1
|
De Vincentiis S, Capitanini E, Kira K, Dell'Amico C, Takahashi J, Onorati M, Raudzus F, Raffa V. Mechanical Forces Guide Axon Growth through the Nigrostriatal Pathway in an Organotypic Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500400. [PMID: 40349175 DOI: 10.1002/advs.202500400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/04/2025] [Indexed: 05/14/2025]
Abstract
Reconstructing the nigrostriatal pathway is one of the major challenges in cell replacement therapies for Parkinson's disease due to the lack of enabling technologies capable of guiding the reinnervation of dopaminergic precursors transplanted into the substantia nigra toward the striatum. This paper examines nano-pulling, as a technology to enable the remote manipulation of axonal growth. Specifically, an organotypic model consisting of co-cultures of the substantia nigra and the striatum is developed to demonstrate that when cortical neural progenitors are transplanted into the substantia nigra, nano-pulling can guide and enhance the elongation of neural projections toward the striatum. To provide additional evidence, induced pluripotent stem cell-derived dopaminergic progenitor neurospheres are generated and it is shown that nano-pulling can induce guided growth and promote the maturation of their neural processes. Altogether, this study demonstrates the potential of nano-pulling as an emerging technique to promote directed reinnervation within the central nervous system.
Collapse
Affiliation(s)
| | | | - Karen Kira
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
- Neuronal Signaling and Regeneration Unit, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
- Center for Medical Education and Internationalization (CMEI), Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | | | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Marco Onorati
- Department of Biology, University of Pisa, Pisa, 56126, Italy
| | - Fabian Raudzus
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
- Neuronal Signaling and Regeneration Unit, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
- Center for Medical Education and Internationalization (CMEI), Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Vittoria Raffa
- Department of Biology, University of Pisa, Pisa, 56126, Italy
| |
Collapse
|
2
|
Kamińska K, Grygier B, Regulska M, Procner M, Leśkiewicz M, Szczęch M, Yang J, Bouzga A, Warszyński P, Lasoń W, Szczepanowicz K, Basta-Kaim A. Multilayered Nanocarriers as a New Strategy for Delivering Drugs with Protective and Anti-inflammatory Potential: Studies in Hippocampal Organotypic Cultures Subjected to Experimental Ischemia. Mol Neurobiol 2025; 62:6333-6351. [PMID: 39786699 PMCID: PMC11953135 DOI: 10.1007/s12035-024-04670-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: 08/29/2024] [Accepted: 12/16/2024] [Indexed: 01/12/2025]
Abstract
Oxidative stress and neuroinflammation play a pivotal role in pathomechanisms of brain ischemia. Our research aimed to formulate a nanotheranostic system for delivering carnosic acid as a neuroprotective agent with anti-oxidative and anti-inflammatory properties to ischemic brain tissue, mimicked by organotypic hippocampal cultures (OHCs) exposed to oxygen-glucose deprivation (OGD). In the first part of this study, the nanocarriers were formulated by encapsulating two types of nanocores (nanoemulsion (AOT) and polymeric (PCL)) containing CA into multilayer shells using the sequential adsorption of charged nanoobjects method. The newly designed nanoparticles possessed favorable physicochemical characteristics as reflected by zeta potential and other parameters. Next, we demonstrated that the newly designed gadolinium-containing nanoparticles were not toxic to OHCs and did not affect the detrimental effects of OGD on the viability of the hippocampal cells. Importantly, they readily crossed the artificial blood-brain barrier based on the human cerebral microvascular endothelial (hCMEC/D3) cell line. Furthermore, the PCL-Gd carnosic acid-loaded nanoparticles displayed anti-inflammatory potential, expressed as decreased OGD-induced HIF-1α and IL-1β levels. Results of the molecular study revealed a complex mechanism of the nanoformulation on ischemia-related neuroinflammation in OHCs, including anti-inflammatory protein A20 stimulation and moderate attenuation of the NFκB signaling pathway. Summing up, this study points to acceptable biocompatibility of the newly designed CA-containing theranostic nanoformulation and emphasizes their interaction with inflammatory processes commonly associated with the ischemic brain.
Collapse
Affiliation(s)
- Kinga Kamińska
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Beata Grygier
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Magdalena Regulska
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Magdalena Procner
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Monika Leśkiewicz
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Marta Szczęch
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239, Kraków, Poland
| | - Juan Yang
- SINTEF Material and Chemistry, Forskningsveien 1, NO-0314, Oslo, Norway
| | - Aud Bouzga
- SINTEF Material and Chemistry, Forskningsveien 1, NO-0314, Oslo, Norway
| | - Piotr Warszyński
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239, Kraków, Poland
| | - Władysław Lasoń
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Krzysztof Szczepanowicz
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239, Kraków, Poland.
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland.
| |
Collapse
|
3
|
Rivera JF, Huang H, Weng W, Sohn H, Girasole AE, Li S, Albanese MA, Qin M, Tao C, Klug ME, Rao S, Paletzki R, Herring BE, Kanoski SE, Zhang LI, Gerfen CR, Sabatini BL, Arnold DB. ATLAS: a rationally designed anterograde transsynaptic tracer. Nat Methods 2025; 22:1101-1111. [PMID: 40312509 PMCID: PMC12074993 DOI: 10.1038/s41592-025-02670-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/11/2025] [Indexed: 05/03/2025]
Abstract
Genetically modified rabies virus can map neural circuits retrogradely from genetically determined cells. However, similar tools for anterograde tracing are not available. Here, we describe a method for anterograde transsynaptic tracing from genetically determined neurons based on a rationally designed protein, ATLAS. Expression of ATLAS in neurons causes presynaptic release of a payload composed of an antibody-like protein, AMPA.FingR, which binds to the N terminus of GluA1, and a recombinase. In the synaptic cleft, AMPA.FingR binds to GluA1, causing the payload to be endocytosed into postsynaptic cells and delivered to the nucleus, where it triggers expression of a recombinase-dependent reporter. In mice, ATLAS mediates monosynaptic transneuronal tracing from random or genetically determined cells that is strictly anterograde, synaptic and nontoxic. Moreover, ATLAS-mediated tracing shows activity dependence, suggesting that it can label active circuits underlying specific behaviors. Finally, ATLAS is composed of modular components that can be independently replaced or modified.
Collapse
Affiliation(s)
- Jacqueline F Rivera
- Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA
| | - Haoyang Huang
- Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Weiguang Weng
- Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA
| | - Heesung Sohn
- Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Allison E Girasole
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Shun Li
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Madeline A Albanese
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Melissa Qin
- Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA
| | - Can Tao
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Molly E Klug
- Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA
| | - Sadhna Rao
- Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Ronald Paletzki
- Section on Neuroanatomy, National Institute of Mental Health, Bethesda, MD, USA
| | - Bruce E Herring
- Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Scott E Kanoski
- Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Li I Zhang
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Charles R Gerfen
- Section on Neuroanatomy, National Institute of Mental Health, Bethesda, MD, USA
| | - Bernardo L Sabatini
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Don B Arnold
- Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA.
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA.
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Kennedy L, Morland C, Narum M, Bergersen LH, Rinholm JE. Lactate Receptor HCAR1 Affects Axonal Development and Contributes to Lactate's Protection of Axons and Myelin in Experimental Neonatal Hypoglycemia. eNeuro 2025; 12:ENEURO.0563-24.2025. [PMID: 40345852 PMCID: PMC12124765 DOI: 10.1523/eneuro.0563-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 05/11/2025] Open
Abstract
Lactate plays an important role in brain energy metabolism. It contributes to normal brain development and to neuroprotection in diabetic hypoglycemia, but its role in neonatal hypoglycemia is unclear. Moreover, lactate can work as a signaling substance via the lactate receptor HCAR1 (Hydroxycarboxylic acid receptor 1). Recent studies indicate that HCAR1 is protective in mouse models of neonatal hypoxic ischemia and has a role in metabolic regulation in glial cells during hypoglycemia. Here we have studied potential impacts of HCAR1 on axonal and myelin development in the cerebral cortex and corpus callosum of young (P21) wild-type (WT) mice and HCAR1 KO mice and in cortical organotypic brain slice cultures. The HCAR1 KO mice showed lower axonal area relative to WT in both cortex and corpus callosum. However, the myelin area was unaffected by HCAR1 KO. Using particle and colocalization analysis, we show that HCAR1 KO predominantly reduces axonal size in unmyelinated axons. Using an organotypic brain slice model of neonatal hypoglycemia, we find that lactate protects both axonal and myelin development in hypoglycemia, partially via HCAR1. Lastly, live imaging with a pH-sensitive dye on acute cortical brain slices indicates that cellular lactate uptake is influenced by HCAR1. In conclusion, our findings support a role of HCAR1 in axonal development and in lactate's protective effects in hypoglycemia.
Collapse
Affiliation(s)
- Lauritz Kennedy
- Department of Microbiology, Oslo University Hospital and University of Oslo, Oslo 0373, Norway
- Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo 0372, Norway
| | - Cecilie Morland
- Department of Pharmacy, The Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo 0371, Norway
| | - Martine Narum
- Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo 0372, Norway
| | - Linda H Bergersen
- The Brain and Muscle Energy Group, Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo 0372, Norway
- King Abdullah University of Science and Technology, Kaust Smart-Health, Thuwal 23955, Saudi Arabia
| | - Johanne E Rinholm
- Department of Microbiology, Oslo University Hospital and University of Oslo, Oslo 0373, Norway
- Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo 0372, Norway
| |
Collapse
|
5
|
Simões‐Pires EN, Torrente D, Singh P, Strickland S, Norris EH. Synergistic effects of the Aβ/fibrinogen complex on synaptotoxicity, neuroinflammation, and blood-brain barrier damage in Alzheimer's disease models. Alzheimers Dement 2025; 21:e70119. [PMID: 40344319 PMCID: PMC12061846 DOI: 10.1002/alz.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/16/2024] [Accepted: 01/12/2025] [Indexed: 05/11/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) is characterized by amyloid-beta (Aβ), hyperphosphorylated tau, chronic neuroinflammation, blood-brain barrier (BBB) damage, and synaptic dysfunction, leading to neuronal loss and cognitive deficits. Vascular proteins, including fibrinogen, extravasate into the brain, further contributing to damage and inflammation. Fibrinogen's interaction with Aβ is well-established, but how this interaction contributes to synaptic dysfunction in AD is unknown. METHODS Organotypic hippocampal cultures (OHC) were exposed to Aβ42 oligomers, fibrinogen, or Aβ42/fibrinogen complexes. Synaptotoxicity was analyzed by Western blot. Aβ42 oligomers, fibrinogen, or their complexes were intracerebroventricularly injected into mice. Histopathological AD markers, synaptotoxicity, neuroinflammation, and vascular markers were observed by Western blot and immunofluorescence. RESULTS Aβ42/fibrinogen complexes led to synaptic loss, tau181 phosphorylation, neuroinflammation, and BBB disruption, independent of Mac1/CD11b receptor signaling. Blocking Aβ42/fibrinogen complex formation prevented synaptotoxicity. DISCUSSION These findings indicate that the Aβ42/fibrinogen complex has a synergistic impact on hippocampal synaptotoxicity and neuroinflammation. HIGHLIGHTS Fibrinogen binds to the central region of Aβ, forming a plasmin-resistant complex. The Aβ/fibrinogen complex induces synaptotoxicity, inflammation, and BBB disruption. Synaptotoxicity induced by the complex is independent of Mac1 receptor signaling.
Collapse
Affiliation(s)
- Elisa Nicoloso Simões‐Pires
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Daniel Torrente
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Pradeep Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| |
Collapse
|
6
|
Ziebarth T, Pape N, Nelson JS, van Alphen FI, Kalia M, Meijer HG, Rose CR, Reiner A. Atypical plume-like events contribute to glutamate accumulation in metabolic stress conditions. iScience 2025; 28:112256. [PMID: 40241754 PMCID: PMC12002667 DOI: 10.1016/j.isci.2025.112256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 12/02/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Neural glutamate homeostasis is important for health and disease. Ischemic conditions, like stroke, cause imbalances in glutamate release and uptake due to energy depletion and depolarization. We here used the glutamate sensor SF-iGluSnFR(A184V) to probe how chemical ischemia affects the extracellular glutamate dynamics in slice cultures from mouse cortex. SF-iGluSnFR imaging showed spontaneous glutamate release indicating synchronous network activity, similar to calcium imaging with GCaMP6f. Glutamate imaging further revealed local, atypically large, and long-lasting plume-like release events. Plumes occurred with low frequency, independent of network activity, and persisted in tetrodotoxin (TTX). Blocking glutamate uptake with TFB-TBOA favored plumes, whereas blocking ionotropic glutamate receptors (iGluRs) suppressed plumes. During chemical ischemia plumes became more pronounced, overly abundant and contributed to large-scale glutamate accumulation. Similar plumes were previously observed in cortical spreading depression and migraine models, and they may thus be a more general consequence of glutamate uptake dysfunctions in neurological and neurodegenerative diseases.
