151
|
Isaac Guillén F, Geist MA, Cheng SY, Harris AM, Treviño ME, Brumback AC, Nishiyama H, Howard MA. A novel mouse model for developmental and epileptic encephalopathy by Purkinje cell-specific deletion of Scn1b. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.19.624370. [PMID: 39605540 PMCID: PMC11601654 DOI: 10.1101/2024.11.19.624370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Loss of function variants of SCN1B are associated with a range of developmental and epileptic encephalopathies (DEEs), including Dravet syndrome. These DEEs feature a wide range of severe neurological disabilities, including changes to social, motor, mood, sleep, and cognitive function which are notoriously difficult to treat, and high rates of early mortality. While the symptomology of SCN1B -associated DEEs indicates broad changes in neural function, most research has focused on epilepsy-related brain structures and function. Mechanistic studies of SCN1B / Scn1b have delineated diverse roles in development and adult maintenance of neural function, via cell adhesion, ion channel regulation, and other intra- and extra-cellular actions. However, use of mouse models is limited as knockout of Scn1b , globally and even in some cell-specific models (e.g., Parvalbumin+ interneuron-specific knockout) in adult mice, leads to severe and progressive epilepsy, health deterioration, and 100% mortality within weeks. Here, we report findings of a novel transgenic mouse line in which Scn1b was specifically deleted in cerebellar Purkinje cells. Unlike most existing models, these mice did not show failure to thrive or early mortality. However, we quantified marked decrements to Purkinje cell physiology as well as motor, social, and cognitive dysfunction. Our data indicate that cerebellar Purkinje cells are an important node for dysfunction and neural disabilities in SCN1B -related DEEs, and combined with previous work identify this as a potentially vital site for understanding mechanisms of DEEs and developing therapies that can treat these disorders holistically.
Collapse
|
152
|
Tamura Y, Maeda S, Takahashi H, Aoto Y, Matsuki T, Seki K. GABAergic circuit interaction between central amygdala and bed nucleus of the stria terminalis in lipopolysaccharide-induced despair-like behavior. Physiol Behav 2024; 288:114753. [PMID: 39551417 DOI: 10.1016/j.physbeh.2024.114753] [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: 10/05/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Hyperexcitability of central amygdala (CeA) induces depressive symptoms. The bed nucleus of the stria terminalis (BNST) receives GABAergic input from the CeA. However, it remains unclear whether the GABAergic neurons in the CeA projecting to BNST contribute to major depression. Here, we investigated the roles of GABAergic neurons in CeA and BNST in lipopolysaccharide (LPS)-induced despair-like behavior. We generated adeno-associated virus vectors (AAV) carrying shRNA against Gad67 to knock down GAD67 expression in CeA (Gad67-KD-CeA) or BNST (Gad67-KD-BNST) in C57BL/6J male mice. Despair-like behavior was assessed by tail suspension test (TST) 24 h after LPS administration. Saline-treated Gad67-KD-CeA mice exhibited longer immobility during TST than saline-treated AAV-injected control (AAV-Cont) mice. Although LPS increased immobility time in AAV-Cont mice, it did not affect immobility time in Gad67-KD-CeA mice. While LPS did not affect the immobility time in Gad67-KD-BNST mice, it increased immobility time in AAV-Cont mice. We injected GFP-expressing AAV with a Dlx promoter, specifically expressed in GABAergic neurons, into CeA, and FluoroGold, a retrograde neuronal tracer, into the BNST. GFP signals associated with CeA GABAergic neurons were detected in the BNST, contacting c-fos and GAD67-expressed cells following LPS. We detected the FluoroGold signals in GAD67- and c-fos-expressed neurons in the CeA after LPS administration. Bilateral intra-BNST injection of muscimol (2 pmol), a GABAA receptor agonist, increased immobility time during TST. These findings suggest that LPS-decreased GABAergic activity in the CeA may lead to disinhibition of GABAergic interneurons in the BNST, resulting in GABAA receptor activation and subsequently induces despair-like behavior.
Collapse
Affiliation(s)
- Yuka Tamura
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Sakura Maeda
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Haruna Takahashi
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Yuta Aoto
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Tohru Matsuki
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Aichi 480-0392, Japan.
| | - Kenjiro Seki
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan.
| |
Collapse
|
153
|
Túrós D, Vasiljevic J, Hahn K, Rottenberg S, Valdeolivas A. Chrysalis: decoding tissue compartments in spatial transcriptomics with archetypal analysis. Commun Biol 2024; 7:1520. [PMID: 39550461 PMCID: PMC11569261 DOI: 10.1038/s42003-024-07165-7] [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: 01/16/2024] [Accepted: 10/29/2024] [Indexed: 11/18/2024] Open
Abstract
Dissecting tissue compartments in spatial transcriptomics (ST) remains challenging due to limited spatial resolution and dependence on single-cell reference data. We present Chrysalis, a computational method that rapidly uncovers tissue compartments through spatially variable gene (SVG) detection and archetypal analysis without requiring external reference data. Additionally, it offers a unique visualisation approach for swift tissue characterisation and provides access to the underlying gene expression signatures, enabling the identification of spatially and functionally distinct cellular niches. Chrysalis was evaluated through various benchmarks and validated against deconvolution, independently obtained cell type abundance data, and histopathological annotations, demonstrating superior performance compared to other algorithms on both in silico and real-world test examples. Furthermore, we showcased its versatility across different technologies, such as Visium, Visium HD, Slide-seq, and Stereo-seq.
Collapse
Affiliation(s)
- Demeter Túrós
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| | - Jelica Vasiljevic
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Kerstin Hahn
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland.
| | - Alberto Valdeolivas
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| |
Collapse
|
154
|
Inskeep KA, Crase B, Dayarathna T, Stottmann RW. SMPD4-mediated sphingolipid metabolism regulates brain and primary cilia development. Development 2024; 151:dev202645. [PMID: 39470011 PMCID: PMC11586524 DOI: 10.1242/dev.202645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Genetic variants in multiple sphingolipid biosynthesis genes cause human brain disorders. A recent study looked at people from 12 unrelated families with variants in the gene SMPD4, a neutral sphingomyelinase that metabolizes sphingomyelin into ceramide at an early stage of the biosynthesis pathway. These individuals have severe developmental brain malformations, including microcephaly and cerebellar hypoplasia. The disease mechanism of SMPD4 was not known and so we pursued a new mouse model. We hypothesized that the role of SMPD4 in producing ceramide is important for making primary cilia, a crucial organelle mediating cellular signaling. We found that the mouse model has cerebellar hypoplasia due to failure of Purkinje cell development. Human induced pluripotent stem cells lacking SMPD4 exhibit neural progenitor cell death and have shortened primary cilia, which is rescued by adding exogenous ceramide. SMPD4 production of ceramide is crucial for human brain development.
Collapse
Affiliation(s)
- Katherine A. Inskeep
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Bryan Crase
- Department of Neuroscience, The Ohio State University College of Arts and Sciences, Columbus, OH 43210, USA
| | - Thamara Dayarathna
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Rolf W. Stottmann
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
155
|
Power I, Rivlin M, Shmuely H, Zaiss M, Navon G, Perlman O. In vivo mapping of the chemical exchange relayed nuclear Overhauser effect using deep magnetic resonance fingerprinting. iScience 2024; 27:111209. [PMID: 39569380 PMCID: PMC11576397 DOI: 10.1016/j.isci.2024.111209] [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: 06/08/2024] [Revised: 09/17/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024] Open
Abstract
Noninvasive magnetic resonance imaging (MRI) of the relayed nuclear Overhauser effect (rNOE) constitutes a promising approach for gaining biological insights into various pathologies, including brain cancer, kidney injury, ischemic stroke, and liver disease. However, rNOE imaging is time-consuming and prone to biases stemming from the water T1 and the semisolid magnetization transfer (MT) contrasts. Here, we developed a rapid rNOE quantification approach, combining magnetic resonance fingerprinting (MRF) acquisition with deep-learning-based reconstruction. The method was systematically validated using tissue-mimicking phantoms, wild-type mice (n = 7), and healthy human volunteers (n = 5). In vitro rNOE parameter maps generated by MRF were highly correlated with ground truth (r > 0.98, p < 0.001). Simultaneous mapping of the rNOE and the semisolid MT exchange parameters in mice and humans were in agreement with previously reported literature values. Whole-brain 3D parameter mapping in humans took less than 5 min (282 s for acquisition and less than 2 s for reconstruction). With its demonstrated ability to rapidly extract quantitative molecular maps, deep rNOE-MRF can potentially serve as a valuable tool for the characterization and detection of molecular abnormalities in vivo.
Collapse
Affiliation(s)
- Inbal Power
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Michal Rivlin
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- School of Chemistry, Tel Aviv University, Tel Aviv, Israel
| | - Hagar Shmuely
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Moritz Zaiss
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Gil Navon
- School of Chemistry, Tel Aviv University, Tel Aviv, Israel
| | - Or Perlman
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
156
|
Strohl JJ, Carrión J, Huerta PT. Brain imaging and machine learning reveal uncoupled functional network for contextual threat memory in long sepsis. Sci Rep 2024; 14:27747. [PMID: 39533062 PMCID: PMC11557587 DOI: 10.1038/s41598-024-79259-5] [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: 08/06/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Positron emission tomography (PET) utilizes radiotracers like [18F]fluorodeoxyglucose (FDG) to measure brain activity in health and disease. Performing behavioral tasks between the FDG injection and the PET scan allows the FDG signal to reflect task-related brain networks. Building on this principle, we introduce an approach called behavioral task-associated PET (beta-PET) consisting of two scans: the first after a mouse is familiarized with a conditioning chamber, and the second upon recall of contextual threat. Associative threat conditioning occurs between scans. Beta-PET focuses on brain regions encoding threat memory (e.g., amygdala, prefrontal cortex) and contextual aspects (e.g., hippocampus, subiculum, entorhinal cortex). Our results show that beta-PET identifies a biologically defined network encoding contextual threat memory and its uncoupling in a mouse model of long sepsis. Moreover, machine learning algorithms (linear logistic regression) and ordinal trends analysis demonstrate that beta-PET robustly predicts the behavioral defense response and its breakdown during long sepsis.
Collapse
Affiliation(s)
- Joshua J Strohl
- Laboratory of Immune and Neural Networks, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Joseph Carrión
- Laboratory of Immune and Neural Networks, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Patricio T Huerta
- Laboratory of Immune and Neural Networks, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA.
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY, 11549, USA.
| |
Collapse
|
157
|
Ono K, Jarysta A, Hughes NC, Jukic A, Chang HHV, Deans MR, Eatock RA, Cullen KE, Kindt KS, Tarchini B. Contributions of mirror-image hair cell orientation to mouse otolith organ and zebrafish neuromast function. eLife 2024; 13:RP97674. [PMID: 39531034 PMCID: PMC11556791 DOI: 10.7554/elife.97674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Otolith organs in the inner ear and neuromasts in the fish lateral-line harbor two populations of hair cells oriented to detect stimuli in opposing directions. The underlying mechanism is highly conserved: the transcription factor EMX2 is regionally expressed in just one hair cell population and acts through the receptor GPR156 to reverse cell orientation relative to the other population. In mouse and zebrafish, loss of Emx2 results in sensory organs that harbor only one hair cell orientation and are not innervated properly. In zebrafish, Emx2 also confers hair cells with reduced mechanosensory properties. Here, we leverage mouse and zebrafish models lacking GPR156 to determine how detecting stimuli of opposing directions serves vestibular function, and whether GPR156 has other roles besides orienting hair cells. We find that otolith organs in Gpr156 mouse mutants have normal zonal organization and normal type I-II hair cell distribution and mechano-electrical transduction properties. In contrast, gpr156 zebrafish mutants lack the smaller mechanically evoked signals that characterize Emx2-positive hair cells. Loss of GPR156 does not affect orientation-selectivity of afferents in mouse utricle or zebrafish neuromasts. Consistent with normal otolith organ anatomy and afferent selectivity, Gpr156 mutant mice do not show overt vestibular dysfunction. Instead, performance on two tests that engage otolith organs is significantly altered - swimming and off-vertical-axis rotation. We conclude that GPR156 relays hair cell orientation and transduction information downstream of EMX2, but not selectivity for direction-specific afferents. These results clarify how molecular mechanisms that confer bi-directionality to sensory organs contribute to function, from single hair cell physiology to animal behavior.
Collapse
Affiliation(s)
- Kazuya Ono
- Department of Neurobiology, University of ChicagoChicagoUnited States
| | | | - Natasha C Hughes
- Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Alma Jukic
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Hui Ho Vanessa Chang
- Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Michael R Deans
- Department of Neurobiology, Spencer Fox Eccles School of Medicine, University of UtahSalt Lake CityUnited States
- Department of Otolaryngology - Head & Neck Surgery, Spencer Fox Eccles School of Medicine at the University of UtahSalt Lake CityUnited States
| | - Ruth Anne Eatock
- Department of Neurobiology, University of ChicagoChicagoUnited States
| | - Kathleen E Cullen
- Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins UniversityBaltimoreUnited States
- Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
- Kavli Neuroscience Discovery Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Basile Tarchini
- The Jackson LaboratoryBar HarborUnited States
- Tufts University School of MedicineBostonUnited States
| |
Collapse
|
158
|
Ball G, Oldham S, Kyriakopoulou V, Williams LZJ, Karolis V, Price A, Hutter J, Seal ML, Alexander-Bloch A, Hajnal JV, Edwards AD, Robinson EC, Seidlitz J. Molecular signatures of cortical expansion in the human foetal brain. Nat Commun 2024; 15:9685. [PMID: 39516464 PMCID: PMC11549424 DOI: 10.1038/s41467-024-54034-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The third trimester of human gestation is characterised by rapid increases in brain volume and cortical surface area. Recent studies have revealed a remarkable molecular diversity across the prenatal cortex but little is known about how this diversity translates into the differential rates of cortical expansion observed during gestation. We present a digital resource, μBrain, to facilitate knowledge translation between molecular and anatomical descriptions of the prenatal brain. Using μBrain, we evaluate the molecular signatures of preferentially-expanded cortical regions, quantified in utero using magnetic resonance imaging. Our findings demonstrate a spatial coupling between areal differences in the timing of neurogenesis and rates of neocortical expansion during gestation. We identify genes, upregulated from mid-gestation, that are highly expressed in rapidly expanding neocortex and implicated in genetic disorders with cognitive sequelae. The μBrain atlas provides a tool to comprehensively map early brain development across domains, model systems and resolution scales.
