51
|
West SL, Gerhart ML, Ebner TJ. Wide-field calcium imaging of cortical activation and functional connectivity in externally- and internally-driven locomotion. Nat Commun 2024; 15:7792. [PMID: 39242572 PMCID: PMC11379880 DOI: 10.1038/s41467-024-51816-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/15/2024] [Indexed: 09/09/2024] Open
Abstract
The role of the cerebral cortex in self-initiated versus sensory-driven movements is central to understanding volitional action. Whether the differences in these two movement classes are due to specific cortical areas versus more cortex-wide engagement is debated. Using wide-field Ca2+ imaging, we compared neural dynamics during spontaneous and motorized treadmill locomotion, determining the similarities and differences in cortex-wide activation and functional connectivity (FC). During motorized locomotion, the cortex exhibits greater activation globally prior to and during locomotion starting compared to spontaneous and less during steady-state walking, during stopping, and after termination. Both conditions are characterized by FC increases in anterior secondary motor cortex (M2) nodes and decreases in all other regions. There are also cortex-wide differences; most notably, M2 decreases in FC with all other nodes during motorized stopping and after termination. Therefore, both internally- and externally-generated movements widely engage the cortex, with differences represented in cortex-wide activation and FC patterns.
Collapse
Affiliation(s)
- Sarah L West
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Morgan L Gerhart
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Timothy J Ebner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
52
|
Chen YY, Chang CJ, Liang YW, Tseng HY, Li SJ, Chang CW, Wu YT, Shao HH, Chen PC, Lai ML, Deng WC, Hsu R, Lo YC. Utilizing diffusion tensor imaging as an image biomarker in exploring the therapeutic efficacy of forniceal deep brain stimulation in a mice model of Alzheimer's disease. J Neural Eng 2024; 21:056003. [PMID: 39230033 DOI: 10.1088/1741-2552/ad7322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/15/2024] [Indexed: 09/05/2024]
Abstract
Objective.With prolonged life expectancy, the incidence of memory deficits, especially in Alzheimer's disease (AD), has increased. Although multiple treatments have been evaluated, no promising treatment has been found to date. Deep brain stimulation (DBS) of the fornix area was explored as a possible treatment because the fornix is intimately connected to memory-related areas that are vulnerable in AD; however, a proper imaging biomarker for assessing the therapeutic efficiency of forniceal DBS in AD has not been established.Approach.This study assessed the efficacy and safety of DBS by estimating the optimal intersection volume between the volume of tissue activated and the fornix. Utilizing a gold-electroplating process, the microelectrode's surface area on the neural probe was increased, enhancing charge transfer performance within potential water window limits. Bilateral fornix implantation was conducted in triple-transgenic AD mice (3 × Tg-AD) and wild-type mice (strain: B6129SF1/J), with forniceal DBS administered exclusively to 3 × Tg-AD mice in the DBS-on group. Behavioral tasks, diffusion tensor imaging (DTI), and immunohistochemistry (IHC) were performed in all mice to assess the therapeutic efficacy of forniceal DBS.Main results.The results illustrated that memory deficits and increased anxiety-like behavior in 3 × Tg-AD mice were rescued by forniceal DBS. Furthermore, forniceal DBS positively altered DTI indices, such as increasing fractional anisotropy (FA) and decreasing mean diffusivity (MD), together with reducing microglial cell and astrocyte counts, suggesting a potential causal relationship between revised FA/MD and reduced cell counts in the anterior cingulate cortex, hippocampus, fornix, amygdala, and entorhinal cortex of 3 × Tg-AD mice following forniceal DBS.Significance.The efficacy of forniceal DBS in AD can be indicated by alterations in DTI-based biomarkers reflecting the decreased activation of glial cells, suggesting reduced neural inflammation as evidenced by improvements in memory and anxiety-like behavior.
Collapse
Affiliation(s)
- You-Yin Chen
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, 12F., Education & Research Building, Shuang-Ho Campus, No. 301, Yuantong Rd., New Taipei City 23564, Taiwan, Republic of China
| | - Chih-Ju Chang
- Department of Neurosurgery, Cathay General Hospital, No. 280, Sec. 4, Renai Rd., Taipei 10629, Taiwan, Republic of China
- School of Medicine, Fu Jen Catholic University, No.510, Zhongzheng Rd., New Taipei City 242062, Taiwan, Republic of China
| | - Yao-Wen Liang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
| | - Hsin-Yi Tseng
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, 12F., Education & Research Building, Shuang-Ho Campus, No. 301, Yuantong Rd., New Taipei City 23564, Taiwan, Republic of China
| | - Ssu-Ju Li
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
| | - Ching-Wen Chang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
| | - Yen-Ting Wu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
| | - Huai-Hsuan Shao
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
| | - Po-Chun Chen
- Department of Materials and Mineral Resources Engineering, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan, Republic of China
| | - Ming-Liang Lai
- Graduate Institute of Intellectual Property, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan, Republic of China
| | - Wen-Chun Deng
- Departments of Neurosurgery, Keelung Chang Gung Memorial Hospital, Chang Gung University, No.222, Maijin Rd., Keelung 20400, Taiwan, Republic of China
| | - RuSiou Hsu
- Department of Ophthalmology, Stanford University, 1651 Page Mill Rd., Palo Alto, CA 94304, United States of America
| | - Yu-Chun Lo
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, 12F., Education & Research Building, Shuang-Ho Campus, No. 301, Yuantong Rd., New Taipei City 23564, Taiwan, Republic of China
| |
Collapse
|
53
|
Flores FJ, Dalla Betta I, Tauber J, Schreier DR, Stephen EP, Wilson MA, Brown EN. Electrographic seizures during low-current thalamic deep brain stimulation in mice. Brain Stimul 2024; 17:975-979. [PMID: 39134207 DOI: 10.1016/j.brs.2024.08.002] [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: 05/11/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Deep brain stimulation of the central thalamus (CT-DBS) has potential for modulating states of consciousness, but it can also trigger electrographic seizures, including poly-spike-wave trains (PSWT). OBJECTIVES To report the probability of inducing PSWTs during CT-DBS in awake, freely-moving mice. METHODS Mice were implanted with electrodes to deliver unilateral and bilateral CT-DBS at different frequencies while recording electroencephalogram (EEG). We titrated stimulation current by gradually increasing it at each frequency until a PSWT appeared. Subsequent stimulations to test arousal modulation were performed at the current one step below the current that caused a PSWT during titration. RESULTS In 2.21% of the test stimulations (10 out of 12 mice), CT-DBS caused PSWTs at currents lower than the titrated current, including currents as low as 20 μA. CONCLUSION Our study found a small but significant probability of inducing PSWTs even after titration and at relatively low currents. EEG should be closely monitored for electrographic seizures when performing CT-DBS in both research and clinical settings.
Collapse
Affiliation(s)
- Francisco J Flores
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Boston, 02114, MA, USA; Center for Brains, Minds, and Machines, Massachusetts Institute of Technology, 43 Vassar St, Cambridge, 02139, MA, USA; Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St, Cambridge, 02139, MA, USA.
| | - Isabella Dalla Betta
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Boston, 02114, MA, USA; Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St, Cambridge, 02139, MA, USA.
| | - John Tauber
- Department of Mathematics and Statistics, Boston University, 665 Commonwealth Ave, Boston, 02215, MA, USA.
| | - David R Schreier
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Boston, 02114, MA, USA; Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St, Cambridge, 02139, MA, USA; Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 101 Merrimac St, Boston, 02114, MA, USA; Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Freiburgstrasse 16, Bern, 3010, Switzerland.
| | - Emily P Stephen
- Department of Mathematics and Statistics, Boston University, 665 Commonwealth Ave, Boston, 02215, MA, USA.
| | - Matthew A Wilson
- Center for Brains, Minds, and Machines, Massachusetts Institute of Technology, 43 Vassar St, Cambridge, 02139, MA, USA; Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St, Cambridge, 02139, MA, USA.
| | - Emery N Brown
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Boston, 02114, MA, USA; Center for Brains, Minds, and Machines, Massachusetts Institute of Technology, 43 Vassar St, Cambridge, 02139, MA, USA; Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar St, Cambridge, 02139, MA, USA; Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, 45 Carleton St, Cambridge, 02142, MA, USA.
| |
Collapse
|
54
|
Furutachi S, Franklin AD, Aldea AM, Mrsic-Flogel TD, Hofer SB. Cooperative thalamocortical circuit mechanism for sensory prediction errors. Nature 2024; 633:398-406. [PMID: 39198646 PMCID: PMC11390482 DOI: 10.1038/s41586-024-07851-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/18/2024] [Indexed: 09/01/2024]
Abstract
The brain functions as a prediction machine, utilizing an internal model of the world to anticipate sensations and the outcomes of our actions. Discrepancies between expected and actual events, referred to as prediction errors, are leveraged to update the internal model and guide our attention towards unexpected events1-10. Despite the importance of prediction-error signals for various neural computations across the brain, surprisingly little is known about the neural circuit mechanisms responsible for their implementation. Here we describe a thalamocortical disinhibitory circuit that is required for generating sensory prediction-error signals in mouse primary visual cortex (V1). We show that violating animals' predictions by an unexpected visual stimulus preferentially boosts responses of the layer 2/3 V1 neurons that are most selective for that stimulus. Prediction errors specifically amplify the unexpected visual input, rather than representing non-specific surprise or difference signals about how the visual input deviates from the animal's predictions. This selective amplification is implemented by a cooperative mechanism requiring thalamic input from the pulvinar and cortical vasoactive-intestinal-peptide-expressing (VIP) inhibitory interneurons. In response to prediction errors, VIP neurons inhibit a specific subpopulation of somatostatin-expressing inhibitory interneurons that gate excitatory pulvinar input to V1, resulting in specific pulvinar-driven response amplification of the most stimulus-selective neurons in V1. Therefore, the brain prioritizes unpredicted sensory information by selectively increasing the salience of unpredicted sensory features through the synergistic interaction of thalamic input and neocortical disinhibitory circuits.
Collapse
Affiliation(s)
- Shohei Furutachi
- Sainsbury Wellcome Centre, University College London, London, UK.
| | | | - Andreea M Aldea
- Sainsbury Wellcome Centre, University College London, London, UK
| | | | - Sonja B Hofer
- Sainsbury Wellcome Centre, University College London, London, UK.
| |
Collapse
|
55
|
Criado-Marrero M, Ravi S, Bhaskar E, Barroso D, Pizzi MA, Williams L, Wellington CL, Febo M, Abisambra JF. Age dictates brain functional connectivity and axonal integrity following repetitive mild traumatic brain injuries in mice. Neuroimage 2024; 298:120764. [PMID: 39089604 DOI: 10.1016/j.neuroimage.2024.120764] [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: 04/30/2024] [Revised: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
Traumatic brain injuries (TBI) present a major public health challenge, demanding an in-depth understanding of age-specific symptoms and risk factors. Aging not only significantly influences brain function and plasticity but also elevates the risk of hospitalizations and death following TBIs. Repetitive mild TBIs (rmTBI) compound these issues, resulting in cumulative and long-term brain damage in the brain. In this study, we investigate the impact of age on brain network changes and white matter properties following rmTBI by employing a multi-modal approach that integrates resting-state functional magnetic resonance imaging (rsfMRI), graph theory analysis, diffusion tensor imaging (DTI), and neurite orientation dispersion and density imaging (NODDI). Our hypothesis is that the effects of rmTBI are worsened in aged animals, with this group showing more pronounced alterations in brain connectivity and white matter structure. Utilizing the closed-head impact model of engineered rotational acceleration (CHIMERA) model, we conducted rmTBIs or sham (control) procedures on young (2.5-3-months-old) and aged (22-months-old) male and female mice to model high-risk groups. Functional and structural imaging unveiled age-related reductions in communication efficiency between brain regions, while injuries induced opposhigh-risking effects on the small-world index across age groups, influencing network segregation. Functional connectivity analysis also identified alterations in 79 out of 148 brain regions by age, treatment (sham vs. rmTBI), or their interaction. Injuries exerted pronounced effects on sensory integration areas, including insular and motor cortices. Age-related disruptions in white matter integrity were observed, indicating alterations in various diffusion directions (mean diffusivity, radial diffusivity, axial diffusivity, and fractional anisotropy) and density neurite properties (dispersion index, intracellular and isotropic volume fraction). Neuroinflammation, assessed through Iba-1 and GFAP markers, correlated with higher dispersion in the optic tract, suggesting a neuroinflammatory response in injured aged animals compared to sham aged. These findings offer insight into the interplay between age, injuries, and brain connectivity, shedding light on the long-term consequences of rmTBI.