Collapse
Affiliation(s)
- Tim Ziebarth
- Department of Biology and Biotechnology, Ruhr University Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| | - Nils Pape
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Joel S.E. Nelson
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Fleur I.M. van Alphen
- Department of Applied Mathematics, University of Twente, Drienerlolaan 5, 7522 NB Enschede, the Netherlands
| | - Manu Kalia
- Department of Applied Mathematics, University of Twente, Drienerlolaan 5, 7522 NB Enschede, the Netherlands
| | - Hil G.E. Meijer
- Department of Applied Mathematics, University of Twente, Drienerlolaan 5, 7522 NB Enschede, the Netherlands
| | - Christine R. Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Andreas Reiner
- Department of Biology and Biotechnology, Ruhr University Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| |
Collapse
|
7
|
McNally MA, Lau LA, Granak S, Hike D, Liu X, Yu X, Donahue RA, Chibnik LB, Ortiz JV, Che A, Chavez-Valdez R, Northington FJ, Staley KJ. Ongoing loss of viable neurons for weeks after mild hypoxia-ischaemia. Brain Commun 2025; 7:fcaf153. [PMID: 40297712 PMCID: PMC12034461 DOI: 10.1093/braincomms/fcaf153] [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: 12/17/2024] [Revised: 03/25/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Mild hypoxic-ischaemic encephalopathy is common in neonates, and there are no evidence-based therapies. By school age, 30-40% of those patients experience adverse neurodevelopmental outcomes. The nature and progression of mild injury is poorly understood. We studied the evolution of mild perinatal brain injury using longitudinal two-photon imaging of transgenic fluorescent calcium-sensitive and insensitive proteins to provide a novel readout of neuronal viability and activity at cellular resolution in vitro and in vivo. In vitro, perinatal organotypic hippocampal cultures underwent 15-20 min of oxygen-glucose deprivation. In vivo, mild hypoxia-ischaemia was completed at post-natal day 10 with carotid ligation and 15 min of hypoxia (FiO2, 0.08). Consistent with a mild injury, minimal immediate neuronal death was seen in vitro or in vivo, and there was no volumetric evidence of injury by ex vivo MRI 2.5 weeks after injury (n = 3 pups/group). However, in both the hippocampus and neocortex, these mild injuries resulted in delayed and progressive neuronal loss by the second week after injury compared to controls; measured by fluorophore quenching (n = 6 slices/group in vitro, P < 0.001; n = 8 pups/group in vivo, P < 0.01). Mild hypoxia-ischaemia transiently suppressed cortical network calcium activity in vivo for over 2 h after injury (versus sham, n = 13 pups/group; P < 0.01). No post-injury seizures were seen. By 24 h, network activity fully recovered, and there was no disruption in the development of normal cortical activity for 11 days (n = 8 pups/group). The participation in network activity of individual neurons destined to die in vivo was indistinguishable from those that survived up to 4 days post-injury (n = 8 pups/group). Despite a lack of significant immediate neuronal death and only transient disruptions of network activity, mild perinatal brain injury resulted in a delayed and progressive increase of neuronal death in the hippocampus and neocortex. Neurons that died late were functioning normally for days after injury, suggesting a new pathophysiology of neuronal death after mild injury. Critically, the neurons destined to die late demonstrated multiple biomarkers of viability long after mild injury, suggesting their later death may be modified with neuroprotective interventions.
Collapse
Affiliation(s)
- Melanie A McNally
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lauren A Lau
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| | - Simon Granak
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| | - David Hike
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02129, USA
| | - Xiaochen Liu
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02129, USA
| | - Xin Yu
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02129, USA
| | - Rachel A Donahue
- Department of Medicine, Biostatics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lori B Chibnik
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - John V Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Alicia Che
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Raul Chavez-Valdez
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Frances J Northington
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kevin J Staley
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
8
|
Trejos AJ, Schanne AXF. Evaluation of 5 Intermediate Structural Variations of Microglia Within an Organotypic Hippocampal Slice Model After Regionalized Toxic Injury. Cell Mol Neurobiol 2025; 45:34. [PMID: 40205019 PMCID: PMC11981971 DOI: 10.1007/s10571-025-01545-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 03/16/2025] [Indexed: 04/11/2025]
Abstract
The dendritic cell of the CNS, the microglia (MG), is an initiation point of the immunological response within the post-blood-brain barrier (BBB) compartment. Microglia drastically changes in response to cell stress to a much different non-dendritic morphologies. This investigation postulates that if the first MG responses to toxic injury are isolated and studied in greater morphological detail, there is much to be learned about microglia's metamorphosis from and M2 to an M1 state. The organotypic hippocampal slice was the experimental setting used to investigate microglial response to toxic injury; this isolates dendritic cell to post-BBB cells dynamics from the impact of nonspecific of in vivo blood-derived signaling. Within the context of biochemically verified precise toxic cell injury/death (induced with mercury or cyanide in combination with 2-deoxy-glucose) to a specific region within the hippocampal slice, MG's morphological response was evaluated. There was up to 35% increase in microglia activation proximally to injury (CA3 region) and no changes distally (DG region) when compared to control slices treated with PBS. Maximum microglia activation consisted of a 3 plus-fold increase in the distance between the nucleus membrane and the cell membrane, which underscores an extensive and quantifiable amount of membrane rearrangement. This quantification can be applied to contemporaneous AI image analysis algorithms to demarcate and quantify relative MG activation in and around a site of injury. In between baseline and activated MG morphologies, 5 intermediate morphologies (or structural variations) are described as it relates to its cell body, nucleus, and dendrites. The result from this study reconciles details of MG's structure to its holistic characteristics in relation to parenchymal cell stress.
Collapse
Affiliation(s)
- A Jesus Trejos
- St. John's University Queens, Jamaica, NY, USA.
- Regeneron Pharmaceuticals, Tarrytown, NY, USA.
| | | |
Collapse
|
9
|
Liu B, Buonomano DV. Ex vivo cortical circuits learn to predict and spontaneously replay temporal patterns. Nat Commun 2025; 16:3179. [PMID: 40185714 PMCID: PMC11971321 DOI: 10.1038/s41467-025-58013-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 03/07/2025] [Indexed: 04/07/2025] Open
Abstract
It has been proposed that prediction and timing are computational primitives of neocortical microcircuits, specifically, that neural mechanisms are in place to allow neocortical circuits to autonomously learn the temporal structure of external stimuli and generate internal predictions. To test this hypothesis, we trained cortical organotypic slices on two temporal patterns using dual-optical stimulation. After 24-h of training, whole-cell recordings revealed network dynamics consistent with training-specific timed prediction. Unexpectedly, there was replay of the learned temporal structure during spontaneous activity. Furthermore, some neurons exhibited timed prediction errors as revealed by larger responses when the expected stimulus was omitted compared to when it was present. Mechanistically our results indicate that learning relied in part on asymmetric connectivity between distinct neuronal ensembles with temporally-ordered activation. These findings further suggest that local cortical microcircuits are intrinsically capable of learning temporal information and generating predictions, and that the learning rules underlying temporal learning and spontaneous replay can be intrinsic to local cortical microcircuits and not necessarily dependent on top-down interactions.
Collapse
Affiliation(s)
- Benjamin Liu
- Department of Neurobiology, Deparment of Psychology, and Psychology, Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Dean V Buonomano
- Department of Neurobiology, Deparment of Psychology, and Psychology, Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Petshow S, Coblentz A, Hamilton AM, Sarkar D, Anisimova M, Flores JC, Zito K. Activity-dependent regulation of Cdc42 by Ephexin5 drives synapse growth and stabilization. SCIENCE ADVANCES 2025; 11:eadp5782. [PMID: 40138406 PMCID: PMC11939064 DOI: 10.1126/sciadv.adp5782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 02/19/2025] [Indexed: 03/29/2025]
Abstract
Synaptic Rho guanosine triphosphatase (GTPase) guanine nucleotide exchange factors (RhoGEFs) play vital roles in regulating the activity-dependent neuronal plasticity that is critical for learning. Ephexin5, a RhoGEF implicated in the etiology of Alzheimer's disease and Angelman syndrome, was originally reported in neurons as a RhoA-specific GEF that negatively regulates spine synapse density. Here, we show that Ephexin5 activates both RhoA and Cdc42 in the brain. Furthermore, using live imaging of GTPase biosensors, we demonstrate that Ephexin5 regulates activity-dependent Cdc42, but not RhoA, signaling at single synapses. The selectivity of Ephexin5 for Cdc42 activation is regulated by tyrosine phosphorylation, which is regulated by neuronal activity. Last, in contrast to Ephexin5's role in negatively regulating synapse density, we show that, downstream of neuronal activity, Ephexin5 positively regulates synaptic growth and stabilization. Our results support a model in which plasticity-inducing neuronal activity regulates Ephexin5 tyrosine phosphorylation, driving Ephexin5-mediated activation of Cdc42 and the spine structural growth and stabilization vital for learning.
Collapse
Affiliation(s)
- Samuel Petshow
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Azariah Coblentz
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Andrew M. Hamilton
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Dipannita Sarkar
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Margarita Anisimova
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Juan C. Flores
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Karen Zito
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| |
Collapse
|
11
|
Ahmed AA, Alegret N, Almeida B, Alvarez-Puebla R, Andrews AM, Ballerini L, Barrios-Capuchino JJ, Becker C, Blick RH, Bonakdar S, Chakraborty I, Chen X, Cheon J, Chilla G, Coelho Conceicao AL, Delehanty J, Dulle M, Efros AL, Epple M, Fedyk M, Feliu N, Feng M, Fernández-Chacón R, Fernandez-Cuesta I, Fertig N, Förster S, Garrido JA, George M, Guse AH, Hampp N, Harberts J, Han J, Heekeren HR, Hofmann UG, Holzapfel M, Hosseinkazemi H, Huang Y, Huber P, Hyeon T, Ingebrandt S, Ienca M, Iske A, Kang Y, Kasieczka G, Kim DH, Kostarelos K, Lee JH, Lin KW, Liu S, Liu X, Liu Y, Lohr C, Mailänder V, Maffongelli L, Megahed S, Mews A, Mutas M, Nack L, Nakatsuka N, Oertner TG, Offenhäusser A, Oheim M, Otange B, Otto F, Patrono E, Peng B, Picchiotti A, Pierini F, Pötter-Nerger M, Pozzi M, Pralle A, Prato M, Qi B, Ramos-Cabrer P, Genger UR, Ritter N, Rittner M, Roy S, Santoro F, Schuck NW, Schulz F, Şeker E, Skiba M, Sosniok M, Stephan H, Wang R, Wang T, Wegner KD, Weiss PS, Xu M, Yang C, Zargarian SS, Zeng Y, Zhou Y, Zhu D, Zierold R, Parak WJ. Interfacing with the Brain: How Nanotechnology Can Contribute. ACS NANO 2025; 19:10630-10717. [PMID: 40063703 PMCID: PMC11948619 DOI: 10.1021/acsnano.4c10525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 03/26/2025]
Abstract
Interfacing artificial devices with the human brain is the central goal of neurotechnology. Yet, our imaginations are often limited by currently available paradigms and technologies. Suggestions for brain-machine interfaces have changed over time, along with the available technology. Mechanical levers and cable winches were used to move parts of the brain during the mechanical age. Sophisticated electronic wiring and remote control have arisen during the electronic age, ultimately leading to plug-and-play computer interfaces. Nonetheless, our brains are so complex that these visions, until recently, largely remained unreachable dreams. The general problem, thus far, is that most of our technology is mechanically and/or electrically engineered, whereas the brain is a living, dynamic entity. As a result, these worlds are difficult to interface with one another. Nanotechnology, which encompasses engineered solid-state objects and integrated circuits, excels at small length scales of single to a few hundred nanometers and, thus, matches the sizes of biomolecules, biomolecular assemblies, and parts of cells. Consequently, we envision nanomaterials and nanotools as opportunities to interface with the brain in alternative ways. Here, we review the existing literature on the use of nanotechnology in brain-machine interfaces and look forward in discussing perspectives and limitations based on the authors' expertise across a range of complementary disciplines─from neuroscience, engineering, physics, and chemistry to biology and medicine, computer science and mathematics, and social science and jurisprudence. We focus on nanotechnology but also include information from related fields when useful and complementary.