Collapse
Affiliation(s)
- G Ball
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| | - S Oldham
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Australia
| | - V Kyriakopoulou
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - L Z J Williams
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - V Karolis
- Centre for the Developing Brain, King's College London, London, UK
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - A Price
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - J Hutter
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - M L Seal
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - A Alexander-Bloch
- Department of Child and Adolescent Psychiatry and Behavioral Sciences, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - J V Hajnal
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - A D Edwards
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - E C Robinson
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - J Seidlitz
- Department of Child and Adolescent Psychiatry and Behavioral Sciences, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
159
|
Yang J, Wang L, Liu L, Zheng X. GraphPCA: a fast and interpretable dimension reduction algorithm for spatial transcriptomics data. Genome Biol 2024; 25:287. [PMID: 39511664 PMCID: PMC11545739 DOI: 10.1186/s13059-024-03429-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
The rapid advancement of spatial transcriptomics technologies has revolutionized our understanding of cell heterogeneity and intricate spatial structures within tissues and organs. However, the high dimensionality and noise in spatial transcriptomic data present significant challenges for downstream data analyses. Here, we develop GraphPCA, an interpretable and quasi-linear dimension reduction algorithm that leverages the strengths of graphical regularization and principal component analysis. Comprehensive evaluations on simulated and multi-resolution spatial transcriptomic datasets generated from various platforms demonstrate the capacity of GraphPCA to enhance downstream analysis tasks including spatial domain detection, denoising, and trajectory inference compared to other state-of-the-art methods.
Collapse
Affiliation(s)
- Jiyuan Yang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Wang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The Guangxi Key Laboratory of Intelligent Precision Medicine, Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lin Liu
- Institute of Natural Sciences, MOE-LSC, School of Mathematical Sciences, CMA-Shanghai, SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University and Shanghai Artificial Intelligence Laboratory, Shanghai, China
| | - Xiaoqi Zheng
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
160
|
Martin KA, Papadoyannis ES, Schiavo JK, Fadaei SS, Issa HA, Song SC, Valencia SO, Temiz NZ, McGinley MJ, McCormick DA, Froemke RC. Vagus nerve stimulation recruits the central cholinergic system to enhance perceptual learning. Nat Neurosci 2024; 27:2152-2166. [PMID: 39284963 PMCID: PMC11932732 DOI: 10.1038/s41593-024-01767-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 08/15/2024] [Indexed: 11/07/2024]
Abstract
Perception can be refined by experience, up to certain limits. It is unclear whether perceptual limits are absolute or could be partially overcome via enhanced neuromodulation and/or plasticity. Recent studies suggest that peripheral nerve stimulation, specifically vagus nerve stimulation (VNS), can alter neural activity and augment experience-dependent plasticity, although little is known about central mechanisms recruited by VNS. Here we developed an auditory discrimination task for mice implanted with a VNS electrode. VNS applied during behavior gradually improved discrimination abilities beyond the level achieved by training alone. Two-photon imaging revealed VNS induced changes to auditory cortical responses and activated cortically projecting cholinergic axons. Anatomical and optogenetic experiments indicated that VNS can enhance task performance through activation of the central cholinergic system. These results highlight the importance of cholinergic modulation for the efficacy of VNS and may contribute to further refinement of VNS methodology for clinical conditions.
Collapse
Affiliation(s)
- Kathleen A Martin
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Eleni S Papadoyannis
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Jennifer K Schiavo
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Saba Shokat Fadaei
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Habon A Issa
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Soomin C Song
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Sofia Orrey Valencia
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Nesibe Z Temiz
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Matthew J McGinley
- Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | | | - Robert C Froemke
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.
- Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
161
|
Castro J, Maddern J, Erickson A, Harrington AM, Brierley SM. Peripheral and central neuroplasticity in a mouse model of endometriosis. J Neurochem 2024; 168:3777-3800. [PMID: 37165846 DOI: 10.1111/jnc.15843] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023]
Abstract
Chronic pelvic pain (CPP) is the most debilitating symptom of gynaecological disorders such as endometriosis. However, it remains unclear how sensory neurons from pelvic organs affected by endometriosis, such as the female reproductive tract, detect and transmit nociceptive events and how these signals are processed within the central nervous system (CNS). Using a previously characterized mouse model of endometriosis, we investigated whether the increased pain sensitivity occurring in endometriosis could be attributed to (i) changes in mechanosensory properties of sensory afferents innervating the reproductive tract, (ii) alterations in sensory input from reproductive organs to the spinal cord or (iii) neuroinflammation and sensitization of spinal neural circuits. Mechanosensitivity of vagina-innervating primary afferents was examined using an ex vivo single-unit extracellular recording preparation. Nociceptive signalling from the vagina to the spinal cord was quantified by phosphorylated MAP kinase ERK1/2 immunoreactivity. Immunohistochemistry was used to determine glial and neuronal circuit alterations within the spinal cord. We found that sensory afferents innervating the rostral, but not caudal portions of the mouse vagina, developed mechanical hypersensitivity in endometriosis. Nociceptive signalling from the vagina to the spinal cord was significantly enhanced in mice with endometriosis. Moreover, mice with endometriosis developed microgliosis, astrogliosis and enhanced substance P neurokinin-1 receptor immunoreactivity within the spinal cord, suggesting the development of neuroinflammation and sensitization of spinal circuitry in endometriosis. These results demonstrate endometriosis-induced neuroplasticity occurring at both peripheral and central sites of sensory afferent pathways. These findings may help to explain the altered sensitivity to pain in endometriosis and provide a novel platform for targeted pain relief treatments for this debilitating disorder.
Collapse
Affiliation(s)
- Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jessica Maddern
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Andelain Erickson
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Andrea M Harrington
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
162
|
van Hoof S, Kreye J, Cordero-Gómez C, Hoffmann J, Momsen Reincke S, Sánchez-Sendin E, Duong SL, Upadhya M, Dhangar D, Michór P, Woodhall GL, Küpper M, Oder A, Kuchling J, Koch SP, Mueller S, Boehm-Sturm P, von Kries JP, Finke C, Kirschstein T, Wright SK, Prüss H. Human cerebrospinal fluid monoclonal CASPR2 autoantibodies induce changes in electrophysiology, functional MRI, and behavior in rodent models. Brain Behav Immun 2024; 122:266-278. [PMID: 39142424 DOI: 10.1016/j.bbi.2024.08.027] [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: 03/21/2024] [Revised: 07/02/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024] Open
Abstract
Anti-contactin associated protein receptor 2 (CASPR2) encephalitis is a severe autoimmune encephalitis with a variable clinical phenotype including behavioral abnormalities, cognitive decline, epileptic seizures, peripheral nerve hyperexcitability and neuropathic pain. The detailed mechanisms of how CASPR2 autoantibodies lead to synaptic dysfunction and clinical symptoms are largely unknown. Aiming for analyses from the molecular to the clinical level, we isolated antibody-secreting cells from the cerebrospinal fluid of two patients with CASPR2 encephalitis. From these we cloned four anti-CASPR2 human monoclonal autoantibodies (mAbs) with strong binding to brain and peripheral nerves. All were highly hypermutated and mainly of the IgG4 subclass. Mutagenesis studies determined selective binding to the discoidin domain of CASPR2. Surface plasmon resonance revealed affinities with dissociation constants KD in the pico- to nanomolar range. CASPR2 mAbs interrupted the interaction of CASPR2 with its binding partner contactin 2 in vitro and were internalized after binding to CASPR2-expressing cells. Electrophysiological recordings of rat hippocampal slices after stereotactic injection of CASPR2 mAbs showed characteristic afterpotentials following electrical stimulation. In vivo experiments with intracerebroventricular administration of human CASPR2 mAbs into mice and rats showed EEG-recorded brain hyperexcitability but no spontaneous recurrent seizures. Behavioral assessment of infused mice showed a subtle clinical phenotype, mainly affecting sociability. Mouse brain MRI exhibited markedly reduced resting-state functional connectivity without short-term structural changes. Together, the experimental data support the direct pathogenicity of CASPR2 autoantibodies. The minimally invasive EEG and MRI techniques applied here may serve as novel objective, quantifiable tools for improved animal models, in particular for subtle neuropsychiatric phenotypes or repeated measurements.
Collapse
Affiliation(s)
- Scott van Hoof
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Helmholtz Innovation Lab BaoBab (Brain Antibody-omics and B-cell Lab), Berlin, Germany
| | - Jakob Kreye
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Helmholtz Innovation Lab BaoBab (Brain Antibody-omics and B-cell Lab), Berlin, Germany; Department of Pediatric Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - César Cordero-Gómez
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Julius Hoffmann
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - S Momsen Reincke
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Elisa Sánchez-Sendin
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Helmholtz Innovation Lab BaoBab (Brain Antibody-omics and B-cell Lab), Berlin, Germany
| | - Sophie L Duong
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Manoj Upadhya
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Divya Dhangar
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Paulina Michór
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Gavin L Woodhall
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Maraike Küpper
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany, Center of Transdisciplinary Neurosciences Rostock (CTNR), Germany
| | - Andreas Oder
- Screening Unit, Leibniz Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Joseph Kuchling
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Neurocure Cluster of Excellence, NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Stefan Paul Koch
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Center for Stroke Research Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité - Universitätsmedizin Berlin, Germany; Charité 3R, Replace, Reduce, Refine, Charité - Universitätsmedizin Berlin, Germany
| | - Susanne Mueller
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Center for Stroke Research Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité - Universitätsmedizin Berlin, Germany; Charité 3R, Replace, Reduce, Refine, Charité - Universitätsmedizin Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Center for Stroke Research Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité - Universitätsmedizin Berlin, Germany; Charité 3R, Replace, Reduce, Refine, Charité - Universitätsmedizin Berlin, Germany
| | - Jens Peter von Kries
- Screening Unit, Leibniz Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Carsten Finke
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Neurocure Cluster of Excellence, NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany, Center of Transdisciplinary Neurosciences Rostock (CTNR), Germany
| | - Sukhvir K Wright
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK; Department of Paediatric Neurology, The Birmingham Women's and Children's Hospital National Health Service Foundation Trust, Birmingham, UK
| | - Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Helmholtz Innovation Lab BaoBab (Brain Antibody-omics and B-cell Lab), Berlin, Germany.
| |
Collapse
|
163
|
Cao J, Zheng Z, Sun D, Chen X, Cheng R, Lv T, An Y, Zheng J, Song J, Wu L, Yang C. Decoder-seq enhances mRNA capture efficiency in spatial RNA sequencing. Nat Biotechnol 2024; 42:1735-1746. [PMID: 38228777 DOI: 10.1038/s41587-023-02086-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024]
Abstract
Spatial transcriptomics technologies with high resolution often lack high sensitivity in mRNA detection. Here we report a dendrimeric DNA coordinate barcoding design for spatial RNA sequencing (Decoder-seq), which offers both high sensitivity and high resolution. Decoder-seq combines dendrimeric nanosubstrates with microfluidic coordinate barcoding to generate spatial arrays with a DNA density approximately ten times higher than previously reported methods while maintaining flexibility in resolution. We show that the high RNA capture efficiency of Decoder-seq improved the detection of lowly expressed olfactory receptor (Olfr) genes in mouse olfactory bulbs and contributed to the discovery of a unique layer enrichment pattern for two Olfr genes. The near-cellular resolution provided by Decoder-seq has enabled the construction of a spatial single-cell atlas of the mouse hippocampus, revealing dendrite-enriched mRNAs in neurons. When applying Decoder-seq to human renal cell carcinomas, we dissected the heterogeneous tumor microenvironment across different cancer subtypes and identified spatial gradient-expressed genes related to epithelial-mesenchymal transition with the potential to predict tumor prognosis and progression.
Collapse
Affiliation(s)
- Jiao Cao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhong Zheng
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Sun
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Chen
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Cheng
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianpeng Lv
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu An
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junhua Zheng
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jia Song
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lingling Wu
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chaoyong Yang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, State Key Laboratory of Physical Chemical of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China.
| |
Collapse
|
164
|
Grimm C, Duss SN, Privitera M, Munn BR, Karalis N, Frässle S, Wilhelm M, Patriarchi T, Razansky D, Wenderoth N, Shine JM, Bohacek J, Zerbi V. Tonic and burst-like locus coeruleus stimulation distinctly shift network activity across the cortical hierarchy. Nat Neurosci 2024; 27:2167-2177. [PMID: 39284964 PMCID: PMC11537968 DOI: 10.1038/s41593-024-01755-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 08/07/2024] [Indexed: 11/07/2024]
Abstract
Noradrenaline (NA) release from the locus coeruleus (LC) changes activity and connectivity in neuronal networks across the brain, modulating multiple behavioral states. NA release is mediated by both tonic and burst-like LC activity. However, it is unknown whether the functional changes in target areas depend on these firing patterns. Using optogenetics, photometry, electrophysiology and functional magnetic resonance imaging in mice, we show that tonic and burst-like LC firing patterns elicit brain responses that hinge on their distinct NA release dynamics. During moderate tonic LC activation, NA release engages regions associated with associative processing, while burst-like stimulation biases the brain toward sensory processing. These activation patterns locally couple with increased astrocytic and inhibitory activity and change the brain's topological configuration in line with the hierarchical organization of the cerebral cortex. Together, these findings reveal how the LC-NA system achieves a nuanced regulation of global circuit operations.
Collapse
Affiliation(s)
- Christina Grimm
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuro-X institute, School of Engineering (STI), EPFL, Lausanne, Switzerland
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
| | - Sian N Duss
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Mattia Privitera
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Brandon R Munn
- School of Physics, The University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Nikolaos Karalis
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stefan Frässle
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zürich & ETH Zürich, Zürich, Switzerland
| | - Maria Wilhelm
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Tommaso Patriarchi
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
- Chemical Neuropharmacology, Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Daniel Razansky
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zürich, Zürich, Switzerland
- Institute of Biological and Medical Imaging (IBMI), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Nicole Wenderoth
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - James M Shine
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland.
| | - Valerio Zerbi
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
- Neuro-X institute, School of Engineering (STI), EPFL, Lausanne, Switzerland.
