1
|
Junaid M, Lee EJ, Lim SB. Single-cell and spatial omics: exploring hypothalamic heterogeneity. Neural Regen Res 2025; 20:1525-1540. [PMID: 38993130 PMCID: PMC11688568 DOI: 10.4103/nrr.nrr-d-24-00231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
Elucidating the complex dynamic cellular organization in the hypothalamus is critical for understanding its role in coordinating fundamental body functions. Over the past decade, single-cell and spatial omics technologies have significantly evolved, overcoming initial technical challenges in capturing and analyzing individual cells. These high-throughput omics technologies now offer a remarkable opportunity to comprehend the complex spatiotemporal patterns of transcriptional diversity and cell-type characteristics across the entire hypothalamus. Current single-cell and single-nucleus RNA sequencing methods comprehensively quantify gene expression by exploring distinct phenotypes across various subregions of the hypothalamus. However, single-cell/single-nucleus RNA sequencing requires isolating the cell/nuclei from the tissue, potentially resulting in the loss of spatial information concerning neuronal networks. Spatial transcriptomics methods, by bypassing the cell dissociation, can elucidate the intricate spatial organization of neural networks through their imaging and sequencing technologies. In this review, we highlight the applicative value of single-cell and spatial transcriptomics in exploring the complex molecular-genetic diversity of hypothalamic cell types, driven by recent high-throughput achievements.
Collapse
Affiliation(s)
- Muhammad Junaid
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| | - Eun Jeong Lee
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Su Bin Lim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| |
Collapse
|
2
|
Bueno D, Schäfer MK, Wang S, Schmeisser MJ, Methner A. NECAB family of neuronal calcium-binding proteins in health and disease. Neural Regen Res 2025; 20:1236-1243. [PMID: 38934399 PMCID: PMC11624857 DOI: 10.4103/nrr.nrr-d-24-00094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/18/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
The N-terminal EF-hand calcium-binding proteins 1-3 (NECAB1-3) constitute a family of predominantly neuronal proteins characterized by the presence of at least one EF-hand calcium-binding domain and a functionally less well characterized C-terminal antibiotic biosynthesis monooxygenase domain. All three family members were initially discovered due to their interactions with other proteins. NECAB1 associates with synaptotagmin-1, a critical neuronal protein involved in membrane trafficking and synaptic vesicle exocytosis. NECAB2 interacts with predominantly striatal G-protein-coupled receptors, while NECAB3 partners with amyloid-β A4 precursor protein-binding family A members 2 and 3, key regulators of amyloid-β production. This demonstrates the capacity of the family for interactions with various classes of proteins. NECAB proteins exhibit distinct subcellular localizations: NECAB1 is found in the nucleus and cytosol, NECAB2 resides in endosomes and the plasma membrane, and NECAB3 is present in the endoplasmic reticulum and Golgi apparatus. The antibiotic biosynthesis monooxygenase domain, an evolutionarily ancient component, is akin to atypical heme oxygenases in prokaryotes but is not well-characterized in vertebrates. Prokaryotic antibiotic biosynthesis monooxygenase domains typically form dimers, suggesting that calcium-mediated conformational changes in NECAB proteins may induce antibiotic biosynthesis monooxygenase domain dimerization, potentially activating some enzymatic properties. However, the substrate for this enzymatic activity remains uncertain. Alternatively, calcium-mediated conformational changes might influence protein interactions or the subcellular localization of NECAB proteins by controlling the availability of protein-protein interaction domains situated between the EF hands and the antibiotic biosynthesis monooxygenase domain. This review summarizes what is known about genomic organization, tissue expression, intracellular localization, interaction partners, and the physiological and pathophysiological role of the NECAB family.
Collapse
Affiliation(s)
- Diones Bueno
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Michael K.E. Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Sudena Wang
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Michael J. Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Axel Methner
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
3
|
Jing J, Hu M, Ngodup T, Ma Q, Lau SNN, Ljungberg MC, McGinley MJ, Trussell LO, Jiang X. Molecular logic for cellular specializations that initiate the auditory parallel processing pathways. Nat Commun 2025; 16:489. [PMID: 39788966 PMCID: PMC11717940 DOI: 10.1038/s41467-024-55257-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: 04/18/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
The cochlear nuclear complex (CN), the starting point for all central auditory processing, encompasses a suite of neuronal cell types highly specialized for neural coding of acoustic signals. However, the molecular logic governing these specializations remains unknown. By combining single-nucleus RNA sequencing and Patch-seq analysis, we reveal a set of transcriptionally distinct cell populations encompassing all previously observed types and discover multiple hitherto unknown subtypes with anatomical and physiological identity. The resulting comprehensive cell-type taxonomy reconciles anatomical position, morphological, physiological, and molecular criteria, enabling the determination of the molecular basis of the specialized cellular phenotypes in the CN. In particular, CN cell-type identity is encoded in a transcriptional architecture that orchestrates functionally congruent expression across a small set of gene families to customize projection patterns, input-output synaptic communication, and biophysical features required for encoding distinct aspects of acoustic signals. This high-resolution account of cellular heterogeneity from the molecular to the circuit level reveals the molecular logic driving cellular specializations, thus enabling the genetic dissection of auditory processing and hearing disorders with a high specificity.
Collapse
Affiliation(s)
- Junzhan Jing
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ming Hu
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Tenzin Ngodup
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, USA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-HNS, University of Washington, Seattle, WA, USA
| | - Qianqian Ma
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shu-Ning Natalie Lau
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - M Cecilia Ljungberg
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Matthew J McGinley
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| | - Laurence O Trussell
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, USA.
| | - Xiaolong Jiang
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
4
|
Wegmann L, Haas HL, Sergeeva OA. Comparative analysis of adenosine 1 receptor expression and function in hippocampal and hypothalamic neurons. Inflamm Res 2025; 74:11. [PMID: 39775928 PMCID: PMC11711771 DOI: 10.1007/s00011-024-01980-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Adenosine, an ATP degradation product, is a sleep pressure factor. The adenosine 1 receptor (A1R) reports sleep need. Histaminergic neurons (HN) of the tuberomamillary nucleus (TMN) fire exclusively during wakefulness and promote arousal. All of them express GABAA receptors and are inhibited by GABA. Does adenosine contribute to their silencing? SUBJECTS AND TREATMENT Responses to adenosine were studied in mouse brain slices and primary dissociated cultures. For HN identification single-cell (sc)RT-PCR, reporter protein and pharmacology were used. Hippocampal Dentate Gyrus granular layer cells (DGgc) were studied in parallel. METHODS Firing frequency was recorded in patch-clamp configuration or by microelectrode arrays. A1R-expression was studied by scRT-PCR and semiquantitative PCR. RESULTS Most DGgc were inhibited through A1R, detected with scRT-PCR in 7 out of 10 PDZd2-positive DGgc; all HN were A1R negative. One HN out of 25 was inhibited by adenosine. The A1R mRNA level in the hippocampus was 6 times higher than in the caudal (posterior) hypothalamus. Response to adenosine was weaker in hypothalamic compared to hippocampal cultures. CONCLUSIONS Most HN are not inhibited by adenosine.
Collapse
Affiliation(s)
- Lea Wegmann
- Medical Faculty and University Hospital, Institute of Neural and Sensory Physiology, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
- Medical Faculty and University Hospital, Institute of Clinical Neurosciences and Medical Psychology, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Helmut L Haas
- Medical Faculty and University Hospital, Institute of Neural and Sensory Physiology, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Olga A Sergeeva
- Medical Faculty and University Hospital, Institute of Neural and Sensory Physiology, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.
- Medical Faculty and University Hospital, Institute of Clinical Neurosciences and Medical Psychology, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.
| |
Collapse
|
5
|
Yang Q, Abdulla A, Farooq M, Ishikawa Y, Liu SJ. Emotional stress increases GluA2 expression and potentiates fear memory via adenylyl cyclase 5. Cell Rep 2025; 44:115180. [PMID: 39786995 DOI: 10.1016/j.celrep.2024.115180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 11/20/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Stress can alter behavior and contributes to psychiatric disorders by regulating the expression of the GluA2 AMPA receptor subunit. We have previously shown in mice that exposure to predator odor stress elevates GluA2 transcription in cerebellar molecular layer interneurons (MLIs), and MLI activity is required for fear memory consolidation. Here, we identified the critical involvement of adenylyl cyclase 5, in both the stress-induced increase in GluA2 in MLIs and the enhancement of fear memory. We found that noradrenaline release during predator odor stress activates AC5 and downstream PKA-CREB signaling. This pathway interacts synergistically with α1-adrenergic receptors to promote synaptic GluA2 expression in MLIs. At a behavioral level, predator odor stress potentiates associative fear memory, and this is abolished in AC5 knockout mice, suggesting that AC5-dependent plasticity is required for enhanced memory formation. Therefore, AC5 is a promising pharmacological target for preventing stress-enhanced fear memory.
Collapse
Affiliation(s)
- Qian Yang
- Department of Cell Biology and Anatomy, LSUHSC, New Orleans, LA 70112, USA
| | - Ahmad Abdulla
- Department of Cell Biology and Anatomy, LSUHSC, New Orleans, LA 70112, USA; Southeast Louisiana VA Healthcare System, New Orleans, LA 70119, USA
| | - Muhammad Farooq
- Department of Cell Biology and Anatomy, LSUHSC, New Orleans, LA 70112, USA; Southeast Louisiana VA Healthcare System, New Orleans, LA 70119, USA
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Siqiong June Liu
- Department of Cell Biology and Anatomy, LSUHSC, New Orleans, LA 70112, USA; Southeast Louisiana VA Healthcare System, New Orleans, LA 70119, USA.
| |
Collapse
|
6
|
Wang B, Zhang Y, Li H, Dou H, Guo Y, Deng Y. Biologically inspired heterogeneous learning for accurate, efficient and low-latency neural network. Natl Sci Rev 2025; 12:nwae301. [PMID: 39758128 PMCID: PMC11697980 DOI: 10.1093/nsr/nwae301] [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: 05/30/2024] [Revised: 07/24/2024] [Accepted: 08/08/2024] [Indexed: 01/07/2025] Open
Abstract
The pursuit of artificial neural networks that mirror the accuracy, efficiency and low latency of biological neural networks remains a cornerstone of artificial intelligence (AI) research. Here, we incorporated recent neuroscientific findings of self-inhibiting autapse and neuron heterogeneity for innovating a spiking neural network (SNN) with enhanced learning and memorizing capacities. A bi-level programming paradigm was formulated to respectively learn neuron-level biophysical variables and network-level synapse weights for nested heterogeneous learning. We successfully demonstrated that our biologically inspired neuron model could reproduce neural statistics at both individual and group levels, contributing to the effective decoding of brain-computer interface data. Furthermore, the heterogeneous SNN showed higher accuracy (1%-10% improvement), superior efficiency (maximal 17.83-fold reduction in energy) and lower latency (maximal 5-fold improvement) in performing several AI tasks. For the first time, we benchmarked SNN for conducting cell type identification from scRNA-seq data. The proposed model correctly identified very rare cell types associated with severe brain diseases where typical SNNs failed.
Collapse
Affiliation(s)
- Bo Wang
- School of Astronautics, Beihang University, Beijing 100191, China
| | - Yuxuan Zhang
- School of Astronautics, Beihang University, Beijing 100191, China
| | - Hongjue Li
- School of Astronautics, Beihang University, Beijing 100191, China
| | - Hongkun Dou
- School of Astronautics, Beihang University, Beijing 100191, China
| | - Yuchen Guo
- Institute for Brain and Cognitive Sciences, BNRist, Tsinghua University, Beijing 100084, China
| | - Yue Deng
- School of Astronautics, Beihang University, Beijing 100191, China
- School of Artificial Intelligence, Beihang University, Beijing 100191, China
| |
Collapse
|
7
|
He JQ, Yuan RL, Jiang YT, Peng Y, Ye JR, Wang SS, Li LQ, Ruan Y, Li PY, Yan X, He WB, Li G, Chu SF, Zhang Z, Chen NH. Esculetin facilitates post-stroke rehabilitation by inhibiting CKLF1-mediated neutrophil infiltration. Acta Pharmacol Sin 2025; 46:52-65. [PMID: 39112771 PMCID: PMC11697434 DOI: 10.1038/s41401-024-01352-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 07/02/2024] [Indexed: 01/04/2025] Open
Abstract
Esculetin (ESC) is a coumarin-derived phytochemical prevalent in traditional Chinese medicine that exhibits anti-acute ischemic stroke activities. Our previous studies demonstrate that CKLF1 is a potential anti-stroke target for coumarin-derived compound. In this study we investigated whether CKLF1 was involved in the neuroprotective effects of ESC against photothrombotic stroke in mice. The mice were treated with ESC (20, 40 or 80 mg·kg-1·d-1, i.g.) for two weeks. The therapeutic effect of ESC was assessed using MRI, neurological function evaluation, and a range of behavioral tests on D1, 3, 7 and 14 of ESC administration. We showed that oral administration of ESC dose-dependently reduced the cerebral infarction volume within one week after stroke, improved behavioral performance, and alleviated neuropathological damage within two weeks. Functional MRI revealed that ESC significantly enhanced the abnormal low-frequency fluctuation (ALFF) value of the motor cortex and promoted functional connectivity between the supplementary motor area (SMA) and multiple brain regions. We demonstrated that ESC significantly reduced the protein levels of CKLF1 and CCR5, as well as the CKLF1/CCR5 protein complex in the peri-infarcted area. We showed that ESC (0.1-10 μM) dose-dependently blocked CKLF1-induced chemotactic movement of neutrophils in the Transwell assay, reducing the interaction of CKLF1/CCR5 on the surface of neutrophils, thereby reducing neutrophil infiltration, and decreasing the expression of ICAM-1, VCAM-1 and MMP-9 in the peri-infarct tissue. Knockout of CKLF1 reduced brain infarction volume and motor dysfunction after stroke but also negated the anti-stroke efficacy and neutrophil infiltration of ESC. These results suggest that the efficacy of ESC in promoting post-stroke neural repair depends on its inhibition on CKLF1-mediated neutrophil infiltration, which offering novel perspectives for elucidating the therapeutic properties of coumarins.