Collapse
Affiliation(s)
- Abdullah
A. A. Ahmed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Department
of Physics, Faculty of Applied Science, Thamar University, Dhamar 87246, Yemen
| | - Nuria Alegret
- Biogipuzkoa
HRI, Paseo Dr. Begiristain
s/n, 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bethany Almeida
- Department
of Chemical and Biomolecular Engineering, Clarkson University, Potsdam, New York 13699, United States
| | - Ramón Alvarez-Puebla
- Universitat
Rovira i Virgili, 43007 Tarragona, Spain
- ICREA, 08010 Barcelona, Spain
| | - Anne M. Andrews
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- Neuroscience
Interdepartmental Program, University of
California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience
& Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Laura Ballerini
- Neuroscience
Area, International School for Advanced
Studies (SISSA/ISAS), Trieste 34136, Italy
| | | | - Charline Becker
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Robert H. Blick
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Shahin Bonakdar
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- National
Cell Bank Department, Pasteur Institute
of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Indranath Chakraborty
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Nano Science and Technology, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Xiaodong Chen
- Innovative
Center for Flexible Devices (iFLEX), Max Planck − NTU Joint
Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jinwoo Cheon
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
- Department
of Chemistry, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Gerwin Chilla
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - James Delehanty
- U.S. Naval
Research Laboratory, Washington, D.C. 20375, United States
| | - Martin Dulle
- JCNS-1, Forschungszentrum
Jülich, 52428 Jülich, Germany
| | | | - Matthias Epple
- Inorganic
Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, 45117 Essen, Germany
| | - Mark Fedyk
- Center
for Neuroengineering and Medicine, UC Davis, Sacramento, California 95817, United States
| | - Neus Feliu
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Miao Feng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Rafael Fernández-Chacón
- Instituto
de Biomedicina de Sevilla (IBiS), Hospital
Universitario Virgen del Rocío/Consejo Superior de Investigaciones
Científicas/Universidad de Sevilla, 41013 Seville, Spain
- Departamento
de Fisiología Médica y Biofísica, Facultad de
Medicina, Universidad de Sevilla, CIBERNED,
ISCIII, 41013 Seville, Spain
| | | | - Niels Fertig
- Nanion
Technologies GmbH, 80339 München, Germany
| | | | - Jose A. Garrido
- ICREA, 08010 Barcelona, Spain
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
| | | | - Andreas H. Guse
- The Calcium
Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Norbert Hampp
- Fachbereich
Chemie, Universität Marburg, 35032 Marburg, Germany
| | - Jann Harberts
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Drug Delivery,
Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node
of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
| | - Jili Han
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Hauke R. Heekeren
- Executive
University Board, Universität Hamburg, 20148 Hamburg Germany
| | - Ulrich G. Hofmann
- Section
for Neuroelectronic Systems, Department for Neurosurgery, University Medical Center Freiburg, 79108 Freiburg, Germany
- Faculty
of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Malte Holzapfel
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | | | - Yalan Huang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Patrick Huber
- Institute
for Materials and X-ray Physics, Hamburg
University of Technology, 21073 Hamburg, Germany
- Center
for X-ray and Nano Science CXNS, Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Taeghwan Hyeon
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Sven Ingebrandt
- Institute
of Materials in Electrical Engineering 1, RWTH Aachen University, 52074 Aachen, Germany
| | - Marcello Ienca
- Institute
for Ethics and History of Medicine, School of Medicine and Health, Technische Universität München (TUM), 81675 München, Germany
| | - Armin Iske
- Fachbereich
Mathematik, Universität Hamburg, 20146 Hamburg, Germany
| | - Yanan Kang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Dae-Hyeong Kim
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kostas Kostarelos
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
- Centre
for Nanotechnology in Medicine, Faculty of Biology, Medicine &
Health and The National Graphene Institute, University of Manchester, Manchester M13 9PL, United
Kingdom
| | - Jae-Hyun Lee
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Kai-Wei Lin
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sijin Liu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yang Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Christian Lohr
- Fachbereich
Biologie, Universität Hamburg, 20146 Hamburg, Germany
| | - Volker Mailänder
- Department
of Dermatology, Center for Translational Nanomedicine, Universitätsmedizin der Johannes-Gutenberg,
Universität Mainz, 55131 Mainz, Germany
- Max Planck
Institute for Polymer Research, Ackermannweg 10, 55129 Mainz, Germany
| | - Laura Maffongelli
- Institute
of Medical Psychology, University of Lübeck, 23562 Lübeck, Germany
| | - Saad Megahed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Physics
Department, Faculty of Science, Al-Azhar
University, 4434104 Cairo, Egypt
| | - Alf Mews
- Fachbereich
Chemie, Universität Hamburg, 20146 Hamburg, Germany
| | - Marina Mutas
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Leroy Nack
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Nako Nakatsuka
- Laboratory
of Chemical Nanotechnology (CHEMINA), Neuro-X
Institute, École Polytechnique Fédérale de Lausanne
(EPFL), Geneva CH-1202, Switzerland
| | - Thomas G. Oertner
- Institute
for Synaptic Neuroscience, University Medical
Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andreas Offenhäusser
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Martin Oheim
- Université
Paris Cité, CNRS, Saints Pères
Paris Institute for the Neurosciences, 75006 Paris, France
| | - Ben Otange
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Ferdinand Otto
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Enrico Patrono
- Institute
of Physiology, Czech Academy of Sciences, Prague 12000, Czech Republic
| | - Bo Peng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Filippo Pierini
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Monika Pötter-Nerger
- Head and
Neurocenter, Department of Neurology, University
Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Pozzi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Arnd Pralle
- University
at Buffalo, Department of Physics, Buffalo, New York 14260, United States
| | - Maurizio Prato
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Department
of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bing Qi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Life Sciences, Southern University of
Science and Technology, Shenzhen, 518055, China
| | - Pedro Ramos-Cabrer
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Ute Resch Genger
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Norbert Ritter
- Executive
Faculty Board, Faculty for Mathematics, Informatics and Natural Sciences, Universität Hamburg, 20345 Hamburg, Germany
| | - Marten Rittner
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sathi Roy
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
- Department
of Mechanical Engineering, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Francesca Santoro
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
- Faculty
of Electrical Engineering and Information Technology, RWTH Aachen, 52074 Aachen, Germany
| | - Nicolas W. Schuck
- Institute
of Psychology, Universität Hamburg, 20146 Hamburg, Germany
- Max Planck
Research Group NeuroCode, Max Planck Institute
for Human Development, 14195 Berlin, Germany
- Max Planck
UCL Centre for Computational Psychiatry and Ageing Research, 14195 Berlin, Germany
| | - Florian Schulz
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Erkin Şeker
- University
of California, Davis, Davis, California 95616, United States
| | - Marvin Skiba
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Martin Sosniok
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Holger Stephan
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, 01328 Dresden, Germany
| | - Ruixia Wang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Ting Wang
- State Key
Laboratory of Organic Electronics and Information Displays & Jiangsu
Key Laboratory for Biosensors, Institute of Advanced Materials (IAM),
Jiangsu National Synergetic Innovation Center for Advanced Materials
(SICAM), Nanjing University of Posts and
Telecommunications, Nanjing 210023, China
| | - K. David Wegner
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Paul S. Weiss
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Materials Science and Engineering, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| | - Ming Xu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chenxi Yang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Seyed Shahrooz Zargarian
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Yuan Zeng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yaofeng Zhou
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Dingcheng Zhu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- College
of Material, Chemistry and Chemical Engineering, Key Laboratory of
Organosilicon Chemistry and Material Technology, Ministry of Education,
Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, China
| | - Robert Zierold
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | |
Collapse
|
12
|
Arizono M, Idziak A, Nägerl UV. Live STED imaging of functional neuroanatomy. Nat Protoc 2025:10.1038/s41596-024-01132-6. [PMID: 40087378 DOI: 10.1038/s41596-024-01132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 12/11/2024] [Indexed: 03/17/2025]
Abstract
In the mammalian brain, a large network of excitable and modulatory cells efficiently processes, analyzes and stores vast amounts of information. The brain's anatomy influences the flow of neural information between neurons and glia, from which all thought, emotion and action arises. Consequently, one of the grand challenges in neuroscience is to uncover the finest structural details of the brain in the context of its overall architecture. Recent developments in microscopy and biosensors have enabled the investigation of brain microstructure and function with unprecedented specificity and resolution, dendritic spines being an exemplary case, which has provided deep insights into neuronal mechanisms of higher brain function, such as learning and memory. As diffraction-limited light microscopy methods cannot resolve the fine details of brain cells (the 'anatomical ground truth'), electron microscopy is used instead to contextualize functional signals. This approach can be quite unsatisfying given the fragility and dynamic nature of the structures under investigation. We have recently developed a method for combining super-resolution stimulated emission depletion microscopy with functional measurements in brain slices, offering nanoscale resolution in functioning brain structures. We describe how to concurrently perform morphological and functional imaging with a confocal STED microscope. Specifically, the procedure guides the user on how to record astrocytic Ca2+ signals at tripartite synapses, outlining a framework for analyzing structure-function relationships of brain cells at nanoscale resolution. The imaging requires 2-3 h and the image analysis between 2 h and 2 d.
Collapse
Affiliation(s)
- Misa Arizono
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, CNRS UMR 5297, Bordeaux, France.
- The Hakubi Center for Advanced Research, Kyoto University, Kyoto, Japan.
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Agata Idziak
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, CNRS UMR 5297, Bordeaux, France
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - U Valentin Nägerl
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, CNRS UMR 5297, Bordeaux, France.
- Department of Anatomy and Cell Biology, University Medical Center, Georg-August-University of Göttingen, Göttingen, Germany.
| |
Collapse
|
13
|
Radoszkiewicz K, Bzinkowska A, Sypecka M, Sulejczak D, Ferrari D, Gelati M, Luigi Vescovi A, Sarnowska A. Unraveling the impact of human cerebrospinal fluid on human neural stem cell fate. Front Cell Dev Biol 2025; 13:1527557. [PMID: 40162096 PMCID: PMC11950821 DOI: 10.3389/fcell.2025.1527557] [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: 11/13/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Human neural stem/progenitor cells (hNSCs) can potentially treat neurological diseases, but their low survival and proliferation rates after transplantation remain challenging. In our study, we preincubated hNSCs with the human cerebrospinal fluid (CSF) to obtain closer to the physiological brain environment and to assess NSC fate and their therapeutic abilities in vitro, ex vivo, and in vivo. We observed significant changes in the differentiation, migratory, and secretory potential of CSF-treated hNSCs, as well as their elevated neuroprotective potential after co-culture with ischemically damaged by oxygen-glucose deprivation (OGD) organotypic rat hippocampal slices culture (OHC) in comparison to the cells cultured in the standard conditions. Next, we investigated their survival and anti-inflammatory abilities in an in vivo ouabain-induced stroke model. This study highlighted and confirmed the critical importance of nutritional supplementation in maintaining NSC culture and enhancing its therapeutic properties.
Collapse
Affiliation(s)
- Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Aleksandra Bzinkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Monika Sypecka
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Daniela Ferrari
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Maurizio Gelati
- UPTA Unit Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Angelo Luigi Vescovi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
- IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
14
|
Toti A, Lucarini E, Ferrara V, Parisio C, Ciampi C, Gerace E, Micheli L, Margiotta F, Venturi D, Mello T, Lacal PM, Graziani G, Mannaioni G, Ghelardini C, Di Cesare Mannelli L. The dual role of VEGF-A in a complex in vitro model of oxaliplatin-induced neurotoxicity: Pain-related and neuroprotective effects. Neurotherapeutics 2025; 22:e00532. [PMID: 39939241 PMCID: PMC12014407 DOI: 10.1016/j.neurot.2025.e00532] [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: 08/13/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/14/2025] Open
Abstract
Vascular endothelial growth factor (VEGF)-A is a main player in the development of neuropathic pain induced by chemotherapy and the pharmacological blockade of VEGF receptor (VEGFR) subtype 1 is a pain killer strategy. Interestingly, VEGF-A has been demonstrated to have also neuroprotective properties. The aim of the study was to investigate the neuroprotective role of VEGF-A against oxaliplatin neurotoxicity, attempting to discriminate pain-related and restorative signaling pathways. We used rat organotypic spinal cord slices treated with oxaliplatin, as an in vitro model to study chemotherapy-induced toxicity. In this model, 10 μM oxaliplatin caused a time-dependent release of VEGF-A, which was reduced by the astrocyte inhibitor fluorocitrate. Moreover, glia inhibition exacerbated oxaliplatin-induced cytotoxicity in a VEGF-A sensitive manner. Treatment with VEGF165b, the main isoform of VEGF-A, prevented the oxaliplatin-induced neuronal damage (indicated by NeuN staining) and astrocyte activation (indicated by GFAP staining). In addition, the blockade of VEGFR-2 by the selective antibody DC101 blunted the protective action of VEGF165b. In the same model, VEGF165b increased the release of molecules relevant in pain signaling, like substance P and CGRP, as well as the mRNA expression of glutamate transporters (EAAT1 and EAAT2), similarly to oxaliplatin and these effects were prevented by the selective VEGFR-1 blocker antibody D16F7. In conclusion, VEGF-A plays a dichotomic role in an in vitro model of chemotherapy-induced toxicity, either promoting neuroprotection or triggering pain mediators release, depending on which of its two receptors is activated. The selective management of VEGF-A signaling is suggested as a therapeutic approach.