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland.
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
165
|
Vingan I, Phatarpekar S, Tung VSK, Hernández AI, Evgrafov OV, Alarcon JM. Spatially resolved transcriptomic signatures of hippocampal subregions and Arc-expressing ensembles in active place avoidance memory. Front Mol Neurosci 2024; 17:1386239. [PMID: 39544521 PMCID: PMC11560897 DOI: 10.3389/fnmol.2024.1386239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/20/2024] [Indexed: 11/17/2024] Open
Abstract
The rodent hippocampus is a spatially organized neuronal network that supports the formation of spatial and episodic memories. We conducted bulk RNA sequencing and spatial transcriptomics experiments to measure gene expression changes in the dorsal hippocampus following the recall of active place avoidance (APA) memory. Through bulk RNA sequencing, we examined the gene expression changes following memory recall across the functionally distinct subregions of the dorsal hippocampus. We found that recall induced differentially expressed genes (DEGs) in the CA1 and CA3 hippocampal subregions were enriched with genes involved in synaptic transmission and synaptic plasticity, while DEGs in the dentate gyrus (DG) were enriched with genes involved in energy balance and ribosomal function. Through spatial transcriptomics, we examined gene expression changes following memory recall across an array of spots encompassing putative memory-associated neuronal ensembles marked by the expression of the IEGs Arc, Egr1, and c-Jun. Within samples from both trained and untrained mice, the subpopulations of spatial transcriptomic spots marked by these IEGs were transcriptomically and spatially distinct from one another. DEGs detected between Arc + and Arc- spots exclusively in the trained mouse were enriched in several memory-related gene ontology terms, including "regulation of synaptic plasticity" and "memory." Our results suggest that APA memory recall is supported by regionalized transcriptomic profiles separating the CA1 and CA3 from the DG, transcriptionally and spatially distinct IEG expressing spatial transcriptomic spots, and biological processes related to synaptic plasticity as a defining the difference between Arc + and Arc- spatial transcriptomic spots.
Collapse
Affiliation(s)
- Isaac Vingan
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Shwetha Phatarpekar
- Institute for Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Victoria Sook Keng Tung
- School of Graduate Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Alejandro Iván Hernández
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Oleg V. Evgrafov
- Institute for Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- School of Graduate Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States
| | - Juan Marcos Alarcon
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| |
Collapse
|
166
|
Kashiwagi M, Beck G, Kanuka M, Arai Y, Tanaka K, Tatsuzawa C, Koga Y, Saito YC, Takagi M, Oishi Y, Sakaguchi M, Baba K, Ikuno M, Yamakado H, Takahashi R, Yanagisawa M, Murayama S, Sakurai T, Sakai K, Nakagawa Y, Watanabe M, Mochizuki H, Hayashi Y. A pontine-medullary loop crucial for REM sleep and its deficit in Parkinson's disease. Cell 2024; 187:6272-6289.e21. [PMID: 39303715 DOI: 10.1016/j.cell.2024.08.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/22/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024]
Abstract
Identifying the properties of the rapid eye movement (REM) sleep circuitry and its relation to diseases has been challenging due to the neuronal heterogeneity of the brainstem. Here, we show in mice that neurons in the pontine sublaterodorsal tegmentum (SubLDT) that express corticotropin-releasing hormone-binding protein (Crhbp+ neurons) and project to the medulla promote REM sleep. Within the medullary area receiving projections from Crhbp+ neurons, neurons expressing nitric oxide synthase 1 (Nos1+ neurons) project to the SubLDT and promote REM sleep, suggesting a positively interacting loop between the pons and the medulla operating as a core REM sleep circuit. Nos1+ neurons also project to areas that control wide forebrain activity. Ablating Crhbp+ neurons reduces sleep and impairs REM sleep atonia. In Parkinson's disease patients with REM sleep behavior disorders, CRHBP-immunoreactive neurons are largely reduced and contain pathologic α-synuclein, providing insight into the mechanisms underlying the sleep deficits characterizing this disease.
Collapse
Affiliation(s)
- Mitsuaki Kashiwagi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Goichi Beck
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mika Kanuka
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshifumi Arai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kaeko Tanaka
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Chika Tatsuzawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yumiko Koga
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki C Saito
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Marina Takagi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yo Oishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Masanori Sakaguchi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kousuke Baba
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masashi Ikuno
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 605-8507, Japan
| | - Hodaka Yamakado
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 605-8507, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 605-8507, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Japan Life Science Center for Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shigeo Murayama
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka 565-0871, Japan; Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi-Ku, Tokyo 173-0015, Japan
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kazuya Sakai
- Integrative Physiology of the Brain Arousal System, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR5292, School of Medicine, Claude Bernard University Lyon 1, 69373 Lyon, France
| | - Yoshimi Nakagawa
- Division of Complex Biosystem Research Institute of Natural Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yu Hayashi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
167
|
Gungor Aydin A, Lemenze A, Bieszczad KM. Functional diversities within neurons and astrocytes in the adult rat auditory cortex revealed by single-nucleus RNA sequencing. Sci Rep 2024; 14:25314. [PMID: 39455606 PMCID: PMC11511993 DOI: 10.1038/s41598-024-74732-7] [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: 04/24/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
The mammalian cerebral cortex is composed of a rich diversity of cell types. Sensory cortical cells are organized into networks that rely on their functional diversity to ultimately carry out a variety of sophisticated cognitive functions for perception, learning, and memory. The auditory cortex (AC) has been most extensively studied for its experience-dependent effects, including for perceptual learning and associative memory. Here, we used single-nucleus RNA sequencing (snRNA-seq) in the AC of the adult rat to investigate the breadth of transcriptionally diverse cell types that likely support the role of AC in experience-dependent functions. A variety of unique excitatory and inhibitory neuron subtypes were identified that harbor unique transcriptional profiles of genes with putative relevance for the adaptive neuroplasticity of cortical microcircuits. In addition, we report for the first time a diversity of astrocytes in AC that may represent functionally unique subtypes, including those that could integrate experience-dependent adult neuroplasticity at cortical synapses. Together, these results pave the way for building models of how cortical neurons work in concert with astrocytes to fulfill dynamic and experience-dependent cognitive functions.
Collapse
Affiliation(s)
- Aysegul Gungor Aydin
- Department of Psychology-Behavioral and Systems Neuroscience, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
| | - Alexander Lemenze
- Department of Pathology, Immunology, and Laboratory Medicine, Rutgers University, Newark, NJ, 07103, USA
| | - Kasia M Bieszczad
- Department of Psychology-Behavioral and Systems Neuroscience, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
- Rutgers Center for Cognitive Science (RuCCS), Rutgers University, Piscataway, NJ, 08854, USA.
- Department of Otolaryngology-Head and Neck Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
168
|
Douma EH, Stoop J, Lingl MVR, Smidt MP, van der Heide LP. Phosphodiesterase inhibition and Gucy2C activation enhance tyrosine hydroxylase Ser40 phosphorylation and improve 6-hydroxydopamine-induced motor deficits. Cell Biosci 2024; 14:132. [PMID: 39456033 PMCID: PMC11515495 DOI: 10.1186/s13578-024-01312-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Parkinson's disease is characterized by a progressive loss of dopaminergic neurons in the nigrostriatal pathway, leading to dopamine deficiency and motor impairments. Current treatments, such as L-DOPA, provide symptomatic relief but result in off-target effects and diminished efficacy over time. This study explores an alternative approach by investigating the activation of tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis. Specifically, we explore the effects of phosphodiesterase (PDE) inhibition and guanylate cyclase-C (GUCY2C) activation on tyrosine hydroxylase Ser40 phosphorylation and their impact on motor behavior in a 6-hydroxydopamine (6-OHDA) Parkinson's disease model. RESULTS Our findings demonstrate that increasing cyclic nucleotide levels through PDE inhibition and GUCY2C activation significantly enhances tyrosine hydroxylase Ser40 phosphorylation. In a Pitx3-deficient mouse model, which mimics the loss of dopaminergic neurons seen in Parkinson's disease, Ser40 phosphorylation remained manipulable despite reduced tyrosine hydroxylase protein levels. Moreover, we observed no evidence of tyrosine hydroxylase degradation due to Ser40 phosphorylation, challenging previous reports. Furthermore, both PDE inhibition and GUCY2C activation resulted in improved motor behavior in the 6-OHDA Parkinson's disease mouse model, highlighting the potential therapeutic benefits of these approaches. CONCLUSIONS This study underscores the therapeutic potential of enhancing tyrosine hydroxylase Ser40 phosphorylation to improve motor function in Parkinson's disease. Both PDE inhibition and GUCY2C activation represent promising non-invasive strategies to modulate endogenous dopamine biosynthesis and address motor deficits. These findings suggest that targeting cyclic nucleotide pathways could lead to novel therapeutic approaches, either as standalone treatments or in combination with existing therapies like L-DOPA, aiming to provide more durable symptom relief and potentially mitigate neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Erik H Douma
- Macrobian-Biotech B.V., Science Park 904, 1098 XH, Amsterdam, The Netherlands
- Parkinnova Therapeutics B.V., Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Jesse Stoop
- Macrobian-Biotech B.V., Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Matthijs V R Lingl
- Swammerdam Institute for Life Sciences, University of Amsterdam, Room C3.104, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Marten P Smidt
- Swammerdam Institute for Life Sciences, University of Amsterdam, Room C3.104, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Lars P van der Heide
- Swammerdam Institute for Life Sciences, University of Amsterdam, Room C3.104, Science Park 904, 1098 XH, Amsterdam, The Netherlands.
| |
Collapse
|
169
|
Klimczak P, Alcaide J, Gramuntell Y, Castillo-Gómez E, Varea E, Perez-Rando M, Nacher J. Long-term effects of a double hit murine model for schizophrenia on parvalbumin expressing cells and plasticity-related molecules in the thalamic reticular nucleus and the habenula. Transl Psychiatry 2024; 14:450. [PMID: 39448557 PMCID: PMC11502763 DOI: 10.1038/s41398-024-03166-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
The exposure to aversive experiences during early-life affects brain maturation and induces changes in behavior. Additionally, when these experiences coincide with subtle neurodevelopmental alterations, they may contribute to the emergence of psychiatric disorders, such as schizophrenia. Studies in patients and animal models have identified changes in parvalbumin (PV) expressing inhibitory neurons, highlighting their significance in the etiology of this disorder. Most studies have been focused on the cortex, but PV+ neurons also provide inhibitory input to diencephalic regions, particularly to the thalamus (through cells in the thalamic reticular nucleus, TRN) and the habenula. Remarkably, alterations in both nuclei have been described in schizophrenia. Some of these changes in PV+ cells may be mediated by perineuronal nets (PNN), specialized regions of the extracellular matrix that often surround them and regulate their synaptic input and activity. Interestingly, the physiological maturation and integration of PV+ neurons, which involves the assembly of PNN, occurs during early postnatal life. Plasticity molecules associated to inhibitory neurons, such as PSA-NCAM, or NMDA receptors (NMDAR) can also influence the structure and function of these cells. Growing evidence also indicates that glial cells regulate the physiology of PV+ neurons by influencing their maturation and modulating their synaptic connectivity. To explore the impact of early-life aversive experiences and concomitant subtle neurodevelopmental alterations on diencephalic PV+ cells, we analyzed adult male mice subjected to a double-hit model (DHM) of schizophrenia, combining a single injection of an NMDAR antagonist at P7 and post-weaning social isolation. We observed that exploratory behavior, PV+ neurons and their associated PNN, as well as PSA-NCAM and NMDAR expression and glial cells, in the TRN and the habenula were affected by the DHM or one of its factors. To our knowledge, this is the first report on such alterations in these diencephalic structures in an animal model combining neurodevelopmental alterations and early-life stress during adolescence. Our findings complement previous work on PV+ neurons in cortical regions and underscore the importance of studying diencephalic inhibitory networks and their intricate interactions with aversive experiences and neurodevelopmental alterations during early life in the context of schizophrenia.
Collapse
Affiliation(s)
- Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Julia Alcaide
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Esther Castillo-Gómez
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain
| | - Emilio Varea
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
| | - Marta Perez-Rando
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain.
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain.
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain.
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain.
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain.
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain.
| |
Collapse
|
170
|
Rubio-Teves M, Martín-Correa P, Alonso-Martínez C, Casas-Torremocha D, García-Amado M, Timonidis N, Sheiban FJ, Bakker R, Tiesinga P, Porrero C, Clascá F. Beyond Barrels: Diverse Thalamocortical Projection Motifs in the Mouse Ventral Posterior Complex. J Neurosci 2024; 44:e1096242024. [PMID: 39197940 PMCID: PMC11502235 DOI: 10.1523/jneurosci.1096-24.2024] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/29/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Thalamocortical pathways from the rodent ventral posterior (VP) thalamic complex to the somatosensory cerebral cortex areas are a key model in modern neuroscience. However, beyond the intensively studied projection from medial VP (VPM) to the primary somatosensory area (S1), the wiring of these pathways remains poorly characterized. We combined micropopulation tract-tracing and single-cell transfection experiments to map the pathways arising from different portions of the VP complex in male mice. We found that pathways originating from different VP regions show differences in area/lamina arborization pattern and axonal varicosity size. Neurons from the rostral VPM subnucleus innervate trigeminal S1 in point-to-point fashion. In contrast, a caudal VPM subnucleus innervates heavily and topographically second somatosensory area (S2), but not S1. Neurons in a third, intermediate VPM subnucleus innervate through branched axons both S1 and S2, with markedly different laminar patterns in each area. A small anterodorsal subnucleus selectively innervates dysgranular S1. The parvicellular VPM subnucleus selectively targets the insular cortex and adjacent portions of S1 and S2. Neurons in the rostral part of the lateral VP nucleus (VPL) innervate spinal S1, while caudal VPL neurons simultaneously target S1 and S2. Rostral and caudal VP nuclei show complementary patterns of calcium-binding protein expression. In addition to the cortex, neurons in caudal VP subnuclei target the sensorimotor striatum. Our finding of a massive projection from VP to S2 separate from the VP projections to S1 adds critical anatomical evidence to the notion that different somatosensory submodalities are processed in parallel in S1 and S2.