Collapse
Affiliation(s)
- Marangelie Criado-Marrero
- Center for Translational Research in Neurodegenerative Disease (CTRND), University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Sakthivel Ravi
- Center for Translational Research in Neurodegenerative Disease (CTRND), University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Ekta Bhaskar
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; Department of Computer of Information Science and Engineering (CISE), University of Florida, Gainesville, FL 32610, USA
| | - Daylin Barroso
- Center for Translational Research in Neurodegenerative Disease (CTRND), University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Michael A Pizzi
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA; Brain Injury Rehabilitation and Neuroresilience (BRAIN) Center University of Florida, Gainesville, FL 32610, USA; Department of Neurology, University of Florida, Gainesville, FL 32610, USA
| | - Lakiesha Williams
- J. Crayton Pruitt Family Department of Biomedical Engineering, Gainesville, FL 32610, USA
| | - Cheryl L Wellington
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Marcelo Febo
- McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA; Department of Psychiatry, University of Florida, Gainesville, FL 32610, USA; Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Jose Francisco Abisambra
- Center for Translational Research in Neurodegenerative Disease (CTRND), University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA; Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA; Brain Injury Rehabilitation and Neuroresilience (BRAIN) Center University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
56
|
Fleischer M, Szepanowski RD, Pesara V, Bihorac JS, Oehler B, Dobrev D, Kleinschnitz C, Fender AC. Direct neuronal protection by the protease-activated receptor PAR4 antagonist ML354 after experimental stroke in mice. Br J Pharmacol 2024; 181:3364-3379. [PMID: 38760890 DOI: 10.1111/bph.16415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/03/2024] [Accepted: 03/22/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND AND PURPOSE Thrombo-inflammation is a key feature of stroke pathophysiology and provides multiple candidate drug targets. Thrombin exerts coagulation-independent actions via protease-activated receptors (PAR), of which PAR1 has been implicated in stroke-associated neuroinflammation. The role of PAR4 in this context is less clear. This study examined if the selective PAR4 antagonist ML354 provides neuroprotection in experimental stroke and explored the underlying mechanisms. EXPERIMENTAL APPROACH Mouse primary cortical neurons were exposed to oxygen-glucose deprivation (OGD) and simulated reperfusion ± ML354. For comparison, functional Ca2+-imaging was performed upon acute stimulation with a PAR4 activating peptide or glutamate. Male mice underwent sham operation or transient middle cerebral artery occlusion (tMCAO), with ML354 or vehicle treatment beginning at recanalization. A subset of mice received a platelet-depleting antibody. Stroke size and functional outcomes were assessed. Abundance of target genes, proteins, and cell markers was determined in cultured cells and tissues by qPCR, immunoblotting, and immunofluorescence. KEY RESULTS Stroke up-regulated PAR4 expression in cortical neurons in vitro and in vivo. OGD augments spontaneous and PAR4-mediated neuronal activity; ML354 suppresses OGD-induced neuronal excitotoxicity and apoptosis. ML354 applied in vivo after tMCAO reduced infarct size, apoptotic markers, macrophage accumulation, and interleukin-1β expression. Platelet depletion did not affect infarct size in mice with tMCAO ± ML354. CONCLUSIONS AND IMPLICATIONS Selective PAR4 inhibition during reperfusion improves infarct size and neurological function after experimental stroke by blunting neuronal excitability, apoptosis, and local inflammation. PAR4 antagonists may provide additional neuroprotective benefits in patients with acute stroke beyond their canonical antiplatelet action.
Collapse
Affiliation(s)
- Michael Fleischer
- Department of Neurology, Center for Translational Neuro- and Behavioral Science (C-TNBS), University Hospital Essen, Essen, Germany
| | - Rebecca D Szepanowski
- Department of Neurology, Center for Translational Neuro- and Behavioral Science (C-TNBS), University Hospital Essen, Essen, Germany
| | - Valeria Pesara
- Department of Neurology, Center for Translational Neuro- and Behavioral Science (C-TNBS), University Hospital Essen, Essen, Germany
| | - Julia Sophie Bihorac
- Department of Neurology, Center for Translational Neuro- and Behavioral Science (C-TNBS), University Hospital Essen, Essen, Germany
| | - Beatrice Oehler
- Department of Anaesthesiology, University of Heidelberg, Heidelberg, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, University Hospital Essen, Essen, Germany
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine and Research Center, Montréal Heart Institute and Université de Montréal, Montréal, Canada
| | - Christoph Kleinschnitz
- Department of Neurology, Center for Translational Neuro- and Behavioral Science (C-TNBS), University Hospital Essen, Essen, Germany
| | - Anke C Fender
- Institute of Pharmacology, University Hospital Essen, Essen, Germany
| |
Collapse
|
57
|
Nitzan N, Buzsáki G. Physiological characteristics of neurons in the mammillary bodies align with topographical organization of subicular inputs. Cell Rep 2024; 43:114539. [PMID: 39052483 PMCID: PMC11475818 DOI: 10.1016/j.celrep.2024.114539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/20/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
The mammillary bodies (MBOs), a group of hypothalamic nuclei, play a pivotal role in memory formation and spatial navigation. They receive extensive inputs from the hippocampus through the fornix, but the physiological significance of these connections remains poorly understood. Damage to the MBOs is associated with various forms of anterograde amnesia. However, information about the physiological characteristics of the MBO is limited, primarily due to the limited number of studies that have directly monitored MBO activity along with population patterns of its upstream partners. Employing large-scale silicon probe recording in mice, we characterize MBO activity and its interaction with the subiculum across various brain states. We find that MBO cells are highly diverse in their relationship to theta, ripple, and slow oscillations. Several of the physiological features are inherited by the topographically organized inputs to MBO cells. Our study provides insights into the functional organization of the MBOs.
Collapse
Affiliation(s)
- Noam Nitzan
- New York University Neuroscience Institute, New York University, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10016, USA
| | - György Buzsáki
- New York University Neuroscience Institute, New York University, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10016, USA.
| |
Collapse
|
58
|
Brooks ER, Moorman AR, Bhattacharya B, Prudhomme I, Land M, Alcorn HL, Sharma R, Pe’er D, Zallen JA. A single-cell atlas of spatial and temporal gene expression in the mouse cranial neural plate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.25.609458. [PMID: 39229123 PMCID: PMC11370589 DOI: 10.1101/2024.08.25.609458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The formation of the mammalian brain requires regionalization and morphogenesis of the cranial neural plate, which transforms from an epithelial sheet into a closed tube that provides the structural foundation for neural patterning and circuit formation. Sonic hedgehog (SHH) signaling is important for cranial neural plate patterning and closure, but the transcriptional changes that give rise to the spatially regulated cell fates and behaviors that build the cranial neural tube have not been systematically analyzed. Here we used single-cell RNA sequencing to generate an atlas of gene expression at six consecutive stages of cranial neural tube closure in the mouse embryo. Ordering transcriptional profiles relative to the major axes of gene expression predicted spatially regulated expression of 870 genes along the anterior-posterior and mediolateral axes of the cranial neural plate and reproduced known expression patterns with over 85% accuracy. Single-cell RNA sequencing of embryos with activated SHH signaling revealed distinct SHH-regulated transcriptional programs in the developing forebrain, midbrain, and hindbrain, suggesting a complex interplay between anterior-posterior and mediolateral patterning systems. These results define a spatiotemporally resolved map of gene expression during cranial neural tube closure and provide a resource for investigating the transcriptional events that drive early mammalian brain development.
Collapse
Affiliation(s)
- Eric R. Brooks
- HHMI and Developmental Biology Program, Sloan Kettering Institute
- North Carolina State University
| | - Andrew R. Moorman
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | | | - Ian Prudhomme
- HHMI and Developmental Biology Program, Sloan Kettering Institute
| | - Max Land
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | | | - Roshan Sharma
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | - Dana Pe’er
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | | |
Collapse
|
59
|
Chan AE, Anderson JQ, Grigsby KB, Jensen BE, Ryabinin AE, Ozburn AR. Sex differences in nucleus accumbens core circuitry engaged by binge-like ethanol drinking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608144. [PMID: 39229134 PMCID: PMC11370393 DOI: 10.1101/2024.08.15.608144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Growing parity in Alcohol Use Disorder (AUD) diagnoses in men and women necessitates consideration of sex as a biological variable. In humans and rodents, the nucleus accumbens core (NAcc) regulates alcohol binge drinking, a risk factor for developing AUD. We labeled NAcc inputs with a viral retrograde tracer and quantified whole-brain c-Fos to determine the regions and NAcc inputs differentially engaged in male and female mice during binge-like ethanol drinking. We found that binge-like ethanol drinking females had 129 brain areas with greater c-Fos than males. Moreover, ethanol engaged more NAcc inputs in binge-like ethanol drinking females (as compared with males), including GABAergic and glutamatergic inputs. Relative to water controls, ethanol increased network modularity and decreased connectivity in both sexes and did so more dramatically in males. These results demonstrate that early-stage binge-like ethanol drinking engages brain regions and NAcc-inputs and alters network dynamics in a sex-specific manner.
Collapse
Affiliation(s)
- Amy E Chan
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Justin Q Anderson
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Kolter B Grigsby
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Bryan E Jensen
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Andrey E Ryabinin
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
| | - Angela R Ozburn
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| |
Collapse
|
60
|
Fay MG, Lang PJ, Denu DS, O’Connor NJ, Haydock B, Blaisdell J, Roussel N, Wilson A, Aronson SM, Angstman PJ, Gong C, Butola T, Devinsky O, Basu J, Tomer R, Glaser JR. ClearScope: a fully integrated light sheet theta microscope for sub-cellular resolution imaging without lateral size constraints. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608141. [PMID: 39229056 PMCID: PMC11370359 DOI: 10.1101/2024.08.15.608141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Three-dimensional (3D) ex vivo imaging of cleared intact brains of animal models and large human and non-human primate postmortem brain specimens is important for understanding the physiological neural network connectivity patterns and the pathological alterations underlying neuropsychiatric and neurological disorders. Light-sheet microscopy has emerged as a highly effective imaging modality for rapid high-resolution imaging of large cleared samples. However, the orthogonal arrangements of illumination and detection optics in light sheet microscopy limits the size of specimen that can be imaged. Recently developed light sheet theta microscopy (LSTM) technology addressed this by utilizing a unique arrangement of two illumination light paths oblique to the detection light path, while allowing perpendicular arrangement of the detection light path relative to the specimen surface. Here, we report development of a next-generation, fully integrated, and user-friendly LSTM system for rapid sub-cellular resolution imaging uniformly throughout a large specimen without constraining the lateral (XY) size. In addition, we provide a seamlessly integrated workflow for image acquisition, data storage, pre- and post-processing, enhancement, and quantitative analysis. We demonstrate the system performance by high-resolution 3D imaging of intact mouse brains and human brain samples, and complete data analysis including digital neuron tracing, vessel reconstruction and design-based stereological analysis in 3D. This technically enhanced and user-friendly LSTM implementation will enable rapid quantitative mapping of molecular and cellular features of interests in diverse types of very large samples.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Cheng Gong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Tanvi Butola
- Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine; New York City, 10016, USA
- Comprehensive Epilepsy Center, Department of Neurology, New York University Grossman School of Medicine; New York City, 10016, USA
| | - Orrin Devinsky
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine; New York City, 10016, USA
- Department of Psychiatry, New York University Grossman School of Medicine; New York City, 10016, USA
- Comprehensive Epilepsy Center, Department of Neurology, New York University Grossman School of Medicine; New York City, 10016, USA
- Department of Neurosurgery, New York University Grossman School of Medicine; New York City, 10016, USA
| | - Jayeeta Basu
- Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine; New York City, 10016, USA
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Raju Tomer
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | |
Collapse
|
61
|
Wu Z, Kondo A, McGrady M, Baker EAG, Chidester B, Wu E, Rahim MK, Bracey NA, Charu V, Cho RJ, Cheng JB, Afkarian M, Zou J, Mayer AT, Trevino AE. Discovery and generalization of tissue structures from spatial omics data. CELL REPORTS METHODS 2024; 4:100838. [PMID: 39127044 PMCID: PMC11384092 DOI: 10.1016/j.crmeth.2024.100838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/15/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
Tissues are organized into anatomical and functional units at different scales. New technologies for high-dimensional molecular profiling in situ have enabled the characterization of structure-function relationships in increasing molecular detail. However, it remains a challenge to consistently identify key functional units across experiments, tissues, and disease contexts, a task that demands extensive manual annotation. Here, we present spatial cellular graph partitioning (SCGP), a flexible method for the unsupervised annotation of tissue structures. We further present a reference-query extension pipeline, SCGP-Extension, that generalizes reference tissue structure labels to previously unseen samples, performing data integration and tissue structure discovery. Our experiments demonstrate reliable, robust partitioning of spatial data in a wide variety of contexts and best-in-class accuracy in identifying expertly annotated structures. Downstream analysis on SCGP-identified tissue structures reveals disease-relevant insights regarding diabetic kidney disease, skin disorder, and neoplastic diseases, underscoring its potential to drive biological insight and discovery from spatial datasets.