Collapse
Affiliation(s)
- A Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - E Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - V Ferrara
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - C Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - C Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - E Gerace
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy; Department of Health Sciences, University of Florence, Florence, Italy
| | - L Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy.
| | - F Margiotta
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - D Venturi
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - T Mello
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - G Graziani
- Department of Systems Medicine, Pharmacology Section, University of Rome Tor Vergata, Rome, Italy
| | - G Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - C Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - L Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
15
|
Reinders NR, van der Spek SJF, Klaassen RV, Koymans KJ, MacGillavry HD, Smit AB, Kessels HW. Amyloid-β-Driven Synaptic Deficits Are Mediated by Synaptic Removal of GluA3-Containing AMPA Receptors. J Neurosci 2025; 45:e0393242024. [PMID: 39779375 PMCID: PMC11867010 DOI: 10.1523/jneurosci.0393-24.2024] [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: 02/28/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
The detrimental effects of oligomeric amyloid-β (Aβ) on synapses are considered the leading cause for cognitive deficits in Alzheimer's disease. However, through which mechanism Aβ oligomers impair synaptic structure and function remains unknown. Here, we used electrophysiology and amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) imaging on mouse and rat neurons to demonstrate that GluA3 expression in neurons lacking GluA3 is sufficient to resensitize their synapses to the damaging effects of Aβ, indicating that GluA3-containing AMPARs at synapses are necessary and sufficient for Aβ to induce synaptic deficits. We found that Aβ oligomers trigger the endocytosis of GluA3 and promote its translocation toward endolysosomal compartments for degradation. Mechanistically, these Aβ-driven effects critically depend on the PDZ-binding motif of GluA3. A single point mutation in the GluA3 PDZ-binding motif prevented Aβ-driven effects and rendered synapses fully resistant to the effects of Aβ. Correspondingly, proteomics on synaptosome fractions from APP/PS1-transgenic mice revealed a selective reduction of GluA3 at an early age. These findings support a model where the endocytosis and lysosomal degradation of GluA3-containing AMPARs are a critical early step in the cascade of events through which Aβ accumulation causes a loss of synapses.
Collapse
Affiliation(s)
- Niels R Reinders
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105 BA, The Netherlands
- Swammerdam Institute of Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam 1098 XH, The Netherlands
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Sophie J F van der Spek
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Remco V Klaassen
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Karin J Koymans
- Swammerdam Institute of Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam 1098 XH, The Netherlands
| | - Harold D MacGillavry
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - August B Smit
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Helmut W Kessels
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105 BA, The Netherlands
- Swammerdam Institute of Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam 1098 XH, The Netherlands
| |
Collapse
|
16
|
Steindl A, Valiente M. Potential of ex vivo organotypic slice cultures in neuro-oncology. Neuro Oncol 2025; 27:338-351. [PMID: 39504579 PMCID: PMC11812025 DOI: 10.1093/neuonc/noae195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Over recent decades, in vitro and in vivo models have significantly advanced brain cancer research; however, each presents distinct challenges for accurately mimicking in situ conditions. In response, organotypic slice cultures have emerged as a promising model recapitulating precisely specific in vivo phenotypes through an ex vivo approach. Ex vivo organotypic brain slice models can integrate biological relevance and patient-specific variability early in drug discovery, thereby aiming for more precise treatment stratification. However, the challenges of obtaining representative fresh brain tissue, ensuring reproducibility, and maintaining essential central nervous system (CNS)-specific conditions reflecting the in situ situation over time have limited the direct application of ex vivo organotypic slice cultures in robust clinical trials. In this review, we explore the benefits and possible limitations of ex vivo organotypic brain slice cultures in neuro-oncological research. Additionally, we share insights from clinical experts in neuro-oncology on how to overcome these current limitations and improve the practical application of organotypic brain slice cultures beyond academic research.
Collapse
Affiliation(s)
- Ariane Steindl
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
17
|
Hanauske T, Koretz CC, Jungenitz T, Roeper J, Drakew A, Deller T. Electrophysiologically calibrated optogenetic stimulation of dentate granule cells mitigates dendritic spine loss in denervated organotypic entorhino-hippocampal slice cultures. Sci Rep 2025; 15:4563. [PMID: 39915664 PMCID: PMC11802742 DOI: 10.1038/s41598-025-88536-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Organotypic slice cultures (OTCs) are versatile tools for studying long-term structure-function relationships of neurons within a defined network (e.g. hippocampus). We developed a method for repeated experimenter-controlled activation of hippocampal granule cells (GCs) in OTCs within the incubator. After several days of contact-free photonic stimulation, we were able to ameliorate entorhinal denervation-induced structural damage in GCs. To achieve this outcome, we had to calibrate the intensity and duration of optogenetic (light) pulses using whole-cell electrophysiological recordings and multi-cell calcium imaging. Our findings showed that ChR2-expressing cells generated action potentials (APs) or calcium transients in response to illumination but were otherwise functionally indistinguishable from non-transduced GCs within the same neural circuit. However, the threshold for AP firing in single GCs varied based on the stimulus light intensity and the expression levels of ChR2. This information allowed us to calibrate light intensity for chronic stimulations. Denervated GCs exhibited significant spine loss four days post-denervation, but this detrimental effect was mitigated when AP firing was induced at a physiological GC bursting rate. Phototoxic damage caused by chronic light exposure was significantly reduced if illuminated with longer wavelength and by adding antioxidants to the culture medium. Our study presents a versatile approach for concurrent non-invasive manipulation and observation of neural circuit activity and remodeling in vitro.
Collapse
Affiliation(s)
- Tijana Hanauske
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| | - Carolin Christina Koretz
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
- Institute for Neurophysiology, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Tassilo Jungenitz
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Jochen Roeper
- Institute for Neurophysiology, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Alexander Drakew
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Thomas Deller
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| |
Collapse
|
18
|
de Sousa VM, Almeida ÁMAN, Ferreira RS, dos Santos BL, da Silva VDA, David JM, dos Santos CC, Costa SL. The Flavonoid Agathisflavone Attenuates Glia Activation After Mechanical Injury of Cortical Tissue and Negatively Regulates Both NRLP3 and IL-1β Expression. Int J Mol Sci 2025; 26:1275. [PMID: 39941042 PMCID: PMC11818122 DOI: 10.3390/ijms26031275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Traumatic brain injury (TBI) has a complex and multifactorial pathology and is a major cause of death and disability for humans. Immediately after TBI, astrocytes and microglia react with complex morphological and functional changes known as reactive gliosis to form a glial scar in the area immediately adjacent to the lesion, which is the major barrier to neuronal regeneration. The flavonoid agathisflavone (bis-apigenin), present in Poincianella pyramidalis leaves, has been shown to have neuroprotective, neurogenic, and anti-inflammatory effects, demonstrated in vitro models of glutamate-induced toxicity, neuroinflammation, and demyelination. In this study, we evaluated the effect and mechanisms of agathisflavone in neuronal integrity and in the modulation of gliosis in an ex vivo model of TBI. For this, microdissections from the encephalon of Wistar rats (P6-8) were prepared and subjected to mechanical injury (MI) and treated or not with daily agathisflavone (5 μM) for 3 days. Astrocyte reactivity was investigated by measuring mRNA and expression of GFAP protein in the lesioned area by immunofluorescence and Western blot. The proportion of microglia was determined by immunofluorescence for Iba-1; mRNA expression for inflammasome NRPL3 and interleukin-1 beta (IL-1β) was determined by RT-qPCR. It was observed that lesions in the cortical tissue induced astrocytes overexpressing GFAP in the typical glial scar formed and that agathisflavone modulated GFAP expression at the transcriptional and post-transcriptional levels, which was associated with a reduction of the glial scar. MI induced an increase in the proportion of microglia (Iba-1+), which was not observed in agathisflavone-treated cultures. Moreover, the flavonoid modulated negatively both the NRLP3 and IL-1β mRNA expression that was increased in the lesioned area of the tissue. These findings support the regulatory properties of agathisflavone in the control of the inflammatory response in glial cells, which can impact neuroprotection and should be considered for future studies for TB and other pathological conditions of the central nervous system.
Collapse
Affiliation(s)
- Verônica Moreira de Sousa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| | - Áurea Maria Alves Nunes Almeida
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| | - Rafael Short Ferreira
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| | - Balbino Lino dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
- College of Nursing, Federal University of Vale do São Francisco, Petrolina 56304-917, PB, Brazil
| | - Victor Diogenes Amara da Silva
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| | - Jorge Mauricio David
- Department of General and Inorganic Chemistry, Institute of Chemistry, Federal University of Bahia, Salvador 40170-110, BA, Brazil;
| | - Cleonice Creusa dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| |
Collapse
|
19
|
Schickel E, Bender T, Kaysan L, Hufgard S, Mayer M, Grosshans DR, Thielemann C, Schroeder IS. Human cerebral organoids model tumor infiltration and migration supported by astrocytes in an autologous setting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635456. [PMID: 39974912 PMCID: PMC11838324 DOI: 10.1101/2025.01.29.635456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Efforts to achieve precise and efficient tumor targeting of highly malignant brain tumors are constrained by the dearth of appropriate models to study the effects and potential side effects of radiation, chemotherapy, and immunotherapy on the most complex human organ, the brain. We established a cerebral organoid model of brain tumorigenesis in an autologous setting by overexpressing c-MYC as one of the most common oncogenes in brain tumors. GFP + /c-MYC high cells were isolated from tumor organoids and used in two different culture approaches: assembloids comprising of a normal cerebral organoid with a GFP + /c-MYC high tumor sphere and co-culture of cerebral organoid slices at air-liquid interface with GFP + /c-MYC high cells. GFP + /c-MYC high cells used in both approaches exhibited tumor-like properties, including overexpression of the c-MYC oncogene, high proliferative and invasive potential, and an immature phenotype as evidenced by increased expression of Ki-67, VIM, and CD133. Organoids and organoid slices served as suitable scaffolds for infiltrating tumor-like cells. Using our highly reproducible and powerful model system that allows long-term culture, we demonstrated that the migratory and infiltrative potential of tumor-like cells is shaped by the environment in which glia cells provide support to tumor-like cells.
Collapse
|
20
|
Humpel C. Long-term live-cell imaging of GFAP+ astroglia and laminin+ vessels in organotypic mouse brain slices using microcontact printing. Front Cell Neurosci 2025; 19:1540150. [PMID: 39935610 PMCID: PMC11808140 DOI: 10.3389/fncel.2025.1540150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
Organotypic brain slices are three-dimensional, 150-μm-thick sections derived from postnatal day 10 mice that can be cultured for several weeks in vitro. However, these slices pose challenges for live-cell imaging due to their thickness, particularly without access to expensive two-photon microscopy. In this study, we present an innovative method to label and visualize specific brain cell populations in living slices. Using microcontact printing, antibodies are applied directly onto the slices in a controlled 400-μm-diameter pattern. Astrocytes are labeled with glial fibrillary acidic protein (GFAP), and vessels are labeled with laminin. Subsequently, slices are incubated with secondary fluorescent antibodies (green fluorescent Alexa-488 or red fluorescent Alexa-546) and visualized using an inverted fluorescence microscope. This approach offers a cost-effective and detailed visualization technique for astroglia and vessels in living brain slices, enabling investigation to be conducted over several weeks.
Collapse
Affiliation(s)
- Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
21
|
Radoszkiewicz K, Rybkowska P, Szymanska M, Krzesniak NE, Sarnowska A. The influence of biomimetic conditions on neurogenic and neuroprotective properties of dedifferentiated fat cells. Stem Cells 2025; 43:sxae066. [PMID: 39576128 PMCID: PMC11811640 DOI: 10.1093/stmcls/sxae066] [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: 05/06/2024] [Accepted: 09/30/2024] [Indexed: 02/12/2025]
Abstract
In the era of a constantly growing number of reports on the therapeutic properties of dedifferentiated, ontogenetically rejuvenated cells and their use in the treatment of neurological diseases, the optimization of their derivation and long-term culture methods seem to be crucial. One of the solutions is seen in the use of dedifferentiated fat cells (DFATs) that are characterized by a greater homogeneity. Moreover, these cells seem to possess a higher expression of transcriptional factors necessary to maintain pluripotency (stemness-related transcriptional factors) as well as a greater ability to differentiate in vitro into 3 embryonic germ layers, and a high proliferative potential in comparison to adipose stem/stromal cells. However, the neurogenic and neuroprotective potential of DFATs is still insufficiently understood; hence, our research goal was to contribute to our current knowledge of the subject. To recreate the brain's physiological (biomimetic) conditions, the cells were cultured at 5% oxygen concentration. The neural differentiation capacity of DFATs was assessed in the presence of the N21 supplement containing the factors that are typically found in the natural environment of the neural cell niche or in the presence of cerebrospinal fluid and under various spatial conditions (microprinting). The neuroprotective properties of DFATs were assessed using the coculture method with the ischemically damaged nerve tissue.