Collapse
Affiliation(s)
- Jia-Qi He
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ruo-Lan Yuan
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yu-Tong Jiang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ye Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jun-Rui Ye
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Sha-Sha Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Li-Qing Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yuan Ruan
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Pei-Yi Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xu Yan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Bin He
- National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Gang Li
- Graduate school, Inner Mongolian Medical University, Hohhot, 010110, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
8
|
Choi D, Paré J, Dravid S, Smith Y. Ultrastructural Localization of Glutamate Delta Receptor 1 in the Rodent and Primate Lateral Habenula. J Comp Neurol 2025; 533:e70019. [PMID: 39794140 PMCID: PMC11723828 DOI: 10.1002/cne.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/19/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
Glutamate delta receptor 1 (GluD1) is a unique synaptogenic molecule expressed at excitatory and inhibitory synapses. The lateral habenula (LHb), a subcortical structure that regulates negative reward prediction error and major monoaminergic systems, is enriched in GluD1. LHb dysfunction has been implicated in psychiatric disorders such as depression and schizophrenia, both of which are associated with GRID1, the gene that encodes GluD1. Thus, disruption in GluD1 synaptic signaling may contribute to LHb dysfunction and the pathophysiology of LHb-associated disorders. Despite its strong cellular expression, little is known about the subsynaptic and subcellular localization of GluD1 in LHb neurons. Given that GluD1 is involved in the development and/or regulation of glutamatergic and GABAergic synapses in various brain regions, a detailed map of GluD1 synaptic localization is essential to elucidate its role in the LHb. To address this issue, we used immunoelectron microscopy methods in rodents and monkeys. In both species, GluD1 immunoreactivity was primarily expressed in dendritic profiles, with lower expression in somata, spines, and glial elements. Pre- and post-embedding immunogold experiments revealed strong GluD1 expression in the core of symmetric GABAergic synapses. Albeit less frequent, GluD1 was also found at the edges (i.e., perisynaptic) of asymmetric, putative glutamatergic synapses. Through the combination of anterograde tracing with immunogold labeling in rats, we showed that axon terminals from the entopeduncular nucleus and the lateral hypothalamus express postsynaptic GluD1 immunolabeling in the LHb. Our findings suggest that GluD1 may play a critical role in modulating GABAergic transmission in the rodent and primate LHb.
Collapse
Affiliation(s)
- Diane Choi
- Graduate Program in Molecular and Systems PharmacologyEmory UniversityAtlantaGeorgiaUSA
- Emory National Primate Research CenterEmory UniversityAtlantaGeorgiaUSA
| | - Jean‐Francois Paré
- Department of NeurologyEmory UniversityAtlantaGeorgiaUSA
- Emory National Primate Research CenterEmory UniversityAtlantaGeorgiaUSA
| | - Shashank Dravid
- Department of Psychiatry and Behavioral SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Yoland Smith
- Graduate Program in Molecular and Systems PharmacologyEmory UniversityAtlantaGeorgiaUSA
- Department of NeurologyEmory UniversityAtlantaGeorgiaUSA
- Emory National Primate Research CenterEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
9
|
Hua Y, Habicher J, Carl M, Manuel R, Boije H. Novel Transgenic Zebrafish Lines to Study the CHRNA3-B4-A5 Gene Cluster. Dev Neurobiol 2025; 85:e22956. [PMID: 39686588 DOI: 10.1002/dneu.22956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 09/13/2024] [Accepted: 11/05/2024] [Indexed: 12/18/2024]
Abstract
Acetylcholine (ACh), a vital neurotransmitter for both the peripheral (PNS) and central nervous systems (CNS), signals through nicotinic ACh receptors (nAChRs) and muscarinic ACh receptors (mAChR). Here, we explore the expression patterns of three nAChR subunits, chrna3, chrnb4, and chrna5, which are located in an evolutionary conserved cluster. This close genomic positioning, in a range of vertebrates, may indicate co-functionality and/or co-expression. Through novel transgenic zebrafish lines, we observe widespread expression within both the PNS and CNS. In the PNS, we observed expression of chrna3tdTomato, chrnb4eGFP, and chrna5tdTomato in the intestinal enteric nervous system; chrna5tdTomato and chrnb4eGFP in sensory ganglia of the lateral line; and chrnb4eGFP in the ear. In the CNS, the expression of chrnb4eGFP and chrna5tdTomato was found in the retina, all three expressed in diverse regions of the brain, where a portion of chrna3tdTomato and chrnb4eGFP cells were found to be inhibitory efferent neurons projecting to the lateral line. Within the spinal cord, we identify distinct populations of chrna3tdTomato-, chrnb4eGFP-, and chrna5tdTomato-expressing neurons within the locomotor network, including dmrt3a-expressing interneurons and mnx1-expressing motor neurons. Notably, three to four primary motor neurons per hemisegment were labeled by both chrna3tdTomato and chrnb4eGFP. Interestingly, we identified an sl-type secondary motor neuron per hemisegement that strongly expressed chrna5tdTomato and co-expressed chrnb4eGFP. These transgenic lines provide insights into the potential roles of nAChRs within the locomotor network and open avenues for exploring their role in nicotine exposure and addiction in a range of tissues throughout the nervous system.
Collapse
Affiliation(s)
- Yuanqi Hua
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Judith Habicher
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Matthias Carl
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Remy Manuel
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Henrik Boije
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
Hua Y, Zhang Y, Guo Z, Bian S, Zhang Y. ImSpiRE: image feature-aided spatial resolution enhancement method. SCIENCE CHINA. LIFE SCIENCES 2025; 68:272-283. [PMID: 39327391 DOI: 10.1007/s11427-023-2636-9] [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: 04/09/2024] [Accepted: 05/31/2024] [Indexed: 09/28/2024]
Abstract
The resolution of most spatially resolved transcriptomic technologies usually cannot attain the single-cell level, limiting their applications in biological discoveries. Here, we introduce ImSpiRE, an image feature-aided spatial resolution enhancement method for in situ capturing spatial transcriptome. Taking the information stored in histological images, ImSpiRE solves an optimal transport problem to redistribute the expression profiles of spots to construct new transcriptional profiles with enhanced resolution, together with extending the gene expression profiles into unmeasured regions. Applications to multiple datasets confirm that ImSpiRE can enhance spatial resolution to the subspot level while contributing to the discovery of tissue domains, signaling communication patterns, and spatiotemporal characterization.
Collapse
Affiliation(s)
- Yuwei Hua
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Institute for Regenerative Medicine, Department of Neurosurgery, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yizhi Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Institute for Regenerative Medicine, Department of Neurosurgery, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhenming Guo
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Institute for Regenerative Medicine, Department of Neurosurgery, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shan Bian
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Institute for Regenerative Medicine, Department of Neurosurgery, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yong Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Institute for Regenerative Medicine, Department of Neurosurgery, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
11
|
Chilaparasetti AN, Thai A, Gao P, Xu X, Gopi M. RegBoost: Enhancing mouse brain image registration using geometric priors and Laplacian interpolation. Neuroimage 2025; 305:120981. [PMID: 39732220 DOI: 10.1016/j.neuroimage.2024.120981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 11/18/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024] Open
Abstract
We show in this work that incorporating geometric features and geometry processing algorithms for mouse brain image registration broadens the applicability of registration algorithms and improves the registration accuracy of existing methods. We introduce the preprocessing and postprocessing steps in our proposed framework as RegBoost. We develop a method to align the axis of 3D image stacks by detecting the central planes that pass symmetrically through the image volumes. We then find geometric contours by defining external and internal structures to facilitate image correspondences. We establish Dirichlet boundary conditions at these correspondences and find the displacement map throughout the volume using Laplacian interpolation. We discuss the challenges in our standalone framework and demonstrate how our new approaches can improve the results of existing image registration methods. We expect our new approach and algorithms will have critical applications in brain mapping projects.
Collapse
Affiliation(s)
| | - Andy Thai
- Department of Computer Science, University of California, Irvine, Irvine, CA 92617, USA.
| | - Pan Gao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92617, USA.
| | - Xiangmin Xu
- Department of Computer Science, University of California, Irvine, Irvine, CA 92617, USA; Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92617, USA.
| | - M Gopi
- Department of Computer Science, University of California, Irvine, Irvine, CA 92617, USA.
| |
Collapse
|
12
|
Kim H, Park G, Shin HG, Kwon D, Kim H, Baek IY, Nam MH, Cho IJ, Kim J, Seong J. Optogenetic Control of Dopamine Receptor 2 Reveals a Novel Aspect of Dopaminergic Neurotransmission in Motor Function. J Neurosci 2025; 45:e1473242024. [PMID: 39562043 DOI: 10.1523/jneurosci.1473-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
Dopaminergic neurotransmission plays a crucial role in motor function through the coordination of dopamine receptor (DRD) subtypes, such as DRD1 and DRD2, thus the functional imbalance of these receptors can lead to Parkinson's disease. However, due to the complexity of dopaminergic circuits in the brain, it is limited to investigating the individual functions of each DRD subtype in specific brain regions. Here, we developed a light-responsive chimeric DRD2, OptoDRD2, which can selectively activate DRD2-like signaling pathways with spatiotemporal resolution. OptoDRD2 was designed to include the light-sensitive component of rhodopsin and the intracellular signaling domain of DRD2. Upon illumination with blue light, OptoDRD2 triggered DRD2-like signaling pathways, such as Gαi/o subtype recruitment, a decrease in cAMP levels, and ERK phosphorylation. To explore unknown roles of DRD2 in glutamatergic cell populations of basal ganglia circuitry, OptoDRD2 was genetically expressed in excitatory neurons in lateral globus pallidus (LGP) of the male mouse brain. The optogenetic stimulation of OptoDRD2 in the LGP region affected a wide range of locomotion-related parameters, such as increased frequency of movement and decreased immobility time, resulting in the facilitation of motor function of living male mice. Therefore, our findings indicate a potentially novel role for DRD2 in the excitatory neurons of the LGP region, suggesting that OptoDRD2 can be a valuable tool enabling the investigation of unknown roles of DRD2 at specific cell types or brain regions.
Collapse
Affiliation(s)
- Hyunbin Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Neuroscience Research Institute, Medical Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Geunhong Park
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Hyo Geun Shin
- School of Electronic and Electrical Engineering, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Duwan Kwon
- Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Heejung Kim
- Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Neuroscience Research Institute, Medical Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - In-Yeop Baek
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Il-Joo Cho
- Departments of Convergence Medicine, Korea University, Seoul 02841, Republic of Korea
- Anatomy, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Jeongjin Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jihye Seong
- Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Neuroscience Research Institute, Medical Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
13
|
Cashion JM, Brown LS, Morris GP, Fortune AJ, Courtney JM, Makowiecki K, Premilovac D, Cullen CL, Young KM, Sutherland BA. Pericyte ablation causes hypoactivity and reactive gliosis in adult mice. Brain Behav Immun 2025; 123:681-696. [PMID: 39406266 DOI: 10.1016/j.bbi.2024.10.014] [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: 10/23/2023] [Revised: 09/16/2024] [Accepted: 10/11/2024] [Indexed: 10/22/2024] Open
Abstract
Capillary pericytes are important regulators of cerebral blood flow, blood-brain barrier integrity and neuroinflammation, but can become lost or dysfunctional in disease. The consequences of pericyte loss or dysfunction is extremely difficult to discern when it forms one component of a complex disease process. To evaluate this directly, we examined the effect of adult pericyte loss on mouse voluntary movement and motor function, and physiological responses such as hypoxia, blood-brain barrier (BBB) integrity and glial reactivity. Tamoxifen delivery to Pdgfrβ-CreERT2:: Rosa26-DTA transgenic mice was titrated to produce a dose-dependent ablation of pericytes in vivo. 100mg/kg of tamoxifen ablated approximately half of all brain pericytes, while two consecutive daily doses of 300mg/kg tamoxifen ablated >80% of brain pericytes. In the open field test, mice with ∼50% pericyte loss spent more time immobile and travelled half the distance of control mice. Mice with >80% pericyte ablation also slipped more frequently while performing the beam walk task. Our histopathological analyses of the brain revealed that blood vessel density was unchanged, but vessel lumen width was increased. Pericyte-ablated mice also exhibited: mild BBB disruption; increased neuronal hypoxia; astrogliosis and increased IBA1+ immunoreactivity, suggestive of microgliosis and/or macrophage infiltration. Our results highlight the importance of pericytes in the brain, as pericyte loss can directly compromise brain health and induce behavioural alterations in mice.