Collapse
Affiliation(s)
- Zhenqin Wu
- Enable Medicine, Menlo Park, CA 94025, USA.
| | | | | | | | | | - Eric Wu
- Enable Medicine, Menlo Park, CA 94025, USA; Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | | | - Nathan A Bracey
- Institute of Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA
| | - Vivek Charu
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Raymond J Cho
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey B Cheng
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA; Department of Dermatology, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Maryam Afkarian
- Division of Nephrology, Department of Medicine, University of California, Davis, Davis, CA 95618, USA
| | - James Zou
- Enable Medicine, Menlo Park, CA 94025, USA; Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
62
|
Catto F, Dadgar-Kiani E, Kirschenbaum D, Economides A, Reuss AM, Trevisan C, Caredio D, Mirzet D, Frick L, Weber-Stadlbauer U, Litvinov S, Koumoutsakos P, Hyung Lee J, Aguzzi A. Quantitative 3D histochemistry reveals region-specific amyloid-β reduction by the antidiabetic drug netoglitazone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608042. [PMID: 39185170 PMCID: PMC11343181 DOI: 10.1101/2024.08.15.608042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
A hallmark of Alzheimer's disease (AD) is the extracellular aggregation of toxic amyloid-beta (Aβ) peptides in form of plaques. Here, we identify netoglitazone, an antidiabetic compound previously tested in humans, as an Aβ aggregation antagonist. Netoglitazone improved cognition and reduced microglia activity in a mouse model of AD. Using quantitative whole-brain three-dimensional histology (Q3D), we precisely identified brain regions where netoglitazone reduced the number and size of Aβ plaques. We demonstrate the utility of Q3D in preclinical drug evaluation for AD by providing a high-resolution brain-wide view of drug efficacy. Applying Q3D has the potential to improve pre-clinical drug evaluation by providing information that can help identify mechanisms leading to brain region-specific drug efficacy.
Collapse
Affiliation(s)
- Francesca Catto
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
- IMAI MedTech, Wagistrasse 18, 8952 Schlieren, Zurich, Switzerland
| | - Ehsan Dadgar-Kiani
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 260, 8057 Zürich
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| | - Daniel Kirschenbaum
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Athena Economides
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Anna Maria Reuss
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Chiara Trevisan
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Davide Caredio
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Delic Mirzet
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Lukas Frick
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 260, 8057 Zürich
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| | - Sergey Litvinov
- Computational Science and Engineering Laboratory, ETH Zürich, Clausiusstrasse 33, 8092, Zurich, Switzerland
- Computational Science and Engineering Laboratory, Harvard University, Cambridge, MA 02138, United States
| | - Petros Koumoutsakos
- Computational Science and Engineering Laboratory, Harvard University, Cambridge, MA 02138, United States
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Electrical Engineering, Stanford University, CA 94305, USA
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| |
Collapse
|
63
|
Zhao Y, Zhang J, Yu H, Hou X, Zhang J. Noninvasive microvascular imaging in newborn rats using high-frequency ultrafast ultrasound. Neuroimage 2024; 297:120738. [PMID: 39009248 DOI: 10.1016/j.neuroimage.2024.120738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024] Open
Abstract
Ultrasound imaging stands as the predominant modality for neonatal health assessment, with recent advancements in ultrafast Doppler (μDoppler) technology offering significant promise in fields such as neonatal brain imaging. Combining μDoppler with high-frequency ultrasound (HF-μDoppler) presents a potential efficient avenue to enhance in vivo microvascular imaging in small animals, notably newborn rats, a crucial preclinical animal model for neonatal disease and development research. It is necessary to verify the imaging performance of HF-μDoppler in preclinical trials. This study investigates the microvascular imaging capabilities of HF-μDoppler using a 30 MHz high-frequency linear array probe in newborn rats. Results demonstrate the clarity of cerebral microvascular imaging in rats aged 1 to 7 postnatal days, extending to whole-body microvascular imaging, encompassing the central nervous system, including the brain and spinal cord. In conclusion, HF-μDoppler technology emerges as a reliable imaging tool, offering a new perspective for preclinical investigations into neonatal diseases and development.
Collapse
Affiliation(s)
- Yunlong Zhao
- Department of Pediatrics, Peking University First Hospital, Beijing, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Jiabin Zhang
- College of Future Technology, Peking University, Beijing, China.
| | - Hao Yu
- College of Engineering, Peking University, Beijing, China
| | - Xinlin Hou
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jue Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; College of Engineering, Peking University, Beijing, China
| |
Collapse
|
64
|
Han X, Maharjan S, Chen J, Zhao Y, Qi Y, White LE, Johnson GA, Wang N. High-resolution diffusion magnetic resonance imaging and spatial-transcriptomic in developing mouse brain. Neuroimage 2024; 297:120734. [PMID: 39032791 PMCID: PMC11377129 DOI: 10.1016/j.neuroimage.2024.120734] [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: 01/04/2024] [Revised: 07/06/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024] Open
Abstract
Brain development is a highly complex process regulated by numerous genes at the molecular and cellular levels. Brain tissue exhibits serial microstructural changes during the development process. High-resolution diffusion magnetic resonance imaging (dMRI) affords a unique opportunity to probe these changes in the developing brain non-destructively. In this study, we acquired multi-shell dMRI datasets at 32 µm isotropic resolution to investigate the tissue microstructure alterations, which we believe to be the highest spatial resolution dMRI datasets obtained for postnatal mouse brains. We adapted the Allen Developing Mouse Brain Atlas (ADMBA) to integrate quantitative MRI metrics and spatial transcriptomics. Diffusion tensor imaging (DTI), diffusion kurtosis imaging (DKI), and neurite orientation dispersion and density imaging (NODDI) metrics were used to quantify brain development at different postnatal days. We demonstrated that the differential evolutions of fiber orientation distributions contribute to the distinct development patterns in white matter (WM) and gray matter (GM). Furthermore, the genes enriched in the nervous system that regulate brain structure and function were expressed in spatial correlation with age-matched dMRI. This study is the first one providing high-resolution dMRI, including DTI, DKI, and NODDI models, to trace mouse brain microstructural changes in WM and GM during postnatal development. This study also highlighted the genotype-phenotype correlation of spatial transcriptomics and dMRI, which may improve our understanding of brain microstructure changes at the molecular level.
Collapse
Affiliation(s)
- Xinyue Han
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA; Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Surendra Maharjan
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA
| | - Jie Chen
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA
| | - Yi Zhao
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN, USA
| | - Yi Qi
- Center for In Vivo Microscopy, Department of Radiology, Duke University, Durham, NC, USA
| | - Leonard E White
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - G Allan Johnson
- Center for In Vivo Microscopy, Department of Radiology, Duke University, Durham, NC, USA; Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nian Wang
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA; Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA; Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
65
|
Curic D, Ashby DM, McGirr A, Davidsen J. Existence of multiple transitions of the critical state due to anesthetics. Nat Commun 2024; 15:7025. [PMID: 39147749 PMCID: PMC11327335 DOI: 10.1038/s41467-024-51399-2] [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: 02/03/2023] [Accepted: 08/05/2024] [Indexed: 08/17/2024] Open
Abstract
Scale-free statistics of coordinated neuronal activity, suggesting a universal operating mechanism across spatio-temporal scales, have been proposed as a necessary condition of healthy resting-state brain activity. Recent studies have focused on anesthetic agents to induce distinct neural states in which consciousness is altered to understand the importance of critical dynamics. However, variation in experimental techniques, species, and anesthetics, have made comparisons across studies difficult. Here we conduct a survey of several common anesthetics (isoflurane, pentobarbital, ketamine) at multiple dosages, using calcium wide-field optical imaging of the mouse cortex. We show that while low-dose anesthesia largely preserves scale-free statistics, surgical plane anesthesia induces multiple dynamical modes, most of which do not maintain critical avalanche dynamics. Our findings indicate multiple pathways away from default critical dynamics associated with quiet wakefulness, not only reflecting differences between these common anesthetics but also showing significant variations in individual responses. This is suggestive of a non-trivial relationship between criticality and the underlying state of the subject.
Collapse
Affiliation(s)
- Davor Curic
- Complexity Science Group, Department of Physics and Astronomy, University of Calgary, Calgary, Alberta, T2N 1N4, Canada.
| | - Donovan M Ashby
- Department of Psychiatry, University of Calgary, Calgary, AB, T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Mathison Centre for Mental Health Research and Education, Calgary, AB, Canada
| | - Alexander McGirr
- Department of Psychiatry, University of Calgary, Calgary, AB, T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Mathison Centre for Mental Health Research and Education, Calgary, AB, Canada
| | - Jörn Davidsen
- Complexity Science Group, Department of Physics and Astronomy, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
66
|
Papale AE, Harish M, Paletzki RF, O'Connor NJ, Eastwood BS, Seal RP, Williamson RS, Gerfen CR, Hooks BM. Symmetry in Frontal But Not Motor and Somatosensory Cortical Projections. J Neurosci 2024; 44:e1195232024. [PMID: 38937102 PMCID: PMC11326871 DOI: 10.1523/jneurosci.1195-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 04/29/2024] [Accepted: 05/05/2024] [Indexed: 06/29/2024] Open
Abstract
The neocortex and striatum are topographically organized for sensory and motor functions. While sensory and motor areas are lateralized for touch and motor control, respectively, frontal areas are involved in decision-making, where lateralization of function may be less important. This study contrasted the topographic precision of cell-type-specific ipsilateral and contralateral cortical projections while varying the injection site location in transgenic mice of both sexes. While sensory cortical areas had strongly topographic outputs to the ipsilateral cortex and striatum, they were weaker and not as topographically precise to contralateral targets. The motor cortex had somewhat stronger projections but still relatively weak contralateral topography. In contrast, frontal cortical areas had high degrees of topographic similarity for both ipsilateral and contralateral projections to the cortex and striatum. Corticothalamic organization is mainly ipsilateral, with weaker, more medial contralateral projections. Corticostriatal computations might integrate input outside closed basal ganglia loops using contralateral projections, enabling the two hemispheres to act as a unit to converge on one result in motor planning and decision-making.
Collapse
Affiliation(s)
- Andrew E Papale
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Madhumita Harish
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Ronald F Paletzki
- Laboratory of Systems Neuroscience, National Institute of Mental Health, Bethesda, Maryland 20892
| | | | | | - Rebecca P Seal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Ross S Williamson
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Charles R Gerfen
- Laboratory of Systems Neuroscience, National Institute of Mental Health, Bethesda, Maryland 20892
| | - Bryan M Hooks
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
67
|
Forte N, Marfella B, Nicois A, Palomba L, Paris D, Motta A, Pina Mollica M, Di Marzo V, Cristino L. The short-chain fatty acid acetate modulates orexin/hypocretin neurons: A novel mechanism in gut-brain axis regulation of energy homeostasis and feeding. Biochem Pharmacol 2024; 226:116383. [PMID: 38908530 DOI: 10.1016/j.bcp.2024.116383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/24/2024]
Abstract
The short-chain fatty acids (SCFAs) acetate, propionate and butyrate, the major products of intestinal microbial fermentation of dietary fibres, are involved in fine-tuning brain functions via the gut-brain axis. However, the effects of SCFAs in the hypothalamic neuronal network regulating several autonomic-brain functions are still unknown. Using NMR spectroscopy, we detected a reduction in brain acetate concentrations in the hypothalamus of obese leptin knockout ob/ob mice compared to lean wild-type littermates. Therefore, we investigated the effect of acetate on orexin/hypocretin neurons (hereafter referred as OX or OX-A neurons), a subset of hypothalamic neurons regulating energy homeostasis, which we have characterized in previous studies to be over-activated by the lack of leptin and enhancement of endocannabinoid tone in the hypothalamus of ob/ob mice. We found that acetate reduces food-intake in concomitance with a reduction of orexin neuronal activity in ob/ob mice. This was demonstrated by evaluating food-intake behaviour and orexin-A/c-FOS immunoreactivity coupled with patch-clamp recordings in Hcrt-eGFP neurons, quantification of prepro-orexin mRNA, and immunolabeling of GPR-43, the main acetate receptor. Our data provide new insights into the mechanisms of the effects of chronic dietary supplementation with acetate, or complex carbohydrates, on energy intake and body weight, which may be partly mediated by inhibition of orexinergic neuron activity.
Collapse
Affiliation(s)
- Nicola Forte
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Brenda Marfella
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy; Department of Biology, University of Naples Federico II, Naples, Italy
| | - Alessandro Nicois
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Letizia Palomba
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Debora Paris
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, Naples, Italy; Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126 Naples, Italy; Task Force on Microbiome Studies, University of Naples Federico II, 80138 Naples, Italy
| | - Vincenzo Di Marzo
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Faculty of Medicine and Faculty of Agricultural and Food Sciences, Université Laval, Québec City, QC, Canada.
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy.
| |
Collapse
|
68
|
Bracha S, Johnson HJ, Pranckevicius NA, Catto F, Economides AE, Litvinov S, Hassi K, Rigoli MT, Cheroni C, Bonfanti M, Valenti A, Stucchi S, Attreya S, Ross PD, Walsh D, Malachi N, Livne H, Eshel R, Krupalnik V, Levin D, Cobb S, Koumoutsakos P, Caporale N, Testa G, Aguzzi A, Koshy AA, Sheiner L, Rechavi O. Engineering Toxoplasma gondii secretion systems for intracellular delivery of multiple large therapeutic proteins to neurons. Nat Microbiol 2024; 9:2051-2072. [PMID: 39075233 PMCID: PMC11306108 DOI: 10.1038/s41564-024-01750-6] [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: 12/02/2023] [Accepted: 06/05/2024] [Indexed: 07/31/2024]
Abstract
Delivering macromolecules across biological barriers such as the blood-brain barrier limits their application in vivo. Previous work has demonstrated that Toxoplasma gondii, a parasite that naturally travels from the human gut to the central nervous system (CNS), can deliver proteins to host cells. Here we engineered T. gondii's endogenous secretion systems, the rhoptries and dense granules, to deliver multiple large (>100 kDa) therapeutic proteins into neurons via translational fusions to toxofilin and GRA16. We demonstrate delivery in cultured cells, brain organoids and in vivo, and probe protein activity using imaging, pull-down assays, scRNA-seq and fluorescent reporters. We demonstrate robust delivery after intraperitoneal administration in mice and characterize 3D distribution throughout the brain. As proof of concept, we demonstrate GRA16-mediated brain delivery of the MeCP2 protein, a putative therapeutic target for Rett syndrome. By characterizing the potential and current limitations of the system, we aim to guide future improvements that will be required for broader application.