Collapse
Affiliation(s)
- Mario Rubio-Teves
- Department of Anatomy & Neuroscience, Autónoma de Madrid University, Madrid E28029, Spain
| | - Pablo Martín-Correa
- Department of Anatomy & Neuroscience, Autónoma de Madrid University, Madrid E28029, Spain
| | - Carmen Alonso-Martínez
- Department of Anatomy & Neuroscience, Autónoma de Madrid University, Madrid E28029, Spain
| | - Diana Casas-Torremocha
- Department of Anatomy & Neuroscience, Autónoma de Madrid University, Madrid E28029, Spain
| | - María García-Amado
- Department of Anatomy & Neuroscience, Autónoma de Madrid University, Madrid E28029, Spain
| | - Nestor Timonidis
- Department of Neuroinformatics, Donders Centre for Neuroscience, Radboud University Nijmegen, Nijmegen 6525 AJ, The Netherlands
| | - Francesco J Sheiban
- NearLab, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan 20133, Italy
| | - Rembrandt Bakker
- Department of Neuroinformatics, Donders Centre for Neuroscience, Radboud University Nijmegen, Nijmegen 6525 AJ, The Netherlands
- Inst. of Neuroscience and Medicine (INM-6) and Inst. for Advanced Simulation (IAS-6) and JARA BRAIN Inst. I, Julich Research Centre, Jülich 52428, Germany
| | - Paul Tiesinga
- Department of Neuroinformatics, Donders Centre for Neuroscience, Radboud University Nijmegen, Nijmegen 6525 AJ, The Netherlands
| | - César Porrero
- Department of Anatomy & Neuroscience, Autónoma de Madrid University, Madrid E28029, Spain
| | - Francisco Clascá
- Department of Anatomy & Neuroscience, Autónoma de Madrid University, Madrid E28029, Spain
| |
Collapse
|
171
|
Vatsa N, Brynildsen JK, Goralski TM, Kurgat K, Meyerdirk L, Breton L, DeWeerd D, Brasseur L, Turner L, Becker K, Gallik KL, Bassett DS, Henderson MX. Network analysis of α-synuclein pathology progression reveals p21-activated kinases as regulators of vulnerability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619411. [PMID: 39484617 PMCID: PMC11526907 DOI: 10.1101/2024.10.22.619411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
α-Synuclein misfolding and progressive accumulation drives a pathogenic process in Parkinson's disease. To understand cellular and network vulnerability to α-synuclein pathology, we developed a framework to quantify network-level vulnerability and identify new therapeutic targets at the cellular level. Full brain α-synuclein pathology was mapped in mice over 9 months. Empirical pathology data was compared to theoretical pathology estimates from a diffusion model of pathology progression along anatomical connections. Unexplained variance in the model enabled us to derive regional vulnerability that we compared to regional gene expression. We identified gene expression patterns that relate to regional vulnerability, including 12 kinases that were enriched in vulnerable regions. Among these, an inhibitor of group II PAKs demonstrated protection from neuron death and α-synuclein pathology, even after delayed compound treatment. This study provides a framework for the derivation of cellular vulnerability from network-based studies and identifies a promising therapeutic pathway for Parkinson's disease.
Collapse
Affiliation(s)
- Naman Vatsa
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Julia K. Brynildsen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas M. Goralski
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Kevin Kurgat
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Lindsay Meyerdirk
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Libby Breton
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniella DeWeerd
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura Brasseur
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | | | | | | | - Dani S. Bassett
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Electrical & Systems Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Santa Fe Institute, Santa Fe, NM, USA
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Michael X. Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Lead Contact
| |
Collapse
|
172
|
Jun S, Park H, Kim M, Kang S, Kim T, Kim D, Yamamoto Y, Tanaka-Yamamoto K. Increased understanding of complex neuronal circuits in the cerebellar cortex. Front Cell Neurosci 2024; 18:1487362. [PMID: 39497921 PMCID: PMC11532081 DOI: 10.3389/fncel.2024.1487362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/27/2024] [Indexed: 11/07/2024] Open
Abstract
The prevailing belief has been that the fundamental structures of cerebellar neuronal circuits, consisting of a few major neuron types, are simple and well understood. Given that the cerebellum has long been known to be crucial for motor behaviors, these simple yet organized circuit structures seemed beneficial for theoretical studies proposing neural mechanisms underlying cerebellar motor functions and learning. On the other hand, experimental studies using advanced techniques have revealed numerous structural properties that were not traditionally defined. These include subdivided neuronal types and their circuit structures, feedback pathways from output Purkinje cells, and the multidimensional organization of neuronal interactions. With the recent recognition of the cerebellar involvement in non-motor functions, it is possible that these newly identified structural properties, which are potentially capable of generating greater complexity than previously recognized, are associated with increased information capacity. This, in turn, could contribute to the wide range of cerebellar functions. However, it remains largely unknown how such structural properties contribute to cerebellar neural computations through the regulation of neuronal activity or synaptic transmissions. To promote further research into cerebellar circuit structures and their functional significance, we aim to summarize the newly identified structural properties of the cerebellar cortex and discuss future research directions concerning cerebellar circuit structures and their potential functions.
Collapse
Affiliation(s)
- Soyoung Jun
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Heeyoun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Muwoong Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Seulgi Kang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Taehyeong Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Integrated Biomedical and Life Sciences, Korea University, Seoul, Republic of Korea
| | - Daun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Life Science, Korea University, Seoul, Republic of Korea
| | - Yukio Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Keiko Tanaka-Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| |
Collapse
|
173
|
Li HS, Tan YT, Zhang XF. Enhancing spatial domain detection in spatial transcriptomics with EnSDD. Commun Biol 2024; 7:1358. [PMID: 39433947 PMCID: PMC11494180 DOI: 10.1038/s42003-024-07001-y] [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: 05/30/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Advancements in spatial transcriptomics have transformed our understanding of organ function and tissue microenvironment. However, accurately identifying spatial domains to depict genome heterogeneity and cellular interactions remains a challenge. In this study, we propose EnSDD (Ensemble-learning for Spatial Domain Detection), a method that ingeniously integrates eight state-of-the-art spatial domain detection methods to automatically identify spatial domains. A key innovation of EnSDD is its dynamic weighting mechanism within the ensemble learning process, which optimizes the contribution of each base model and provides a performance evaluation metric without the need for ground truth data. By leveraging the spatial domains identified through EnSDD, we incorporate the detection of domain-specific spatially variable genes and the spatial distribution of cell types, thereby providing deeper insights into tissue heterogeneity. We validate EnSDD across diverse spatial transcriptomics datasets from various tissue organizational structures. Our results demonstrate that EnSDD significantly enhances spatial domain identification accuracy, identifies genes with spatial expression patterns, and reveals domain-specific cell type enrichment patterns, offering invaluable insights into tissue spatial heterogeneity and regionalization.
Collapse
Affiliation(s)
- Hui-Sheng Li
- School of Mathematical Sciences, Zhejiang University of Technology, Hangzhou, 310023, China
| | - Yu-Ting Tan
- School of Mathematics and Statistics, and Hubei Key Lab-Math. Sci., Central China Normal University, Wuhan, 430079, China
| | - Xiao-Fei Zhang
- School of Mathematics and Statistics, and Hubei Key Lab-Math. Sci., Central China Normal University, Wuhan, 430079, China.
- Key Laboratory of Nonlinear Analysis & Applications (Ministry of Education), Central China Normal University, Wuhan, 430079, China.
| |
Collapse
|
174
|
Kronman FN, Liwang JK, Betty R, Vanselow DJ, Wu YT, Tustison NJ, Bhandiwad A, Manjila SB, Minteer JA, Shin D, Lee CH, Patil R, Duda JT, Xue J, Lin Y, Cheng KC, Puelles L, Gee JC, Zhang J, Ng L, Kim Y. Developmental mouse brain common coordinate framework. Nat Commun 2024; 15:9072. [PMID: 39433760 PMCID: PMC11494176 DOI: 10.1038/s41467-024-53254-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
3D brain atlases are key resources to understand the brain's spatial organization and promote interoperability across different studies. However, unlike the adult mouse brain, the lack of developing mouse brain 3D reference atlases hinders advancements in understanding brain development. Here, we present a 3D developmental common coordinate framework (DevCCF) spanning embryonic day (E)11.5, E13.5, E15.5, E18.5, and postnatal day (P)4, P14, and P56, featuring undistorted morphologically averaged atlas templates created from magnetic resonance imaging and co-registered high-resolution light sheet fluorescence microscopy templates. The DevCCF with 3D anatomical segmentations can be downloaded or explored via an interactive 3D web-visualizer. As a use case, we utilize the DevCCF to unveil GABAergic neuron emergence in embryonic brains. Moreover, we map the Allen CCFv3 and spatial transcriptome cell-type data to our stereotaxic P56 atlas. In summary, the DevCCF is an openly accessible resource for multi-study data integration to advance our understanding of brain development.
Collapse
Affiliation(s)
- Fae N Kronman
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Josephine K Liwang
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Rebecca Betty
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Daniel J Vanselow
- Department of Pathology, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Nicholas J Tustison
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | | | - Steffy B Manjila
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Jennifer A Minteer
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Donghui Shin
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Choong Heon Lee
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Rohan Patil
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Jeffrey T Duda
- Department of Radiology, Penn Image Computing and Science Lab, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jian Xue
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yingxi Lin
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Keith C Cheng
- Department of Pathology, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, Universidad de Murcia, and Murcia Arrixaca Institute for Biomedical Research (IMIB), Murcia, Spain
| | - James C Gee
- Department of Radiology, Penn Image Computing and Science Lab, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jiangyang Zhang
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA.
| |
Collapse
|
175
|
Giffen KP, Liu H, Yamane KL, Li Y, Chen L, Kramer KL, Zallocchi M, He DZ. Molecular specializations underlying phenotypic differences in inner ear hair cells of zebrafish and mice. Front Neurol 2024; 15:1437558. [PMID: 39484049 PMCID: PMC11524865 DOI: 10.3389/fneur.2024.1437558] [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: 05/23/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction Hair cells (HCs) are the sensory receptors of the auditory and vestibular systems in the inner ears of vertebrates that selectively transduce mechanical stimuli into electrical activity. Although all HCs have the hallmark stereocilia bundle for mechanotransduction, HCs in non-mammals and mammals differ in their molecular specialization in the apical, basolateral, and synaptic membranes. HCs of non-mammals, such as zebrafish (zHCs), are electrically tuned to specific frequencies and possess an active process in the stereocilia bundle to amplify sound signals. Mammalian HCs, in contrast, are not electrically tuned and achieve amplification by somatic motility of outer HCs (OHCs). Methods To understand the genetic mechanisms underlying differences between adult zebrafish and mammalian HCs, we compared their RNA-seq-characterized transcriptomes, focusing on protein-coding orthologous genes related to HC specialization. Results There was considerable shared expression of gene orthologs among the HCs, including those genes associated with mechanotransduction, ion transport/channels, and synaptic signaling. However, there were some notable differences in expression among zHCs, OHCs, and inner HCs (IHCs), which likely underlie the distinctive physiological properties of each cell type. For example, OHCs highly express Slc26a5 which encodes the motor protein prestin that contributes to OHC electromotility. However, zHCs have only weak expression of slc26a5, and subsequently showed no voltage-dependent electromotility when measured. Notably, the zHCs expressed more paralogous genes including those associated with HC-specific functions and transcriptional activity, though it is unknown whether they have functions similar to their mammalian counterparts. There was overlap in the expressed genes associated with a known hearing phenotype. Discussion Our analyses unveil substantial differences in gene expression patterns that may explain phenotypic specialization of zebrafish and mouse HCs. This dataset also includes several protein-coding genes to further the functional characterization of HCs and study of HC evolution from non-mammals to mammals.
Collapse
Affiliation(s)
- Kimberlee P. Giffen
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Department of Basic Sciences, Augusta University/University of Georgia Medical Partnership, Athens, GA, United States
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Huizhan Liu
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Kacey L. Yamane
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Yi Li
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
- Department of Otorhinolaryngology, Beijing Tongren Hospital, Beijing Capital Medical University, Beijing, China
| | - Lei Chen
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
| | - Kenneth L. Kramer
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Marisa Zallocchi
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - David Z. He
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
176
|
Grouza V, Bagheri H, Liu H, Tuznik M, Wu Z, Robinson N, Siminovitch KA, Peterson AC, Rudko DA. Ultra-high-resolution mapping of myelin and g-ratio in a panel of Mbp enhancer-edited mouse strains using microstructural MRI. Neuroimage 2024; 300:120850. [PMID: 39260782 DOI: 10.1016/j.neuroimage.2024.120850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/27/2024] [Accepted: 09/09/2024] [Indexed: 09/13/2024] Open
Abstract
Non-invasive myelin water fraction (MWF) and g-ratio mapping using microstructural MRI have the potential to offer critical insights into brain microstructure and our understanding of neuroplasticity and neuroinflammation. By leveraging a unique panel of variably hypomyelinating mouse strains, we validated a high-resolution, model-free image reconstruction method for whole-brain MWF mapping. Further, by employing a bipolar gradient echo MRI sequence, we achieved high spatial resolution and robust mapping of MWF and g-ratio across the whole mouse brain. Our regional white matter-tract specific analyses demonstrated a graded decrease in MWF in white matter tracts which correlated strongly with myelin basic protein gene (Mbp) mRNA levels. Using these measures, we derived the first sensitive calibrations between MWF and Mbp mRNA in the mouse. Minimal changes in axonal density supported our hypothesis that observed MWF alterations stem from hypomyelination. Overall, our work strongly emphasizes the potential of non-invasive, MRI-derived MWF and g-ratio modeling for both preclinical model validation and ultimately translation to humans.