Collapse
Affiliation(s)
- Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02‐106 Warsaw, Poland
| | - Paulina Rybkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02‐106 Warsaw, Poland
| | - Magdalena Szymanska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02‐106 Warsaw, Poland
| | - Natalia Ewa Krzesniak
- Department of Plastic and Reconstructive Surgery, Centre of Postgraduate Medical Education, Prof. W. Orlowski Memorial Hospital, 00‐416 Warsaw, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02‐106 Warsaw, Poland
| |
Collapse
|
22
|
Flores JC, Sarkar D, Zito K. A synapse-specific refractory period for plasticity at individual dendritic spines. Proc Natl Acad Sci U S A 2025; 122:e2410433122. [PMID: 39772745 PMCID: PMC11745398 DOI: 10.1073/pnas.2410433122] [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: 05/24/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
How newly formed memories are preserved while brain plasticity is ongoing has been a source of debate. One idea is that synapses which experienced recent plasticity become resistant to further plasticity, a type of metaplasticity often referred to as saturation. Here, we probe the local dendritic mechanisms that limit plasticity at recently potentiated synapses. We show that recently potentiated individual synapses exhibit a synapse-specific refractory period for further potentiation. We further found that the refractory period is associated with reduced postsynaptic CaMKII signaling; however, stronger synaptic activation fully restored CaMKII signaling but only partially restored the ability for further plasticity. Importantly, the refractory period is released after one hour, a timing that coincides with the enrichment of several postsynaptic proteins to preplasticity levels. Notably, increasing the level of the postsynaptic scaffolding protein, PSD95, but not of PSD93, overcomes the refractory period. Our results support a model in which potentiation at a single synapse is sufficient to initiate a synapse-specific refractory period that persists until key postsynaptic proteins regain their steady-state synaptic levels.
Collapse
Affiliation(s)
- Juan C. Flores
- Center for Neuroscience, University of California, Davis, CA95618
| | - Dipannita Sarkar
- Center for Neuroscience, University of California, Davis, CA95618
| | - Karen Zito
- Center for Neuroscience, University of California, Davis, CA95618
| |
Collapse
|
23
|
Fink R, Imai S, Gockel N, Lauer G, Renken K, Wietek J, Lamothe-Molina PJ, Fuhrmann F, Mittag M, Ziebarth T, Canziani A, Kubitschke M, Kistmacher V, Kretschmer A, Sebastian E, Schmitz D, Terai T, Gründemann J, Hassan S, Patriarchi T, Reiner A, Fuhrmann M, Campbell RE, Masseck OA. PinkyCaMP a mScarlet-based calcium sensor with exceptional brightness, photostability, and multiplexing capabilities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.16.628673. [PMID: 39763884 PMCID: PMC11702558 DOI: 10.1101/2024.12.16.628673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Genetically encoded calcium (Ca2+) indicators (GECIs) are widely used for imaging neuronal activity, yet current limitations of existing red fluorescent GECIs have constrained their applicability. The inherently dim fluorescence and low signal-to-noise ratio of red-shifted GECIs have posed significant challenges. More critically, several red-fluorescent GECIs exhibit photoswitching when exposed to blue light, thereby limiting their applicability in all-optical experimental approaches. Here, we present the development of PinkyCaMP, the first mScarlet-based Ca2+ sensor that outperforms current red fluorescent sensors in brightness, photostability, signal-to-noise ratio, and compatibility with optogenetics and neurotransmitter imaging. PinkyCaMP is well-tolerated by neurons, showing no toxicity or aggregation, both in vitro and in vivo. All imaging approaches, including single-photon excitation methods such as fiber photometry, widefield imaging, miniscope imaging, as well as two-photon imaging in awake mice, are fully compatible with PinkyCaMP.
Collapse
Affiliation(s)
- Ryan Fink
- Synthetic Biology, University of Bremen, Bremen, Germany
| | - Shosei Imai
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Nala Gockel
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - German Lauer
- Cellular Neurobiology, Department of Biology and Biotechnology, Ruhr University Bochum, Germany
| | - Kim Renken
- Synthetic Biology, University of Bremen, Bremen, Germany
| | - Jonas Wietek
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Neuroscience Research Center, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, 10117 Berlin, Germany
| | | | - Falko Fuhrmann
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Manuel Mittag
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Tim Ziebarth
- Cellular Neurobiology, Department of Biology and Biotechnology, Ruhr University Bochum, Germany
| | - Annika Canziani
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | | | | | - Anny Kretschmer
- Network Dysfunction, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Eva Sebastian
- Neural Circuit Computation, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Neuroscience Research Center, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, 10117 Berlin, Germany
- Network Dysfunction, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Einstein Center for Neuroscience, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, NeuroCure Cluster of Excellence, Berlin, Germany
- Humboldt-Universität zu Berlin, Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Takuya Terai
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Jan Gründemann
- Neural Circuit Computation, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Sami Hassan
- System Neurobiology,University of Bremen, Bremen, Germany
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, University and ETH Zürich, Switzerland
| | - Andreas Reiner
- Cellular Neurobiology, Department of Biology and Biotechnology, Ruhr University Bochum, Germany
| | - Martin Fuhrmann
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Robert E Campbell
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- CERVO Brain Research Center and Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Québec, Canada
| | | |
Collapse
|
24
|
Duan X, Zhang C, Wu Y, Ju J, Xu Z, Li X, Liu Y, Ohdah S, Constantin OM, Pan Y, Lu Z, Wang C, Chen X, Gee CE, Nagel G, Hou ST, Gao S, Song K. Suppression of epileptic seizures by transcranial activation of K +-selective channelrhodopsin. Nat Commun 2025; 16:559. [PMID: 39789018 PMCID: PMC11718177 DOI: 10.1038/s41467-025-55818-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Optogenetics is a valuable tool for studying the mechanisms of neurological diseases and is now being developed for therapeutic applications. In rodents and macaques, improved channelrhodopsins have been applied to achieve transcranial optogenetic stimulation. While transcranial photoexcitation of neurons has been achieved, noninvasive optogenetic inhibition for treating hyperexcitability-induced neurological disorders has remained elusive. There is a critical need for effective inhibitory optogenetic tools that are highly light-sensitive and capable of suppressing neuronal activity in deep brain tissue. In this study, we developed a highly sensitive moderately K+-selective channelrhodopsin (HcKCR1-hs) by molecular engineering of the recently discovered Hyphochytrium catenoides kalium (potassium) channelrhodopsin 1. Transcranial activation of HcKCR1-hs significantly prolongs the time to the first seizure, increases survival, and decreases seizure activity in several status epilepticus mouse models. Our approach for transcranial optogenetic inhibition of neural hyperactivity may be adapted for cell type-specific neuromodulation in both basic and preclinical settings.
Collapse
Affiliation(s)
- Xiaodong Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Chong Zhang
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany
| | - Yujie Wu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jun Ju
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhe Xu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Xuanyi Li
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yao Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Schugofa Ohdah
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Oana M Constantin
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Yifan Pan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhonghua Lu
- Research Center for Primate Neuromodulation and Neuroimaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cheng Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaojing Chen
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Christine E Gee
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Georg Nagel
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany
| | - Sheng-Tao Hou
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Shiqiang Gao
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany.
| | - Kun Song
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
25
|
Lee EJ, Lee MJ, Ryu YJ, Nam SH, Kim R, Song S, Park K, Park YJ, Kim JI, Koh SH, Chang MS. Neuroplasticity therapy using glia-like cells derived from human mesenchymal stem cells for the recovery of cerebral infarction sequelae. Mol Ther 2025; 33:356-374. [PMID: 39563032 PMCID: PMC11764092 DOI: 10.1016/j.ymthe.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/12/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
Despite a dramatic increase in ischemic stroke incidence worldwide, effective therapies for attenuating sequelae of cerebral infarction are lacking. This study investigates the use of human mesenchymal stem cells (hMSCs) induced toward glia-like cells (ghMSCs) to ameliorate chronic sequelae resulting from cerebral infarction. Transcriptome analysis demonstrated that ghMSCs exhibited astrocytic characteristics, and assessments conducted ex vivo using organotypic brain slice cultures demonstrated that ghMSCs exhibited superior neuroregenerative and neuroprotective activity against ischemic damage compared to hMSCs. The observed beneficial effects of ghMSCs were diminished by pre-treatment with a CXCR2 antagonist, indicating a direct role for CXCR2 signaling. Studies conducted in rats subjected to cerebral infarction demonstrated that ghMSCs restored neurobehavioral functions and reduced chronic brain infarction in a dose-dependent manner when transplanted at the subacute-to-chronic phase. These beneficial impacts were also inhibited by a CXCR2 antagonist. Molecular analyses confirmed that increased neuroplasticity contributed to ghMSCs' neuroregenerative effects. These data indicate that ghMSCs hold promise for treating refractory sequelae resulting from cerebral infarction by enhancing neuroplasticity and identify CXCR2 signaling as an important mediator of ghMSCs' mechanism of action.
Collapse
Affiliation(s)
- Eun Ji Lee
- Department of Translational Medical Science, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Republic of Korea
| | - Min-Ju Lee
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea
| | - Ye Jin Ryu
- Department of Translational Medical Science, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Republic of Korea
| | - Sang-Hyeon Nam
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea
| | - Rokhyun Kim
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sehyeon Song
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea; Interdisciplinary Program in Neuroscience, Seoul National University College of Natural Sciences, Seoul 08826, Republic of Korea
| | - Kyunghyuk Park
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| | - Young Jun Park
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| | - Jong-Il Kim
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Seong-Ho Koh
- Department of Translational Medical Science, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Republic of Korea; Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Gyeonggi-do 11923, Republic of Korea.
| | - Mi-Sook Chang
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea; Interdisciplinary Program in Neuroscience, Seoul National University College of Natural Sciences, Seoul 08826, Republic of Korea; Neuroscience Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
26
|
Normoyle KP, Lillis KP, Egawa K, McNally MA, Paulchakrabarti M, Coudhury BP, Lau L, Shiu FH, Staley KJ. Displacement of extracellular chloride by immobile anionic constituents of the brain's extracellular matrix. J Physiol 2025; 603:353-378. [PMID: 39621449 PMCID: PMC11747837 DOI: 10.1113/jp285463] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/01/2024] [Indexed: 01/19/2025] Open
Abstract
GABA is the primary inhibitory neurotransmitter. Membrane currents evoked by GABAA receptor activation have uniquely small driving forces: their reversal potential (EGABA) is very close to the resting membrane potential. As a consequence, GABAA currents can flow in either direction, depending on both the membrane potential and the local intra and extracellular concentrations of the primary permeant ion, chloride (Cl). Local cytoplasmic Cl concentrations vary widely because of displacement of mobile Cl ions by relatively immobile anions. Here, we use new reporters of extracellular chloride (Cl- o) to demonstrate that Cl is displaced in the extracellular space by high and spatially heterogenous concentrations of immobile anions including sulfated glycosaminoglycans (sGAGs). Cl- o varies widely, and the mean Cl- o is only half the canonical concentration (i.e. the Cl concentration in the cerebrospinal fluid). These unexpectedly low and heterogenous Cl- o domains provide a mechanism to link the varied but highly stable distribution of sGAGs and other immobile anions in the brain's extracellular space to neuronal signal processing via the effects on the amplitude and direction of GABAA transmembrane Cl currents. KEY POINTS: Extracellular chloride concentrations in the brain were measured using a new chloride-sensitive organic fluorophore and two-photon fluorescence lifetime imaging. In vivo, the extracellular chloride concentration was spatially heterogenous and only half of the cerebrospinal fluid chloride concentration Stable displacement of extracellular chloride by immobile extracellular anions was responsible for the low extracellular chloride concentration The changes in extracellular chloride were of sufficient magnitude to alter the conductance and reversal potential of GABAA chloride currents The stability of the extracellular matrix, the impact of the component immobile anions, including sulfated glycosaminoglycans on extracellular chloride concentrations, and the consequent effect on GABAA signalling suggests a previously unappreciated mechanism for modulating GABAA signalling.