Collapse
Affiliation(s)
- Jake M Cashion
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Lachlan S Brown
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Gary P Morris
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Alastair J Fortune
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Jo-Maree Courtney
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kalina Makowiecki
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Dino Premilovac
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.
| |
Collapse
|
14
|
Parag RR, Yamamoto T, Saito K, Zhu D, Yang L, Van Meir EG. Novel Isoforms of Adhesion G Protein-Coupled Receptor B1 (ADGRB1/BAI1) Generated from an Alternative Promoter in Intron 17. Mol Neurobiol 2025; 62:900-917. [PMID: 38941066 PMCID: PMC11711277 DOI: 10.1007/s12035-024-04293-3] [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: 02/24/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024]
Abstract
Brain-specific angiogenesis inhibitor 1 (BAI1) belongs to the adhesion G-protein-coupled receptors, which exhibit large multi-domain extracellular N termini that mediate cell-cell and cell-matrix interactions. To explore the existence of BAI1 isoforms, we queried genomic datasets for markers of active chromatin and new transcript variants in the ADGRB1 (adhesion G-protein-coupled receptor B1) gene. Two major types of mRNAs were identified in human/mouse brain, those with a start codon in exon 2 encoding a full-length protein of a predicted size of 173.5/173.3 kDa and shorter transcripts starting from alternative exons at the intron 17/exon 18 boundary with new or exon 19 start codons, predicting two shorter isoforms of 76.9/76.4 and 70.8/70.5 kDa, respectively. Immunoblots on wild-type and Adgrb1 exon 2-deleted mice, reverse transcription PCR, and promoter-luciferase reporter assay confirmed that the shorter isoforms originate from an alternative promoter in intron 17. The shorter BAI1 isoforms lack most of the N terminus and are very close in structure to the truncated BAI1 isoform generated through GPS processing from the full-length receptor. The cleaved BAI1 isoform has a 19 amino acid extracellular stalk that may serve as a receptor agonist, while the alternative transcripts generate BAI1 isoforms with extracellular N termini of 5 or 60 amino acids. Further studies are warranted to compare the functions of these isoforms and examine the distinct roles they play in different tissues and cell types.
Collapse
Affiliation(s)
- Rashed Rezwan Parag
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, Heersink School of Medicine, University of Alabama at Birmingham (UAB), WTI 520E, 1824 6th Avenue South, Birmingham, AL, 35233, USA
- Graduate Biomedical Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Takahiro Yamamoto
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, Heersink School of Medicine, University of Alabama at Birmingham (UAB), WTI 520E, 1824 6th Avenue South, Birmingham, AL, 35233, USA
- Department of Neurosurgery, Kumamoto University, Kumamoto, Japan
| | - Kiyotaka Saito
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, Heersink School of Medicine, University of Alabama at Birmingham (UAB), WTI 520E, 1824 6th Avenue South, Birmingham, AL, 35233, USA
| | - Dan Zhu
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Liquan Yang
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Erwin G Van Meir
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, Heersink School of Medicine, University of Alabama at Birmingham (UAB), WTI 520E, 1824 6th Avenue South, Birmingham, AL, 35233, USA.
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
| |
Collapse
|
15
|
Cipurko D, Ueda T, Mei L, Chevrier N. Repurposing large-format microarrays for scalable spatial transcriptomics. Nat Methods 2025; 22:145-155. [PMID: 39562752 DOI: 10.1038/s41592-024-02501-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/08/2024] [Indexed: 11/21/2024]
Abstract
Spatiomolecular analyses are key to study tissue functions and malfunctions. However, we lack profiling tools for spatial transcriptomics that are easy to adopt, low cost and scalable in terms of sample size and number. Here, we describe a method, Array-seq, to repurpose classical oligonucleotide microarrays for spatial transcriptomics profiling. We generate Array-seq slides from microarrays carrying custom-design probes that contain common sequences flanking unique barcodes at known coordinates. Then we perform a simple, two-step reaction that produces mRNA capture probes across all spots on the microarray. We demonstrate that Array-seq yields spatial transcriptomes with high detection sensitivity and localization specificity using histological sections from mouse tissues as test systems. Moreover, we show that the large surface area of Array-seq slides yields spatial transcriptomes (i) at high throughput by profiling multi-organ sections, (ii) in three dimensions by processing serial sections from one sample, and (iii) across whole human organs. Thus, by combining classical DNA microarrays and next-generation sequencing, we have created a simple and flexible platform for spatiomolecular studies of small-to-large specimens at scale.
Collapse
Affiliation(s)
- Denis Cipurko
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Medical Scientist Training Program, University of Chicago, Chicago, IL, USA
| | - Tatsuki Ueda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Linghan Mei
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
16
|
Dominguez G, Wu Y, Zhou J. Epigenetic Regulation and Neurodevelopmental Disorders: From MeCP2 to the TCF20/PHF14 Complex. Genes (Basel) 2024; 15:1653. [PMID: 39766920 DOI: 10.3390/genes15121653] [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: 12/01/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) affect approximately 15% of children and adolescents worldwide. This group of disorders is often polygenic with varying risk factors, with many associated genes converging on shared molecular pathways, including chromatin regulation and transcriptional control. Understanding how NDD-associated chromatin regulators and protein complexes orchestrate these regulatory pathways is crucial for elucidating NDD pathogenesis and developing targeted therapeutic strategies. Recently, the TCF20/PHF14 chromatin complex was identified in the mammalian brain, expanding the list of chromatin regulatory remodelers implicated in NDDs. This complex-which includes MeCP2, RAI1, TCF20, PHF14, and HMG20A-plays a vital role in epigenetic and transcriptional regulation. METHODS We review and summarize current research and clinical reports pertaining to the different components of the MeCP2-interacting TCF20/PHF14 complex. We examine the NDDs associated with the TCF20/PHF14 complex, explore the molecular and neuronal functions of its components, and discuss emerging therapeutic strategies targeting this complex to mitigate symptoms, with broader applicability to other NDDs. RESULTS Mutations in the genes encoding the components of the MeCP2-interacting TCF20/PHF14 complex have been linked to various NDDs, underscoring its critical contribution to brain development and NDD pathogenesis. CONCLUSIONS The MeCP2-interacting TCF20/PHF14 complex and its associated NDDs could serve as a model system to provide insight into the interplay between epigenetic regulation and NDD pathogenesis.
Collapse
Affiliation(s)
- Gaea Dominguez
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yongji Wu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jian Zhou
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
17
|
Kidd BM, Varholick JA, Tuyn DM, Kamat PK, Simon ZD, Liu L, Mekler MP, Pompilus M, Bubenik JL, Davenport ML, Carter HA, Grudny MM, Barbazuk WB, Doré S, Febo M, Candelario-Jalil E, Maden M, Swanson MS. Stroke-induced neuroplasticity in spiny mice in the absence of tissue regeneration. NPJ Regen Med 2024; 9:41. [PMID: 39706830 DOI: 10.1038/s41536-024-00386-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024] Open
Abstract
Stroke is a major cause of disability for adults over 40 years of age. While research into animal models has prioritized treatments aimed at diminishing post-stroke damage, no studies have investigated the response to a severe stroke injury in a highly regenerative adult mammal. Here we investigate the effects of transient ischemia on adult spiny mice, Acomys cahirinus, due to their ability to regenerate multiple tissues without scarring. Transient middle cerebral artery occlusion was performed and Acomys showed rapid behavioral recovery post-stroke yet failed to regenerate impacted brain regions. An Acomys brain atlas in combination with functional (f)MRI demonstrated recovery coincides with neuroplasticity. The strength and quality of the global connectome are preserved post-injury with distinct contralateral and ipsilateral brain regions compensating for lost tissue. Thus, we propose Acomys recovers functionally from an ischemic stroke injury not by tissue regeneration but by altering its brain connectome.
Collapse
Affiliation(s)
- Benjamin M Kidd
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Justin A Varholick
- Department of Biology, College of Liberal Arts and Sciences and the Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Dana M Tuyn
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Pradip K Kamat
- Departments of Anesthesiology, Neurology, Psychology, and Pharmaceutics, Center for Translational Research in Neurodegenerative Disease, and the College of Medicine, University of Florida, Gainesville, FL, USA
| | - Zachary D Simon
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Psychiatry and the McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lei Liu
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Mackenzie P Mekler
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Marjory Pompilus
- Department of Psychiatry and the McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jodi L Bubenik
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Mackenzie L Davenport
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Helmut A Carter
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Matteo M Grudny
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Psychiatry and the McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - W Brad Barbazuk
- Department of Biology, College of Liberal Arts and Sciences and the Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Sylvain Doré
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Departments of Anesthesiology, Neurology, Psychology, and Pharmaceutics, Center for Translational Research in Neurodegenerative Disease, and the College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Psychiatry and the McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Marcelo Febo
- Department of Psychiatry and the McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Malcolm Maden
- Department of Biology, College of Liberal Arts and Sciences and the Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
18
|
Saenz C, Fernandez G, Llovera R, Tolosa MJ, Cantel S, Fehrentz JA, Mackie K, Leggio L, Zigman J, De Francesco PN, Perello M. Growth hormone secretagogue receptor and cannabinoid receptor type 1 intersection in the mouse brain. Brain Struct Funct 2024; 230:15. [PMID: 39702649 DOI: 10.1007/s00429-024-02876-3] [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: 08/01/2024] [Accepted: 10/14/2024] [Indexed: 12/21/2024]
Abstract
The growth hormone secretagogue receptor (GHSR) and the cannabinoid receptor type 1 (CB1R) are G-protein coupled receptors highly expressed in the brain and involved in critical regulatory processes, such as energy homeostasis, appetite control, reward, and stress responses. GHSR mediates the effects of both ghrelin and liver-expressed antimicrobial peptide 2, while CB1R is targeted by cannabinoids. Strikingly, both receptors mediate their effects by acting on common brain areas and their individual roles have been well characterized. However, the potential for their co-expression in the same neuronal subsets remains largely unexplored. Here, we aim to map the cell populations where GHSR and CB1R might converge, hypothesizing that their co-expression in specific brain circuits could mediate integrated physiological responses. By utilizing two complementary labeling techniques-GHSR-eGFP mice and Fr-ghrelin labeling of GHSR+ cells-along with specific CB1R immunostaining, we sought to visualize and quantify potential areas of overlap. Also, we analyzed several cell RNA sequencing datasets to estimate the fraction of brain cells expressing both GPCRs and their phenotype. Our neuroanatomical studies revealed evident overlap of GHSR+ and CB1R+ signals in specific neuronal subsets mainly located in the cerebral cortex, hippocampus and the amygdala. Transcriptomic analysis revealed specific subsets of Ghsr+/Cnr1+ glutamatergic neurons in the hippocampus and amygdala, as well as different subtypes of Ghsr+/Cnr1+ neurons in the midbrain, hypothalamus, pons, and medulla. Thus, we revealed that GHSR and CB1R interact differentially across specific regions of the mouse brain, providing new insights into how these receptors' actions are integrated. Current findings may open new avenues for dual therapeutic interventions in metabolic disorders, obesity, and psychiatric conditions.
Collapse
Affiliation(s)
- Camila Saenz
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Ramiro Llovera
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - María J Tolosa
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Kenneth Mackie
- Department of Psychological & Brain Sciences, Indiana University, Bloomington Indiana, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Jeffrey Zigman
- Center for Hypothalamic Research and Division of Endocrinology & Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pablo N De Francesco
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina.
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina.
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
19
|
Harke J, Lee JR, Nguyen SC, Arab A, Rakowiecki SM, Hugelier S, Paliou C, Rauseo A, Yunker R, Xu K, Yao Y, Lakadamyali M, Andrey G, Epstein DJ, Joyce EF. Multiple allelic configurations govern long-range Shh enhancer-promoter communication in the embryonic forebrain. Mol Cell 2024; 84:4698-4710.e6. [PMID: 39579767 DOI: 10.1016/j.molcel.2024.10.042] [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: 09/13/2023] [Revised: 08/30/2024] [Accepted: 10/30/2024] [Indexed: 11/25/2024]
Abstract
Developmental gene regulation requires input from enhancers spread over large genomic distances. Our understanding of long-range enhancer-promoter (E-P) communication, characterized as loops, remains incomplete without addressing the role of intervening chromatin. Here, we examine the topology of the entire Sonic hedgehog (Shh) regulatory domain in individual alleles from the mouse embryonic forebrain. Through sequential Oligopaint labeling and super-resolution microscopy, we find that the Shh locus maintains a compact structure that adopts several diverse configurations independent of Shh expression. The most frequent configuration contained distal E-P contacts at the expense of those more proximal to Shh, consistent with an interconnected loop. Genetic perturbations demonstrate that this long-range E-P communication operates by Shh-expression-independent and dependent mechanisms, involving CTCF binding sites and active enhancers, respectively. We propose a model whereby gene regulatory elements secure long-range E-P interactions amid an inherent architectural framework to coordinate spatiotemporal patterns of gene expression.
