ventrolateral periaqueductal gray (vlPAG) is a key brain area within the descending pain modulatory pathway and an important target for opioid-induced analgesia. The vlPAG contains heterogeneous neurons with respect to neurotransmitter content, receptor and channel expression, and in vivo response to noxious stimuli. This study characterizes intrinsic membrane properties of vlPAG neurons to identify neuron types that respond to inflammation and determine whether the pain-responsive neurons are inhibited by opioids. Surveying 382 neurons identified four neuron types with distinct intrinsic firing patterns: Phasic (48%), Tonic (33%), Onset (10%), and Random (9%). Mu-opioid receptor (MOR) expression was determined by the ability of a selective MOR agonist (DAMGO) to activate G protein-coupled inwardly rectifying potassium channel (GIRK) currents. Opioid-sensitive neurons were observed within each neuron type. Opioid sensitivity did not correlate with other intrinsic firing features, including low-threshold spiking that has been previously proposed to identify opioid-sensitive GABAergic neurons in the vlPAG of mice. Complete Freund's adjuvant (CFA)-induced acute inflammation (2 h) had no effect on vlPAG neuron firing patterns. However, persistent inflammation (5-7 days) selectively activated Phasic neurons through a significant reduction in their firing threshold. Opioid-sensitive neurons were strongly activated compared with the opioid-insensitive Phasic neurons. Overall, this study provides a framework to further identify neurons activated by persistent inflammation so that they may be targeted for future pain therapies.NEW & NOTEWORTHY Intrinsic firing properties define four distinct vlPAG neuron populations, and a subset of each population expresses MORs coupled to GIRK channels. Persistent, but not acute, inflammation selectively activates opioid-sensitive Phasic vlPAG neurons. Although the vlPAG is known to contribute to the descending inhibition of pain, the activation of a single physiologically defined neuron type in the presence of persistent inflammation represents a mechanism by which the vlPAG participates in descending facilitation of pain.
Collapse
Affiliation(s)
- Kylie B McPherson
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon, United States
- The Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Courtney A Bouchet
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon, United States
- The Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Basile Coutens
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon, United States
| | - Susan L Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
49
|
Oyaga MR, Serra I, Kurup D, Koekkoek SKE, Badura A. Delay eyeblink conditioning performance and brain-wide c-Fos expression in male and female mice. Open Biol 2023; 13:220121. [PMID: 37161289 PMCID: PMC10170203 DOI: 10.1098/rsob.220121] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Delay eyeblink conditioning has been extensively used to study associative learning and the cerebellar circuits underlying this task have been largely identified. However, there is a little knowledge on how factors such as strain, sex and innate behaviour influence performance during this type of learning. In this study, we used male and female mice of C57BL/6J (B6) and B6CBAF1 strains to investigate the effect of sex, strain and locomotion in delay eyeblink conditioning. We performed a short and a long delay eyeblink conditioning paradigm and used a c-Fos immunostaining approach to explore the involvement of different brain areas in this task. We found that both B6 and B6CBAF1 females reach higher learning scores compared to males in the initial stages of learning. This sex-dependent difference was no longer present as the learning progressed. Moreover, we found a strong positive correlation between learning scores and voluntary locomotion irrespective of the training duration. c-Fos immunostainings after the short paradigm showed positive correlations between c-Fos expression and learning scores in the cerebellar cortex and brainstem, as well as previously unreported areas. By contrast, after the long paradigm, c-Fos expression was only significantly elevated in the brainstem. Taken together, we show that differences in voluntary locomotion and activity across brain areas correlate with performance in delay eyeblink conditioning across strains and sexes.
Collapse
Affiliation(s)
- Maria Roa Oyaga
- Department of Neuroscience, Erasmus MC, 3000 Rotterdam, the Netherlands
| | - Ines Serra
- Department of Neuroscience, Erasmus MC, 3000 Rotterdam, the Netherlands
| | - Devika Kurup
- Department of Neuroscience, Erasmus MC, 3000 Rotterdam, the Netherlands
| | | | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC, 3000 Rotterdam, the Netherlands
- Netherlands Institute of Neuroscience, Royal Dutch Academy for Arts and Sciences, Amsterdam 1105 BA, the Netherlands
| |
Collapse
|
50
|
Hageter J, Starkey J, Horstick EJ. Thalamic regulation of a visual critical period and motor behavior. Cell Rep 2023; 42:112287. [PMID: 36952349 PMCID: PMC10514242 DOI: 10.1016/j.celrep.2023.112287] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/02/2023] [Accepted: 03/03/2023] [Indexed: 03/24/2023] Open
Abstract
During the visual critical period (CP), sensory experience refines the structure and function of visual circuits. The basis of this plasticity was long thought to be limited to cortical circuits, but recently described thalamic plasticity challenges this dogma and demonstrates greater complexity underlying visual plasticity. Yet how visual experience modulates thalamic neurons or how the thalamus modulates CP timing is incompletely understood. Using a larval zebrafish, thalamus-centric ocular dominance model, we show functional changes in the thalamus and a role of inhibitory signaling to establish CP timing using a combination of functional imaging, optogenetics, and pharmacology. Hemisphere-specific changes in genetically defined thalamic neurons correlate with changes in visuomotor behavior, establishing a role of thalamic plasticity in modulating motor performance. Our work demonstrates that visual plasticity is broadly conserved and that visual experience leads to neuron-level functional changes in the thalamus that require inhibitory signaling to establish critical period timing.
Collapse
Affiliation(s)
- John Hageter
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA
| | - Jacob Starkey
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA
| | - Eric J Horstick
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA; Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|