Collapse
Affiliation(s)
- Kieran P Normoyle
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Kyle P Lillis
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Kiyoshi Egawa
- Department of Medicine, Hokaiddo University, Sapporo, Hokaiddo, Japan
| | - Melanie A McNally
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | | | - Biswa P Coudhury
- GlycoAnalytics Core, University of California San Diego, La Jolla, CA, USA
| | - Lauren Lau
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Fu Hung Shiu
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Kevin J Staley
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Rodríguez-Bodero A, Bonifazi P, Tønnesen J, Encinas-Pérez JM. Picrotoxin-Induced Epileptogenic Hippocampal Organotypic Slice Cultures (hOTCs). Methods Mol Biol 2025; 2899:367-388. [PMID: 40067635 DOI: 10.1007/978-1-0716-4386-0_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
Cultured organotypic hippocampal slices (hOTCs) have become increasingly popular as a model for studying brain function. This model offers significant advantages over traditional in vitro methods, as they allow the examination of mid to long-term manipulations while preserving the structure of the dentate gyrus (DG) in the hippocampus. In this chapter, we focus on a protocol based on hOTCs of mouse entorhinal cortex and hippocampus, which by integrating techniques such as retroviral injections, immunohistochemistry, and microscopy imaging, physiological or pathological processes can be easily investigated.
Collapse
Affiliation(s)
- Ane Rodríguez-Bodero
- The Neurogenesis, Neuroinflammation and Network Dynamics Lab (3ND), Achucarro Basque Center for Neuroscience, Leioa, Spain.
| | - Paolo Bonifazi
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Computational Neuroimaging Laboratory, BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Jan Tønnesen
- The Neurogenesis, Neuroinflammation and Network Dynamics Lab (3ND), Achucarro Basque Center for Neuroscience, Leioa, Spain
- BioFisika Institute (CSIC, UPV/EHU), Leioa, Spain
| | - Juan Manuel Encinas-Pérez
- The Neurogenesis, Neuroinflammation and Network Dynamics Lab (3ND), Achucarro Basque Center for Neuroscience, Leioa, Spain.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
- University of the Basque Country (UPV/EHU) Scientific Campus UPV/EHU, Barrio Sarriena, Leioa, Spain.
| |
Collapse
|
28
|
Bosch M, Castro J, Sur M, Hayashi Y. Photomarking Relocalization Technique for Correlated Two-Photon and Electron Microscopy Imaging of Single Stimulated Synapses. Methods Mol Biol 2025; 2910:145-175. [PMID: 40220099 DOI: 10.1007/978-1-0716-4446-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Synapses learn and remember by persistent modifications of their internal structures and composition but, due to their small size, it is difficult to observe these changes at the ultrastructural level in real time. Two-photon fluorescence microscopy (2PM) allows time-course live imaging of individual synapses but lacks ultrastructural resolution. Electron microscopy (EM) allows the ultrastructural imaging of subcellular components but cannot detect fluorescence and lacks temporal resolution. Here we describe a combination of procedures designed to achieve the correlated imaging of the same individual synapse under both 2PM and EM. This technique permits the selective stimulation and live imaging of a single dendritic spine and the subsequent localization of the same spine in EM ultrathin serial sections. Landmarks created through a photomarking method based on the 2-photon-induced precipitation of an electrodense compound are used to unequivocally localize the stimulated synapse. This technique was developed to image, for the first time, the ultrastructure of the postsynaptic density in which long-term potentiation was selectively induced just seconds or minutes before, but it can be applied for the study of any biological process that requires the precise relocalization of micron-wide structures for their correlated imaging with 2PM and EM.
Collapse
Affiliation(s)
- Miquel Bosch
- RIKEN-MIT Neuroscience Research Center, Saitama, Japan.
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Universitat Internacional de Catalunya, Barcelona, Spain.
| | - Jorge Castro
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- MBF Bioscience, Williston, VT, USA
| | - Mriganka Sur
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yasunori Hayashi
- RIKEN-MIT Neuroscience Research Center, Saitama, Japan
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Brain Science Institute, RIKEN, Wako, Saitama, Japan
- Saitama University Brain Science Institute, Saitama University, Saitama, Japan
- School of Life Science, South China Normal University, Guangzhou, China
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
29
|
Nagumanova A, Seeholzer LR, Di Benedetto B. Cortical Organotypic Brain Slice Cultures to Examine Sex- and Age-Dependent Astrocyte-Mediated Synaptic Phagocytosis. Methods Mol Biol 2025; 2896:203-214. [PMID: 40111607 DOI: 10.1007/978-1-0716-4366-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Astrocytes, the most abundant glial cells in the brain, are an integral part of the synaptic compartment and contribute to synaptic pruning, a key process for refining neural circuits during early postnatal development (PND). Dysregulations in this process are implicated in various neuropsychiatric disorders, including major depressive disorder (MDD). To investigate astrocyte functions in a physiologically relevatpdelnt context, organotypic brain slice cultures (OBSCs) offer a powerful model, reproducing more closely in vivo conditions than traditional cell cultures and preserving complex brain architecture and interactions. Here, we present OBSCs as an ex vivo culturing method to provide a platform to explore astrocyte-mediated synaptic pruning dynamics in the rat prefrontal cortex (PFC) during PND. Our approach is based on assessing the role of MEGF10, a key protein involved in synaptic pruning, alongside the synaptic markers synaptophysin and PSD95, using Western blotting to analyze the expression levels of these markers in the cortex of developing rat pups. Additionally, we combine immunofluorescence staining with confocal imaging and IMARIS 9.8 software-assisted analysis to investigate the colocalization of the lysosomal marker LAMP1 with synaptic and astrocytic markers to evaluate the precise rate of synaptic engulfment. The methods presented here allow a deeper examination of an astrocyte-mediated synaptic remodeling in healthy and pathophysiological conditions.
Collapse
Affiliation(s)
- Anastasiia Nagumanova
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Lea R Seeholzer
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Barbara Di Benedetto
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany.
- Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
30
|
Zhou J, Greenfield AL, Loudermilk RP, Bartley CM, Chen C, Chen X, Leroux MA, Lu Y, Necula D, Ngo TT, Tran BT, Honma PS, Lauderdale K, Zhao C, Zhou X, Wang H, Nicoll RA, Wang C, Paz JT, Palop JJ, Wilson MR, Pleasure SJ. Disrupted callosal connectivity underlies long-lasting sensory-motor deficits in an NMDA receptor antibody encephalitis mouse model. J Clin Invest 2024; 135:e173493. [PMID: 39739422 PMCID: PMC11870732 DOI: 10.1172/jci173493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
N-methyl-d-aspartate (NMDA) receptor-mediated autoimmune encephalitis (NMDAR-AE) frequently results in persistent sensory-motor deficits, especially in children, yet the underlying mechanisms remain unclear. This study investigated the long-term effects of exposure to a patient-derived GluN1-specific mAb during a critical developmental period (from postnatal day 3 to day 12) in mice. We observed long-lasting sensory-motor deficits characteristic of NMDAR-AE, along with permanent changes in callosal axons within the primary somatosensory cortex (S1) in adulthood, including increased terminal branch complexity. This complexity was associated with paroxysmal recruitment of neurons in S1 in response to callosal stimulation. Particularly during complex motor tasks, mAb3-treated mice exhibited significantly reduced interhemispheric functional connectivity between S1 regions, consistent with pronounced sensory-motor behavioral deficits. These findings suggest that transient exposure to anti-GluN1 mAb during a critical developmental window may lead to irreversible morphological and functional changes in callosal axons, which could significantly impair sensory-motor integration and contribute to long-lasting sensory-motor deficits. Our study establishes a new model of NMDAR-AE and identifies novel cellular and network-level mechanisms underlying persistent sensory-motor deficits in this context. These insights lay the foundation for future research into molecular mechanisms and the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Neurology
- Weill Institute for Neurosciences
- Center for Encephalitis and Meningitis, and
| | | | | | - Christopher M. Bartley
- Weill Institute for Neurosciences
- Center for Encephalitis and Meningitis, and
- Department of Psychiatry and Behavioral Sciences, UCSF, San Francisco, California, USA
| | - Chun Chen
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Xiumin Chen
- Department of Neurology and Institute of Neuroscience of Soochow University, Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | | | - Yujun Lu
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Pudong New Area, Shanghai, China
| | - Deanna Necula
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
- Neuroscience Graduate Program, UCSF, San Francisco, California, USA
| | - Thomas T. Ngo
- Department of Neurology
- Weill Institute for Neurosciences
- Center for Encephalitis and Meningitis, and
| | - Baouyen T. Tran
- Department of Neurology
- Weill Institute for Neurosciences
- Center for Encephalitis and Meningitis, and
| | - Patrick S. Honma
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
- Neuroscience Graduate Program, UCSF, San Francisco, California, USA
| | - Kelli Lauderdale
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Chao Zhao
- Center for Data Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Hong Wang
- Department of Neurology
- Weill Institute for Neurosciences
| | - Roger A. Nicoll
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology. Department of Physiology, UCSF, San Francisco, California, USA
| | - Cong Wang
- Institute of Rehabilitation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Queensland Brain Institute, The University of Queensland, St. Lucia, Brisbane, Australia
| | - Jeanne T. Paz
- Department of Neurology
- Weill Institute for Neurosciences
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
- Neuroscience Graduate Program, UCSF, San Francisco, California, USA
| | - Jorge J. Palop
- Department of Neurology
- Weill Institute for Neurosciences
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
- Neuroscience Graduate Program, UCSF, San Francisco, California, USA
| | - Michael R. Wilson
- Department of Neurology
- Weill Institute for Neurosciences
- Center for Encephalitis and Meningitis, and
| | - Samuel J. Pleasure
- Department of Neurology
- Weill Institute for Neurosciences
- Center for Encephalitis and Meningitis, and
- Programs in Neuroscience and Developmental Stem Cell Biology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Kavli Institute for Fundamental Neuroscience, San Francisco, California, USA
| |
Collapse
|
31
|
McNally MA, Lau LA, Granak S, Hike D, Liu X, Yu X, Donahue RA, Chibnik LB, Ortiz JV, Che A, Northington F, Staley K. Ongoing loss of viable neurons for weeks after mild perinatal hypoxia-ischemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.19.629457. [PMID: 39763962 PMCID: PMC11702593 DOI: 10.1101/2024.12.19.629457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Mild hypoxic-ischemic encephalopathy is common in neonates with no evidence-based therapies, and 30-40% of patients experience adverse outcomes. The nature and progression of mild injury is poorly understood. Thus, we studied the evolution of mild perinatal brain injury using longitudinal two-photon imaging of transgenic fluorescent proteins as a novel readout of neuronal viability and activity at cellular resolution. In vitro, perinatal murine organotypic hippocampal cultures underwent 15-20 minutes of oxygen-glucose deprivation. In vivo, mild hypoxia-ischemia was completed in post-natal day 10 mouse pups of both sexes with carotid ligation and 15 minutes of hypoxia. Consistent with a mild injury, minimal immediate neuronal death was seen and there was no volumetric evidence of injury by ex vivo MRI 2.5 weeks after injury. In both the hippocampus and neocortex, these mild injuries resulted in a significantly delayed and progressive neuronal loss in the second week after injury, measured by fluorophore quenching. Mild hypoxia-ischemia transiently suppressed cortical network activity followed by normal maturation. No post-injury seizures were seen. The participation in network activity of individual neurons destined to die was indistinguishable from those that survived for 4 days post-injury. In conclusion, our results showed that mild perinatal brain injury resulted in a prolonged increase of neuronal death. Neurons that died late were functioning normally for days after injury, suggesting a new pathophysiology of neuronal death. Critically, the neurons destined to die late demonstrated multiple biomarkers of viability long after mild injury, suggesting their later death may be modified with neuroprotective interventions. SIGNIFICANCE STATEMENT Neonatal encephalopathy due to peripartum hypoxia-ischemia (HI) is a major cause of neonatal mortality and morbidity worldwide. Of these infants, most are categorized as having mild HI. Infants with mild HI have significant long-term disabilities. There are currently no evidence-based therapies, largely because the progression and pathophysiology of mild injury is poorly understood. We have identified, for the first time, that mild perinatal HI results in a delayed and prolonged increase in neuronal death. The cortical and hippocampal neurons that die over a week after injury participate normally in neural network activity and exhibit robust viability for many days after injury, indicating a novel pathophysiology of neuronal death. Clinically, these data suggest an extended therapeutic window for mild perinatal HI.
Collapse
|
32
|
Szczepankiewicz AA, Parobczak K, Zaręba-Kozioł M, Ruszczycki B, Bijata M, Trzaskoma P, Hajnowski G, Holm-Kaczmarek D, Włodarczyk J, Sas-Nowosielska H, Wilczyński GM, Rędowicz MJ, Magalska A. Neuronal activation affects the organization and protein composition of the nuclear speckles. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119829. [PMID: 39197592 DOI: 10.1016/j.bbamcr.2024.119829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Nuclear speckles, also known as interchromatin granule clusters (IGCs), are subnuclear domains highly enriched in proteins involved in transcription and mRNA metabolism and, until recently, have been regarded primarily as their storage and modification hubs. However, several recent studies on non-neuronal cell types indicate that nuclear speckles may directly contribute to gene expression as some of the active genes have been shown to associate with these structures. Neuronal activity is one of the key transcriptional regulators and may lead to the rearrangement of some nuclear bodies. Notably, the impact of neuronal activation on IGC/nuclear speckles organization and function remains unexplored. To address this research gap, we examined whether and how neuronal stimulation affects the organization of these bodies in granular neurons from the rat hippocampal formation. Our findings demonstrate that neuronal stimulation induces morphological and proteomic remodelling of the nuclear speckles under both in vitro and in vivo conditions. Importantly, these changes are not associated with cellular stress or cell death but are dependent on transcription and splicing.