Collapse
Affiliation(s)
- Jailynn Harke
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeewon R Lee
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Son C Nguyen
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arian Arab
- Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Staci M Rakowiecki
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Siewert Hugelier
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christina Paliou
- Andalusian Center for Developmental Biology (CABD), Spanish National Research Council, Pablo de Olavide University, Andalusian Regional Government, Seville, Spain
| | - Antonella Rauseo
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Rebecca Yunker
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kellen Xu
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Yao Yao
- Department of Animal and Dairy Science, Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Melike Lakadamyali
- Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guillaume Andrey
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Douglas J Epstein
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Eric F Joyce
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Hardy M, Chen Y, Baram TZ, Justice NJ. Targeting corticotropin-releasing hormone receptor type 1 (Crhr1) neurons: validating the specificity of a novel transgenic Crhr1-FlpO mouse. Brain Struct Funct 2024; 230:12. [PMID: 39692887 DOI: 10.1007/s00429-024-02879-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/05/2024] [Indexed: 12/19/2024]
Abstract
Corticotropin-releasing hormone (CRH) signaling through its cognate receptors, CRHR1 and CRHR2, contributes to diverse stress-related functions in the mammalian brain. Whereas CRHR2 is predominantly expressed in choroid plexus and blood vessels, CRHR1 is abundantly expressed in neurons in discrete brain regions, including the neocortex, hippocampus and nucleus accumbens. Activation of CRHR1 influences motivated behaviors, emotional states, and learning and memory. However, it is unknown whether alterations in CRHR1 signaling contribute to aberrant motivated behaviors observed, for example, in stressful contexts. These questions require tools to manipulate CRHR1 selectively. Here we describe and validate a novel Crhr1-FlpO mouse. Using bacterial artificial chromosome (BAC) transgenesis, we engineered a transgenic mouse that expresses FlpO recombinase in CRHR1-expressing cells. We used two independent methods to assess the specificity of FlpO to CRHR1-expressing cells. First, we injected Crhr1-FlpO mice with Flp-dependent viruses expressing fluorescent reporter molecules. Additionally, we crossed the Crhr1-FlpO mouse with a transgenic Flp-dependent reporter mouse. CRHR1 and reporter molecules were identified using immunocytochemistry and visualized via confocal microscopy in several brain regions in which CRHR1 expression and function is established. Expression of Flp-dependent viral constructs was highly specific to CRHR1-expressing cells in all regions examined (over 90% co-localization). In accord, robust and specific expression of the Flp-dependent transgenic reporter was observed in a reporter mouse, recapitulating endogenous CRHR1 expression. The Crhr1-FlpO mouse enables selective genetic access to CRHR1-expressing cells within the mouse brain. When combined with Cre-lox or site-specific recombinases, the mouse facilitates intersectional manipulations of CRHR1-expressing neurons.
Collapse
Affiliation(s)
- Mason Hardy
- Department of Anatomy & Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - Yuncai Chen
- Department of Anatomy & Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Anatomy & Neurobiology, University of California-Irvine, Irvine, CA, USA.
- Departments of Pediatrics and Neurology, University of California-Irvine, Irvine, CA, USA.
| | - Nicholas J Justice
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Sciences Center-Houston, Houston, TX, USA
| |
Collapse
|
21
|
Sun ED, Zhou OY, Hauptschein M, Rappoport N, Xu L, Navarro Negredo P, Liu L, Rando TA, Zou J, Brunet A. Spatial transcriptomic clocks reveal cell proximity effects in brain ageing. Nature 2024:10.1038/s41586-024-08334-8. [PMID: 39695234 DOI: 10.1038/s41586-024-08334-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
Old age is associated with a decline in cognitive function and an increase in neurodegenerative disease risk1. Brain ageing is complex and is accompanied by many cellular changes2. Furthermore, the influence that aged cells have on neighbouring cells and how this contributes to tissue decline is unknown. More generally, the tools to systematically address this question in ageing tissues have not yet been developed. Here we generate a spatially resolved single-cell transcriptomics brain atlas of 4.2 million cells from 20 distinct ages across the adult lifespan and across two rejuvenating interventions-exercise and partial reprogramming. We build spatial ageing clocks, machine learning models trained on this spatial transcriptomics atlas, to identify spatial and cell-type-specific transcriptomic fingerprints of ageing, rejuvenation and disease, including for rare cell types. Using spatial ageing clocks and deep learning, we find that T cells, which increasingly infiltrate the brain with age, have a marked pro-ageing proximity effect on neighbouring cells. Surprisingly, neural stem cells have a strong pro-rejuvenating proximity effect on neighbouring cells. We also identify potential mediators of the pro-ageing effect of T cells and the pro-rejuvenating effect of neural stem cells on their neighbours. These results suggest that rare cell types can have a potent influence on their neighbours and could be targeted to counter tissue ageing. Spatial ageing clocks represent a useful tool for studying cell-cell interactions in spatial contexts and should allow scalable assessment of the efficacy of interventions for ageing and disease.
Collapse
Affiliation(s)
- Eric D Sun
- Biomedical Data Science Graduate Program, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Olivia Y Zhou
- Department of Genetics, Stanford University, Stanford, CA, USA
- Biophysics Graduate Program, Stanford University, Stanford, CA, USA
- Medical Scientist Training Program, Stanford University, Stanford, CA, USA
| | - Max Hauptschein
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Lucy Xu
- Department of Genetics, Stanford University, Stanford, CA, USA
- Biology Graduate Program, Stanford University, Stanford, CA, USA
| | | | - Ling Liu
- Department of Neurology, Stanford University, Stanford, CA, USA
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, UCLA, Los Angeles, CA, USA
| | - Thomas A Rando
- Department of Neurology, Stanford University, Stanford, CA, USA
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, UCLA, Los Angeles, CA, USA
| | - James Zou
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Glenn Center for the Biology of Aging, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
22
|
Elhabbari K, Sireci S, Rothermel M, Brunert D. Olfactory deficits in aging and Alzheimer's-spotlight on inhibitory interneurons. Front Neurosci 2024; 18:1503069. [PMID: 39737436 PMCID: PMC11683112 DOI: 10.3389/fnins.2024.1503069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
Cognitive function in healthy aging and neurodegenerative diseases like Alzheimer's disease (AD) correlates to olfactory performance. Aging and disease progression both show marked olfactory deficits in humans and rodents. As a clear understanding of what causes olfactory deficits is still missing, research on this topic is paramount to diagnostics and early intervention therapy. A recent development of this research is focusing on GABAergic interneurons. Both aging and AD show a change in excitation/inhibition balance, indicating reduced inhibitory network functions. In the olfactory system, inhibition has an especially prominent role in processing information, as the olfactory bulb (OB), the first relay station of olfactory information in the brain, contains an unusually high number of inhibitory interneurons. This review summarizes the current knowledge on inhibitory interneurons at the level of the OB and the primary olfactory cortices to gain an overview of how these neurons might influence olfactory behavior. We also compare changes in interneuron composition in different olfactory brain areas between healthy aging and AD as the most common neurodegenerative disease. We find that pathophysiological changes in olfactory areas mirror findings from hippocampal and cortical regions that describe a marked cell loss for GABAergic interneurons in AD but not aging. Rather than differences in brain areas, differences in vulnerability were shown for different interneuron populations through all olfactory regions, with somatostatin-positive cells most strongly affected.
Collapse
Affiliation(s)
| | | | | | - Daniela Brunert
- Institute of Physiology, RG Neurophysiology and Optogenetics, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
23
|
Alitalo O, Kohtala S, Rosenholm M, Saarreharju R, González-Hernández G, Sarparanta M, Rozov S, Rantamäki T. Nitrous oxide induces hypothermia and TrkB activation: Maintenance of body temperature abolishes antidepressant-like effects in mice. Neuropharmacology 2024; 261:110172. [PMID: 39362627 DOI: 10.1016/j.neuropharm.2024.110172] [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: 08/20/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
Recent studies indicate that nitrous oxide (N2O), a gaseous anesthetic and an NMDA (N-methyl-D-aspartate) receptor antagonist, produces rapid antidepressant effect in patients suffering from treatment-resistant depression. Our recent work implies that hypothermia and reduced energy expenditure are connected with antidepressant-induced activation of TrkB neurotrophin receptors - a key regulator of synaptic plasticity. In this study, we demonstrate that a brief exposure to N2O leads to a drop in body temperature following the treatment, which is linked to decreased locomotor activity; enhanced slow-wave electroencephalographic activity; reduced brain glucose utilization; and increased phosphorylation of TrkB, GSK3β (glycogen synthase kinase 3β), and p70S6K (a kinase downstream of mTor (mammalian target of rapamycin)) in the medial prefrontal cortex of adult male mice. Moreover, preventing the hypothermic response in a chronic corticosterone stress model of depression attenuated the antidepressant-like behavioral effects of N2O in the saccharin preference test. These findings indicate that N2O treatment modulates TrkB signaling and related neurotrophic signaling pathways in a temperature-dependent manner, suggesting that the phenomenon driving TrkB activation - altered thermoregulation and energy expenditure - is linked to antidepressant-like behavioral responses.
Collapse
Affiliation(s)
- Okko Alitalo
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Samuel Kohtala
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Marko Rosenholm
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland; Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Roosa Saarreharju
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Gemma González-Hernández
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland; Neuropsychopharmacology and Psychobiology Research Group, Department of Neurosciences, University of Cádiz, Cádiz, Spain
| | | | - Stanislav Rozov
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Tomi Rantamäki
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|
24
|
Wu L, Liu X, Jiang Q, Li M, Liang M, Wang S, Wang R, Su L, Ni T, Dong N, Zhu L, Guan F, Zhu J, Zhang W, Wu M, Chen Y, Chen T, Wang B. Methamphetamine-induced impairment of memory and fleeting neuroinflammation: Profiling mRNA changes in mouse hippocampus following short-term and long-term exposure. Neuropharmacology 2024; 261:110175. [PMID: 39357738 DOI: 10.1016/j.neuropharm.2024.110175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/20/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Methamphetamine (METH) has been implicated in inducing memory impairment, but the precise mechanisms underlying this effect remain unclear. Current research often limits itself to singular models or focuses on individual gene or protein functions, which hampers a comprehensive understanding of the underlying mechanisms. In this study, we established three METH mouse exposure models, extracted hippocampal nuclei, and utilized RNA sequencing to analyze changes in mRNA expression profiles. Our results indicate that METH significantly impairs the learning and memory capabilities of mice. Additionally, we observed that METH-induced inflammatory responses occur in the early phase and do not further exacerbate with repeated injections. However, RNA sequencing revealed the persistent enrichment of inflammatory pathway molecules, which correlated with worsened behaviors. This suggests that although METH-induced neuroinflammation plays a critical role in learning and memory impairment, the continued enrichment of inflammatory pathway molecules is associated with behavioral outcomes. These findings provide crucial evidence for the potential application of immune intervention in METH-related disorders.
Collapse
Affiliation(s)
- Laiqiang Wu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Xiaorui Liu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Qingchen Jiang
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ming Li
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Min Liang
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Shuai Wang
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Rui Wang
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Linlan Su
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Tong Ni
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Nan Dong
- School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Li Zhu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Fanglin Guan
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Jie Zhu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Wen Zhang
- Department of Pathology, Northwest Women's and Children's Hospital, Xi'an, China
| | - Min Wu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Yanjiong Chen
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Teng Chen
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China.
| | - Biao Wang
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.
| |
Collapse
|
25
|
Watanabe S, Lee S, Harb M, Nouraein S, Raisley E, Li H, Buitrago N, Pforr B, Szablowski JO. Monitoring in vivo transcription with synthetic serum markers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627810. [PMID: 39713329 PMCID: PMC11661152 DOI: 10.1101/2024.12.10.627810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Understanding transcription profiles of living tissues is critical for biology and medicine. However, measurement of the transcript levels is typically done in homogenized tissues post-mortem. Here, we present a new platform that enables non-invasive monitoring of specific mRNA levels in vivo , without tissue destruction. We achieved this by combining two cutting-edge tools - synthetic serum markers, called Released Markers of Activity ( RMAs ), and RNA-based sensors of transcription. We call this platform IN-vivo Tracking of ACtive Transcription, or INTACT . In INTACT, when the target mRNA is expressed, the RNA sensor detects it and triggers the production and release of RMA reporters into the blood. Once in blood, the RMAs can be easily measured through a simple blood draw. Our data shows that INTACT can measure transcription of transgenes, as well as endogenous transcripts, such as c-Fos or Arc , both in vivo in the brain and in tissue culture. INTACT enables simple measurement of transcript level histories in genetically-targetable cell populations of living animals.