Collapse
Affiliation(s)
- Shahar Bracha
- Department of Neurobiology, Biochemistry and Biophysics, Wise Faculty of Life Sciences and Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel.
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA.
| | - Hannah J Johnson
- Neuroscience Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA
- Departments of Neurology and Immunobiology, College of Medicine, and BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Nicole A Pranckevicius
- Centre for Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Francesca Catto
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Athena E Economides
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sergey Litvinov
- Computational Science and Engineering Laboratory, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Karoliina Hassi
- Centre for Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Marco Tullio Rigoli
- Human Technopole, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Cristina Cheroni
- Human Technopole, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | | | - Alessia Valenti
- Human Technopole, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Sarah Stucchi
- Human Technopole, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Shruti Attreya
- Undergraduate Biology Research Program, University of Arizona, Tucson, AZ, USA
| | - Paul D Ross
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Daniel Walsh
- Centre for Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | | | | | | | - Stuart Cobb
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Petros Koumoutsakos
- Computational Science and Engineering Laboratory, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Nicolò Caporale
- Human Technopole, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Giuseppe Testa
- Human Technopole, Milan, Italy.
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Anita A Koshy
- Departments of Neurology and Immunobiology, College of Medicine, and BIO5 Institute, University of Arizona, Tucson, AZ, USA.
| | - Lilach Sheiner
- Centre for Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| | - Oded Rechavi
- Department of Neurobiology, Biochemistry and Biophysics, Wise Faculty of Life Sciences and Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
69
|
Araki S, Onishi I, Ikoma Y, Matsui K. Astrocyte switch to the hyperactive mode. Glia 2024; 72:1418-1434. [PMID: 38591259 DOI: 10.1002/glia.24537] [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: 01/23/2024] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 04/10/2024]
Abstract
Increasing pieces of evidence have suggested that astrocyte function has a strong influence on neuronal activity and plasticity, both in physiological and pathophysiological situations. In epilepsy, astrocytes have been shown to respond to epileptic neuronal seizures; however, whether they can act as a trigger for seizures has not been determined. Here, using the copper implantation method, spontaneous neuronal hyperactivity episodes were reliably induced during the week following implantation. With near 24-h continuous recording for over 1 week of the local field potential with in vivo electrophysiology and astrocyte cytosolic Ca2+ with the fiber photometry method, spontaneous occurrences of seizure episodes were captured. Approximately 1 day after the implantation, isolated aberrant astrocyte Ca2+ events were often observed before they were accompanied by neuronal hyperactivity, suggesting the role of astrocytes in epileptogenesis. Within a single developed episode, astrocyte Ca2+ increase preceded the neuronal hyperactivity by ~20 s, suggesting that actions originating from astrocytes could be the trigger for the occurrence of epileptic seizures. Astrocyte-specific stimulation by channelrhodopsin-2 or deep-brain direct current stimulation was capable of inducing neuronal hyperactivity. Injection of an astrocyte-specific metabolic inhibitor, fluorocitrate, was able to significantly reduce the magnitude of spontaneously occurring neuronal hyperactivity. These results suggest that astrocytes have a role in triggering individual seizures and the reciprocal astrocyte-neuron interactions likely amplify and exacerbate seizures. Therefore, future epilepsy treatment could be targeted at astrocytes to achieve epilepsy control.
Collapse
Affiliation(s)
- Shun Araki
- Super-network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Ichinosuke Onishi
- Super-network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yoko Ikoma
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
70
|
Marrero K, Aruljothi K, Delgadillo C, Kabbara S, Swatch L, Zagha E. Goal-directed learning is multidimensional and accompanied by diverse and widespread changes in neocortical signaling. Cereb Cortex 2024; 34:bhae328. [PMID: 39110412 PMCID: PMC11304966 DOI: 10.1093/cercor/bhae328] [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: 05/08/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
New tasks are often learned in stages with each stage reflecting a different learning challenge. Accordingly, each learning stage is likely mediated by distinct neuronal processes. And yet, most rodent studies of the neuronal correlates of goal-directed learning focus on individual outcome measures and individual brain regions. Here, we longitudinally studied mice from naïve to expert performance in a head-fixed, operant conditioning whisker discrimination task. In addition to tracking the primary behavioral outcome of stimulus discrimination, we tracked and compared an array of object-based and temporal-based behavioral measures. These behavioral analyses identify multiple, partially overlapping learning stages in this task, consistent with initial response implementation, early stimulus-response generalization, and late response inhibition. To begin to understand the neuronal foundations of these learning processes, we performed widefield Ca2+ imaging of dorsal neocortex throughout learning and correlated behavioral measures with neuronal activity. We found distinct and widespread correlations between neocortical activation patterns and various behavioral measures. For example, improvements in sensory discrimination correlated with target stimulus evoked activations of response-related cortices along with distractor stimulus evoked global cortical suppression. Our study reveals multidimensional learning for a simple goal-directed learning task and generates hypotheses for the neuronal modulations underlying these various learning processes.
Collapse
Affiliation(s)
- Krista Marrero
- Neuroscience Graduate Program, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Krithiga Aruljothi
- Department of Psychology, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Christian Delgadillo
- Division of Biomedical Sciences, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Sarah Kabbara
- Neuroscience Graduate Program, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Lovleen Swatch
- College of Natural & Agricultural Sciences, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Edward Zagha
- Neuroscience Graduate Program, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
- Department of Psychology, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| |
Collapse
|
71
|
Jiang S, Song B, Liu Z, Shen S, Qian W, Sun J, Chen G, Zhu Y. Neuronal activity in the anterior paraventricular nucleus of thalamus positively correlated with sweetener consumption in mice. Neurosci Res 2024; 205:16-26. [PMID: 38364907 DOI: 10.1016/j.neures.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/02/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024]
Abstract
Although the brain can discriminate between various sweet substances, the underlying neural mechanisms of this complex behavior remain elusive. This study examines the role of the anterior paraventricular nucleus of the thalamus (aPVT) in governing sweet preference in mice. We fed the mice six different diets with equal sweetness for six weeks: control diet (CD), high sucrose diet (HSD), high stevioside diet (HSSD), high xylitol diet (HXD), high glycyrrhizin diet (HGD), and high mogroside diet (HMD). The mice exhibited a marked preference specifically for the HSD and HSSD. Following consumption of these diets, c-Fos expression levels in the aPVT were significantly higher in these two groups compared to the others. Utilizing fiber photometry calcium imaging, we observed rapid activation of aPVT neurons in response to sucrose and stevioside intake, but not to xylitol or water. Our findings suggest that aPVT activity aligns with sweet preference in mice, and notably, stevioside is the sole plant-based sweetener that elicits an aPVT response comparable to that of sucrose.
Collapse
Affiliation(s)
- Shaolei Jiang
- Key Laboratory of Optical Technology and Instrument for Medicine, Ministry of Education, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; The Brain Cognition and Brain Diseases Institute of Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen 518055, China
| | - Bo Song
- Key Laboratory of Optical Technology and Instrument for Medicine, Ministry of Education, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Zhongdong Liu
- Grain College, Henan University of Technology, Zhengzhou 450001, China; Instituto de Física da Universidade de São Paulo, Sã o Paulo 05508-090, Brazil
| | - Shuifa Shen
- Hefei lnstitutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; School of Intelligent Manufacturing, Zhejiang Guangsha Vocational and Technical University of Construction, Jinhua 322100, China
| | - Weiliang Qian
- Instituto de Física da Universidade de São Paulo, Sã o Paulo 05508-090, Brazil
| | - Jing Sun
- Department of Anesthesiology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, No.2004 Hongli Road, Shenzhen 518028, China
| | - Gaowei Chen
- The Brain Cognition and Brain Diseases Institute of Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen 518055, China.
| | - Yingjie Zhu
- The Brain Cognition and Brain Diseases Institute of Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen 518055, China.
| |
Collapse
|
72
|
Rudelt L, González Marx D, Spitzner FP, Cramer B, Zierenberg J, Priesemann V. Signatures of hierarchical temporal processing in the mouse visual system. PLoS Comput Biol 2024; 20:e1012355. [PMID: 39173067 PMCID: PMC11373856 DOI: 10.1371/journal.pcbi.1012355] [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: 01/17/2024] [Revised: 09/04/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
A core challenge for the brain is to process information across various timescales. This could be achieved by a hierarchical organization of temporal processing through intrinsic mechanisms (e.g., recurrent coupling or adaptation), but recent evidence from spike recordings of the rodent visual system seems to conflict with this hypothesis. Here, we used an optimized information-theoretic and classical autocorrelation analysis to show that information- and correlation timescales of spiking activity increase along the anatomical hierarchy of the mouse visual system under visual stimulation, while information-theoretic predictability decreases. Moreover, intrinsic timescales for spontaneous activity displayed a similar hierarchy, whereas the hierarchy of predictability was stimulus-dependent. We could reproduce these observations in a basic recurrent network model with correlated sensory input. Our findings suggest that the rodent visual system employs intrinsic mechanisms to achieve longer integration for higher cortical areas, while simultaneously reducing predictability for an efficient neural code.
Collapse
Affiliation(s)
- Lucas Rudelt
- Max-Planck-Institute for Dynamics and Self-Organization, Göttingen, Germany
- Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
| | - Daniel González Marx
- Max-Planck-Institute for Dynamics and Self-Organization, Göttingen, Germany
- Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
| | - F Paul Spitzner
- Max-Planck-Institute for Dynamics and Self-Organization, Göttingen, Germany
- Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
| | - Benjamin Cramer
- Kirchhoff-Institute for Physics, Heidelberg University, Heidelberg, Germany
| | - Johannes Zierenberg
- Max-Planck-Institute for Dynamics and Self-Organization, Göttingen, Germany
- Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
| | - Viola Priesemann
- Max-Planck-Institute for Dynamics and Self-Organization, Göttingen, Germany
- Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
- Bernstein Center for Computational Neuroscience (BCCN), Göttingen, Germany
| |
Collapse
|
73
|
Bennett HC, Zhang Q, Wu YT, Manjila SB, Chon U, Shin D, Vanselow DJ, Pi HJ, Drew PJ, Kim Y. Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. Nat Commun 2024; 15:6398. [PMID: 39080289 PMCID: PMC11289283 DOI: 10.1038/s41467-024-50559-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Aging is frequently associated with compromised cerebrovasculature and pericytes. However, we do not know how normal aging differentially impacts vascular structure and function in different brain areas. Here we utilize mesoscale microscopy methods and in vivo imaging to determine detailed changes in aged murine cerebrovascular networks. Whole-brain vascular tracing shows an overall ~10% decrease in vascular length and branching density with ~7% increase in vascular radii in aged brains. Light sheet imaging with 3D immunolabeling reveals increased arteriole tortuosity of aged brains. Notably, vasculature and pericyte densities show selective and significant reductions in the deep cortical layers, hippocampal network, and basal forebrain areas. We find increased blood extravasation, implying compromised blood-brain barrier function in aged brains. Moreover, in vivo imaging in awake mice demonstrates reduced baseline and on-demand blood oxygenation despite relatively intact neurovascular coupling. Collectively, we uncover regional vulnerabilities of cerebrovascular network and physiological changes that can mediate cognitive decline in normal aging.
Collapse
Affiliation(s)
- Hannah C Bennett
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Qingguang Zhang
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Neurosurgery, Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Steffy B Manjila
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Uree Chon
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Neurosciences Graduate Program, Stanford University, Stanford, CA, 94305, USA
| | - Donghui Shin
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Daniel J Vanselow
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Hyun-Jae Pi
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Patrick J Drew
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, Biology, and Neurosurgery, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA.
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
74
|
Gliko O, Mallory M, Dalley R, Gala R, Gornet J, Zeng H, Sorensen SA, Sümbül U. High-throughput analysis of dendrite and axonal arbors reveals transcriptomic correlates of neuroanatomy. Nat Commun 2024; 15:6337. [PMID: 39068160 PMCID: PMC11283452 DOI: 10.1038/s41467-024-50728-9] [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: 12/01/2023] [Accepted: 07/16/2024] [Indexed: 07/30/2024] Open
Abstract
Neuronal anatomy is central to the organization and function of brain cell types. However, anatomical variability within apparently homogeneous populations of cells can obscure such insights. Here, we report large-scale automation of neuronal morphology reconstruction and analysis on a dataset of 813 inhibitory neurons characterized using the Patch-seq method, which enables measurement of multiple properties from individual neurons, including local morphology and transcriptional signature. We demonstrate that these automated reconstructions can be used in the same manner as manual reconstructions to understand the relationship between some, but not all, cellular properties used to define cell types. We uncover gene expression correlates of laminar innervation on multiple transcriptomically defined neuronal subclasses and types. In particular, our results reveal correlates of the variability in Layer 1 (L1) axonal innervation in a transcriptomically defined subpopulation of Martinotti cells in the adult mouse neocortex.