Collapse
Affiliation(s)
- Andrzej Antoni Szczepankiewicz
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Kamil Parobczak
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Monika Zaręba-Kozioł
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Błażej Ruszczycki
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; AGH University of Krakow, Faculty of Physics and Applied Computer Science, Department of Medical Physics and Biophysics, al. A. Mickiewicza 30, 30-059 Krakow, Poland
| | - Monika Bijata
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Paweł Trzaskoma
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Grzegorz Hajnowski
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Dagmara Holm-Kaczmarek
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Jakub Włodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Hanna Sas-Nowosielska
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Grzegorz Marek Wilczyński
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Adriana Magalska
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| |
Collapse
|
33
|
Collantes-Fernández E, Horcajo P, Benavides J, Sánchez-Sánchez R, Blanco-Murcia J, Montaner-Da Torre S, Hecker YP, Ortega-Mora LM, Pastor-Fernández I. Evaluating the suitability of placental bovine explants for ex vivo modelling of host-pathogen interactions in Neospora caninum infections. Theriogenology 2024; 230:305-313. [PMID: 39368452 DOI: 10.1016/j.theriogenology.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/07/2024]
Abstract
Bovine abortions, often caused by infectious agents like Neospora caninum, inflict substantial economic losses. Studying host-pathogen interactions in pregnant cows is challenging, and existing cell cultures lack the intricate complexity of real tissues. To bridge the gap between in vitro and in vivo models, we explored the use of cryopreserved bovine placental explants. Building upon our successful development of protocols for obtaining, culturing, and cryopreserving sheep placental explants, we applied these methods to bovine tissues. Here, we compared fresh and cryopreserved bovine explants, evaluating their integrity and functionality over culture time. Additionally, we investigated their susceptibility to N. caninum infection. Our findings revealed that bovine explants deteriorate faster in culture compared to sheep explants, exhibiting diminished viability and function. Cryopreservation further exacerbated this deterioration. While fresh explants were successfully infected with N. caninum, parasite replication was limited. Notably, cryopreservation reduced infection efficiency. This pioneering work paves the way for developing ex vivo models to study reproductive pathogens in cattle. However, further optimization of the model is essential. These improved models will have the potential to significantly reduce the reliance on animals in research.
Collapse
Affiliation(s)
- Esther Collantes-Fernández
- SALUVET Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Pilar Horcajo
- SALUVET Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | | | - Roberto Sánchez-Sánchez
- SALUVET Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Javier Blanco-Murcia
- SALUVET Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain; Animal Medicine and Surgery Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Sandra Montaner-Da Torre
- SALUVET Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Yanina P Hecker
- SALUVET Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Luis Miguel Ortega-Mora
- SALUVET Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Iván Pastor-Fernández
- SALUVET Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain; Parasitology Unit, Microbiology and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, Spain.
| |
Collapse
|
34
|
Borovac J, Rai J, Valencia M, Li H, Georgiou J, Collingridge GL, Takao K, Okamoto K. Optogenetic elevation of postsynaptic cGMP in the hippocampal dentate gyrus enhances LTP and modifies mouse behaviors. Front Mol Neurosci 2024; 17:1479360. [PMID: 39660172 PMCID: PMC11629205 DOI: 10.3389/fnmol.2024.1479360] [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: 08/12/2024] [Accepted: 10/18/2024] [Indexed: 12/12/2024] Open
Abstract
A major intracellular messenger implicated in synaptic plasticity and cognitive functions both in health and disease is cyclic GMP (cGMP). Utilizing a photoactivatable guanylyl cyclase (BlgC) actuator to increase cGMP in dentate granule neurons of the hippocampus by light, we studied the effects of spatiotemporal cGMP elevations in synaptic and cognitive functions. At medial perforant path to dentate gyrus (MPP-DG) synapses, we found enhanced long-term potentiation (LTP) of synaptic responses when postsynaptic cGMP was elevated during the induction period. Basal synaptic transmission and the paired-pulse ratio were unaffected, suggesting the cGMP effect on LTP was postsynaptic in origin. In behaving mice implanted with a fiber optic and wireless LED device, their performance following DG photoactivation (5-10 min) was studied in a variety of behavioral tasks. There were enhancements in reference memory and social behavior within tens of minutes following DG BlgC photoactivation, and with time (hours), an anxiogenic effect developed. Thus, postsynaptic cGMP elevations, specifically in the DG and specifically during conditions that evoke synaptic plasticity or during experience, are able to rapidly modify synaptic strength and behavioral responses, respectively. The optogenetics technology and new roles for cGMP in the DG may have applications in brain disorders that are impacted by dysregulated cGMP signaling, such as Alzheimer's disease.
Collapse
Affiliation(s)
- Jelena Borovac
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Behavioral Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Jayant Rai
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Megan Valencia
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Hang Li
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - John Georgiou
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Graham L. Collingridge
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- TANZ Centre for Research in Neurodegenerative Diseases (CRND), University of Toronto, Toronto, ON, Canada
| | - Keizo Takao
- Department of Behavioral Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama, Japan
- Research Centre for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Kenichi Okamoto
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
35
|
Xiong W, Qin M, Zhong H. PKA regulation of neuronal function requires the dissociation of catalytic subunits from regulatory subunits. eLife 2024; 13:RP93766. [PMID: 39508822 PMCID: PMC11542917 DOI: 10.7554/elife.93766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
Protein kinase A (PKA) plays essential roles in diverse cellular functions. However, the spatiotemporal dynamics of endogenous PKA upon activation remain debated. The classical model predicts that PKA catalytic subunits dissociate from regulatory subunits in the presence of cAMP, whereas a second model proposes that catalytic subunits remain associated with regulatory subunits following physiological activation. Here, we report that different PKA subtypes, as defined by the regulatory subunit, exhibit distinct subcellular localization at rest in CA1 neurons of cultured hippocampal slices. Nevertheless, when all tested PKA subtypes are activated by norepinephrine, presumably via the β-adrenergic receptor, catalytic subunits translocate to dendritic spines but regulatory subunits remain unmoved. These differential spatial dynamics between the subunits indicate that at least a significant fraction of PKA dissociates. Furthermore, PKA-dependent regulation of synaptic plasticity and transmission can be supported only by wildtype, dissociable PKA, but not by inseparable PKA. These results indicate that endogenous PKA regulatory and catalytic subunits dissociate to achieve PKA function in neurons.
Collapse
Affiliation(s)
- Weihong Xiong
- Vollum Institute, Oregon Health and Science UniversityPortlandUnited States
| | - Maozhen Qin
- Vollum Institute, Oregon Health and Science UniversityPortlandUnited States
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science UniversityPortlandUnited States
| |
Collapse
|
36
|
Jain A, Nakahata Y, Pancani T, Watabe T, Rusina P, South K, Adachi K, Yan L, Simorowski N, Furukawa H, Yasuda R. Dendritic, delayed, stochastic CaMKII activation in behavioural time scale plasticity. Nature 2024; 635:151-159. [PMID: 39385027 PMCID: PMC11540904 DOI: 10.1038/s41586-024-08021-8] [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: 08/08/2023] [Accepted: 09/05/2024] [Indexed: 10/11/2024]
Abstract
Behavioural time scale plasticity (BTSP) is non-Hebbian plasticity induced by integrating presynaptic and postsynaptic components separated by a behaviourally relevant time scale (seconds)1. BTSP in hippocampal CA1 neurons underlies place cell formation. However, the molecular mechanisms that enable synapse-specific plasticity on a behavioural time scale are unknown. Here we show that BTSP can be induced in a single dendritic spine using two-photon glutamate uncaging paired with postsynaptic current injection temporally separated by a behavioural time scale. Using an improved Ca2+/calmodulin-dependent kinase II (CaMKII) sensor, we did not detect CaMKII activation during this BTSP induction. Instead, we observed dendritic, delayed and stochastic CaMKII activation (DDSC) associated with Ca2+ influx and plateau potentials 10-100 s after BTSP induction. DDSC required both presynaptic and postsynaptic activity, which suggests that CaMKII can integrate these two signals. Also, optogenetically blocking CaMKII 15-30 s after the BTSP protocol inhibited synaptic potentiation, which indicated that DDSC is an essential mechanism of BTSP. IP3-dependent intracellular Ca2+ release facilitated both DDSC and BTSP. Thus, our study suggests that non-synapse-specific CaMKII activation provides an instructive signal with an extensive time window over tens of seconds during BTSP.
Collapse
Affiliation(s)
- Anant Jain
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- Centre for High Impact Neuroscience and Translational Applications (CHINTA), TCG CREST, Kolkata, India
| | - Yoshihisa Nakahata
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Tristano Pancani
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Tetsuya Watabe
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Polina Rusina
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Kelly South
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Kengo Adachi
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Long Yan
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Noriko Simorowski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| |
Collapse
|
37
|
Browner D, Adamatzky A. Micro-electrode array recording of extracellular electrical potentials of liquid static surface fermented Hericium erinaceus. Biosystems 2024; 245:105298. [PMID: 39159880 DOI: 10.1016/j.biosystems.2024.105298] [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: 06/10/2024] [Revised: 08/06/2024] [Accepted: 08/11/2024] [Indexed: 08/21/2024]
Abstract
Hericium erinaceus is a basidiomycetes fungus with previously uncharacterised extracellular electrophysiology. Here, we present results of recordings of the electrical potentials of fungal biofilms of this species using microelectrode arrays (MEAs). In particular, we focused on modelling the temporal and spatial progression of the low frequency (≤ 1 Hz) potentials. Culture media control studies showed that the electrical potential activity results from the growth and subsequent spiking behaviours of the mycelium extracellular matrices. An antifungal assay using nystatin suspension, 10,000 unit/mL in DPBS, provided evidence for the biological origin of electrical potentials due to targeting of the selective permeability of the cell membrane and subsequent cessation of electrical activity. Conversely, injection of L-glutamic acid increased the combined multi-channel mean firing rate from 0.04 Hz to 0.1 Hz. Analysis of bursting and spatial propagation of the extracellular signals are also presented.
Collapse
|
38
|
Herstel LJ, Wierenga CJ. Distinct Modulation of I h by Synaptic Potentiation in Excitatory and Inhibitory Neurons. eNeuro 2024; 11:ENEURO.0185-24.2024. [PMID: 39406481 PMCID: PMC11574699 DOI: 10.1523/eneuro.0185-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 11/15/2024] Open
Abstract
Selective modifications in the expression or function of dendritic ion channels regulate the propagation of synaptic inputs and determine the intrinsic excitability of a neuron. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels open upon membrane hyperpolarization and conduct a depolarizing inward current (I h). HCN channels are enriched in the dendrites of hippocampal pyramidal neurons where they regulate the integration of synaptic inputs. Synaptic plasticity can bidirectionally modify dendritic HCN channels in excitatory neurons depending on the strength of synaptic potentiation. In inhibitory neurons, however, the dendritic expression and modulation of HCN channels are largely unknown. In this study, we systematically compared the modulation of I h by synaptic potentiation in hippocampal CA1 pyramidal neurons and stratum radiatum (sRad) interneurons in mouse organotypic cultures. I h properties were similar in inhibitory and excitatory neurons and contributed to resting membrane potential and action potential firing. We found that in sRad interneurons, HCN channels were downregulated after synaptic plasticity, irrespective of the strength of synaptic potentiation. This suggests differential regulation of I h in excitatory and inhibitory neurons, possibly signifying their distinct role in network activity.