Collapse
|
26
|
Yang L, Fang LZ, Lynch MR, Xu CS, Hahm HJ, Zhang Y, Heitmeier MR, Costa VD, Samineni VK, Creed MC. Transcriptomic landscape of mammalian ventral pallidum at single-cell resolution. SCIENCE ADVANCES 2024; 10:eadq6017. [PMID: 39661664 PMCID: PMC11633743 DOI: 10.1126/sciadv.adq6017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
The ventral pallidum (VP) is critical for motivated behaviors. While contemporary work has begun to elucidate the functional diversity of VP neurons, the molecular heterogeneity underlying this functional diversity remains incompletely understood. We used single-nucleus RNA sequencing and in situ hybridization to define the transcriptional taxonomy of VP cell types in mice, macaques, and baboons. We found transcriptional conservation between all three species, within the broader neurochemical cell types. Unique dopaminoceptive and cholinergic subclusters were identified and conserved across both primate species but had no homolog in mice. This harmonized consensus VP cellular atlas will pave the way for understanding the structure and function of the VP and identified key neuropeptides, neurotransmitters, and neurotransmitter receptors that could be targeted within specific VP cell types for functional investigations.
Collapse
Affiliation(s)
- Lite Yang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Lisa Z. Fang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Michelle R. Lynch
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- NINDS Neuroscience Postbaccalaureate Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Chang S. Xu
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah J. Hahm
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yufen Zhang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Monique R. Heitmeier
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Vincent D. Costa
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
- Division of Developmental and Cognitive Neuroscience, Emory National Primate Research Center, Atlanta, GA, USA
| | - Vijay K. Samineni
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Meaghan C. Creed
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
27
|
Yaguchi K, Miyamichi K, Tasaka GI. Flexible adjustment of oxytocin neuron activity in mouse dams revealed by microendoscopy. SCIENCE ADVANCES 2024; 10:eadt1555. [PMID: 39671475 PMCID: PMC11641008 DOI: 10.1126/sciadv.adt1555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/08/2024] [Indexed: 12/15/2024]
Abstract
Oxytocin (OT) neurons in the hypothalamic paraventricular nucleus (PVH) play an important role in various physiological and behavioral processes, including the initiation of milk ejection and the regulation of maternal behaviors. However, their activity patterns at the single-cell level remain poorly understood. Using microendoscopic Ca2+ imaging in freely moving mouse dams, we demonstrate highly correlated pulsatile activity among individual OT neurons during lactation. The number of OT neurons engaged in the pulsatile activity significantly increased, along with a broadening of individual waveforms in the mid-lactation stage. Notably, only ~10% of the imaged OT neurons exhibited a significantly elevated response during pup retrieval, a hallmark of maternal behaviors, with a magnitude smaller than that observed during lactation. Collectively, these findings demonstrate the utility of microendoscopic imaging for PVH OT neurons and highlight the flexible adjustments of their individual activity patterns in freely behaving mouse dams.
Collapse
Affiliation(s)
- Kasane Yaguchi
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Kazunari Miyamichi
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Gen-ichi Tasaka
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
- Japan Science and Technology Agency, PRESTO, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
28
|
Milisav F, Bazinet V, Betzel RF, Misic B. A simulated annealing algorithm for randomizing weighted networks. NATURE COMPUTATIONAL SCIENCE 2024:10.1038/s43588-024-00735-z. [PMID: 39658626 DOI: 10.1038/s43588-024-00735-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 11/01/2024] [Indexed: 12/12/2024]
Abstract
Scientific discovery in connectomics relies on network null models. The prominence of network features is conventionally evaluated against null distributions estimated using randomized networks. Modern imaging technologies provide an increasingly rich array of biologically meaningful edge weights. Despite the prevalence of weighted graph analysis in connectomics, randomization models that only preserve binary node degree remain most widely used. Here we propose a simulated annealing procedure for generating randomized networks that preserve weighted degree (strength) sequences. We show that the procedure outperforms other rewiring algorithms and generalizes to multiple network formats, including directed and signed networks, as well as diverse real-world networks. Throughout, we use morphospace representation to assess the sampling behavior of the algorithm and the variability of the resulting ensemble. Finally, we show that accurate strength preservation yields different inferences about brain network organization. Collectively, this work provides a simple but powerful method to analyze richly detailed next-generation connectomics datasets.
Collapse
Affiliation(s)
- Filip Milisav
- Montréal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Vincent Bazinet
- Montréal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Richard F Betzel
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Bratislav Misic
- Montréal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
29
|
Harima Y, Tsurutani M, Yamada S, Uchida S, Inada K, Hagihara M, Irie S, Shigeta M, Abe T, Inoue YU, Inoue T, Miyamichi K. Parallel labeled-line organization of sympathetic outflow for selective organ regulation in mice. Nat Commun 2024; 15:10478. [PMID: 39658565 PMCID: PMC11631959 DOI: 10.1038/s41467-024-54928-1] [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: 05/11/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024] Open
Abstract
The sympathetic nervous system is crucial for responding to environmental changes. This regulation is coordinated by the spinal sympathetic preganglionic neurons (SPNs), innervating both postganglionic neurons and the adrenal gland. Despite decades of research supporting the concept of selective control within this system, the neural circuit organization responsible for the output specificity remains poorly understood. Here, by combining recent single-cell transcriptome data with viral-genetic toolkits in mice, we identify two subtypes of SPNs in the lower thoracic spinal cord, defined at the molecular level, exhibiting nonoverlapping patterns of innervation: one specifically projecting to the celiac/superior mesenteric ganglia, and the other targeting the adrenal grand. Chemogenetic manipulations on these distinct SPN subtypes revealed selective impacts on the motility of the gastrointestinal tracts or glucose metabolism mediated by the adrenal gland, respectively. This molecularly delineated parallel labeled-line organization in sympathetic outflows presents a potential avenue for selectively manipulating organ functions.
Collapse
Affiliation(s)
- Yukiko Harima
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
| | - Masafumi Tsurutani
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Serika Yamada
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Shuntaro Uchida
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Kengo Inada
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Mitsue Hagihara
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Satsuki Irie
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Mayo Shigeta
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Yukiko U Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Takayoshi Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Kazunari Miyamichi
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
| |
Collapse
|
30
|
Khan AH, Bagley JR, LaPierre N, Gonzalez-Figueroa C, Spencer TC, Choudhury M, Xiao X, Eskin E, Jentsch JD, Smith DJ. Differing genetics of saline and cocaine self-administration in the hybrid mouse diversity panel. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626933. [PMID: 39713377 PMCID: PMC11661131 DOI: 10.1101/2024.12.04.626933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
To identify genes involved in regulating the behavioral and brain transcriptomic response to the potentially addictive drug cocaine, we performed genome-wide association studies (GWASs) for intravenous self-administration of cocaine or saline (as a control) over 10 days using a panel of inbred and recombinant inbred mice. A linear mixed model increased statistical power for these longitudinal data and identified 145 loci for responding when saline only was delivered, compared to 17 for the corresponding cocaine GWAS. Only one locus overlapped. Transcriptome-wide association studies (TWASs) using RNA-Seq data from the medial frontal cortex and nucleus accumbens identified 5031434O11Rik and Zfp60 as significant for saline self-administration. Two other genes, Myh4 and Npc1, were nominated based on proximity to loci for multiple endpoints or a cis locus regulating expression. All four genes have previously been implicated in locomotor activity. Our results indicate distinct genetic bases for saline and cocaine self-administration, and suggest some common genes for saline self-administration and locomotor activity.
Collapse
Affiliation(s)
- Arshad H Khan
- Department of Molecular and Medical Pharmacology, Geffen School of Medicine, UCLA, Los Angeles, CA 90095
- Current address: Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048
| | - Jared R Bagley
- Department of Psychology, Binghamton University, Binghamton, NY 13902
- Current address: Department of Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902
| | - Nathan LaPierre
- Department of Computer Science, UCLA, Los Angeles, CA 90095
- Current address: Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | | | - Tadeo C Spencer
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095
| | - Mudra Choudhury
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095
- Current address: Sanford Burnham Prebys, La Jolla, CA 92037
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095
| | - Eleazar Eskin
- Department of Computational Medicine, UCLA, Los Angeles, CA 90095
| | - James D Jentsch
- Department of Psychology, Binghamton University, Binghamton, NY 13902
| | - Desmond J Smith
- Department of Molecular and Medical Pharmacology, Geffen School of Medicine, UCLA, Los Angeles, CA 90095
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Box 951735, 23-151A CHS, Los Angeles, CA 90095-1735
| |
Collapse
|
31
|
Morrison LM, Huang H, Handler HP, Fu M, Jones DM, Bushart DD, Pappas SS, Orr HT, Shakkottai VG. Increased intrinsic membrane excitability is associated with olivary hypertrophy in spinocerebellar ataxia type 1. Hum Mol Genet 2024; 33:2159-2176. [PMID: 39475127 PMCID: PMC11630738 DOI: 10.1093/hmg/ddae146] [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: 06/17/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024] Open
Abstract
One of the characteristic regions of brainstem degeneration across multiple spinocerebellar ataxias (SCAs) is the inferior olive (IO), a medullary nucleus that plays a key role in motor learning. The vulnerability of IO neurons remains a poorly-understood area of SCA pathology. In this work, we address this by evaluating IO disease in SCA1, a prototypic inherited olivopontocerebellar atrophy, using the genetically-precise SCA1 knock-in (SCA1-KI) mouse. We find that these mice exhibit olivary hypertrophy, a phenotype reminiscent of a degenerative disorder known as hypertrophic olivary degeneration (HOD). Similar to early stages of HOD, SCA1-KI IO neurons display early dendritic lengthening and later somatic expansion without frank cell loss. Though HOD is known to be caused by brainstem lesions that disrupt IO inhibitory innervation, we observe no loss of inhibitory terminals in the SCA1-KI IO. Additionally, we find that a separate mouse model of SCA1 in which mutant ATXN1 is expressed solely in cerebellar Purkinje cells shows no evidence of olivary hypertrophy. Patch-clamp recordings from brainstem slices indicate that SCA1-KI IO neurons are hyperexcitable, generating spike trains in response to membrane depolarization. Transcriptome analysis further reveals reduced medullary expression of ion channels responsible for IO neuron spike afterhyperpolarization (AHP)-a result that appears to have a functional consequence, as SCA1-KI IO neuron spikes exhibit a diminished AHP. These findings suggest that expression of mutant ATXN1 in IO neurons results in an HOD-like olivary hypertrophy, in association with increased intrinsic membrane excitability and ion channel transcriptional dysregulation.
Collapse
Affiliation(s)
- Logan M Morrison
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd. Dallas, TX 75390, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., TX 75390, United States
| | - Haoran Huang
- Medical Scientist Training Program, The Ohio State University, Columbus, OH 43210, United States
- College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Hillary P Handler
- Molecular Diagnostics Laboratory, University of Minnesota Fairview Medical Center, Minneapolis, MN 55455, United States
| | - Min Fu
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd. Dallas, TX 75390, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., TX 75390, United States
| | - Deborah M Jones
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., TX 75390, United States
| | - David D Bushart
- College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Samuel S Pappas
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd. Dallas, TX 75390, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., TX 75390, United States
| | - Harry T Orr
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, 420 Delaware Street SE, MN 55455, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, United States
| | - Vikram G Shakkottai
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd. Dallas, TX 75390, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., TX 75390, United States
| |
Collapse
|
32
|
Wang Z, Li L, Li M, Lu Z, Qin L, Naumann RK, Wang H. Chemogenetic Modulation of Preoptic Gabre Neurons Decreases Body Temperature and Heart Rate. Int J Mol Sci 2024; 25:13061. [PMID: 39684772 DOI: 10.3390/ijms252313061] [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: 09/30/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 12/18/2024] Open
Abstract
The preoptic area of the hypothalamus is critical for regulation of brain-body interaction, including circuits that control vital signs such as body temperature and heart rate. The preoptic area contains approximately 70 molecularly distinct cell types. The Gabre gene is expressed in a subset of preoptic area cell types. It encodes the GABA receptor ε-subunit, which is thought to confer resistance to anesthetics at the molecular level, but the function of Gabre cells in the brain remains largely unknown. We generated and have extensively characterized a Gabre-cre knock-in mouse line and used chemogenetic tools to interrogate the function of Gabre cells in the preoptic area. Comparison with macaque GABRE expression revealed the conserved character of Gabre cells in the preoptic area. In awake mice, we found that chemogenetic activation of Gabre neurons in the preoptic area reduced body temperature, whereas chemogenetic inhibition had no effect. Furthermore, chemogenetic inhibition of Gabre neurons in the preoptic area decreased the heart rate, whereas chemogenetic activation had no effect under isoflurane anesthesia. These findings suggest an important role of preoptic Gabre neurons in maintaining vital signs such as body temperature and heart rate during wakefulness and under anesthesia.