Collapse
Affiliation(s)
| | | | | | | | - James Gornet
- California Institute of Technology, Pasadena, CA, USA
| | | | | | | |
Collapse
|
75
|
Jones EAA, Low IIC, Cho FS, Giocomo LM. Entorhinal cortex represents task-relevant remote locations independent of CA1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.604815. [PMID: 39091781 PMCID: PMC11291150 DOI: 10.1101/2024.07.23.604815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Neurons can collectively represent the current sensory experience while an animal is exploring its environment or remote experiences while the animal is immobile. These remote representations can reflect learned associations1-3 and be required for learning4. Neurons in the medial entorhinal cortex (MEC) reflect the animal's current location during movement5, but little is known about what MEC neurons collectively represent during immobility. Here, we recorded thousands of neurons in superficial MEC and dorsal CA1 as mice learned to associate two pairs of rewarded locations. We found that during immobility, the MEC neural population frequently represented positions far from the animal's location, which we defined as 'non-local coding'. Cells with spatial firing fields at remote locations drove non-local coding, even as cells representing the current position remained active. While MEC non-local coding has been reported during sharp-wave ripples in downstream CA16, we observed non-local coding more often outside of ripples. In fact, CA1 activity was less coordinated with MEC during non-local coding. We further observed that non-local coding was pertinent to the task, as MEC preferentially represented remote task-relevant locations at appropriate times, while rarely representing task-irrelevant locations. Together, this work raises the possibility that MEC non-local coding could strengthen associations between locations independently from CA1.
Collapse
Affiliation(s)
- Emily A. Aery Jones
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Isabel I. C. Low
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Frances S. Cho
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Lisa M. Giocomo
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
76
|
Etxeberria A, Shen YAA, Vito S, Silverman SM, Imperio J, Lalehzadeh G, Soung AL, Du C, Xie L, Choy MK, Hsiao YC, Ngu H, Cho CH, Ghosh S, Novikova G, Rezzonico MG, Leahey R, Weber M, Gogineni A, Elstrott J, Xiong M, Greene JJ, Stark KL, Chan P, Roth GA, Adrian M, Li Q, Choi M, Wong WR, Sandoval W, Foreman O, Nugent AA, Friedman BA, Sadekar S, Hötzel I, Hansen DV, Chih B, Yuen TJ, Weimer RM, Easton A, Meilandt WJ, Bohlen CJ. Neutral or Detrimental Effects of TREM2 Agonist Antibodies in Preclinical Models of Alzheimer's Disease and Multiple Sclerosis. J Neurosci 2024; 44:e2347232024. [PMID: 38830764 PMCID: PMC11255434 DOI: 10.1523/jneurosci.2347-23.2024] [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: 12/18/2023] [Revised: 05/06/2024] [Accepted: 05/25/2024] [Indexed: 06/05/2024] Open
Abstract
Human genetics and preclinical studies have identified key contributions of TREM2 to several neurodegenerative conditions, inspiring efforts to modulate TREM2 therapeutically. Here, we characterize the activities of three TREM2 agonist antibodies in multiple mixed-sex mouse models of Alzheimer's disease (AD) pathology and remyelination. Receptor activation and downstream signaling are explored in vitro, and active dose ranges are determined in vivo based on pharmacodynamic responses from microglia. For mice bearing amyloid-β (Aβ) pathology (PS2APP) or combined Aβ and tau pathology (TauPS2APP), chronic TREM2 agonist antibody treatment had limited impact on microglia engagement with pathology, overall pathology burden, or downstream neuronal damage. For mice with demyelinating injuries triggered acutely with lysolecithin, TREM2 agonist antibodies unexpectedly disrupted injury resolution. Likewise, TREM2 agonist antibodies limited myelin recovery for mice experiencing chronic demyelination from cuprizone. We highlight the contributions of dose timing and frequency across models. These results introduce important considerations for future TREM2-targeting approaches.
Collapse
Affiliation(s)
- Ainhoa Etxeberria
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Yun-An A Shen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Stephen Vito
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Sean M Silverman
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Jose Imperio
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Guita Lalehzadeh
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Allison L Soung
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Changchun Du
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Luke Xie
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Man Kin Choy
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Yi-Chun Hsiao
- Antibody Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Hai Ngu
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Chang Hoon Cho
- Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, California 94080
| | - Soumitra Ghosh
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Gloriia Novikova
- Bioinformatics, Genentech, Inc., South San Francisco, California 94080
| | | | - Rebecca Leahey
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Martin Weber
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Alvin Gogineni
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Justin Elstrott
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Monica Xiong
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Jacob J Greene
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Kimberly L Stark
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Pamela Chan
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Gillie A Roth
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., South San Francisco, California 94080
| | - Max Adrian
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Qingling Li
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Meena Choi
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Weng Ruh Wong
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Wendy Sandoval
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Oded Foreman
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Alicia A Nugent
- Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, California 94080
| | - Brad A Friedman
- Bioinformatics, Genentech, Inc., South San Francisco, California 94080
| | - Shraddha Sadekar
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., South San Francisco, California 94080
| | - Isidro Hötzel
- Antibody Engineering, Genentech, Inc., South San Francisco, California 94080
| | - David V Hansen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Ben Chih
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Tracy J Yuen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Robby M Weimer
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Amy Easton
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - William J Meilandt
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Christopher J Bohlen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
77
|
Bouabid S, Zhang L, Vu MAT, Tang K, Graham BM, Noggle CA, Howe MW. Spatially organized striatum-wide acetylcholine dynamics for the learning and extinction of Pavlovian cues and actions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602947. [PMID: 39071401 PMCID: PMC11275942 DOI: 10.1101/2024.07.10.602947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Striatal acetylcholine (ACh) has been linked to behavioral flexibility. A key component of flexibility is down-regulating responding as valued cues and actions become decoupled from positive outcomes. We used array fiber photometry in mice to investigate how ACh release across the striatum evolves during learning and extinction of Pavlovian associations. Changes in multi-phasic release to cues and consummatory actions were bi-directional and region-specific. Following extinction, increases in cue-evoked ACh release emerged in the anterior dorsal striatum (aDS) which preceded a down-regulation of anticipatory behavior. Silencing ACh release from cholinergic interneurons in the aDS blocked behavioral extinction. Dopamine release dipped below baseline for down-shifted cues, but glutamate input onto cholinergic interneurons did not change, suggesting an intrastriatal mechanism for the emergence of ACh increases. Our large-scale mapping of striatal ACh dynamics during learning pinpoints region-specific elevations in ACh release positioned to down-regulate behavior during extinction, a central feature of flexible behavior.
Collapse
Affiliation(s)
- Safa Bouabid
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Liangzhu Zhang
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Mai-Anh T. Vu
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Kylie Tang
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Benjamin M. Graham
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Christian A. Noggle
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Mark W. Howe
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| |
Collapse
|
78
|
Jin M, Ogundare SO, Lanio M, Sorid S, Whye AR, Santos SL, Franceschini A, Denny CA. A SMARTR workflow for multi-ensemble atlas mapping and brain-wide network analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603299. [PMID: 39071434 PMCID: PMC11275872 DOI: 10.1101/2024.07.12.603299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
In the last decade, activity-dependent strategies for labelling multiple immediate early gene (IEG) ensembles in mice have generated unprecedented insight into the mechanisms of memory encoding, storage, and retrieval. However, few strategies exist for brain-wide mapping of multiple ensembles, including their overlapping population, and none incorporate capabilities for downstream network analysis. Here, we introduce a scalable workflow to analyze traditionally coronally-sectioned datasets produced by activity-dependent tagging systems. Intrinsic to this pipeline is simple multi-ensemble atlas registration and statistical testing in R (SMARTR), an R package which wraps mapping capabilities with functions for statistical analysis and network visualization. We demonstrate the versatility of SMARTR by mapping the ensembles underlying the acquisition and expression of learned helplessness (LH), a robust stress model. Applying network analysis, we find that exposure to inescapable shock (IS), compared to context training (CT), results in decreased centrality of regions engaged in spatial and contextual processing and higher influence of regions involved in somatosensory and affective processing. During LH expression, the substantia nigra emerges as a highly influential region which shows a functional reversal following IS, indicating a possible regulatory function of motor activity during helplessness. We also report that IS results in a robust decrease in reactivation activity across a number of cortical, hippocampal, and amygdalar regions, indicating suppression of ensemble reactivation may be a neurobiological signature of LH. These results highlight the emergent insights uniquely garnered by applying our analysis approach to multiple ensemble datasets and demonstrate the strength of our workflow as a hypothesis-generating toolkit.
Collapse
Affiliation(s)
- Michelle Jin
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
- Neurobiology and Behavior (NB&B) Graduate Program, Columbia University, New York, NY, 10027, USA
| | - Simon O. Ogundare
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
- Columbia College, New York, NY, 10027, USA
| | - Marcos Lanio
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
- Adult Neurology Residency Program, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| | | | - Alicia R. Whye
- Columbia College, New York, NY, 10027, USA
- Tri-Institutional MD-PhD Program, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sofia Leal Santos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
| | - Alessandra Franceschini
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Christine. A. Denny
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| |
Collapse
|
79
|
Tanner J, Faskowitz J, Teixeira AS, Seguin C, Coletta L, Gozzi A, Mišić B, Betzel RF. A multi-modal, asymmetric, weighted, and signed description of anatomical connectivity. Nat Commun 2024; 15:5865. [PMID: 38997282 PMCID: PMC11245624 DOI: 10.1038/s41467-024-50248-6] [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: 08/08/2023] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
The macroscale connectome is the network of physical, white-matter tracts between brain areas. The connections are generally weighted and their values interpreted as measures of communication efficacy. In most applications, weights are either assigned based on imaging features-e.g. diffusion parameters-or inferred using statistical models. In reality, the ground-truth weights are unknown, motivating the exploration of alternative edge weighting schemes. Here, we explore a multi-modal, regression-based model that endows reconstructed fiber tracts with directed and signed weights. We find that the model fits observed data well, outperforming a suite of null models. The estimated weights are subject-specific and highly reliable, even when fit using relatively few training samples, and the networks maintain a number of desirable features. In summary, we offer a simple framework for weighting connectome data, demonstrating both its ease of implementation while benchmarking its utility for typical connectome analyses, including graph theoretic modeling and brain-behavior associations.
Collapse
Affiliation(s)
- Jacob Tanner
- Cognitive Science Program, Indiana University, Bloomington, IN, USA
- School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA
| | - Joshua Faskowitz
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Andreia Sofia Teixeira
- LASIGE, Departamento de Informática, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Caio Seguin
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | | | - Alessandro Gozzi
- Functional Neuroimaging Lab, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy
| | - Bratislav Mišić
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, Canada
| | - Richard F Betzel
- Cognitive Science Program, Indiana University, Bloomington, IN, USA.
- School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
80
|
Ding Y, Huang Y, Gao P, Thai A, Chilaparasetti AN, Gopi M, Xu X, Li C. Brain image data processing using collaborative data workflows on Texera. Front Neural Circuits 2024; 18:1398884. [PMID: 39050044 PMCID: PMC11266044 DOI: 10.3389/fncir.2024.1398884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
In the realm of neuroscience, mapping the three-dimensional (3D) neural circuitry and architecture of the brain is important for advancing our understanding of neural circuit organization and function. This study presents a novel pipeline that transforms mouse brain samples into detailed 3D brain models using a collaborative data analytics platform called "Texera." The user-friendly Texera platform allows for effective interdisciplinary collaboration between team members in neuroscience, computer vision, and data processing. Our pipeline utilizes the tile images from a serial two-photon tomography/TissueCyte system, then stitches tile images into brain section images, and constructs 3D whole-brain image datasets. The resulting 3D data supports downstream analyses, including 3D whole-brain registration, atlas-based segmentation, cell counting, and high-resolution volumetric visualization. Using this platform, we implemented specialized optimization methods and obtained significant performance enhancement in workflow operations. We expect the neuroscience community can adopt our approach for large-scale image-based data processing and analysis.
Collapse
Affiliation(s)
- Yunyan Ding
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
| | - Yicong Huang
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
| | - Pan Gao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Andy Thai
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
| | | | - M. Gopi
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
| | - Xiangmin Xu
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, United States
| | - Chen Li
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
81
|
Lambert T, Niknejad HR, Kil D, Brunner C, Nuttin B, Montaldo G, Urban A. Functional ultrasound imaging and neuronal activity: how accurate is the spatiotemporal match? BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602912. [PMID: 39026833 PMCID: PMC11257620 DOI: 10.1101/2024.07.10.602912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Over the last decade, functional ultrasound (fUS) has risen as a critical tool in functional neuroimaging, leveraging hemodynamic changes to infer neural activity indirectly. Recent studies have established a strong correlation between neural spike rates (SR) and functional ultrasound signals. However, understanding their spatial distribution and variability across different brain areas is required to thoroughly interpret fUS signals. In this regard, we conducted simultaneous fUS imaging and Neuropixels recordings during stimulus-evoked activity in awake mice within three regions the visual pathway. Our findings indicate that the temporal dynamics of fUS and SR signals are linearly correlated, though the correlation coefficients vary among visual regions. Conversely, the spatial correlation between the two signals remains consistent across all regions with a spread of approximately 300 micrometers. Finally, we introduce a model that integrates the spatial and temporal components of the fUS signal, allowing for a more accurate interpretation of fUS images.