Collapse
Affiliation(s)
- Vladimir Grouza
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Hooman Bagheri
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Hanwen Liu
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Marius Tuznik
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Zhe Wu
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Nicole Robinson
- Histology Innovation Platform, Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Katherine A Siminovitch
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Mount Sinai Hospital, Lunenfeld-Tanenbaum and Toronto General Hospital Research Institutes, Toronto, Ontario, Canada
| | - Alan C Peterson
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Department of Human Genetics, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - David A Rudko
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
177
|
Huerta PT, Strohl JJ, Carrión J. Brain imaging and machine learning reveal uncoupled functional network for contextual threat memory in long sepsis. RESEARCH SQUARE 2024:rs.3.rs-4870916. [PMID: 39483911 PMCID: PMC11527171 DOI: 10.21203/rs.3.rs-4870916/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Positron emission tomography (PET) is a highly sensitive tool for studying physiology and metabolism through positron-emitting radionuclides that label molecular targets in the body with unparalleled specificity, without disturbing their biological function. Here, we introduce a small-animal technique called behavioral task-associated PET (beta-PET) consisting of two scans: the first after a mouse is familiarized with a conditioning chamber, and the second upon recall of contextual threat. Associative threat conditioning occurs between the scans. Beta-PET focuses on brain regions encoding threat memory (e.g., amygdala, prefrontal cortex) and contextual aspects (e.g., hippocampus, subiculum, entorhinal cortex). Our results show that beta-PET identifies a biologically defined functional network encoding contextual threat memory and its uncoupling in a mouse model of long sepsis. Moreover, machine learning algorithms (linear logistic regression) and ordinal trends analysis demonstrate that beta-PET robustly predicts the behavioral defense response and its breakdown during long sepsis.
Collapse
Affiliation(s)
| | | | - Joseph Carrión
- Feinstein Institutes for Medical Research, Northwell Health
| |
Collapse
|
178
|
Westi EW, Molhemi S, Hansen CT, Skoven CS, Knopper RW, Ahmad DA, Rindshøj MB, Ameen AO, Hansen B, Kohlmeier KA, Aldana BI. Comprehensive Analysis of the 5xFAD Mouse Model of Alzheimer's Disease Using dMRI, Immunohistochemistry, and Neuronal and Glial Functional Metabolic Mapping. Biomolecules 2024; 14:1294. [PMID: 39456227 PMCID: PMC11505609 DOI: 10.3390/biom14101294] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by complex interactions between neuropathological markers, metabolic dysregulation, and structural brain changes. In this study, we utilized a multimodal approach, combining immunohistochemistry, functional metabolic mapping, and microstructure sensitive diffusion MRI (dMRI) to progressively investigate these interactions in the 5xFAD mouse model of AD. Our analysis revealed age-dependent and region-specific accumulation of key AD markers, including amyloid-beta (Aβ), GFAP, and IBA1, with significant differences observed between the hippocampal formation and upper and lower regions of the cortex by 6 months of age. Functional metabolic mapping validated localized disruptions in energy metabolism, with glucose hypometabolism in the hippocampus and impaired astrocytic metabolism in the cortex. Notably, increased cortical glutaminolysis suggested a shift in microglial metabolism, reflecting an adaptive response to neuroinflammatory processes. While dMRI showed no significant microstructural differences between 5xFAD and wild-type controls, the study highlights the importance of metabolic alterations as critical events in AD pathology. These findings emphasize the need for targeted therapeutic strategies addressing specific metabolic disturbances and underscore the potential of integrating advanced imaging with metabolic and molecular analyses to advance our understanding of AD progression.
Collapse
Affiliation(s)
- Emil W. Westi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (E.W.W.); (C.T.H.); (D.A.A.); (M.B.R.); (A.O.A.); (K.A.K.)
| | - Saba Molhemi
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; (S.M.); (C.S.S.); (R.W.K.); (B.H.)
| | - Caroline Termøhlen Hansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (E.W.W.); (C.T.H.); (D.A.A.); (M.B.R.); (A.O.A.); (K.A.K.)
| | - Christian Stald Skoven
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; (S.M.); (C.S.S.); (R.W.K.); (B.H.)
| | - Rasmus West Knopper
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; (S.M.); (C.S.S.); (R.W.K.); (B.H.)
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100040, China
| | - Dashne Amein Ahmad
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (E.W.W.); (C.T.H.); (D.A.A.); (M.B.R.); (A.O.A.); (K.A.K.)
| | - Maja B. Rindshøj
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (E.W.W.); (C.T.H.); (D.A.A.); (M.B.R.); (A.O.A.); (K.A.K.)
| | - Aishat O. Ameen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (E.W.W.); (C.T.H.); (D.A.A.); (M.B.R.); (A.O.A.); (K.A.K.)
| | - Brian Hansen
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; (S.M.); (C.S.S.); (R.W.K.); (B.H.)
| | - Kristi A. Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (E.W.W.); (C.T.H.); (D.A.A.); (M.B.R.); (A.O.A.); (K.A.K.)
| | - Blanca I. Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (E.W.W.); (C.T.H.); (D.A.A.); (M.B.R.); (A.O.A.); (K.A.K.)
| |
Collapse
|
179
|
Manoharan VT, Abdelkareem A, Gill G, Brown S, Gillmor A, Hall C, Seo H, Narta K, Grewal S, Dang NH, Ahn BY, Osz K, Lun X, Mah L, Zemp F, Mahoney D, Senger DL, Chan JA, Morrissy AS. Spatiotemporal modeling reveals high-resolution invasion states in glioblastoma. Genome Biol 2024; 25:264. [PMID: 39390467 PMCID: PMC11465563 DOI: 10.1186/s13059-024-03407-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 09/29/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Diffuse invasion of glioblastoma cells through normal brain tissue is a key contributor to tumor aggressiveness, resistance to conventional therapies, and dismal prognosis in patients. A deeper understanding of how components of the tumor microenvironment (TME) contribute to overall tumor organization and to programs of invasion may reveal opportunities for improved therapeutic strategies. RESULTS Towards this goal, we apply a novel computational workflow to a spatiotemporally profiled GBM xenograft cohort, leveraging the ability to distinguish human tumor from mouse TME to overcome previous limitations in the analysis of diffuse invasion. Our analytic approach, based on unsupervised deconvolution, performs reference-free discovery of cell types and cell activities within the complete GBM ecosystem. We present a comprehensive catalogue of 15 tumor cell programs set within the spatiotemporal context of 90 mouse brain and TME cell types, cell activities, and anatomic structures. Distinct tumor programs related to invasion align with routes of perivascular, white matter, and parenchymal invasion. Furthermore, sub-modules of genes serving as program network hubs are highly prognostic in GBM patients. CONCLUSION The compendium of programs presented here provides a basis for rational targeting of tumor and/or TME components. We anticipate that our approach will facilitate an ecosystem-level understanding of the immediate and long-term consequences of such perturbations, including the identification of compensatory programs that will inform improved combinatorial therapies.
Collapse
Affiliation(s)
- Varsha Thoppey Manoharan
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Aly Abdelkareem
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Gurveer Gill
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Samuel Brown
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Aaron Gillmor
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Courtney Hall
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Heewon Seo
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Kiran Narta
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Sean Grewal
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Ngoc Ha Dang
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Bo Young Ahn
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Kata Osz
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Xueqing Lun
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Laura Mah
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Franz Zemp
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Douglas Mahoney
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Donna L Senger
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.
| | - Jennifer A Chan
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada.
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| | - A Sorana Morrissy
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada.
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
180
|
Huchede P, Meyer S, Berthelot C, Hamadou M, Bertrand-Chapel A, Rakotomalala A, Manceau L, Tomine J, Lespinasse N, Lewandowski P, Cordier-Bussat M, Broutier L, Dutour A, Rochet I, Blay JY, Degletagne C, Attignon V, Montero-Carcaboso A, Le Grand M, Pasquier E, Vasiljevic A, Gilardi-Hebenstreit P, Meignan S, Leblond P, Ribes V, Cosset E, Castets M. BMP2 and BMP7 cooperate with H3.3K27M to promote quiescence and invasiveness in pediatric diffuse midline gliomas. eLife 2024; 12:RP91313. [PMID: 39373720 PMCID: PMC11458179 DOI: 10.7554/elife.91313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024] Open
Abstract
Pediatric diffuse midline gliomas (pDMG) are an aggressive type of childhood cancer with a fatal outcome. Their major epigenetic determinism has become clear, notably with the identification of K27M mutations in histone H3. However, the synergistic oncogenic mechanisms that induce and maintain tumor cell phenotype have yet to be deciphered. In 20 to 30% of cases, these tumors have an altered BMP signaling pathway with an oncogenic mutation on the BMP type I receptor ALK2, encoded by ACVR1. However, the potential impact of the BMP pathway in tumors non-mutated for ACVR1 is less clear. By integrating bulk, single-cell, and spatial transcriptomic data, we show here that the BMP signaling pathway is activated at similar levels between ACVR1 wild-type and mutant tumors and identify BMP2 and BMP7 as putative activators of the pathway in a specific subpopulation of cells. By using both pediatric isogenic glioma lines genetically modified to overexpress H3.3K27M and patients-derived DIPG cell lines, we demonstrate that BMP2/7 synergizes with H3.3K27M to induce a transcriptomic rewiring associated with a quiescent but invasive cell state. These data suggest a generic oncogenic role for the BMP pathway in gliomagenesis of pDMG and pave the way for specific targeting of downstream effectors mediating the K27M/BMP crosstalk.
Collapse
Affiliation(s)
- Paul Huchede
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Swann Meyer
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Clement Berthelot
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Maud Hamadou
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Adrien Bertrand-Chapel
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Andria Rakotomalala
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER Cancer Heterogeneity Plasticity and Resistance to Therapies, Centre Oscar LambretLilleFrance
| | - Line Manceau
- Université Paris Cité, CNRS, Institut Jacques MonodParisFrance
| | - Julia Tomine
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Nicolas Lespinasse
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Paul Lewandowski
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER Cancer Heterogeneity Plasticity and Resistance to Therapies, Centre Oscar LambretLilleFrance
| | - Martine Cordier-Bussat
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Laura Broutier
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Aurelie Dutour
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Isabelle Rochet
- Multisite Institute of Pathology, Groupement Hospitalier Est du CHU de Lyon, Hôpital Femme-Mère EnfantBronFrance
| | - Jean-Yves Blay
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | | | | | - Angel Montero-Carcaboso
- Preclinical Therapeutics and Drug Delivery Research Program, Department of Oncology, Hospital Sant Joan de DéuBarcelonaSpain
| | - Marion Le Grand
- Centre de Recherche en Cancérologie de Marseille (CRCM), Université Aix-Marseille, Institut Paoli- Calmettes, Centre de Lutte Contre le Cancer de la région PACA, INSERM 1068, CNRS 7258MarseilleFrance
| | - Eddy Pasquier
- Centre de Recherche en Cancérologie de Marseille (CRCM), Université Aix-Marseille, Institut Paoli- Calmettes, Centre de Lutte Contre le Cancer de la région PACA, INSERM 1068, CNRS 7258MarseilleFrance
| | - Alexandre Vasiljevic
- Multisite Institute of Pathology, Groupement Hospitalier Est du CHU de Lyon, Hôpital Femme-Mère EnfantBronFrance
| | | | - Samuel Meignan
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER Cancer Heterogeneity Plasticity and Resistance to Therapies, Centre Oscar LambretLilleFrance
| | - Pierre Leblond
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
- Department of Pediatric Oncology, Institute of Pediatric Hematology and Oncology (IHOPe), Centre Léon BérardLyonFrance
| | - Vanessa Ribes
- Université Paris Cité, CNRS, Institut Jacques MonodParisFrance
| | - Erika Cosset
- GLIMMER Of lIght (GLIoblastoma MetabolisM, HetERogeneity, and OrganoIds) team, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Marie Castets
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| |
Collapse
|
181
|
Pavešković M, De-Paula RB, Ojelade SA, Tantry EK, Kochukov MY, Bao S, Veeraragavan S, Garza AR, Srivastava S, Song SY, Fujita M, Duong DM, Bennett DA, De Jager PL, Seyfried NT, Dickinson ME, Heaney JD, Arenkiel BR, Shulman JM. Alzheimer's disease risk gene CD2AP is a dose-sensitive determinant of synaptic structure and plasticity. Hum Mol Genet 2024; 33:1815-1832. [PMID: 39146503 PMCID: PMC11458016 DOI: 10.1093/hmg/ddae115] [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: 02/27/2024] [Revised: 06/15/2024] [Indexed: 08/17/2024] Open
Abstract
CD2-Associated protein (CD2AP) is a candidate susceptibility gene for Alzheimer's disease, but its role in the mammalian central nervous system remains largely unknown. We show that CD2AP protein is broadly expressed in the adult mouse brain, including within cortical and hippocampal neurons, where it is detected at pre-synaptic terminals. Deletion of Cd2ap altered dendritic branching and spine density, and impaired ubiquitin-proteasome system activity. Moreover, in mice harboring either one or two copies of a germline Cd2ap null allele, we noted increased paired-pulse facilitation at hippocampal Schaffer-collateral synapses, consistent with a haploinsufficient requirement for pre-synaptic release. Whereas conditional Cd2ap knockout in the brain revealed no gross behavioral deficits in either 3.5- or 12-month-old mice, Cd2ap heterozygous mice demonstrated subtle impairments in discrimination learning using a touchscreen task. Based on unbiased proteomics, partial or complete loss of Cd2ap triggered perturbation of proteins with roles in protein folding, lipid metabolism, proteostasis, and synaptic function. Overall, our results reveal conserved, dose-sensitive requirements for CD2AP in the maintenance of neuronal structure and function, including synaptic homeostasis and plasticity, and inform our understanding of possible cell-type specific mechanisms in Alzheimer's Disease.