Collapse
Affiliation(s)
- Ziyue Wang
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Lanxiang Li
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Miao Li
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Zhonghua Lu
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Lihua Qin
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Robert Konrad Naumann
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Hong Wang
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| |
Collapse
|
33
|
Lu Z, Chan SW, Jiang B, Cui D, Sakata I, Sakai T, Huang X, Liu JYH, Chan TWD, Rudd JA. Action of cocaine- and amphetamine-regulated transcript (CART) peptide to attenuate cisplatin-induced emesis in Suncus murinus (house musk shrew). Eur J Pharmacol 2024; 984:177072. [PMID: 39447859 DOI: 10.1016/j.ejphar.2024.177072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Cocaine- and amphetamine-regulated transcript (CART) peptide is a brain-gut neuropeptide that has been implicated in a range of physiological functions including appetite, which is disturbed during chemotherapy. The aims of the present study were to identify the distribution and expression of CART mRNA and CART peptide, and to examine the potential of CART (55-102) to attenuate cisplatin-induced emesis in Suncus murinus. CART mRNA and peptide were detected throughout the entire brain, including the forebrain, hypothalamus, and brainstem, and also in the gut wall and stomach. In conscious, freely moving animals, intracerebroventricular administration of CART (55-102) did not modulate food and water intake or alter locomotor activity when administered alone. Cisplatin induced emesis and upregulated the expression of CART mRNA in the brainstem. However, CART (55-102) reduced the number of cisplatin-induced retches. Both CART (55-102) and cisplatin increased the number of c-Fos positive cells in the nucleus tractus solitarius, lateral hypothalamus, paraventricular hypothalamus, and bed nucleus of the stria terminalis (BNST), compared to saline-treated animals, whereas cisplatin also induced c-Fos expression in the area postrema (AP), arcuate nucleus, and central nucleus of the amygdala. Pre-treatment with CART (55-102) significantly attenuated the increased c-Fos positive cells in the BNST and AP. These data indicate that CART mRNA and peptide were localized to regions involved in reward/enforcement, emotion, feeding and emesis. The anti-emetic effect of CART (55-102) against cisplatin-induced emesis may involve both the forebrain limbic system and the brainstem.
Collapse
Affiliation(s)
- Zengbing Lu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sze Wa Chan
- School of Health Sciences, Saint Francis University, Hong Kong SAR, China.
| | - Bin Jiang
- School of Health Sciences, Saint Francis University, Hong Kong SAR, China
| | - Dexuan Cui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ichiro Sakata
- Graduate School of Science and Engineering, Saitama University, Japan
| | - Takafumi Sakai
- Graduate School of Science and Engineering, Saitama University, Japan
| | - Xiaofei Huang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Health Sciences, Saint Francis University, Hong Kong SAR, China
| | - Julia Yuen Hang Liu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tak Wah Dominic Chan
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - John A Rudd
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
34
|
Patiño M, Rossa MA, Lagos WN, Patne NS, Callaway EM. Transcriptomic cell-type specificity of local cortical circuits. Neuron 2024; 112:3851-3866.e4. [PMID: 39353431 PMCID: PMC11624072 DOI: 10.1016/j.neuron.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/02/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
Complex neocortical functions rely on networks of diverse excitatory and inhibitory neurons. While local connectivity rules between major neuronal subclasses have been established, the specificity of connections at the level of transcriptomic subtypes remains unclear. We introduce single transcriptome assisted rabies tracing (START), a method combining monosynaptic rabies tracing and single-nuclei RNA sequencing to identify transcriptomic cell types, providing inputs to defined neuron populations. We employ START to transcriptomically characterize inhibitory neurons providing monosynaptic input to 5 different layer-specific excitatory cortical neuron populations in mouse primary visual cortex (V1). At the subclass level, we observe results consistent with findings from prior studies that resolve neuronal subclasses using antibody staining, transgenic mouse lines, and morphological reconstruction. With improved neuronal subtype granularity achieved with START, we demonstrate transcriptomic subtype specificity of inhibitory inputs to various excitatory neuron subclasses. These results establish local connectivity rules at the resolution of transcriptomic inhibitory cell types.
Collapse
Affiliation(s)
- Maribel Patiño
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA; Medical Scientist Training Program, University of California, San Diego, La Jolla, CA, USA
| | - Marley A Rossa
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Willian Nuñez Lagos
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Neelakshi S Patne
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neuroscience Graduate Program, Boston University, Boston, MA, USA
| | - Edward M Callaway
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
35
|
Perera C, Cruz R, Shemesh N, Carvalho T, Thomas DL, Wells J, Ianuș A. Non-invasive MRI of blood-cerebrospinal fluid-barrier function in a mouse model of Alzheimer's disease: a potential biomarker of early pathology. Fluids Barriers CNS 2024; 21:97. [PMID: 39633378 PMCID: PMC11616325 DOI: 10.1186/s12987-024-00597-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Choroid plexus (CP) or blood-cerebrospinal fluid-barrier (BCSFB) is a unique functional tissue which lines the brain's fluid-filled ventricles, with a crucial role in CSF production and clearance. BCSFB dysfunction is thought to contribute to toxic protein build-up in neurodegenerative disorders, including Alzheimer's disease (AD). However, the dynamics of this process remain unknown, mainly due to the paucity of in-vivo methods for assessing CP function. METHODS We harness recent developments in Arterial Spin Labelling MRI to measure water delivery across the BCSFB as a proxy for CP function, as well as cerebral blood flow (CBF), at different stages of AD in the widely used triple transgenic mouse model (3xTg), with ages between 8 and 32 weeks. We further compared the MRI results with Y-maze behaviour testing, and histologically validated the expected pathological changes, which recapitulate both amyloid and tau deposition. RESULTS Total BCSFB-mediated water delivery is significantly higher in 3xTg mice (> 50%) from 8 weeks (preclinical stage), an increase which is not explained by differences in ventricular volumes, while tissue parameters such as CBF and T1 are not different between groups at all ages. Behaviour differences between the groups were observed starting at 20 weeks, especially in terms of locomotion, with 3xTg animals showing a significantly smaller number of arm entries in the Y-maze. CONCLUSIONS Our work strongly suggests the involvement of CP in the early stages of AD, before the onset of symptoms and behavioural changes, providing a potential biomarker of pathology.
Collapse
Affiliation(s)
- Charith Perera
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Renata Cruz
- Champalimaud Research, Champalimaud Foundation, Av. Brasilia, Lisbon, 1400-038, Portugal
| | - Noam Shemesh
- Champalimaud Research, Champalimaud Foundation, Av. Brasilia, Lisbon, 1400-038, Portugal
| | - Tânia Carvalho
- Champalimaud Research, Champalimaud Foundation, Av. Brasilia, Lisbon, 1400-038, Portugal
| | - David L Thomas
- Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Dementia Research Centre, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, UK
| | - Jack Wells
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Andrada Ianuș
- Champalimaud Research, Champalimaud Foundation, Av. Brasilia, Lisbon, 1400-038, Portugal.
- King's College London, School of Biomedical Engineering and Imaging Sciences, Imaging Physics and Engineering Research Department; Cancer Imaging Research Department, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK.
| |
Collapse
|
36
|
Jackson HD, Cotler MJ, Saunders GW, Cornelssen CA, West PJ, Metcalf CS, Wilcox KS, Cima MJ. Intracerebral delivery of antiseizure medications by microinvasive neural implants. Brain 2024; 147:4147-4156. [PMID: 39241108 DOI: 10.1093/brain/awae282] [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: 04/16/2024] [Revised: 07/20/2024] [Accepted: 08/19/2024] [Indexed: 09/08/2024] Open
Abstract
Focal epilepsy is a difficult disease to treat as two-thirds of patients will not respond to oral anti-seizure medications (ASMs) or have severe off-target effects that lead to drug discontinuation. Current non-pharmaceutical treatment methods (resection or ablation) are underutilized due to the associated morbidities, invasive nature and inaccessibility of seizure foci. Less invasive non-ablative modalities may potentially offer an alternative. Targeting the seizure focus in this way may avoid unassociated critical brain structures to preserve function and alleviate seizure burden. Here we report use of an implantable, miniaturized neural drug delivery system [microinvasive neural implant infusion platform (MINI)] to administer ASMs directly to the seizure focus in a mouse model of temporal lobe epilepsy. We examined the effect local delivery of phenobarbital and valproate had on focal seizures, as well as adverse effects, and compared this to systemic delivery. We show that local delivery of phenobarbital and valproate using our chronic implants significantly reduced focal seizures at all doses given. Furthermore, we show that local delivery of these compounds resulted in no adverse effects to motor function, whereas systemic delivery resulted in significant motor impairment. The results of this study demonstrate the potential of ASM micro dosing to the epileptic focus as a treatment option for people with drug resistant epilepsy. This technology could also be applied to a variety of disease states, enabling a deeper understanding of focal drug delivery in the treatment of neurological disorders.
Collapse
Affiliation(s)
- Hannah D Jackson
- Harvard-MIT Program in Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Max J Cotler
- Harvard-MIT Program in Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gerald W Saunders
- Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Carena A Cornelssen
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Peter J West
- Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT 84112, USA
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Cameron S Metcalf
- Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT 84112, USA
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Karen S Wilcox
- Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Michael J Cima
- Department of Materials Science and Engineering, Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
37
|
MacDonald DI, Jayabalan M, Seaman J, Balaji R, Nickolls A, Chesler A. Pain persists in mice lacking both Substance P and CGRPα signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.15.567208. [PMID: 38076807 PMCID: PMC10705526 DOI: 10.1101/2023.11.15.567208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The neuropeptides Substance P and CGRPα have long been thought important for pain sensation. Both peptides and their receptors are expressed at high levels in pain-responsive neurons from the periphery to the brain making them attractive therapeutic targets. However, drugs targeting these pathways individually did not relieve pain in clinical trials. Since Substance P and CGRPα are extensively co-expressed we hypothesized that their simultaneous inhibition would be required for effective analgesia. We therefore generated Tac1 and Calca double knockout (DKO) mice and assessed their behavior using a wide range of pain-relevant assays. As expected, Substance P and CGRPα peptides were undetectable throughout the nervous system of DKO mice. To our surprise, these animals displayed largely intact responses to mechanical, thermal, chemical, and visceral pain stimuli, as well as itch. Moreover, chronic inflammatory pain and neurogenic inflammation were unaffected by loss of the two peptides. Finally, neuropathic pain evoked by nerve injury or chemotherapy treatment was also preserved in peptide-deficient mice. Thus, our results demonstrate that even in combination, Substance P and CGRPα are not required for the transmission of acute and chronic pain.
Collapse
Affiliation(s)
- Donald Iain MacDonald
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Monessha Jayabalan
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Jonathan Seaman
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Rakshita Balaji
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Alec Nickolls
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Alexander Chesler
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| |
Collapse
|
38
|
Yang CX, Sin DD, Ng RT. SMART: spatial transcriptomics deconvolution using marker-gene-assisted topic model. Genome Biol 2024; 25:304. [PMID: 39623485 PMCID: PMC11610197 DOI: 10.1186/s13059-024-03441-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/20/2024] [Indexed: 12/06/2024] Open
Abstract
While spatial transcriptomics offer valuable insights into gene expression patterns within the spatial context of tissue, many technologies do not have a single-cell resolution. Here, we present SMART, a marker gene-assisted deconvolution method that simultaneously infers the cell type-specific gene expression profile and the cellular composition at each spot. Using multiple datasets, we show that SMART outperforms the existing methods in realistic settings. It also provides a two-stage approach to enhance its performance on cell subtypes. The covariate model of SMART enables the identification of cell type-specific differentially expressed genes across conditions, elucidating biological changes at a single-cell-type resolution.
Collapse
Affiliation(s)
- Chen Xi Yang
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada.
- Department of Bioinformatics, Faculty of Science, University of British Columbia, Vancouver, BC, Canada.
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Raymond T Ng
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- Department of Bioinformatics, Faculty of Science, University of British Columbia, Vancouver, BC, Canada
- Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
39
|
Nandi A, Nakano M, Brašić JR, Brinson ZS, Kitzmiller K, Mathur A, Mohamed M, Roberts J, Wong DF, Kuwabara H. Improved Quantification of MicroPET/CT Imaging Using CT-derived Scaling Factors. Mol Imaging Biol 2024; 26:1016-1026. [PMID: 39313673 DOI: 10.1007/s11307-024-01947-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE Combined micro-PET/CT scanners are widely employed to investigate models of brain disorders in rodents using PET-based coregistration. We examined if CT-based coregistration could improve estimates of brain dimensions and consequently estimates of nondisplaceable binding potential (BPND) in rodent PET studies. PROCEDURES PET and CT scans were acquired on 5 female and 5 male CD-1 mice with 3-[18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB), a radiotracer for the metabotropic glutamate receptor subtype 5 (mGluR5). In the proposed PET/CT (PTCT) approach, the tracer-specific standard volume was dimension-customized to each animal using the scaling factors from CT-to-standard CT coregistration to simplify PET-to-standard PET coregistration (i.e., 3 CT- and 6 PET-derived parameters). For comparison, conventional PET-based coregistration was performed with 9 (PT9) or 12 (PT12) parameters. PET frames were transferred to the standard space by the three approaches (PTCT, PT9, and PT12) to obtain regional time-activity curves (TACs) and BPND in 14 standard volumes of interest (VOIs). Lastly, CT images of the animals were transferred to the standard space by CT-based parameters from PTCT and with the scaling factors replaced with those from PET-based PT9 to evaluate agreement of the skull to the standard CT. RESULTS The PET-based approaches showed various degrees of underestimations of scaling factors in the posterior-anterior-direction compared to PTCT, which resulted in negatively proportional overestimation of radioactivity in the cerebellum (reference region) up to 20%, and proportional, more prominent underestimation of BPND in target regions down to -50%. The skulls of individual animals agreed with the standard skull for scaling factors from PTCT but not for the scaling factors from PT9, which suggested inaccuracy of the latter. CONCLUSIONS The results indicated that conventional PET-based coregistration approaches could yield biased estimates of BPND in proportion to errors of brain dimensions when applied to tracers for which the cerebellum serves as reference region. The proposed PTCT provides evidence of a quantitative improvement over PET-based approaches for brain studies using micro-PET/CT scanners.