Collapse
Affiliation(s)
- Théo Lambert
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Hamid Reza Niknejad
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
- University Medical Center Utrecht, Utrecht, The Netherlands (current affiliation)
| | - Dries Kil
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Clément Brunner
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Bart Nuttin
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | - Gabriel Montaldo
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Alan Urban
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
82
|
Gruver KM, Jiao JWY, Fields E, Song S, Sjöström PJ, Watt AJ. Structured connectivity in the output of the cerebellar cortex. Nat Commun 2024; 15:5563. [PMID: 38982047 PMCID: PMC11233638 DOI: 10.1038/s41467-024-49339-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/28/2024] [Indexed: 07/11/2024] Open
Abstract
The spatial organization of a neuronal circuit is critically important for its function since the location of neurons is often associated with function. In the cerebellum, the major output of the cerebellar cortex are synapses made from Purkinje cells onto neurons in the cerebellar nuclei, yet little has been known about the spatial organization of these synapses. We explored this question using whole-cell electrophysiology and optogenetics in acute sagittal cerebellar slices to produce spatial connectivity maps of cerebellar cortical output in mice. We observed non-random connectivity where Purkinje cell inputs clustered in cerebellar transverse zones: while many nuclear neurons received inputs from a single zone, several multi-zonal connectivity motifs were also observed. Single neurons receiving input from all four zones were overrepresented in our data. These findings reveal that the output of the cerebellar cortex is spatially structured and represents a locus for multimodal integration in the cerebellum.
Collapse
Affiliation(s)
- Kim M Gruver
- Department of Biology, McGill University, Montréal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jenny W Y Jiao
- Department of Biology, McGill University, Montréal, QC, Canada
| | - Eviatar Fields
- Department of Biology, McGill University, Montréal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
| | - Sen Song
- Laboratory of Brain and Intelligence and Department of Biomedical Engineering, Tsinghua University, Beijing, China
| | - Per Jesper Sjöström
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Alanna J Watt
- Department of Biology, McGill University, Montréal, QC, Canada.
| |
Collapse
|
83
|
Doran PR, Fomin-Thunemann N, Tang RP, Balog D, Zimmerman B, Kılıç K, Martin EA, Kura S, Fisher HP, Chabbott G, Herbert J, Rauscher BC, Jiang JX, Sakadzic S, Boas DA, Devor A, Chen IA, Thunemann M. Widefield in vivo imaging system with two fluorescence and two reflectance channels, a single sCMOS detector, and shielded illumination. NEUROPHOTONICS 2024; 11:034310. [PMID: 38881627 PMCID: PMC11177117 DOI: 10.1117/1.nph.11.3.034310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024]
Abstract
Significance Widefield microscopy of the entire dorsal part of mouse cerebral cortex enables large-scale ("mesoscopic") imaging of different aspects of neuronal activity with spectrally compatible fluorescent indicators as well as hemodynamics via oxy- and deoxyhemoglobin absorption. Versatile and cost-effective imaging systems are needed for large-scale, color-multiplexed imaging of multiple fluorescent and intrinsic contrasts. Aim We aim to develop a system for mesoscopic imaging of two fluorescent and two reflectance channels. Approach Excitation of red and green fluorescence is achieved through epi-illumination. Hemoglobin absorption imaging is achieved using 525- and 625-nm light-emitting diodes positioned around the objective lens. An aluminum hemisphere placed between objective and cranial window provides diffuse illumination of the brain. Signals are recorded sequentially by a single sCMOS detector. Results We demonstrate the performance of our imaging system by recording large-scale spontaneous and stimulus-evoked neuronal, cholinergic, and hemodynamic activity in awake, head-fixed mice with a curved "crystal skull" window expressing the red calcium indicator jRGECO1a and the green acetylcholine sensorGRAB ACh 3.0 . Shielding of illumination light through the aluminum hemisphere enables concurrent recording of pupil diameter changes. Conclusions Our widefield microscope design with a single camera can be used to acquire multiple aspects of brain physiology and is compatible with behavioral readouts of pupil diameter.
Collapse
Affiliation(s)
- Patrick R. Doran
- Boston University, Graduate Program in Biomedical Engineering, Boston, Massachusetts, United States
| | - Natalie Fomin-Thunemann
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Rockwell P. Tang
- Boston University, Graduate Program in Biomedical Engineering, Boston, Massachusetts, United States
| | - Dora Balog
- Boston University, Graduate Program in Biomedical Engineering, Boston, Massachusetts, United States
| | - Bernhard Zimmerman
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Kıvılcım Kılıç
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Emily A. Martin
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Sreekanth Kura
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Harrison P. Fisher
- Boston University, Graduate Program in Biomedical Engineering, Boston, Massachusetts, United States
| | - Grace Chabbott
- Boston University, Undergraduate Program in Biomedical Engineering, Boston, Massachusetts, United States
| | - Joel Herbert
- Boston University, Undergraduate Program in Neuroscience, Boston, Massachusetts, United States
| | - Bradley C. Rauscher
- Boston University, Graduate Program in Biomedical Engineering, Boston, Massachusetts, United States
| | - John X. Jiang
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Sava Sakadzic
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Charlestown, Massachusetts, United States
| | - David A. Boas
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Anna Devor
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Charlestown, Massachusetts, United States
| | - Ichun Anderson Chen
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Martin Thunemann
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| |
Collapse
|
84
|
Dou YN, Liu Y, Ding WQ, Li Q, Zhou H, Li L, Zhao MT, Li ZYQ, Yuan J, Wang XF, Zou WY, Li A, Sun YG. Single-neuron projectome-guided analysis reveals the neural circuit mechanism underlying endogenous opioid antinociception. Natl Sci Rev 2024; 11:nwae195. [PMID: 39045468 PMCID: PMC11264302 DOI: 10.1093/nsr/nwae195] [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: 01/21/2024] [Revised: 05/05/2024] [Accepted: 05/24/2024] [Indexed: 07/25/2024] Open
Abstract
Endogenous opioid antinociception is a self-regulatory mechanism that reduces chronic pain, but its underlying circuit mechanism remains largely unknown. Here, we showed that endogenous opioid antinociception required the activation of mu-opioid receptors (MORs) in GABAergic neurons of the central amygdala nucleus (CEA) in a persistent-hyperalgesia mouse model. Pharmacogenetic suppression of these CEAMOR neurons, which mimics the effect of MOR activation, alleviated the persistent hyperalgesia. Furthermore, single-neuron projection analysis revealed multiple projectome-based subtypes of CEAMOR neurons, each innervating distinct target brain regions. We found that the suppression of axon branches projecting to the parabrachial nucleus (PB) of one subtype of CEAMOR neurons alleviated persistent hyperalgesia, indicating a subtype- and axonal-branch-specific mechanism of action. Further electrophysiological analysis revealed that suppression of a distinct CEA-PB disinhibitory circuit controlled endogenous opioid antinociception. Thus, this study identified the central neural circuit that underlies endogenous opioid antinociception, providing new insight into the endogenous pain modulatory mechanisms.
Collapse
Affiliation(s)
- Yan-Nong Dou
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuan Liu
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Department of Biology, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Lingang Laboratory, Shanghai 200031, China
| | - Wen-Qun Ding
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Li
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hua Zhou
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ling Li
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng-Ting Zhao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zheng-Yi-Qi Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jing Yuan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Xiao-Fei Wang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wang-Yuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Yan-Gang Sun
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
85
|
Rault N, Bergmans T, Delfstra N, Kleijnen BJ, Zeldenrust F, Celikel T. Where Top-Down Meets Bottom-Up: Cell-Type Specific Connectivity Map of the Whisker System. Neuroinformatics 2024; 22:251-268. [PMID: 38767789 PMCID: PMC11329691 DOI: 10.1007/s12021-024-09658-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2024] [Indexed: 05/22/2024]
Abstract
Sensorimotor computation integrates bottom-up world state information with top-down knowledge and task goals to form action plans. In the rodent whisker system, a prime model of active sensing, evidence shows neuromodulatory neurotransmitters shape whisker control, affecting whisking frequency and amplitude. Since neuromodulatory neurotransmitters are mostly released from subcortical nuclei and have long-range projections that reach the rest of the central nervous system, mapping the circuits of top-down neuromodulatory control of sensorimotor nuclei will help to systematically address the mechanisms of active sensing. Therefore, we developed a neuroinformatic target discovery pipeline to mine the Allen Institute's Mouse Brain Connectivity Atlas. Using network connectivity analysis, we identified new putative connections along the whisker system and anatomically confirmed the existence of 42 previously unknown monosynaptic connections. Using this data, we updated the sensorimotor connectivity map of the mouse whisker system and developed the first cell-type-specific map of the network. The map includes 157 projections across 18 principal nuclei of the whisker system and neuromodulatory neurotransmitter-releasing. Performing a graph network analysis of this connectome, we identified cell-type specific hubs, sources, and sinks, provided anatomical evidence for monosynaptic inhibitory projections into all stages of the ascending pathway, and showed that neuromodulatory projections improve network-wide connectivity. These results argue that beyond the modulatory chemical contributions to information processing and transfer in the whisker system, the circuit connectivity features of the neuromodulatory networks position them as nodes of sensory and motor integration.
Collapse
Affiliation(s)
- Nicolas Rault
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands.
| | - Tido Bergmans
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Natasja Delfstra
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | | | - Fleur Zeldenrust
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | | |
Collapse
|
86
|
Mitani TT, Susaki EA, Matsumoto K, Ueda HR. Realization of cellomics to dive into the whole-body or whole-organ cell cloud. Nat Methods 2024; 21:1138-1142. [PMID: 38871985 DOI: 10.1038/s41592-024-02307-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Affiliation(s)
- Tomoki T Mitani
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
- Department of Systems Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Etsuo A Susaki
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Katsuhiko Matsumoto
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hiroki R Ueda
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan.
- Department of Systems Pharmacology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
87
|
Song RX, Miao HT, Jia SY, Li WG, Liu JZ, Zhang W, Xing BR, Zhao JY, Zhang LM, Li XM. Hemorrhagic Shock and Resuscitation Causes Excessive Dopaminergic Signaling in the mPFC and Cognitive Dysfunction. Mol Neurobiol 2024; 61:3899-3910. [PMID: 38041715 DOI: 10.1007/s12035-023-03804-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: 08/22/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023]
Abstract
Peri-operative hemorrhagic shock and resuscitation (HSR), a severe traumatic stress, is closely associated with post-operative anxiety, depression, and cognitive dysfunction, subsequently causing a serious burden on families and society. Following the co-release of corticotropin-releasing factor and catecholamine, traumatic stress activates dopaminergic neurons, increasing the addictive behavior and neurocognitive impairment risks. This study investigates the association between cognitive dysfunction and dopaminergic neurons in the mPFC under HSR conditions. This study established an HSR model by bleeding and re-transfusion in the mice. After HSR exposure, a dopamine D1 receptor antagonist, SKF-83566, was administered intraperitoneally for three consecutive days. Novel object recognition (NOR), conditioned fearing (FC), and conditioned place preference (CPP) were used to assess cognitive changes 16 days after HSR exposure. Local field potential (LFP) in the mPFC was also investigated during the novel object exploration. Compared with the mice exposed to sham, there was a significant decrease in the object recognition index, a reduction in context- and tone-related freezing time, an increase in CPP values, a downregulation of β-power but upregulation of γ-power in the mPFC in the mice exposed to HSR. Moreover, the mice exposed to HSR showed significantly upregulated TH-positive cell number, cleaved caspase-1- and TH-positive cells, and interleukin (IL)-1β/18 expression in the mPFC compared with sham; SKF-83566 could partially reverse these alternations. The HSR caused excessive dopaminergic signaling and cognitive dysfunction in the mPFC, a condition that might be ameliorated using a dopamine D1 receptor inhibitor.
Collapse
Affiliation(s)
- Rong-Xin Song
- Anesthesia and Trauma Research Unit, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Hui-Tao Miao
- Anesthesia and Trauma Research Unit, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Shi-Yan Jia
- Anesthesia and Trauma Research Unit, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Wen-Guang Li
- Graduate School, Hebei Medical University, Shijiazhuang, China
| | - Ji-Zhen Liu
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bao-Rui Xing
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Jian-Yong Zhao
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Li-Min Zhang
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China.
| | - Xiao-Ming Li
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China.
| |
Collapse
|
88
|
Ertürk A. Deep 3D histology powered by tissue clearing, omics and AI. Nat Methods 2024; 21:1153-1165. [PMID: 38997593 DOI: 10.1038/s41592-024-02327-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 05/28/2024] [Indexed: 07/14/2024]
Abstract
To comprehensively understand tissue and organism physiology and pathophysiology, it is essential to create complete three-dimensional (3D) cellular maps. These maps require structural data, such as the 3D configuration and positioning of tissues and cells, and molecular data on the constitution of each cell, spanning from the DNA sequence to protein expression. While single-cell transcriptomics is illuminating the cellular and molecular diversity across species and tissues, the 3D spatial context of these molecular data is often overlooked. Here, I discuss emerging 3D tissue histology techniques that add the missing third spatial dimension to biomedical research. Through innovations in tissue-clearing chemistry, labeling and volumetric imaging that enhance 3D reconstructions and their synergy with molecular techniques, these technologies will provide detailed blueprints of entire organs or organisms at the cellular level. Machine learning, especially deep learning, will be essential for extracting meaningful insights from the vast data. Further development of integrated structural, molecular and computational methods will unlock the full potential of next-generation 3D histology.