Collapse
Affiliation(s)
- Matea Pavešković
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Ruth B De-Paula
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Quantitative and Computational Biology Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Shamsideen A Ojelade
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Evelyne K Tantry
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Mikhail Y Kochukov
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Suyang Bao
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Surabi Veeraragavan
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Alexandra R Garza
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Snigdha Srivastava
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Medical Scientist Training Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Si-Yuan Song
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, United States
| | - Duc M Duong
- Departments of Biochemistry and Neurology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, 600 S. Paulina Street, Chicago, IL 60612, United States
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, United States
| | - Nicholas T Seyfried
- Departments of Biochemistry and Neurology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Mary E Dickinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Benjamin R Arenkiel
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Joshua M Shulman
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Center for Alzheimer’s and Neurodegenerative Diseases, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| |
Collapse
|
182
|
Jing J, Hu M, Ngodup T, Ma Q, Lau SNN, Ljungberg C, McGinley MJ, Trussell LO, Jiang X. Molecular logic for cellular specializations that initiate the auditory parallel processing pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.15.539065. [PMID: 37293040 PMCID: PMC10245571 DOI: 10.1101/2023.05.15.539065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The cochlear nuclear complex (CN), the starting point for all central auditory processing, comprises a suite of neuronal cell types that are highly specialized for neural coding of acoustic signals, yet molecular logic governing cellular 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 new 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 remarkable 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 illustrates molecular logic for cellular specializations and enables genetic dissection of auditory processing and hearing disorders with unprecedented 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
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX,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
| | - 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
|
183
|
Zhao YB, Wang SZ, Guo WT, Wang L, Tang X, Li JN, Xu L, Zhou QX. Hippocampal dipeptidyl peptidase 9 bidirectionally regulates memory associated with synaptic plasticity. J Adv Res 2024:S2090-1232(24)00433-8. [PMID: 39369958 DOI: 10.1016/j.jare.2024.09.031] [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: 02/29/2024] [Revised: 09/11/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024] Open
Abstract
INTRODUCTION Subtypes of the dipeptidyl peptidase (DPP) family, such as DPP4, are reportedly associated with memory impairment. DPP9 is widely distributed in cells throughout the body, including the brain. However, whether DPP9 regulates memory has not yet been elucidated. OBJECTIVES This study aimed to elucidate the role of DPP9 in memory, as well as the underlying molecular mechanism. METHODS We performed immunofluorescence on mouse brains to explore the distribution of DPP9 in different brain regions and used AAV vectors to construct knockdown and overexpression models. The effects of changing DPP9 expression on memory were demonstrated through behavioral experiments. Finally, we used electrophysiology, proteomics and affinity purification mass spectrometry (AP-MS) to study the molecular mechanism by which DPP9 affects memory. RESULTS Here, we report that DPP9, which is found almost exclusively in neurons, is expressed and has enzyme activity in many brain regions, especially in the hippocampus. Hippocampal DPP9 expression increases after fear memory formation. Fear memory was impaired by DPP9 knockdown and enhanced by DPP9 protein overexpression in the hippocampus. According to subsequent hippocampal proteomics, multiple pathways, including the peptidase pathway, which can be bidirectionally regulated by DPP9. DPP9 directly interacts with its enzymatic substrate neuropeptide Y (NPY) in neurons. Hippocampal long-term potentiation (LTP) is also bidirectionally regulated by DPP9. Moreover, inhibiting DPP enzyme activity impaired both LTP and memory. In addition, AP-MS revealed that DPP9-interacting proteins are involved in the functions of dendritic spines and axons. By combining AP-MS and proteomics, DPP9 was shown to play a role in regulating actin functions. CONCLUSION Taken together, our findings reveal that DPP9 affects the CNS not only through enzymatic activity but also through protein-protein interactions. This study provides new insights into the molecular mechanisms of memory and DPP family functions.
Collapse
Affiliation(s)
- Ya-Bo Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences (CAS), Kunming 650223, China
| | - Shi-Zhe Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences (CAS), Kunming 650223, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Wen-Ting Guo
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences (CAS), Kunming 650223, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Le Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences (CAS), Kunming 650223, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xun Tang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences (CAS), Kunming 650223, China
| | - Jin-Nan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences (CAS), Kunming 650223, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences (CAS), Kunming 650223, China; University of Chinese Academy of Sciences, Beijing 101408, China; KIZ-SU Joint Laboratory of Animal Model and Drug Development, China
| | - Qi-Xin Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences (CAS), Kunming 650223, China; University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
184
|
Usmanova DR, Plata G, Vitkup D. Functional Optimization in Distinct Tissues and Conditions Constrains the Rate of Protein Evolution. Mol Biol Evol 2024; 41:msae200. [PMID: 39431545 PMCID: PMC11523136 DOI: 10.1093/molbev/msae200] [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] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 10/22/2024] Open
Abstract
Understanding the main determinants of protein evolution is a fundamental challenge in biology. Despite many decades of active research, the molecular and cellular mechanisms underlying the substantial variability of evolutionary rates across cellular proteins are not currently well understood. It also remains unclear how protein molecular function is optimized in the context of multicellular species and why many proteins, such as enzymes, are only moderately efficient on average. Our analysis of genomics and functional datasets reveals in multiple organisms a strong inverse relationship between the optimality of protein molecular function and the rate of protein evolution. Furthermore, we find that highly expressed proteins tend to be substantially more functionally optimized. These results suggest that cellular expression costs lead to more pronounced functional optimization of abundant proteins and that the purifying selection to maintain high levels of functional optimality significantly slows protein evolution. We observe that in multicellular species both the rate of protein evolution and the degree of protein functional efficiency are primarily affected by expression in several distinct cell types and tissues, specifically, in developed neurons with upregulated synaptic processes in animals and in young and fast-growing tissues in plants. Overall, our analysis reveals how various constraints from the molecular, cellular, and species' levels of biological organization jointly affect the rate of protein evolution and the level of protein functional adaptation.
Collapse
Affiliation(s)
- Dinara R Usmanova
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Germán Plata
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
- BiomEdit, Fishers, IN 46037, USA
| | - Dennis Vitkup
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
- Department of Biomedical Informatics, Columbia University, New York, NY 10032, USA
| |
Collapse
|
185
|
Ishar J, Tam YM, Mages S, Klughammer J. BoReMi: Bokeh-based jupyter-interface for registering spatio-molecular data to related microscopy images. PLoS Comput Biol 2024; 20:e1012504. [PMID: 39374301 PMCID: PMC11486395 DOI: 10.1371/journal.pcbi.1012504] [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: 12/16/2023] [Revised: 10/17/2024] [Accepted: 09/18/2024] [Indexed: 10/09/2024] Open
Abstract
Spatio-molecular data and microscopy images provide complementary information, essential to study structure and function of spatially organised multicellular systems such as healthy or diseased tissues. However, aligning these two types of data can be challenging due to distortions and differences in resolution, orientation, and position. Manual registration is tedious but may be necessary for challenging samples as well as for the generation of ground-truth data sets that enable benchmarking of existing and emerging automated alignment tools. To make the process of manual registration more convenient, efficient, and integrated, we created BoReMi, a python-based, Jupyter-integrated, visual tool that offers all the relevant functionalities for aligning and registering spatio-molecular data and associated microscopy images. We showcase BoReMi's utility using publicly available data and images and make BoReMi as well as an interactive demo available on GitHub.
Collapse
Affiliation(s)
- Jaspreet Ishar
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Yee Man Tam
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany
| | - Simon Mages
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany
| | - Johanna Klughammer
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
186
|
Liu Y, Lin D, Najam SS, Huang S, Song M, Sirakawin C, Zhao C, Jiang H, Konopka W, Herzig S, Vinnikov IA. Functional redundancy between glucocorticoid and mineralocorticoid receptors in mature corticotropin-releasing hormone neurons protects from obesity. Obesity (Silver Spring) 2024; 32:1885-1896. [PMID: 39315404 DOI: 10.1002/oby.24116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVE Here, we aimed to investigate the role of glucocorticoid and mineralocorticoid receptors (GRs and MRs, respectively) in the regulation of energy homeostasis. METHODS We used three mouse models with simultaneous deletion of GRs and MRs in either forebrain neurons, the paraventricular nucleus, or corticotropin-releasing hormone (CRH) neurons and compared them with wild-type controls or isolated knockout groups. In addition to body weight, food intake, energy expenditure, insulin sensitivity, fat/lean mass distribution, and plasma corticosterone levels, we also performed transcriptomic analysis of CRH neurons and assessed their response to melanocortinergic stimulation. RESULTS Similar to global double-knockout models, deletion of GRs and MRs specifically in mature CRH neurons resulted in obesity. Importantly, the latter was accompanied by insulin resistance, but not increased plasma corticosterone levels. Transcriptomic analysis of these neurons revealed upregulation of several genes involved in postsynaptic signal transduction, including the Ptk2b gene, which encodes proline-rich tyrosine kinase 2. Knockout of both nuclear receptors leads to upregulation of Ptk2b in CRH neurons, which results in their diminished responsiveness to melanocortinergic stimulation. CONCLUSIONS Our data demonstrate the functional redundancy of GRs and MRs in CRH neurons to maintain energy homeostasis and prevent obesity. Simultaneous targeting of both receptors might represent an unprecedented approach to counteract obesity.
Collapse
Affiliation(s)
- Yu Liu
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Dongfa Lin
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, China
| | - Syeda Sadia Najam
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Shangyuan Huang
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Muyi Song
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Chaweewan Sirakawin
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Catherine Zhao
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Haixia Jiang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Witold Konopka
- Laboratory of Neuroplasticity and Metabolism, Department of Life Sciences and Biotechnology, Łukasiewicz PORT Polish Center for Technology Development, Wrocław, Poland
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Munich Germany; Joint Heidelberg-IDC Translational Diabetes Unit, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany; Molecular Metabolic Control, Technical University Munich, Munich, Germany; German Center for Diabetes Research, Munich, Germany
| | - Ilya A Vinnikov
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
187
|
Tagliatti E, Bizzotto M, Morini R, Filipello F, Rasile M, Matteoli M. Prenatal drivers of microglia vulnerability in the adult. Immunol Rev 2024; 327:100-110. [PMID: 39508795 DOI: 10.1111/imr.13418] [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] [Indexed: 11/15/2024]
Abstract
Environmental insults during early development heavily affect brain trajectories. Among these, maternal infections, high-fat diet regimens, and sleep disturbances pose a significant risk for neurodevelopmental derangements in the offspring. Notably, scattered evidence is starting to emerge that also paternal lifestyle habits may impact the offspring development. Given their key role in controlling neurogenesis, synaptogenesis and shaping neuronal circuits, microglia represent the most likely suspects of mediating the detrimental effects of prenatal insults. For some of these environmental triggers, like maternal infections, ample literature evidence demonstrates the central role of microglia, also delineating the specific transcriptomic and proteomic profiles induced by these insults. In other contexts, the analysis of microglia is still in its infancy. Fostering these studies is needed to define microglia as potential therapeutic target in the frame of disorders consequent to maternal immune activation.
Collapse
Affiliation(s)
| | | | | | | | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| |
Collapse
|
188
|
Safari MS, Woerl P, Garmsiri C, Weber D, Kwiatkowski M, Hotze M, Kuenkel L, Lang L, Erlacher M, Gelpi E, Hainfellner JA, Baier G, Baier-Bitterlich G, Zur Nedden S. Glucose-1,6-bisphosphate: A new gatekeeper of cerebral mitochondrial pyruvate uptake. Mol Metab 2024; 88:102018. [PMID: 39182844 PMCID: PMC11404074 DOI: 10.1016/j.molmet.2024.102018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024] Open
Abstract
OBJECTIVE Glucose-1,6-bisphosphate (G-1,6-BP), a byproduct of glycolysis that is synthesized by phosphoglucomutase 2 like 1 (PGM2L1), is particularly abundant in neurons. G-1,6-BP is sensitive to the glycolytic flux, due to its dependence on 1,3-bisphosphoglycerate as phosphate donor, and the energy state, due to its degradation by inosine monophosphate-activated phosphomannomutase 1. Since the exact role of this metabolite remains unclear, our aim was to elucidate the specific function of G-1,6-BP in the brain. METHODS The effect of PGM2L1 on neuronal post-ischemic viability was assessed by siRNA-mediated knockdown of PGM2L1 in primary mouse neurons. Acute mouse brain slices were used to correlate the reduction in G-1,6-BP upon ischemia to changes in carbon metabolism by 13C6-glucose tracing. A drug affinity responsive target stability assay was used to test if G-1,6-BP interacts with the mitochondrial pyruvate carrier (MPC) subunits in mouse brain protein extracts. Human embryonic kidney cells expressing a MPC bioluminescence resonance energy transfer sensor were used to analyze how PGM2L1 overexpression affects MPC activity. The effect of G-1,6-BP on mitochondrial pyruvate uptake and oxygen consumption rates was analyzed in isolated mouse brain mitochondria. PGM2L1 and a predicted upstream kinase were overexpressed in a human neuroblastoma cell line and G-1,6-BP levels were measured. RESULTS We found that G-1,6-BP in mouse brain slices was quickly degraded upon ischemia and reperfusion. Knockdown of PGM2L1 in mouse neurons reduced post-ischemic viability, indicating that PGM2L1 plays a neuroprotective role. The reduction in G-1,6-BP upon ischemia was not accompanied by alterations in glycolytic rates but we did see a reduced 13C6-glucose incorporation into citrate, suggesting a potential role in mitochondrial pyruvate uptake or metabolism. Indeed, G-1,6-BP interacted with both MPC subunits and overexpression of PGM2L1 increased MPC activity. G-1,6-BP, at concentrations found in the brain, enhanced mitochondrial pyruvate uptake and pyruvate-induced oxygen consumption rates. Overexpression of a predicted upstream kinase inhibited PGM2L1 activity, showing that besides metabolism, also signaling pathways can regulate G-1,6-BP levels. CONCLUSIONS We provide evidence that G-1,6-BP positively regulates mitochondrial pyruvate uptake and post-ischemic neuronal viability. These compelling data reveal a novel mechanism by which neurons can couple glycolysis-derived pyruvate to the tricarboxylic acid cycle. This process is sensitive to the glycolytic flux, the cell's energetic state, and upstream signaling cascades, offering many regulatory means to fine-tune this critical metabolic step.