Collapse
Affiliation(s)
- Ayon Nandi
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC Room 3243, 601 N. Caroline St, Baltimore, 21287, MD, USA
| | - Masayoshi Nakano
- Clinical Science Division, R&D, Janssen Pharmaceutical K.K, Tokyo, Japan
| | - James Robert Brašić
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC Room 3243, 601 N. Caroline St, Baltimore, 21287, MD, USA
- Department of Psychiatry, New York City Health + Hospitals/Bellevue, New York, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Zabecca S Brinson
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kelly Kitzmiller
- Radiation Oncology, University of Maryland Medical Center, Baltimore, MD, USA
| | - Anil Mathur
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC Room 3243, 601 N. Caroline St, Baltimore, 21287, MD, USA
| | | | - Joshua Roberts
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dean F Wong
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC Room 3243, 601 N. Caroline St, Baltimore, 21287, MD, USA
- Lab of CNS Neuropsychopharmacology And Multimodal Imaging (CNAMI), Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Hiroto Kuwabara
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC Room 3243, 601 N. Caroline St, Baltimore, 21287, MD, USA.
| |
Collapse
|
40
|
Bjerke IE. Data sharing and re-use as a moral imperative in animal research. Eur J Neurosci 2024; 60:7148-7151. [PMID: 39587710 DOI: 10.1111/ejn.16626] [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: 10/16/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
Here, I discuss data sharing and re-use as a moral imperative for research that uses animals in neuroscience. I argue that we are ethically required to make sure that we gain as much knowledge as possible from the animals we use, and that doing so involves making sure that the data from our experiment makes it into the public record of knowledge. I suggest several ways in which journals, grant bodies and policymakers can contribute to making data sharing and re-use mainstream practices.
Collapse
Affiliation(s)
- Ingvild E Bjerke
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
41
|
Du N, Kompotis K, Sato M, Pedron E, Androvic S, Brown S. Behavioural phenotypes of Dicer knockout in the mouse SCN. Eur J Neurosci 2024; 60:6634-6651. [PMID: 39551620 PMCID: PMC11612849 DOI: 10.1111/ejn.16605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 10/15/2024] [Accepted: 10/29/2024] [Indexed: 11/19/2024]
Abstract
The suprachiasmatic nucleus (SCN) is the master clock that directly dictates behavioural rhythms to anticipate the earth's light/dark cycles. Although post-transcriptional regulators called microRNAs have been implicated in physiological SCN function, how the absence of the entire mature miRNome impacts SCN output has not yet been explored. To study the behavioural consequences of miRNA depletion in the SCN, we first generated a mouse model in which Dicer is inactivated in the SCN by crossing Syt10Cre mice with Dicerflox mice to study behavioural consequences of miRNA depletion in the SCN. Loss of all mature miRNAs in the SCN shortened the circadian period length by ~37 minutes at the tissue level and by ~45 minutes at the locomotor activity level. Moreover, knockout animals exhibited a reduction in the precision of the circadian rhythm with more variable activity onsets under both LD 12:12 and DD conditions. We also observed that knockouts with higher onset variations were inclined to develop ultradian rhythms under constant light. In a second mouse model, recombination of Dicerflox via Cre delivery specifically in the SCN resulted in loss of behavioural rhythms in some animals depending on the injection efficiency. Together, our observations highlight the importance of microRNAs for a physiological SCN function and their pivotal role in robust circadian oscillations.
Collapse
Affiliation(s)
- Ngoc‐Hien Du
- Institute of Pharmacology and ToxicologyUniversity of ZurichZurichSwitzerland
- Present address:
Laboratory for Biomedical MicrofluidicsSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | | | - Miho Sato
- Institute of Pharmacology and ToxicologyUniversity of ZurichZurichSwitzerland
| | - Erica Pedron
- Institute of Pharmacology and ToxicologyUniversity of ZurichZurichSwitzerland
| | - Sabrina Androvic
- Institute of Pharmacology and ToxicologyUniversity of ZurichZurichSwitzerland
| | - Steven Brown
- Institute of Pharmacology and ToxicologyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
42
|
Ceder MM, Magnusson KA, Weman HM, Henriksson K, Andréasson L, Lindström T, Wiggins O, Lagerström MC. The mRNA expression profile of glycine receptor subunits alpha 1, alpha 2, alpha 4 and beta in female and male mice. Mol Cell Neurosci 2024; 131:103976. [PMID: 39580061 DOI: 10.1016/j.mcn.2024.103976] [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: 04/25/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 11/25/2024] Open
Abstract
Glycine receptors are ligand-gated chloride-selective channels that control excitability in the central nervous system (CNS). Herein, we have investigated the mRNA expression of the glycine receptor alpha 1 (Glra1), alpha 2 (Glra2), alpha 4 (Glra4) and the beta (Glrb) subunits, in adult female and male mice. Single-cell RNA sequencing data re-analysis of the Zeisel et al. (2018) dataset indicated widespread expression of Glra1, Glra2 and Glrb in the CNS, while only a few cells in the cortex, striatum, thalamus, midbrain and the spinal cord expressed Glra4. Highest occurrence of Glra1, Glra2 and Glrb were found in the brainstem. Moreover, Glra1 and Glrb were revealed to have the highest occurrences in the spinal cord of the investigated subunits. However, both Glra2 and Glrb had a more widespread expression in the CNS compared with Glra1 and Glra4. Bulk quantitative real-time-PCR (qRT-PCR) analysis revealed Glra1 expression in the hypothalamus, thalamus, brainstem and the spinal cord, and widespread, but low, Glra2 and Glrb expression in the CNS. Moreover, Glrb could be detected in a few visceral organs. Additionally, females and males were found to express Glra1, Glra2 and Glrb differently in certain brain areas such as the brainstem. Expression levels of Glra4 were too low to be detected using qRT-PCR. Lastly, RNAscope spatially validated the expression of Glra1, Glra2 and Glrb in the areas indicated by the single-cell and bulk analyses, and further revealed that Glra4 can be detected in the cortex, amygdala, hypothalamus, thalamus, brainstem, especially the cochlear nucleus, and in the spinal cord.
Collapse
Affiliation(s)
- Mikaela M Ceder
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kajsa A Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hannah M Weman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Katharina Henriksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Linn Andréasson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Teresa Lindström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Oskar Wiggins
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Malin C Lagerström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
43
|
Nagai N, Shioiri T, Hatano S, Sugiura N, Watanabe H. Regulatory role of Heparan sulfate in leptin signaling. Cell Signal 2024; 124:111456. [PMID: 39384005 DOI: 10.1016/j.cellsig.2024.111456] [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: 07/17/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Leptin, a hormone mainly secreted by adipocytes, has attracted significant attention since its discovery in 1994. Initially known for its role in appetite suppression and energy regulation, leptin is now recognized for its influence on various physiological processes, including immune response, bone formation, and reproduction. It exerts its effects by binding to receptors and initiating an intracellular signaling cascade. Heparan sulfate (HS) is known to regulate the intracellular signaling of various ligands. HS is present as the glycan portion of HSPGs on cell surfaces and in intercellular spaces, with diverse structures due to extensive sulfation and epimerization. Although HS chains on HSPGs are involved in many physiological processes, the detailed effects of HS chains on leptin signaling are not well understood. This study examined the role of HS chains on HSPGs in leptin signaling using Neuro2A cells expressing the full-length leptin receptor (LepR). We showed that cell surface HS was essential for efficient leptin signaling. Enzymatic degradation of HS significantly reduced leptin-induced phosphorylation of downstream molecules, such as signal transducer and activator of transcription 3 and p44/p42 Mitogen-activated protein kinase. In addition, HS regulated LepR expression and internalization, as treatment with HS-degrading enzymes decreased cell surface LepR. HS was also found to exhibit a weak interaction with LepR. Enzymatic removal of HS enhanced the interaction between LepR and low-density lipoprotein receptor-related protein 1, suggesting that HS negatively regulates this interaction. In conclusion, HS plays a significant role in modulating LepR availability on the cell surface, thereby influencing leptin signaling. These findings provide new insights into the complex regulation of leptin signaling and highlight potential therapeutic targets for metabolic disorders and obesity.
Collapse
Affiliation(s)
- Naoko Nagai
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| | - Tatsumasa Shioiri
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| | - Sonoko Hatano
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| | - Nobuo Sugiura
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| |
Collapse
|
44
|
Cho N, Squair JW, Aureli V, James ND, Bole-Feysot L, Dewany I, Hankov N, Baud L, Leonhartsberger A, Sveistyte K, Skinnider MA, Gautier M, Laskaratos A, Galan K, Goubran M, Ravier J, Merlos F, Batti L, Pages S, Berard N, Intering N, Varescon C, Watrin A, Duguet L, Carda S, Bartholdi KA, Hutson TH, Kathe C, Hodara M, Anderson MA, Draganski B, Demesmaeker R, Asboth L, Barraud Q, Bloch J, Courtine G. Hypothalamic deep brain stimulation augments walking after spinal cord injury. Nat Med 2024; 30:3676-3686. [PMID: 39623087 DOI: 10.1038/s41591-024-03306-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/13/2024] [Indexed: 12/15/2024]
Abstract
A spinal cord injury (SCI) disrupts the neuronal projections from the brain to the region of the spinal cord that produces walking, leading to various degrees of paralysis. Here, we aimed to identify brain regions that steer the recovery of walking after incomplete SCI and that could be targeted to augment this recovery. To uncover these regions, we constructed a space-time brain-wide atlas of transcriptionally active and spinal cord-projecting neurons underlying the recovery of walking after incomplete SCI. Unexpectedly, interrogation of this atlas nominated the lateral hypothalamus (LH). We demonstrate that glutamatergic neurons located in the LH (LHVglut2) contribute to the recovery of walking after incomplete SCI and that augmenting their activity improves walking. We translated this discovery into a deep brain stimulation therapy of the LH (DBSLH) that immediately augmented walking in mice and rats with SCI and durably increased recovery through the reorganization of residual lumbar-terminating projections from brainstem neurons. A pilot clinical study showed that DBSLH immediately improved walking in two participants with incomplete SCI and, in conjunction with rehabilitation, mediated functional recovery that persisted when DBSLH was turned off. There were no serious adverse events related to DBSLH. These results highlight the potential of targeting specific brain regions to maximize the engagement of spinal cord-projecting neurons in the recovery of neurological functions after SCI. Further trials must establish the safety and efficacy profile of DBSLH, including potential changes in body weight, psychological status, hormonal profiles and autonomic functions.
Collapse
Affiliation(s)
- Newton Cho
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Jordan W Squair
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Department of Neurosurgery, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Viviana Aureli
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Department of Neurosurgery, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Nicholas D James
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Léa Bole-Feysot
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Inssia Dewany
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Nicolas Hankov
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Laetitia Baud
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Anna Leonhartsberger
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Kristina Sveistyte
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Michael A Skinnider
- Lewis-Sigler Institute of Integrative Genomics and Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Matthieu Gautier
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Achilleas Laskaratos
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Katia Galan
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Maged Goubran
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jimmy Ravier
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Frederic Merlos
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Laura Batti
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Stéphane Pages
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Nadia Berard
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Nadine Intering
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Camille Varescon
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | | | | | - Stefano Carda
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Kay A Bartholdi
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Thomas H Hutson
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Claudia Kathe
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Michael Hodara
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Mark A Anderson
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Bogdan Draganski
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Robin Demesmaeker
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Leonie Asboth
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Quentin Barraud
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Jocelyne Bloch
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland.
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.
- Department of Neurosurgery, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| | - Grégoire Courtine
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland.
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.
- Department of Neurosurgery, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| |
Collapse
|
45
|
Zhu X, Shi L, Li P, Lu J. Cerebral blood flow patterns induced by photoactivation based on laser speckle contrast imaging. BIOMEDICAL OPTICS EXPRESS 2024; 15:6739-6755. [PMID: 39679412 PMCID: PMC11640580 DOI: 10.1364/boe.541444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Neurovascular coupling (NVC) is crucial for maintaining brain function and holds significant implications for diagnosing neurological disorders. However, the neuron type and spatial specificity in NVC remain poorly understood. In this study, we investigated the spatiotemporal characteristics of local cerebral blood flow (CBF) driven by excitatory (VGLUT2) and inhibitory (VGAT) neurons in the mouse sensorimotor cortex. By integrating optogenetics, wavefront modulation technology, and laser speckle contrast imaging (LSCI), we achieved precise, spatially targeted photoactivation of type-specific neurons and real-time CBF monitoring. We observed three distinct CBF response patterns across different locations: unimodal, bimodal, and biphasic. While unimodal and bimodal patterns were observed in different locations for both neuron types, the biphasic pattern was exclusive to inhibitory neurons. Our results reveal the spatiotemporal complexity of NVC across different neuron types and demonstrate our method's ability to analyze this complexity in detail.