Collapse
Affiliation(s)
- Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany.
- School of Medicine, Koç University, İstanbul, Turkey.
- Deep Piction GmbH, Munich, Germany.
| |
Collapse
|
89
|
Onos KD, Lin PB, Pandey RS, Persohn SA, Burton CP, Miner EW, Eldridge K, Kanyinda JN, Foley KE, Carter GW, Howell GR, Territo PR. Assessment of neurovascular uncoupling: APOE status is a key driver of early metabolic and vascular dysfunction. Alzheimers Dement 2024; 20:4951-4969. [PMID: 38713704 PMCID: PMC11247674 DOI: 10.1002/alz.13842] [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: 12/08/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 05/09/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common cause of dementia worldwide, with apolipoprotein Eε4 (APOEε4) being the strongest genetic risk factor. Current clinical diagnostic imaging focuses on amyloid and tau; however, new methods are needed for earlier detection. METHODS PET imaging was used to assess metabolism-perfusion in both sexes of aging C57BL/6J, and hAPOE mice, and were verified by transcriptomics, and immunopathology. RESULTS All hAPOE strains showed AD phenotype progression by 8 months, with females exhibiting the regional changes, which correlated with GO-term enrichments for glucose metabolism, perfusion, and immunity. Uncoupling analysis revealed APOEε4/ε4 exhibited significant Type-1 uncoupling (↓ glucose uptake, ↑ perfusion) at 8 and 12 months, while APOEε3/ε4 demonstrated Type-2 uncoupling (↑ glucose uptake, ↓ perfusion), while immunopathology confirmed cell specific contributions. DISCUSSION This work highlights APOEε4 status in AD progression manifests as neurovascular uncoupling driven by immunological activation, and may serve as an early diagnostic biomarker. HIGHLIGHTS We developed a novel analytical method to analyze PET imaging of 18F-FDG and 64Cu-PTSM data in both sexes of aging C57BL/6J, and hAPOEε3/ε3, hAPOEε4/ε4, and hAPOEε3/ε4 mice to assess metabolism-perfusion profiles termed neurovascular uncoupling. This analysis revealed APOEε4/ε4 exhibited significant Type-1 uncoupling (decreased glucose uptake, increased perfusion) at 8 and 12 months, while APOEε3/ε4 demonstrated significant Type-2 uncoupling (increased glucose uptake, decreased perfusion) by 8 months which aligns with immunopathology and transcriptomic signatures. This work highlights that there may be different mechanisms underlying age related changes in APOEε4/ε4 compared with APOEε3/ε4. We predict that these changes may be driven by immunological activation and response, and may serve as an early diagnostic biomarker.
Collapse
Affiliation(s)
| | - Peter B. Lin
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Ravi S. Pandey
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Scott A. Persohn
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Charles P. Burton
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Ethan W. Miner
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kierra Eldridge
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | | | - Kate E. Foley
- The Jackson LaboratoryBar HarborMaineUSA
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Gregory W. Carter
- The Jackson LaboratoryBar HarborMaineUSA
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | | | - Paul R. Territo
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of MedicineDivision of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
90
|
Oliver Goral R, Lamb PW, Yakel JL. Acetylcholine Neurons Become Cholinergic during Three Time Windows in the Developing Mouse Brain. eNeuro 2024; 11:ENEURO.0542-23.2024. [PMID: 38942474 PMCID: PMC11253243 DOI: 10.1523/eneuro.0542-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/12/2024] [Accepted: 06/22/2024] [Indexed: 06/30/2024] Open
Abstract
Acetylcholine (ACh) neurons in the central nervous system are required for the coordination of neural network activity during higher brain functions, such as attention, learning, and memory, as well as locomotion. Disturbed cholinergic signaling has been described in many neurodevelopmental and neurodegenerative disorders. Furthermore, cotransmission of other signaling molecules, such as glutamate and GABA, with ACh has been associated with essential roles in brain function or disease. However, it is unknown when ACh neurons become cholinergic during development. Thus, understanding the timeline of how the cholinergic system develops and becomes active in the healthy brain is a crucial part of understanding brain development. To study this, we used transgenic mice to selectively label ACh neurons with tdTomato. We imaged serial sectioned brains and generated whole-brain reconstructions at different time points during pre- and postnatal development. We found three crucial time windows-two in the prenatal and one in the postnatal brain-during which most ACh neuron populations become cholinergic in the brain. We also found that cholinergic gene expression is initiated in cortical ACh interneurons, while the cerebral cortex is innervated by cholinergic projection neurons from the basal forebrain. Taken together, we show that ACh neuron populations are present and become cholinergic before postnatal day 12, which is the onset of major sensory processes, such as hearing and vision. We conclude that the birth of ACh neurons and initiation of cholinergic gene expression are temporally separated during development but highly coordinated by brain anatomical structure.
Collapse
Affiliation(s)
- Rene Oliver Goral
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
- Center on Compulsive Behaviors, National Institutes of Health, Bethesda, Maryland 20892
| | - Patricia W Lamb
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Jerrel L Yakel
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| |
Collapse
|
91
|
Hsu TT, Huang TN, Wang CY, Hsueh YP. Deep brain stimulation of the Tbr1-deficient mouse model of autism spectrum disorder at the basolateral amygdala alters amygdalar connectivity, whole-brain synchronization, and social behaviors. PLoS Biol 2024; 22:e3002646. [PMID: 39012916 PMCID: PMC11280143 DOI: 10.1371/journal.pbio.3002646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/26/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024] Open
Abstract
Autism spectrum disorders (ASDs) are considered neural dysconnectivity syndromes. To better understand ASD and uncover potential treatments, it is imperative to know and dissect the connectivity deficits under conditions of autism. Here, we apply a whole-brain immunostaining and quantification platform to demonstrate impaired structural and functional connectivity and aberrant whole-brain synchronization in a Tbr1+/- autism mouse model. We express a channelrhodopsin variant oChIEF fused with Citrine at the basolateral amygdala (BLA) to outline the axonal projections of BLA neurons. By activating the BLA under blue light theta-burst stimulation (TBS), we then evaluate the effect of BLA activation on C-FOS expression at a whole brain level to represent neural activity. We show that Tbr1 haploinsufficiency almost completely disrupts contralateral BLA axonal projections and results in mistargeting in both ipsilateral and contralateral hemispheres, thereby globally altering BLA functional connectivity. Based on correlated C-FOS expression among brain regions, we further show that Tbr1 deficiency severely disrupts whole-brain synchronization in the absence of salient stimulation. Tbr1+/- and wild-type (WT) mice exhibit opposing responses to TBS-induced amygdalar activation, reducing synchronization in WT mice but enhancing it in Tbr1+/- mice. Whole-brain modular organization and intermodule connectivity are also affected by Tbr1 deficiency and amygdalar activation. Following BLA activation by TBS, the synchronizations of the whole brain and the default mode network, a specific subnetwork highly relevant to ASD, are enhanced in Tbr1+/- mice, implying a potential ameliorating effect of amygdalar stimulation on brain function. Indeed, TBS-mediated BLA activation increases nose-to-nose social interactions of Tbr1+/- mice, strengthening evidence for the role of amygdalar connectivity in social behaviors. Our high-resolution analytical platform reveals the inter- and intrahemispheric connectopathies arising from ASD. Our study emphasizes the defective synchronization at a whole-brain scale caused by Tbr1 deficiency and implies a potential beneficial effect of deep brain stimulation at the amygdala for TBR1-linked autism.
Collapse
Affiliation(s)
- Tsan-Ting Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Tzyy-Nan Huang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Chien-Yao Wang
- Institute of Information Science, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| |
Collapse
|
92
|
Yan Y, Murphy TH. Decoding state-dependent cortical-cerebellar cellular functional connectivity in the mouse brain. Cell Rep 2024; 43:114348. [PMID: 38865245 DOI: 10.1016/j.celrep.2024.114348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 04/16/2024] [Accepted: 05/26/2024] [Indexed: 06/14/2024] Open
Abstract
The cortex and cerebellum form multi-synaptic reciprocal connections. We investigate the functional connectivity between single spiking cerebellar neurons and the population activity of the mouse dorsal cortex using mesoscale imaging. Cortical representations of individual cerebellar neurons vary significantly across different brain states but are drawn from a common set of cortical networks. These cortical-cerebellar connectivity features are observed in mossy fibers and Purkinje cells as well as neurons in different cerebellar lobules, albeit with variations across cell types and regions. Complex spikes of Purkinje cells preferably associate with the sensorimotor cortex, whereas simple spikes display more diverse cortical connectivity patterns. The spontaneous functional connectivity patterns align with cerebellar neurons' functional responses to external stimuli in a modality-specific manner. The tuning properties of subsets of cerebellar neurons differ between anesthesia and awake states, mirrored by state-dependent changes in their long-range functional connectivity patterns with mesoscale cortical activity.
Collapse
Affiliation(s)
- Yuhao Yan
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Timothy H Murphy
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
93
|
Nietz AK, Popa LS, Carter RE, Gerhart ML, Manikonda K, Ranum LP, Ebner TJ. Cerebral cortical functional hyperconnectivity in a mouse model of spinocerebellar ataxia type 8 (SCA8). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599947. [PMID: 38948725 PMCID: PMC11212952 DOI: 10.1101/2024.06.20.599947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Spinocerebellar Ataxia Type 8 (SCA8) is an inherited neurodegenerative disease caused by a bidirectionally expressed CTG●CAG expansion mutation in the ATXN-8 and ATXN8-OS genes. While primarily a motor disorder, psychiatric and cognitive symptoms have been reported. It is difficult to elucidate how the disease alters brain function in areas with little or no degeneration producing both motor and cognitive symptoms. Using transparent polymer skulls and CNS-wide GCaMP6f expression, we studied neocortical networks throughout SCA8 progression using wide-field Ca2+ imaging in a transgenic mouse model of SCA8. We observed that neocortical networks in SCA8+ mice were hyperconnected globally which led to network configurations with increased global efficiency and centrality. At the regional level, significant network changes occurred in nearly all cortical regions, however mainly involved sensory and association cortices. Changes in functional connectivity in anterior motor regions worsened later in the disease. Near perfect decoding of animal genotype was obtained using a generalized linear model based on canonical correlation strengths between activity in cortical regions. The major contributors to decoding were concentrated in the somatosensory, higher visual and retrosplenial cortices and occasionally extended into the motor regions, demonstrating that the areas with the largest network changes are predictive of disease state.
Collapse
Affiliation(s)
- Angela K. Nietz
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Laurentiu S. Popa
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Russell E. Carter
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Morgan L Gerhart
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Keerthi Manikonda
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Laura P.W. Ranum
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Timothy J. Ebner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
94
|
Zheng X, Wu B, Liu Y, Simmons SK, Kim K, Clarke GS, Ashiq A, Park J, Li J, Wang Z, Tong L, Wang Q, Rajamani KT, Muñoz-Castañeda R, Mu S, Qi T, Zhang Y, Ngiam ZC, Ohte N, Hanashima C, Wu Z, Xu X, Levin JZ, Jin X. Massively parallel in vivo Perturb-seq reveals cell-type-specific transcriptional networks in cortical development. Cell 2024; 187:3236-3248.e21. [PMID: 38772369 PMCID: PMC11193654 DOI: 10.1016/j.cell.2024.04.050] [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: 09/08/2023] [Revised: 11/30/2023] [Accepted: 04/30/2024] [Indexed: 05/23/2024]
Abstract
Leveraging AAVs' versatile tropism and labeling capacity, we expanded the scale of in vivo CRISPR screening with single-cell transcriptomic phenotyping across embryonic to adult brains and peripheral nervous systems. Through extensive tests of 86 vectors across AAV serotypes combined with a transposon system, we substantially amplified labeling efficacy and accelerated in vivo gene delivery from weeks to days. Our proof-of-principle in utero screen identified the pleiotropic effects of Foxg1, highlighting its tight regulation of distinct networks essential for cell fate specification of Layer 6 corticothalamic neurons. Notably, our platform can label >6% of cerebral cells, surpassing the current state-of-the-art efficacy at <0.1% by lentivirus, to achieve analysis of over 30,000 cells in one experiment and enable massively parallel in vivo Perturb-seq. Compatible with various phenotypic measurements (single-cell or spatial multi-omics), it presents a flexible approach to interrogate gene function across cell types in vivo, translating gene variants to their causal function.