Collapse
Affiliation(s)
- Motahareh Solina Safari
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Priska Woerl
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Carolin Garmsiri
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Dido Weber
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Marcel Kwiatkowski
- Department of Biochemistry, Institute of Bioanalytic & Intermediary Metabolism, University of Innsbruck, 6020 Innsbruck, Austria
| | - Madlen Hotze
- Department of Biochemistry, Institute of Bioanalytic & Intermediary Metabolism, University of Innsbruck, 6020 Innsbruck, Austria
| | - Louisa Kuenkel
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Luisa Lang
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Matthias Erlacher
- CCB-Biocenter, Institute of Genomics and RNomics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Ellen Gelpi
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes A Hainfellner
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Gottfried Baier
- Institute for Cell Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Gabriele Baier-Bitterlich
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stephanie Zur Nedden
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
189
|
Tang Z, Luo S, Zeng H, Huang J, Sui X, Wu M, Wang X. Search and match across spatial omics samples at single-cell resolution. Nat Methods 2024; 21:1818-1829. [PMID: 39294367 PMCID: PMC11529703 DOI: 10.1038/s41592-024-02410-7] [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: 05/23/2023] [Accepted: 08/12/2024] [Indexed: 09/20/2024]
Abstract
Spatial omics technologies characterize tissue molecular properties with spatial information, but integrating and comparing spatial data across different technologies and modalities is challenging. A comparative analysis tool that can search, match and visualize both similarities and differences of molecular features in space across multiple samples is lacking. To address this, we introduce CAST (cross-sample alignment of spatial omics), a deep graph neural network-based method enabling spatial-to-spatial searching and matching at the single-cell level. CAST aligns tissues based on intrinsic similarities of spatial molecular features and reconstructs spatially resolved single-cell multi-omic profiles. CAST further allows spatially resolved differential analysis (∆Analysis) to pinpoint and visualize disease-associated molecular pathways and cell-cell interactions and single-cell relative translational efficiency profiling to reveal variations in translational control across cell types and regions. CAST serves as an integrative framework for seamless single-cell spatial data searching and matching across technologies, modalities and sample conditions.
Collapse
Affiliation(s)
- Zefang Tang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shuchen Luo
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hu Zeng
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jiahao Huang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Sui
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Morgan Wu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xiao Wang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
190
|
Nie XP, Xu XS, Feng Z, Wang W, Ma C, Yang YX, Li JN, Zhou QX, Xu FQ, Luo MH, Zhou JN, Gong H, Xu L. Depicting Primate-Like Granular Dorsolateral Prefrontal Cortex in the Chinese Tree Shrew. eNeuro 2024; 11:ENEURO.0307-24.2024. [PMID: 39455280 PMCID: PMC11514722 DOI: 10.1523/eneuro.0307-24.2024] [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: 07/10/2024] [Revised: 09/04/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
It remains unknown whether the Chinese tree shrew, regarded as the closest sister of primate, has evolved a dorsolateral prefrontal cortex (dlPFC) comparable with primates that is characterized by a fourth layer (L4) enriched with granular cells and reciprocal connections with the mediodorsal nucleus (MD). Here, we reported that following AAV-hSyn-EGFP expression in the MD neurons, the fluorescence micro-optical sectioning tomography revealed their projection trajectories and targeted brain areas, such as the hippocampus, the corpus striatum, and the dlPFC. Cre-dependent transsynaptic viral tracing identified the MD projection terminals that targeted the L4 of the dlPFC, in which the presence of granular cells was confirmed via cytoarchitectural studies by using the Nissl, Golgi, and vGlut2 stainings. Additionally, the L5/6 of the dlPFC projected back to the MD. These results suggest that the tree shrew has evolved a primate-like dlPFC which can serve as an alternative for studying cognition-related functions of the dlPFC.
Collapse
Affiliation(s)
- Xiu-Peng Nie
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Science Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Kunming 650223, China
| | - Xiao-Shan Xu
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Science Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Kunming 650223, China
| | - Zhao Feng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Wei Wang
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Science Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Kunming 650223, China
| | - Chen Ma
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Science Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Kunming 650223, China
| | - Yue-Xiong Yang
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Science Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Kunming 650223, China
| | - Jin-Nan Li
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Science Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Kunming 650223, China
| | - Qi-Xin Zhou
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Science Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Kunming 650223, China
| | - Fu-Qiang Xu
- Shen Zhen Institute of Advanced Technology, Chinese Academy of Sciences, Xi Li Shen Zhen University Town, Shenzhen 518055, China
- Chinese Academy of Sciences Centre for Excellence in Brain Science and Intelligent Technology, Shanghai 200031, China
| | - Min-Hua Luo
- State Key Laboratory of Virology and Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Jiang-Ning Zhou
- Chinese Academy of Sciences Centre for Excellence in Brain Science and Intelligent Technology, Shanghai 200031, China
- Institute of Brain Science, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Chinese Academy of Science Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Lin Xu
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Science Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Kunming 650223, China
- Chinese Academy of Sciences Centre for Excellence in Brain Science and Intelligent Technology, Shanghai 200031, China
| |
Collapse
|
191
|
Olson RJ, Bartlett L, Sonneborn A, Milton R, Bretton-Granatoor Z, Firdous A, Harris AZ, Abbas AI. Decoupling of cortical activity from behavioral state following administration of the classic psychedelic DOI. Neuropharmacology 2024; 257:110030. [PMID: 38851531 PMCID: PMC11260522 DOI: 10.1016/j.neuropharm.2024.110030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/02/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
Administration or consumption of classic psychedelics (CPs) leads to profound changes in experience which are often described as highly novel and meaningful. They have shown substantial promise in treating depressive symptoms and may be therapeutic in other situations. Although research suggests that the therapeutic response is correlated with the intensity of the experience, the neural circuit basis for the alterations in experience caused by CPs requires further study. The medial prefrontal cortex (mPFC), where CPs have been shown to induce rapid, 5-HT2A receptor-dependent structural and neurophysiological changes, is believed to be a key site of action. To investigate the acute neural circuit changes induced by CPs, we recorded single neurons and local field potentials in the mPFC of freely behaving male mice after administration of the 5-HT2A/2C receptor-selective CP, 2,5-Dimethoxy-4-iodoamphetamine (DOI). We segregated recordings into active and rest periods in order to examine cortical activity during desynchronized (active) and synchronized (rest) states. We found that DOI induced a robust decrease in low frequency power when animals were at rest, attenuating the usual synchronization that occurs during less active behavioral states. DOI also increased broadband gamma power and suppressed activity in fast-spiking neurons in both active and rest periods. Together, these results suggest that the CP DOI induces persistent desynchronization in mPFC, including during rest when mPFC typically exhibits more synchronized activity. This shift in cortical dynamics may in part underlie the longer-lasting effects of CPs on plasticity, and may be critical to their therapeutic properties.
Collapse
Affiliation(s)
- Randall J Olson
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland OR 97239, USA
| | - Lowell Bartlett
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland OR 97239, USA
| | - Alex Sonneborn
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland OR 97239, USA
| | - Russell Milton
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland OR 97239, USA
| | | | - Ayesha Firdous
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10034, USA
| | - Alexander Z Harris
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10034, USA; Department of Psychiatry, Columbia University, New York, NY, 10034, USA
| | - Atheir I Abbas
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland OR 97239, USA; Department of Psychiatry, Oregon Health and Science University, Portland OR 97239, USA; VA Portland Health Care System, Portland OR, 97239, USA.
| |
Collapse
|
192
|
Grattan DR. Does the brain make prolactin? J Neuroendocrinol 2024; 36:e13432. [PMID: 39041379 DOI: 10.1111/jne.13432] [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: 03/17/2024] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024]
Abstract
The prolactin receptor (Prlr) is widely expressed in the brain, particularly in the hypothalamus. Prolactin also has an increasing range of well-characterised effects on central nervous system function. Because of this, over many years, there has been interest in whether the hormone itself is also expressed within the brain, perhaps acting as a neuropeptide to regulate brain function via its receptor in neurons. The aim of this invited review is to critically evaluate the evidence for brain production of prolactin. Unlike the evidence for the Prlr, evidence for brain prolactin is inconsistent and variable. A range of different antibodies have been used, each characterising a different distribution of prolactin-like immunoreactivity. Prolactin mRNA has been detected in the brain, but only at levels markedly lower than seen in the pituitary gland. Importantly, it has largely only been detected by highly sensitive amplification-based techniques, and the extreme sensitivity means there is a risk of false-positive data. Modern in situ hybridisation methods and single-cell RNA sequencing have not provided supporting evidence, but it is hard to prove a negative! Finally, I acknowledge and discuss the possibility that prolactin might be produced in the brain under specific circumstances, such as to promote a neuroprotective response to cell damage. Collectively, however, based on this analysis, I have formed the opinion that brain production of prolactin is unlikely, and even if occurs, it is of little physiological consequence. Most, if not all of the brain actions of prolactin can be explained by pituitary prolactin gaining access to the brain.
Collapse
Affiliation(s)
- David R Grattan
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| |
Collapse
|
193
|
Zouridis IS, Balsamo G, Preston-Ferrer P, Burgalossi A. Anatomical and electrophysiological analysis of the fasciola cinerea of the mouse hippocampus. Hippocampus 2024; 34:528-539. [PMID: 39105449 DOI: 10.1002/hipo.23623] [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: 06/13/2023] [Revised: 05/20/2024] [Accepted: 07/03/2024] [Indexed: 08/07/2024]
Abstract
The hippocampus is considered essential for several forms of declarative memory, including spatial and social memory. Despite the extensive research of the classic subfields of the hippocampus, the fasciola cinerea (FC)-a medially located structure within the hippocampal formation-has remained largely unexplored. In the present study, we performed a morpho-functional characterization of principal neurons in the mouse FC. Using in vivo juxtacellular recording of single neurons, we found that FC neurons are distinct from neighboring CA1 pyramidal cells, both morphologically and electrophysiologically. Specifically, FC neurons displayed non-pyramidal morphology and granule cell-like apical dendrites. Compared to neighboring CA1 pyramidal neurons, FC neurons exhibited more regular in vivo firing patterns and a lower tendency to fire spikes at short interspike intervals. Furthermore, tracing experiments revealed that the FC receives inputs from the lateral but not the medial entorhinal cortex and CA3, and it provides a major intra-hippocampal projection to the septal CA2 and sparser inputs to the distal CA1. Overall, our results indicate that the FC is a morphologically and electrophysiologically distinct subfield of the hippocampal formation; given the established role of CA2 in social memory and seizure initiation, the unique efferent intra-hippocampal connectivity of the FC points to possible roles in social cognition and temporal lobe epilepsy.
Collapse
Affiliation(s)
- Ioannis S Zouridis
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
- Graduate Training Centre of Neuroscience-International Max-Planck Research School (IMPRS), Tübingen, Germany
| | - Giuseppe Balsamo
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
- Graduate Training Centre of Neuroscience-International Max-Planck Research School (IMPRS), Tübingen, Germany
| | - Patricia Preston-Ferrer
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - Andrea Burgalossi
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| |
Collapse
|
194
|
Yang L, Fang LZ, Lynch MR, Xu CS, Hahm H, Zhang Y, Heitmeier MR, Costa V, Samineni VK, Creed MC. Transcriptomic landscape of mammalian ventral pallidum at single-cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595793. [PMID: 38826431 PMCID: PMC11142225 DOI: 10.1101/2024.05.24.595793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
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 snRNA-seq 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 neuro receptors that could be targeted within specific VP cell types for functional investigations. Teaser Genetic identity of ventral pallidum cell types is conserved across rodents and primates at the transcriptional level.
Collapse
|
195
|
Hao S, Zhu X, Huang Z, Yang Q, Liu H, Wu Y, Zhan Y, Dong Y, Li C, Wang H, Haasdijk E, Wu Z, Li S, Yan H, Zhu L, Guo S, Wang Z, Ye A, Lin Y, Cui L, Tan X, Liu H, Wang M, Chen J, Zhong Y, Du W, Wang G, Lai T, Cao M, Yang T, Xu Y, Li L, Yu Q, Zhuang Z, Xia Y, Lei Y, An Y, Cheng M, Zhao Y, Han L, Yuan Y, Song X, Song Y, Gu L, Liu C, Lin X, Wang R, Wang Z, Wang Y, Li S, Li H, Song J, Chen M, Zhou W, Yuan N, Sun S, Wang S, Chen Y, Zheng M, Fang J, Zhang R, Zhang S, Chai Q, Liu J, Wei W, He J, Zhou H, Sun Y, Liu Z, Liu C, Yao J, Liang Z, Xu X, Poo M, Li C, De Zeeuw CI, Shen Z, Liu Z, Liu L, Liu S, Sun Y, Liu C. Cross-species single-cell spatial transcriptomic atlases of the cerebellar cortex. Science 2024; 385:eado3927. [PMID: 39325889 DOI: 10.1126/science.ado3927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 08/14/2024] [Indexed: 09/28/2024]
Abstract
The molecular and cellular organization of the primate cerebellum remains poorly characterized. We obtained single-cell spatial transcriptomic atlases of macaque, marmoset, and mouse cerebella and identified primate-specific cell subtypes, including Purkinje cells and molecular-layer interneurons, that show different expression of the glutamate ionotropic receptor Delta type subunit 2 (GRID2) gene. Distinct gene expression profiles were found in anterior, posterior, and vestibular regions in all species, whereas region-selective gene expression was predominantly observed in the granular layer of primates and in the Purkinje layer of mice. Gene expression gradients in the cerebellar cortex matched well with functional connectivity gradients revealed with awake functional magnetic resonance imaging, with more lobule-specific differences between primates and mice than between two primate species. These comprehensive atlases and comparative analyses provide the basis for understanding cerebellar evolution and function.