Collapse
Affiliation(s)
- Xuan Zhu
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Liang Shi
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Pengcheng Li
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215100, China
| | - Jinling Lu
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
46
|
Bahaaeldin M, Bülte C, Luelsberg F, Kumar S, Kappler J, Völker C, Schilling K, Baader SL. Engrailed-2 and inflammation convergently and independently impinge on cerebellar Purkinje cell differentiation. J Neuroinflammation 2024; 21:306. [PMID: 39609827 PMCID: PMC11603920 DOI: 10.1186/s12974-024-03301-6] [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: 07/29/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024] Open
Abstract
Autism spectrum disorders (ASD) have a complex pathogenesis thought to include both genetic and extrinsic factors. Among the latter, inflammation of the developing brain has recently gained growing attention. However, how genetic predisposition and inflammation might converge to precipitate autistic behavior remains elusive. Cerebellar structure and function are well known to be affected in autism. We therefore used cerebellar slice cultures to probe whether inflammatory stimulation and (over)expression of the autism susceptibility gene Engrailed-2 interact in shaping differentiation of Purkinje cells, key organizers of cerebellar histogenesis and function. We show that lipopolysaccharide treatment reduces Purkinje cell dendritogenesis and that this effect is enhanced by over-expression of Engrailed-2 in these cells. The effects of lipopolysaccharide can be blocked by inhibiting microglia proliferation and also by blocking tumor necrosis factor alpha receptor signaling, suggesting microglia and tumor necrosis factor alpha are major players in this scenario. These findings identify Purkinje cells as a potential integrator of genetic and environmental signals that lead to an autism-associated morphology.
Collapse
Affiliation(s)
- Mohammed Bahaaeldin
- Institute of Anatomy, Anatomy and Cell Biology, University of Bonn, Nussallee 10, 53125, Bonn, Germany
| | - Carolin Bülte
- Institute of Anatomy, Anatomy and Cell Biology, University of Bonn, Nussallee 10, 53125, Bonn, Germany
| | - Fabienne Luelsberg
- Institute of Anatomy, Anatomy and Cell Biology, University of Bonn, Nussallee 10, 53125, Bonn, Germany
| | - Sujeet Kumar
- Institute of Anatomy, Anatomy and Cell Biology, University of Bonn, Nussallee 10, 53125, Bonn, Germany
- National Reference Laboratory for Tuberculosis, ICMR-RMRC, Bhubaneswar, Odisha, India
| | - Joachim Kappler
- Institute of Biochemistry and Molecular Biology, University of Bonn, Nussallee 11, 53125, Bonn, Germany
| | - Christof Völker
- Institute of Biochemistry and Molecular Biology, University of Bonn, Nussallee 11, 53125, Bonn, Germany
| | - Karl Schilling
- Institute of Anatomy, Anatomy and Cell Biology, University of Bonn, Nussallee 10, 53125, Bonn, Germany
| | - Stephan L Baader
- Institute of Anatomy, Anatomy and Cell Biology, University of Bonn, Nussallee 10, 53125, Bonn, Germany.
| |
Collapse
|
47
|
Lv Y, Yuan Y, Zhong X, Yu Q, Lu X, Qu B, Zhao H. Exploration and practice of potential association prediction between diseases and drugs based on Swanson framework and bioinformatics. Sci Rep 2024; 14:29643. [PMID: 39609506 PMCID: PMC11604654 DOI: 10.1038/s41598-024-79587-6] [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: 04/03/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
Compared to traditional intermediate concepts, specific bioinformatics entities are more informative and higher directional. This study is based on the BITOLA system and combines bioinformatics methods to determine the intermediate concept which is key to improve efficiency of Literature-based Knowledge Discovery, proposes the concept of "Swanson framework + Bioinformatics", and conducts practice of Literature-based Knowledge Discovery to improve the scientificity and efficiency of research and development. Firstly, detected the disease related genes (i.e. differentially expressed genes) according to the results of gene functional analysis as intermediate concepts to carry out Literature-based Knowledge Discovery. Taking the disease "Autism Spectrum Disorder (ASD)" as an example, the potential "disease-drug" association was predicted, and the predicted drugs were verified from the perspective of bioinformatics. Two drugs potentially associated with ASD were found: Fish oil and Forskolin, which were closely related to ASD in bioinformatics analysis results and literature verification. The two "disease-drug" association results showed better scientificity. The BIOINF-ABC+ model improves the accuracy of calculations by 76% compared to using the BITOLA system alone. In addition, it also shows high accuracy and credibility in literature verification. The BIOINF-ABC+ model based on the "Swanson framework + Bioinformatics" has good practicality, applicability, and accuracy in conducting "disease-drug" association prediction in the biomedical field, and can be used for mining "disease-drug" relationships.
Collapse
Affiliation(s)
- Yanhua Lv
- Shanxi Medical University, Jinzhong, China.
| | | | | | - Qi Yu
- Shanxi Medical University, Jinzhong, China.
| | - Xuechun Lu
- Second Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Baoqiang Qu
- Institute of Scientific and Technical Information of China, Beijing, China
| | | |
Collapse
|
48
|
Lanoizelet M, Michel L, Lagadec R, Mayeur H, Guichard L, Logeux V, Séverac D, Martin K, Klopp C, Marcellini S, Castillo H, Pollet N, Candal E, Debiais-Thibaud M, Boisvert C, Billoud B, Schubert M, Blader P, Mazan S. Analysis of a shark reveals ancient, Wnt-dependent, habenular asymmetries in vertebrates. Nat Commun 2024; 15:10194. [PMID: 39587074 PMCID: PMC11589584 DOI: 10.1038/s41467-024-54042-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 10/30/2024] [Indexed: 11/27/2024] Open
Abstract
The mode of evolution of left-right asymmetries in the vertebrate habenulae remains largely unknown. Using a transcriptomic approach, we show that in a cartilaginous fish, the catshark Scyliorhinus canicula, habenulae exhibit marked asymmetries, in both their medial and lateral components. Comparisons across vertebrates suggest that those identified in lateral habenulae reflect an ancestral gnathostome trait, partially conserved in lampreys, and independently lost in tetrapods and neopterygians. Asymmetry formation involves distinct mechanisms in the catshark lateral and medial habenulae. Medial habenulae are submitted to a marked, asymmetric temporal regulation of neurogenesis, undetectable in their lateral counterparts. Conversely, asymmetry formation in lateral habenulae results from asymmetric choices of neuronal identity in post-mitotic progenitors, a regulation dependent on the repression of Wnt signaling by Nodal on the left. Based on comparisons with the mouse and the zebrafish, we propose that habenular asymmetry formation involves a recurrent developmental logic across vertebrates, which relies on conserved, temporally regulated genetic programs sequentially shaping choices of neuronal identity on both sides and asymmetrically modified by Wnt activity.
Collapse
Affiliation(s)
- Maxence Lanoizelet
- CNRS, Sorbonne Université, UMR7232-Biologie Intégrative des Organismes Marins, Observatoire Océanologique, Banyuls-sur-Mer, France
| | - Léo Michel
- CNRS, Sorbonne Université, UMR7232-Biologie Intégrative des Organismes Marins, Observatoire Océanologique, Banyuls-sur-Mer, France
| | - Ronan Lagadec
- CNRS, Sorbonne Université, UMR7232-Biologie Intégrative des Organismes Marins, Observatoire Océanologique, Banyuls-sur-Mer, France
| | - Hélène Mayeur
- CNRS, Sorbonne Université, UMR7232-Biologie Intégrative des Organismes Marins, Observatoire Océanologique, Banyuls-sur-Mer, France
| | - Lucile Guichard
- CNRS, Sorbonne Université, UMR7232-Biologie Intégrative des Organismes Marins, Observatoire Océanologique, Banyuls-sur-Mer, France
| | - Valentin Logeux
- Centre de Ressources Biologiques Marines, Sorbonne Université, Observatoire Océanologique, UMS 2348, Banyuls-sur-Mer, France
| | - Dany Séverac
- MGX, Université Montpellier, CNRS, INSERM, Montpellier, France
| | - Kyle Martin
- UK Research and Innovation, Biotechnology and Biological Sciences Research Council, Swindon, UK
| | - Christophe Klopp
- Plateforme Bioinformatique, Genotoul, BioinfoMics, UR875 Biométrie et Intelligence Artificielle, INRAE, Castanet-Tolosan, France
| | - Sylvain Marcellini
- Department of Cell Biology, School of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Héctor Castillo
- Department of Cell Biology, School of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Nicolas Pollet
- Université Paris-Saclay, CNRS, IRD, Évolution, Génomes, Comportement et Écologie, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Eva Candal
- Departament of Functional Biology, CIBUS, Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Catherine Boisvert
- School of Molecular and Life Sciences, Curtin University, Perth, WA, Australia
| | - Bernard Billoud
- UMR8227, CNRS-Sorbonne Université, Station Biologique, Roscoff, France
| | - Michael Schubert
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer, Institut de la Mer de Villefranche, Sorbonne Université, CNRS, Villefranche-sur-Mer, France
| | - Patrick Blader
- Centre de Biologie Intégrative (CBI, FR 3743), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Sylvie Mazan
- CNRS, Sorbonne Université, UMR7232-Biologie Intégrative des Organismes Marins, Observatoire Océanologique, Banyuls-sur-Mer, France.
| |
Collapse
|
49
|
Farmer AL, Febo M, Wilkes BJ, Lewis MH. Environmental Enrichment Attenuates Repetitive Behavior and Alters the Functional Connectivity of Pain and Sensory Pathways in C58 Mice. Cells 2024; 13:1933. [PMID: 39682680 DOI: 10.3390/cells13231933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 12/18/2024] Open
Abstract
Restricted repetitive behaviors (RRB) encompass a variety of inflexible behaviors, which are diagnostic for autism spectrum disorder (ASD). Despite being requisite diagnostic criteria, the neurocircuitry of these behaviors remains poorly understood, limiting treatment development. Studies in translational animal models show environmental enrichment (EE) reduces the expression of RRB, although the underlying mechanisms are largely unknown. This study used functional magnetic resonance imaging to identify functional connectivity alterations associated with RRB and its attenuation by EE in C58 mice, an animal model of RRB. Extensive differences were observed between C58 mice and C57BL/6 control mice. Higher RRB was associated with altered connectivity between the somatosensory network and reticular thalamic nucleus and between striatal and sensory processing regions. Animals housed in EE displayed increased connectivity between the somatosensory network and the anterior pretectal nucleus and hippocampus, as well as reduced connectivity between the visual network and area prostriata. These results suggest aberrant sensory perception is associated with RRB in C58 mice. EE may reduce RRB by altering functional connectivity in pain and visual networks. This study raises questions about the role of sensory processing and pain in RRB development and identifies new potential intervention targets.
Collapse
Affiliation(s)
- Anna L Farmer
- Department of Psychology, University of Florida, Gainesville, FL 32603, USA
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, FL 32611, USA
| | - Bradley J Wilkes
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32608, USA
| | - Mark H Lewis
- Department of Psychology, University of Florida, Gainesville, FL 32603, USA
- Department of Psychiatry, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
50
|
Catanese J, Murakami TC, Catto A, Kenny PJ, Ibañez-Tallon I. Precise 3D Localization of Intracerebral Implants Using a Simple Brain Clearing Method. J Integr Neurosci 2024; 23:207. [PMID: 39613469 DOI: 10.31083/j.jin2311207] [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: 01/22/2024] [Revised: 07/24/2024] [Accepted: 08/05/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Precise localization of intracerebral implants in rodent brains is required for physiological and behavioral studies, particularly if targeting deep brain nuclei. Traditional histological methods, based on manual estimation through sectioning can introduce errors and complicate data interpretation. METHODS Here, we introduce an alternative method based on recent advances in tissue-clearing techniques and light-sheet fluorescence microscopy. This method uses a simplified recipe of the Clear, Unobstructed Brain/Body Imaging Cocktails and Computational Analysis (CUBIC) method, which is a rapid clearing procedure using an aqueous-based solution compatible with fluorescence and fluorescence markers. We demonstrate the utility of this approach in anesthetized transgenic mice expressing channelrhodopsin-2 (ChR2) and enhanced yellow fluorescent fusion (EYFP) protein under the choline acetyltransferase (ChAT) promoter/enhancer regions (ChAT-ChR2-EYFP mice) with implanted linear silicon optrode probes into the midbrain interpeduncular nucleus (IPN). RESULTS By applying the red fluorescent DiD' dye (DiIC18(5) solid (1,1'-Dioctadecyl-3,3,3',3'-Tetramethylindodicarbocyanine, 4-Chlorobenzenesulfonate Salt) to the electrode surface, we precisely visualize the electrode localization in the IPN of ChAT-ChR2-EYFP mice. Three-dimensional brain videos from different orientations highlight the potential of this method. Optogenetic responses recorded from electrodes placed in the IPN validate these findings. CONCLUSIONS This method allows for precise localization of brain implantation sites in transgenic mice expressing cell-specific fluorescence markers. It enables virtual brain slicing in any orientation, making it a useful tool for functional studies in mice.
Collapse
Affiliation(s)
- Julien Catanese
- Laboratory of Molecular Biology, The Rockefeller University, New York, NY 10065, USA
| | - Tatsuya C Murakami
- Laboratory of Molecular Biology, The Rockefeller University, New York, NY 10065, USA
| | - Adam Catto
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ines Ibañez-Tallon
- Laboratory of Molecular Biology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|