Collapse
Affiliation(s)
- Xinhe Zheng
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Boli Wu
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Yuejia Liu
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Sean K Simmons
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kwanho Kim
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Grace S Clarke
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Abdullah Ashiq
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Joshua Park
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Jiwen Li
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Zhilin Wang
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Liqi Tong
- Center for Neural Circuit Mapping, Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92617, USA
| | - Qizhao Wang
- Center for Neural Circuit Mapping, Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92617, USA
| | - Keerthi T Rajamani
- Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Rodrigo Muñoz-Castañeda
- Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Shang Mu
- Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Tianbo Qi
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Yunxiao Zhang
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Zi Chao Ngiam
- Center for Advanced Biomedical Sciences, Waseda University, Tokyo 162-8480, Japan
| | - Naoto Ohte
- Center for Advanced Biomedical Sciences, Waseda University, Tokyo 162-8480, Japan
| | - Carina Hanashima
- Center for Advanced Biomedical Sciences, Waseda University, Tokyo 162-8480, Japan
| | - Zhuhao Wu
- Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Xiangmin Xu
- Center for Neural Circuit Mapping, Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92617, USA
| | - Joshua Z Levin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xin Jin
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA.
| |
Collapse
|
95
|
Mandino F, Horien C, Shen X, Desrosiers-Gregoire G, Luo W, Markicevic M, Constable RX, Papademetris X, Chakravarty MM, Betzel RF, Lake EMR. Multimodal identification of the mouse brain using simultaneous Ca 2+ imaging and fMRI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.594620. [PMID: 38826324 PMCID: PMC11142213 DOI: 10.1101/2024.05.24.594620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Individual differences in neuroimaging are of interest to clinical and cognitive neuroscientists based on their potential for guiding the personalized treatment of various heterogeneous neurological conditions and diseases. Despite many advantages, the workhorse in this arena, BOLD (blood-oxygen-level-dependent) functional magnetic resonance imaging (fMRI) suffers from low spatiotemporal resolution and specificity as well as a propensity for noise and spurious signal corruption. To better understand individual differences in BOLD-fMRI data, we can use animal models where fMRI, alongside complementary but more invasive contrasts, can be accessed. Here, we apply simultaneous wide-field fluorescence calcium imaging and BOLD-fMRI in mice to interrogate individual differences using a connectome-based identification framework adopted from the human fMRI literature. This approach yields high spatiotemporal resolution cell-type specific signals (here, from glia, excitatory, as well as inhibitory interneurons) from the whole cortex. We found mouse multimodal connectome- based identification to be successful and explored various features of these data.
Collapse
|
96
|
van Velthoven CTJ, Gao Y, Kunst M, Lee C, McMillen D, Chakka AB, Casper T, Clark M, Chakrabarty R, Daniel S, Dolbeare T, Ferrer R, Gloe J, Goldy J, Guzman J, Halterman C, Ho W, Huang M, James K, Nguy B, Pham T, Ronellenfitch K, Thomas ED, Torkelson A, Pagan CM, Kruse L, Dee N, Ng L, Waters J, Smith KA, Tasic B, Yao Z, Zeng H. The transcriptomic and spatial organization of telencephalic GABAergic neuronal types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599583. [PMID: 38948843 PMCID: PMC11212977 DOI: 10.1101/2024.06.18.599583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The telencephalon of the mammalian brain comprises multiple regions and circuit pathways that play adaptive and integrative roles in a variety of brain functions. There is a wide array of GABAergic neurons in the telencephalon; they play a multitude of circuit functions, and dysfunction of these neurons has been implicated in diverse brain disorders. In this study, we conducted a systematic and in-depth analysis of the transcriptomic and spatial organization of GABAergic neuronal types in all regions of the mouse telencephalon and their developmental origins. This was accomplished by utilizing 611,423 single-cell transcriptomes from the comprehensive and high-resolution transcriptomic and spatial cell type atlas for the adult whole mouse brain we have generated, supplemented with an additional single-cell RNA-sequencing dataset containing 99,438 high-quality single-cell transcriptomes collected from the pre- and postnatal developing mouse brain. We present a hierarchically organized adult telencephalic GABAergic neuronal cell type taxonomy of 7 classes, 52 subclasses, 284 supertypes, and 1,051 clusters, as well as a corresponding developmental taxonomy of 450 clusters across different ages. Detailed charting efforts reveal extraordinary complexity where relationships among cell types reflect both spatial locations and developmental origins. Transcriptomically and developmentally related cell types can often be found in distant and diverse brain regions indicating that long-distance migration and dispersion is a common characteristic of nearly all classes of telencephalic GABAergic neurons. Additionally, we find various spatial dimensions of both discrete and continuous variations among related cell types that are correlated with gene expression gradients. Lastly, we find that cortical, striatal and some pallidal GABAergic neurons undergo extensive postnatal diversification, whereas septal and most pallidal GABAergic neuronal types emerge simultaneously during the embryonic stage with limited postnatal diversification. Overall, the telencephalic GABAergic cell type taxonomy can serve as a foundational reference for molecular, structural and functional studies of cell types and circuits by the entire community.
Collapse
Affiliation(s)
| | - Yuan Gao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mike Huang
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Beagan Nguy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
97
|
Carraro C, Montgomery JV, Klimmt J, Paquet D, Schultze JL, Beyer MD. Tackling neurodegeneration in vitro with omics: a path towards new targets and drugs. Front Mol Neurosci 2024; 17:1414886. [PMID: 38952421 PMCID: PMC11215216 DOI: 10.3389/fnmol.2024.1414886] [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: 04/09/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024] Open
Abstract
Drug discovery is a generally inefficient and capital-intensive process. For neurodegenerative diseases (NDDs), the development of novel therapeutics is particularly urgent considering the long list of late-stage drug candidate failures. Although our knowledge on the pathogenic mechanisms driving neurodegeneration is growing, additional efforts are required to achieve a better and ultimately complete understanding of the pathophysiological underpinnings of NDDs. Beyond the etiology of NDDs being heterogeneous and multifactorial, this process is further complicated by the fact that current experimental models only partially recapitulate the major phenotypes observed in humans. In such a scenario, multi-omic approaches have the potential to accelerate the identification of new or repurposed drugs against a multitude of the underlying mechanisms driving NDDs. One major advantage for the implementation of multi-omic approaches in the drug discovery process is that these overarching tools are able to disentangle disease states and model perturbations through the comprehensive characterization of distinct molecular layers (i.e., genome, transcriptome, proteome) up to a single-cell resolution. Because of recent advances increasing their affordability and scalability, the use of omics technologies to drive drug discovery is nascent, but rapidly expanding in the neuroscience field. Combined with increasingly advanced in vitro models, which particularly benefited from the introduction of human iPSCs, multi-omics are shaping a new paradigm in drug discovery for NDDs, from disease characterization to therapeutics prediction and experimental screening. In this review, we discuss examples, main advantages and open challenges in the use of multi-omic approaches for the in vitro discovery of targets and therapies against NDDs.
Collapse
Affiliation(s)
- Caterina Carraro
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Jessica V. Montgomery
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), Bonn, Germany
| | - Julien Klimmt
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joachim L. Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE, Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn and West German Genome Center, Bonn, Germany
| | - Marc D. Beyer
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), Bonn, Germany
- PRECISE, Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn and West German Genome Center, Bonn, Germany
- Immunogenomics & Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), Bonn, Germany
| |
Collapse
|
98
|
Birman D, Chapuis G, Faulkner M, Rossant C, Benson J, Catarino JA, Churchland AK, Hu F, Huntenburg JM, Khanal A, Krasniak C, Lau PYP, Meijer GT, Miska NJ, Noel JP, Pan-Vazquez A, Roth N, Schartner M, Socha KZ, Steinmetz NA, Urai AE, Wells MJ, West SJ, Winter O. Interactive data exploration websites for large-scale electrophysiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597950. [PMID: 38915704 PMCID: PMC11195081 DOI: 10.1101/2024.06.07.597950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Methodological advances in neuroscience have enabled the collection of massive datasets which demand innovative approaches for scientific communication. Existing platforms for data storage lack intuitive tools for data exploration, limiting our ability to interact effectively with these brain-wide datasets. We introduce two public websites: (Data and Atlas) developed for the International Brain Laboratory which provide access to millions of behavioral trials and hundreds of thousands of individual neurons. These interfaces allow users to discover both the raw and processed brain-wide data released by the IBL at the scale of the whole brain, individual sessions, trials, and neurons. By hosting these data interfaces as websites they are available cross-platform with no installation. By releasing each site's code as a modular open-source framework, other researchers can easily develop their own web interfaces and explore their own data. As neuroscience datasets continue to expand, customizable web interfaces offer a glimpse into a future of streamlined data exploration and act as blueprints for future tools.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Fei Hu
- University of California Berkeley, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Ganglberger F, Kargl D, Töpfer M, Hernandez-Lallement J, Lawless N, Fernandez-Albert F, Haubensak W, Bühler K. BrainTACO: an explorable multi-scale multi-modal brain transcriptomic and connectivity data resource. Commun Biol 2024; 7:730. [PMID: 38877144 PMCID: PMC11178817 DOI: 10.1038/s42003-024-06355-7] [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: 04/27/2023] [Accepted: 05/20/2024] [Indexed: 06/16/2024] Open
Abstract
Exploring the relationships between genes and brain circuitry can be accelerated by joint analysis of heterogeneous datasets from 3D imaging data, anatomical data, as well as brain networks at varying scales, resolutions, and modalities. Generating an integrated view, beyond the individual resources' original purpose, requires the fusion of these data to a common space, and a visualization that bridges the gap across scales. However, despite ever expanding datasets, few platforms for integration and exploration of this heterogeneous data exist. To this end, we present the BrainTACO (Brain Transcriptomic And Connectivity Data) resource, a selection of heterogeneous, and multi-scale neurobiological data spatially mapped onto a common, hierarchical reference space, combined via a holistic data integration scheme. To access BrainTACO, we extended BrainTrawler, a web-based visual analytics framework for spatial neurobiological data, with comparative visualizations of multiple resources. This enables gene expression dissection of brain networks with, to the best of our knowledge, an unprecedented coverage and allows for the identification of potential genetic drivers of connectivity in both mice and humans that may contribute to the discovery of dysconnectivity phenotypes. Hence, BrainTACO reduces the need for time-consuming manual data aggregation often required for computational analyses in script-based toolboxes, and supports neuroscientists by directly leveraging the data instead of preparing it.
Collapse
Affiliation(s)
- Florian Ganglberger
- Biomedical Image Informatics, VRVis Research Center, Vienna, Austria
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma, Biberach an der Riss, Germany
| | - Dominic Kargl
- Department of Neuronal Cell Biology, Vienna Medical University, Vienna, Austria
| | - Markus Töpfer
- Biomedical Image Informatics, VRVis Research Center, Vienna, Austria
| | - Julien Hernandez-Lallement
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma, Biberach an der Riss, Germany
| | - Nathan Lawless
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma, Biberach an der Riss, Germany
| | - Francesc Fernandez-Albert
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma, Biberach an der Riss, Germany
| | - Wulf Haubensak
- Department of Neuronal Cell Biology, Vienna Medical University, Vienna, Austria
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Katja Bühler
- Biomedical Image Informatics, VRVis Research Center, Vienna, Austria.
| |
Collapse
|
100
|
Huang L, Sun Y, Luo C, Wang W, Shi S, Sun G, Ju P, Chen J. Characterizing defective lipid metabolism in the lateral septum of mice treated with olanzapine: implications for its side effects. Front Pharmacol 2024; 15:1419098. [PMID: 38948475 PMCID: PMC11211371 DOI: 10.3389/fphar.2024.1419098] [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: 04/17/2024] [Accepted: 05/23/2024] [Indexed: 07/02/2024] Open
Abstract
Schizophrenia significantly impacts cognitive and behavioral functions and is primarily treated with second-generation antipsychotics (SGAs) such as olanzapine. Despite their efficacy, these drugs are linked to serious metabolic side effects which can diminish patient compliance, worsen psychiatric symptoms and increase cardiovascular disease risk. This study explores the hypothesis that SGAs affect the molecular determinants of synaptic plasticity and brain activity, particularly focusing on the lateral septum (LS) and its interactions within hypothalamic circuits that regulate feeding and energy expenditure. Utilizing functional ultrasound imaging, RNA sequencing, and weighted gene co-expression network analysis, we identified significant alterations in the functional connection between the hypothalamus and LS, along with changes in gene expression in the LS of mice following prolonged olanzapine exposure. Our analysis revealed a module closely linked to increases in body weight and adiposity, featuring genes primarily involved in lipid metabolism pathways, notably Apoa1, Apoc3, and Apoh. These findings suggest that olanzapine may influence body weight and adiposity through its impact on lipid metabolism-related genes in the LS. Therefore, the neural circuits connecting the LS and LH, along with the accompanying alterations in lipid metabolism, are likely crucial factors contributing to the weight gain and metabolic side effects associated with olanzapine treatment.
Collapse
Affiliation(s)
- Lixuan Huang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Sun
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai, China
| | - Chao Luo
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai, China
| | - Si Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Genmin Sun
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peijun Ju
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai, China
- Shanghai Institute of Traditional Chinese Medicine for Mental Health, Shanghai, China
| | - Jianhua Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai, China
- Shanghai Institute of Traditional Chinese Medicine for Mental Health, Shanghai, China
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|