Collapse
Affiliation(s)
| | - Xiaojia Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Zhi Huang
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Qianqian Yang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hean Liu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yan Wu
- BGI Research, Hangzhou 310030, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yafeng Zhan
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Dong
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- Lingang Laboratory, Shanghai 200031, China
| | - Chao Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - He Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Elize Haasdijk
- Department of Neuroscience, Erasmus MC, 3015 GE Rotterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, 1105 BA Amsterdam, Netherlands
| | - Zihan Wu
- Tencent AI Lab, Shenzhen 518057, China
| | - Shenglong Li
- BGI Research, Hangzhou 310030, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Haotian Yan
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lijing Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | | | - Zefang Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Aojun Ye
- University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Luman Cui
- BGI Research, Shenzhen 518083, China
| | - Xing Tan
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | | | - Mingli Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- Lingang Laboratory, Shanghai 200031, China
| | - Jing Chen
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Yanqing Zhong
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wensi Du
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Guangling Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tingting Lai
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Mengdi Cao
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Yuanfang Xu
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ling Li
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Qian Yu
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | | | - Ying Xia
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ying Lei
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yingjie An
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengnan Cheng
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yun Zhao
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lei Han
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yue Yuan
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xinxiang Song
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yumo Song
- BGI Research, Shenzhen 518083, China
| | - Liqin Gu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Chang Liu
- BGI Research, Shenzhen 518083, China
| | | | - Ruiqi Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | | | - Yang Wang
- BGI Research, Shenzhen 518083, China
| | - Shenyu Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huanhuan Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jingjing Song
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengni Chen
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wanqiu Zhou
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Nini Yuan
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Suhong Sun
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shiwen Wang
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mingyuan Zheng
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiao Fang
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ruiyi Zhang
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shuzhen Zhang
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qinwen Chai
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiabing Liu
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wu Wei
- Lingang Laboratory, Shanghai 200031, China
| | - Jie He
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haibo Zhou
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yangang Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Liu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chuanyu Liu
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China
| | | | - Zhifeng Liang
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xun Xu
- BGI Research, Hangzhou 310030, China
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Muming Poo
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201602, China
| | - Chengyu Li
- Lingang Laboratory, Shanghai 200031, China
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, 3015 GE Rotterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, 1105 BA Amsterdam, Netherlands
| | - Zhiming Shen
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201602, China
| | - Zhiyong Liu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
| | - Yidi Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Cirong Liu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Genetic Evolution & Animal Models, Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
196
|
Otten J, Dan S, Rostin L, Profetto AE, Lardenoije R, Klengel T. Spatial transcriptomics reveals modulation of transcriptional networks across brain regions after auditory threat conditioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614979. [PMID: 39386587 PMCID: PMC11463379 DOI: 10.1101/2024.09.25.614979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Prior research has demonstrated genome-wide transcriptional changes related to fear and anxiety across species, often focusing on individual brain regions or cell types. However, the extent of gene expression differences across brain regions and how these changes interact at the level of transcriptional connectivity remains unclear. To address this, we performed spatial transcriptomics RNAseq analyses in an auditory threat conditioning paradigm in mice. We generated a spatial transcriptomic atlas of a coronal mouse brain section covering cortical and subcortical regions, corresponding to histologically defined regions. Our finding revealed widespread transcriptional responses across all brain regions examined, particularly in the medial and lateral habenula, and the choroid plexus. Network analyses highlighted altered transcriptional connectivity between cortical and subcortical regions, emphasizing the role of steroidogenic factor 1. These results provide new insights into the transcriptional networks involved in auditory threat conditioning, enhancing our understanding of molecular and neural mechanisms underlying fear and anxiety disorders.
Collapse
|
197
|
Drexler R, Drinnenberg A, Gavish A, Yalcin B, Shamardani K, Rogers A, Mancusi R, Taylor KR, Kim YS, Woo PJ, Ravel A, Tatlock E, Ramakrishnan C, Ayala-Sarmiento AE, Pacheco DRF, Siverts L, Daigle TL, Tasic B, Zeng H, Breunig JJ, Deisseroth K, Monje M. Cholinergic Neuronal Activity Promotes Diffuse Midline Glioma Growth through Muscarinic Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.21.614235. [PMID: 39386427 PMCID: PMC11463519 DOI: 10.1101/2024.09.21.614235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Neuronal activity promotes the proliferation of healthy oligodendrocyte precursor cells (OPC) and their malignant counterparts, gliomas. Many gliomas arise from and closely resemble oligodendroglial lineage precursors, including diffuse midline glioma (DMG), a cancer affecting midline structures such as the thalamus, brainstem and spinal cord. In DMG, glutamatergic and GABAergic neuronal activity promotes progression through both paracrine signaling and through bona-fide neuron-to-glioma synapses. However, the putative roles of other neuronal subpopulations - especially neuromodulatory neurons located in the brainstem that project to long-range target sites in midline anatomical locations where DMGs arise - remain largely unexplored. Here, we demonstrate that the activity of cholinergic midbrain neurons modulates both healthy OPC and malignant DMG proliferation in a circuit-specific manner at sites of long-range cholinergic projections. Optogenetic stimulation of the cholinergic pedunculopontine nucleus (PPN) promotes glioma growth in pons, while stimulation of the laterodorsal tegmentum nucleus (LDT) facilitates proliferation in thalamus, consistent with the predominant projection patterns of each cholinergic midbrain nucleus. Reciprocal signaling was evident, as increased activity of cholinergic neurons in the PPN and LDT was observed in pontine DMG-bearing mice. In co-culture, hiPSC-derived cholinergic neurons form neuron-to-glioma networks with DMG cells and robustly promote proliferation. Single-cell RNA sequencing analyses revealed prominent expression of the muscarinic receptor genes CHRM1 and CHRM3 in primary patient DMG samples, particularly enriched in the OPC-like tumor subpopulation. Acetylcholine, the neurotransmitter cholinergic neurons release, exerts a direct effect on DMG tumor cells, promoting increased proliferation and invasion through muscarinic receptors. Pharmacological blockade of M1 and M3 acetylcholine receptors abolished the activity-regulated increase in DMG proliferation in cholinergic neuron-glioma co-culture and in vivo. Taken together, these findings demonstrate that midbrain cholinergic neuron long-range projections to midline structures promote activity-dependent DMG growth through M1 and M3 cholinergic receptors, mirroring a parallel proliferative effect on healthy OPCs.
Collapse
Affiliation(s)
- Richard Drexler
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
- These authors contributed equally
| | - Antonia Drinnenberg
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| | - Avishai Gavish
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Belgin Yalcin
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Kiarash Shamardani
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Abigail Rogers
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Rebecca Mancusi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Kathryn R Taylor
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Yoon Seok Kim
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Pamelyn J Woo
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Alexandre Ravel
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Eva Tatlock
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Alberto E Ayala-Sarmiento
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Joshua J Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford, CA 94305, USA
| |
Collapse
|
198
|
Shen R, Cheng M, Wang W, Fan Q, Yan H, Wen J, Yuan Z, Yao J, Li Y, Yuan J. Graph domain adaptation-based framework for gene expression enhancement and cell type identification in large-scale spatially resolved transcriptomics. Brief Bioinform 2024; 25:bbae576. [PMID: 39508445 PMCID: PMC11541786 DOI: 10.1093/bib/bbae576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/25/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
Spatially resolved transcriptomics (SRT) technologies facilitate gene expression profiling with spatial resolution in a naïve state. Nevertheless, current SRT technologies exhibit limitations, manifesting as either low transcript detection sensitivity or restricted gene throughput. These constraints result in diminished precision and coverage in gene measurement. In response, we introduce SpaGDA, a sophisticated deep learning-based graph domain adaptation framework for both scenarios of gene expression imputation and cell type identification in spatially resolved transcriptomics data by impartially transferring knowledge from reference scRNA-seq data. Systematic benchmarking analyses across several SRT datasets generated from different technologies have demonstrated SpaGDA's superior effectiveness compared to state-of-the-art methods in both scenarios. Further applied to three SRT datasets of different biological contexts, SpaGDA not only better recovers the well-established knowledge sourced from public atlases and existing scientific literature but also yields a more informative spatial expression pattern of genes. Together, these results demonstrate that SpaGDA can be used to overcome the challenges of current SRT data and provide more accurate insights into biological processes or disease development. The SpaGDA is available in https://github.com/shenrb/SpaGDA.
Collapse
Affiliation(s)
- Rongbo Shen
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, No. 1 Xinzao Road, Xinzao Town, Panyu District, Guangzhou 510005, China
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
- Tencent AI Lab, Shenzhen 518000, China
| | - Meiling Cheng
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan 430074, China
| | - Wencang Wang
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Qi Fan
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Huan Yan
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Jiayue Wen
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Handan Road, Shanghai 200433, China
| | | | - Yixue Li
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, No. 1 Xinzao Road, Xinzao Town, Panyu District, Guangzhou 510005, China
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Jiao Yuan
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, No. 1 Xinzao Road, Xinzao Town, Panyu District, Guangzhou 510005, China
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| |
Collapse
|
199
|
Havel V, Kruegel AC, Bechand B, McIntosh S, Stallings L, Hodges A, Wulf MG, Nelson M, Hunkele A, Ansonoff M, Pintar JE, Hwu C, Ople RS, Abi-Gerges N, Zaidi SA, Katritch V, Yang M, Javitch JA, Majumdar S, Hemby SE, Sames D. Oxa-Iboga alkaloids lack cardiac risk and disrupt opioid use in animal models. Nat Commun 2024; 15:8118. [PMID: 39304653 PMCID: PMC11415492 DOI: 10.1038/s41467-024-51856-y] [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: 09/07/2023] [Accepted: 08/16/2024] [Indexed: 09/22/2024] Open
Abstract
Ibogaine and its main metabolite noribogaine provide important molecular prototypes for markedly different treatment of substance use disorders and co-morbid mental health illnesses. However, these compounds present a cardiac safety risk and a highly complex molecular mechanism. We introduce a class of iboga alkaloids - termed oxa-iboga - defined as benzofuran-containing iboga analogs and created via structural editing of the iboga skeleton. The oxa-iboga compounds lack the proarrhythmic adverse effects of ibogaine and noribogaine in primary human cardiomyocytes and show superior efficacy in animal models of opioid use disorder in male rats. They act as potent kappa opioid receptor agonists in vitro and in vivo, but exhibit atypical behavioral features compared to standard kappa opioid agonists. Oxa-noribogaine induces long-lasting suppression of morphine, heroin, and fentanyl intake after a single dose or a short treatment regimen, reversal of persistent opioid-induced hyperalgesia, and suppression of opioid drug seeking in rodent relapse models. As such, oxa-iboga compounds represent mechanistically distinct iboga analogs with therapeutic potential.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Ibogaine/analogs & derivatives
- Ibogaine/pharmacology
- Ibogaine/therapeutic use
- Rats
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Opioid-Related Disorders/drug therapy
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/pharmacology
- Rats, Sprague-Dawley
- Disease Models, Animal
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/genetics
- Alkaloids/pharmacology
- Hyperalgesia/chemically induced
- Hyperalgesia/drug therapy
Collapse
Affiliation(s)
- Václav Havel
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Andrew C Kruegel
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Benjamin Bechand
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Scot McIntosh
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point University, High Point, NC, 27268, USA
| | - Leia Stallings
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point University, High Point, NC, 27268, USA
| | - Alana Hodges
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point University, High Point, NC, 27268, USA
| | - Madalee G Wulf
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Mel Nelson
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, 10032, USA
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Amanda Hunkele
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St Louis and Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Michael Ansonoff
- Department of Neuroscience and Cell Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - John E Pintar
- Department of Neuroscience and Cell Biology, Rutgers University, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University, Piscataway, NJ, 08854, USA
| | - Christopher Hwu
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Rohini S Ople
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St Louis and Washington University School of Medicine, St Louis, MO, 63110, USA
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Najah Abi-Gerges
- AnaBios Corporation, 1155 Island Ave, Suite 200, San Diego, CA, 92101, USA
| | - Saheem A Zaidi
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Chemistry, Bridge Institute, Michelson Center for Convergent Sciences, University of Southern California, Los Angeles, CA, 90089, USA
| | - Vsevolod Katritch
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Chemistry, Bridge Institute, Michelson Center for Convergent Sciences, University of Southern California, Los Angeles, CA, 90089, USA
| | - Mu Yang
- Mouse Neurobehavioral Core facility, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jonathan A Javitch
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, 10032, USA
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Susruta Majumdar
- Department of Neurology and Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St Louis and Washington University School of Medicine, St Louis, MO, 63110, USA
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Scott E Hemby
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point University, High Point, NC, 27268, USA
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, NY, 10027, USA.
- The Zuckerman Mind Brain Behavior Institute at Columbia University, New York, NY, USA.
| |
Collapse
|
200
|
Sultan ZW, Najac M, Raman IM. Control of action potential afterdepolarizations in the inferior olive by inactivating A-type currents through K V4 channels. J Physiol 2024. [PMID: 39303148 PMCID: PMC11922792 DOI: 10.1113/jp286818] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024] Open
Abstract
Neurons of the inferior olive (IO) fire action potentials with large, long-lasting afterdepolarizations (ADPs). Broader ADPs support more spikes in climbing fibre axons and evoke longer bursts of complex spikes in Purkinje cells, which affect the magnitude and sign of cerebellar synaptic plasticity. In the present study, we investigated the ionic mechanisms that regulate IO action potential waveforms by making whole-cell recordings in brainstem slices from C57BL6/J mice. IO spikes evoked from rest had ADPs of ∼30 ms. After 500-ms hyperpolarizations, however, evoked action potentials were brief (1-2 ms), lacking ADPs altogether. Because such preconditioning should maximally recruit depolarizing Ih and T-type currents and minimize repolarizing Ca-dependent currents known to shape the ADP, the rapid action potential downstroke suggested additional, dominant recovery of voltage-gated K currents at negative voltages. Under voltage clamp, outward currents evoked from -98 mV included large, voltage-gated, rapidly inactivating 'A-type' K currents. These currents had a steep availability curve with half-inactivation at -85 mV, suitable for recruitment by small hyperpolarizations. The fast decay time constant increased with depolarization, as is typical of KV4 channels. The KV4 channel blocker AmmTx3 almost eliminated inactivating currents and broadened action potentials evoked from strongly negative potentials by ∼8-fold. Optogenetic stimulation of inhibitory cerebellar nucleo-olivary terminals hyperpolarized IO cells sufficiently to abolish the ADP. The data support the idea that currents through KV4 channels control action potential waveforms in IO cells, shortening ADPs during synaptic inhibition or troughs of membrane potential oscillations, thereby controlling the number of climbing fibre action potentials that propagate to the cerebellum. KEY POINTS: Neurons in the mouse inferior olive (IO) express a large, inactivating, voltage-gated A-type K current carried by KV4 channels. IO action potentials evoked from rest have large, long afterdepolarizations that disappear with pre-spike hyperpolarizations of 5-15 mV. The steep voltage-sensitivity and rapid recovery of KV4 channels regulates the duration of the afterdepolarization over more than one order of magnitude. Factors such as synaptic inhibition are sufficient to recruit KV4 channels and eliminate afterdepolarization (ADP). By controlling the ADP, KV4 channels can set the number of climbing fibre action potentials relayed to the cerebellum and regulate plasticity implicated in motor learning.
Collapse
Affiliation(s)
- Ziyad W Sultan
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA
| | - Marion Najac
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Indira M Raman
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA
| |
Collapse
|