Collapse
Affiliation(s)
- Lotte J Herstel
- Biology Department, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 AJ, the Netherlands
| | - Corette J Wierenga
- Biology Department, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 AJ, the Netherlands
| |
Collapse
|
39
|
Kang W, Siewe AD, Oluigbo CC, Arijesudade MO, Brailoiu E, Undieh AS. Dopamine internalization via Uptake 2 and stimulation of intracellular D 5-receptor-dependent calcium mobilization and CDP-diacylglycerol signaling. Front Pharmacol 2024; 15:1422998. [PMID: 39525629 PMCID: PMC11543475 DOI: 10.3389/fphar.2024.1422998] [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: 04/25/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Dopamine stimulates CDP-diacylglycerol biosynthesis through D1-like receptors, particularly the D5 subtype most of which is intracellularly localized. CDP-diacylglycerol regulates phosphatidylinositol-4,5-bisphosphate-dependent signaling cascades by serving as obligatory substrate for phosphatidylinositol biosynthesis. Here, we used acute and organotypic brain tissues and cultured cells to explore the mechanism by which extracellular dopamine acts to modulate intracellular CDP-diacylglycerol. Dopamine stimulated CDP-diacylglycerol in organotypic and neural cells lacking the presynaptic dopamine transporter, and this action was selectively mimicked by D1-like receptor agonists SKF38393 and SKF83959. Dopaminergic CDP-diacylglycerol stimulation was blocked by decynium-22 which blocks Uptake2-like transporters and by anti-microtubule disrupters of cytoskeletal transport, suggesting transmembrane uptake and guided transport of the ligands to intracellular sites of CDP-diacylglycerol regulation. Fluorescent or radiolabeled dopamine was saturably transported into primary neurons or B35 neuroblastoma cells expressing the plasmamembrane monoamine transporter, PMAT. Microinjection of 10-nM final concentration of dopamine into human D5-receptor-transfected U2-OS cells rapidly and transiently increased cytosolic calcium concentrations by 316%, whereas non-D5-receptor-expressing U2-OS cells showed no response. Given that U2-OS cells natively express PMAT, bath application of 10 μM dopamine slowly increased cytosolic calcium in D5-expressing cells. These observations indicate that dopamine is actively transported by a PMAT-implicated Uptake2-like mechanism into postsynaptic-type dopaminoceptive cells where the monoamine stimulates its intracellular D5-type receptors to mobilize cytosolic calcium and promote CDP-diacylglycerol biosynthesis. This is probably the first demonstration of functional intracellular dopamine receptor coupling in neural tissue, thus challenging the conventional paradigm that postsynaptic dopamine uptake serves merely as a mechanism for deactivating spent or excessive synaptic transmitter.
Collapse
Affiliation(s)
- Wenfei Kang
- Department of Biomedical Sciences, School of Medicine, City University of New York, New York, NY, United States
| | - Arlette Deukam Siewe
- Department of Biomedical Sciences, School of Medicine, City University of New York, New York, NY, United States
| | - Chizurum C. Oluigbo
- Department of Biomedical Sciences, School of Medicine, City University of New York, New York, NY, United States
- Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, United States
| | - Mercy O. Arijesudade
- Department of Biomedical Sciences, School of Medicine, City University of New York, New York, NY, United States
| | - Eugen Brailoiu
- Department of Pharmacology, School of Medicine, Temple University, Philadelphia, PA, United States
| | - Ashiwel S. Undieh
- Department of Biomedical Sciences, School of Medicine, City University of New York, New York, NY, United States
- Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, United States
| |
Collapse
|
40
|
Hoyt KR, Horning P, Georgette Ang P, Karelina K, Obrietan K. Ribosomal S6 kinase signaling regulates neuronal viability during development and confers resistance to excitotoxic cell death in mature neurons. Neuroscience 2024; 558:1-10. [PMID: 39137868 DOI: 10.1016/j.neuroscience.2024.08.012] [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: 07/12/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024]
Abstract
The Ribosomal S6 Kinase (RSK) family of serine/threonine kinases function as key downstream effectors of the MAPK signaling cascade. In the nervous system, RSK signaling plays crucial roles in neuronal development and contributes to activity-dependent neuronal plasticity. This study examined the role of RSK signaling in cell viability during neuronal development and in neuroprotection in the mature nervous system. Using neuronal cell-culture-based profiling, we found that suppressing RSK signaling led to significant cell death in developing primary neuronal cultures. To this end, treatment with the RSK inhibitors BiD1870 or SL0101 on the first day of culturing resulted in over 80% cell death. In contrast, more mature cultures showed attenuated cell death upon RSK inhibition. Inhibition of RSK signaling during early neuronal development also disrupted neurite outgrowth and cell growth. In maturing hippocampal explant cultures, treatment with BiD1870 had minimal effects on cell viability, but led to a striking augmentation of NMDA-induced cell death. Finally, we used the endothelin 1 (ET-1) model of ischemia to examine the neuroprotective effects of RSK signaling in the mature hippocampus in vivo. Notably, in the absence of RSK inhibition, the granule cell layer (GCL) was resistant to the effects of ET-1; However, disruption of RSK signaling (via the microinjection of BiD1870) prior to ET-1 injection triggered cell death within the GCL, thus indicating a neuroprotective role for RSK signaling in the mature nervous system. Together these data reveal distinct, developmentally-defined, roles for RSK signaling in the nervous system.
Collapse
Affiliation(s)
- Kari R Hoyt
- Division of Pharmaceutics and Pharmacology, Ohio State University, Columbus, OH, USA.
| | - Paul Horning
- Department of Neuroscience, Ohio State University, Columbus, OH, USA; Division of Pharmaceutics and Pharmacology, Ohio State University, Columbus, OH, USA
| | - Pia Georgette Ang
- Division of Pharmaceutics and Pharmacology, Ohio State University, Columbus, OH, USA
| | - Kate Karelina
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
41
|
Yilmaz SN, Steiner K, Marksteiner J, Faserl K, Villunger M, Sarg B, Humpel C. From Organotypic Mouse Brain Slices to Human Alzheimer's Plasma Biomarkers: A Focus on Nerve Fiber Outgrowth. Biomolecules 2024; 14:1326. [PMID: 39456259 PMCID: PMC11506054 DOI: 10.3390/biom14101326] [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: 09/26/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by memory loss and progressive deterioration of cognitive functions. Being able to identify reliable biomarkers in easily available body fluids such as blood plasma is vital for the disease. To achieve this, we used a technique that applied human plasma to organotypic brain slice culture via microcontact printing. After a 2-week culture period, we performed immunolabeling for neurofilament and myelin oligodendrocyte glycoprotein (MOG) to visualize newly formed nerve fibers and oligodendrocytes. There was no significant change in the number of new nerve fibers in the AD plasma group compared to the healthy control group, while the length of the produced fibers significantly decreased. A significant increase in the number of MOG+ dots around these new fibers was detected in the patient group. According to our hypothesis, there are factors in the plasma of AD patients that affect the growth of new nerve fibers, which also affect the oligodendrocytes. Based on these findings, we selected the most promising plasma samples and conducted mass spectrometry using a differential approach and we identified three putative biomarkers: aldehyde-dehydrogenase 1A1, alpha-synuclein and protein S100-A4. Our method represents a novel and innovative approach for translating research findings from mouse models to human applications.
Collapse
Affiliation(s)
- Sakir Necat Yilmaz
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.N.Y.); (K.S.)
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, Mersin 33130, Turkey
| | - Katharina Steiner
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.N.Y.); (K.S.)
| | - Josef Marksteiner
- Department of Psychiatry and Psychotherapy A, Hall State Hospital, 6060 Hall in Tirol, Austria;
| | - Klaus Faserl
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Mathias Villunger
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Bettina Sarg
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.N.Y.); (K.S.)
| |
Collapse
|
42
|
Selfe JS, Steyn TJS, Shorer EF, Burman RJ, Düsterwald KM, Kraitzick AZ, Abdelfattah AS, Schreiter ER, Newey SE, Akerman CJ, Raimondo JV. All-optical reporting of inhibitory receptor driving force in the nervous system. Nat Commun 2024; 15:8913. [PMID: 39414774 PMCID: PMC11484818 DOI: 10.1038/s41467-024-53074-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: 01/23/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024] Open
Abstract
Ionic driving forces provide the net electromotive force for ion movement across receptors, channels, and transporters, and are a fundamental property of all cells. In the nervous system, fast synaptic inhibition is mediated by chloride permeable GABAA and glycine receptors, and single-cell intracellular recordings have been the only method for estimating driving forces across these receptors (DFGABAA). Here we present a tool for quantifying inhibitory receptor driving force named ORCHID: all-Optical Reporting of CHloride Ion Driving force. We demonstrate ORCHID's ability to provide accurate, high-throughput measurements of resting and dynamic DFGABAA from genetically targeted cell types over multiple timescales. ORCHID confirms theoretical predictions about the biophysical mechanisms that establish DFGABAA, reveals differences in DFGABAA between neurons and astrocytes, and affords the first in vivo measurements of intact DFGABAA. This work extends our understanding of inhibitory synaptic transmission and demonstrates the potential for all-optical methods to assess ionic driving forces.
Collapse
Affiliation(s)
- Joshua S Selfe
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Teresa J S Steyn
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Eran F Shorer
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Neurology, School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, United States of America
| | - Richard J Burman
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Kira M Düsterwald
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Gatsby Computational Neuroscience Unit, University College London, London, United Kingdom
| | - Ariel Z Kraitzick
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Ahmed S Abdelfattah
- Department of Neuroscience, Brown University, Providence, Rhode Island, United States of America
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island, United States of America
| | - Eric R Schreiter
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, United States of America
| | - Sarah E Newey
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Colin J Akerman
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Joseph V Raimondo
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa.
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa.
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
43
|
Liouta K, Lubas M, Venugopal V, Chabbert J, Jeannière C, Diaz C, Munier M, Tessier B, Claverol S, Favereaux A, Sainlos M, de Wit J, Letellier M, Thoumine O, Chamma I. LRRTM2 controls presynapse nano-organization and AMPA receptor sub-positioning through Neurexin-binding interface. Nat Commun 2024; 15:8807. [PMID: 39394199 PMCID: PMC11470055 DOI: 10.1038/s41467-024-53090-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: 08/08/2023] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
Synapses are organized into nanocolumns that control synaptic transmission efficacy through precise alignment of postsynaptic neurotransmitter receptors and presynaptic release sites. Recent evidence show that Leucine-Rich Repeat Transmembrane protein LRRTM2, highly enriched and confined at synapses, interacts with Neurexins through its C-terminal cap, but the role of this binding interface has not been explored in synapse formation and function. Here, we develop a conditional knock-out mouse model (cKO) to address the molecular mechanisms of LRRTM2 regulation, and its role in synapse organization and function. We show that LRRTM2 cKO specifically impairs excitatory synapse formation and function in mice. Surface expression, synaptic clustering, and membrane dynamics of LRRTM2 are tightly controlled by selective motifs in the C-terminal domain. Conversely, the N-terminal domain controls presynapse nano-organization and postsynapse AMPAR sub-positioning and stabilization through the recently identified Neurexin-binding interface. Thus, we identify LRRTM2 as a central organizer of pre- and post- excitatory synapse nanostructure through interaction with presynaptic Neurexins.
Collapse
Affiliation(s)
- Konstantina Liouta
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Malgorzata Lubas
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Vasika Venugopal
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Julia Chabbert
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Caroline Jeannière
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Candice Diaz
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Matthieu Munier
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Béatrice Tessier
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | | | - Alexandre Favereaux
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Matthieu Sainlos
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Mathieu Letellier
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Olivier Thoumine
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Ingrid Chamma
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France.
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France.
| |
Collapse
|
44
|
Gellhorn Serra M, Meier L, Sauerhering L, Wilhelm J, Kupke A. Organotypic brain slices as a model to study the neurotropism of the highly pathogenic Nipah and Ebola viruses. J Gen Virol 2024; 105. [PMID: 39466030 DOI: 10.1099/jgv.0.002038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
Nipah virus (NiV) and Ebola virus (EBOV) are highly pathogenic zoonotic viruses with case fatality rates of up to 90%. While the brain is a known target organ following NiV infection, involvement of the central nervous system in EBOV-infected patients only became more evident after the West African epidemic in 2013-2016. To gain a deeper comprehension of the neurotropism of NiV and EBOV with respect to target cells, affected brain regions and local inflammatory responses, murine organotypic brain slices (BS) were established and infected. Both NiV and EBOV demonstrated the capacity to infect BS from adult wt mice and mice lacking the receptor for type I IFNs (IFNAR-/-) and targeted various cell types. NiV was observed to replicate in BS derived from both mouse strains, yet no release of infectious particles was detected. In contrast, EBOV replication was limited in both BS models. The release of several pro-inflammatory cytokines and chemokines, including eotaxin, IFN-γ, IL-1α, IL-9, IL-17a and keratinocyte-derived chemokine (KC), was observed in both virus-infected models, suggesting a potential role of the inflammatory response in NiV- or EBOV-induced neuropathology. It is noteworthy that the choroid plexus was identified as a highly susceptible target for EBOV and NiV infection, suggesting that the blood-cerebrospinal fluid barrier may serve as a potential entry point for these viruses.
Collapse
Affiliation(s)
- Michelle Gellhorn Serra
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research (DZIF), TTU Emerging Infections, Giessen, Germany
| | - Lars Meier
- Philipps University Marburg, Institute of Virology, Marburg, Germany
| | - Lucie Sauerhering
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research (DZIF), TTU Emerging Infections, Giessen, Germany
| | - Jochen Wilhelm
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
- Institute for Lung Health, Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Alexandra Kupke
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research (DZIF), TTU Emerging Infections, Giessen, Germany
| |
Collapse
|
45
|
Carretero-Guillén A, Treviño M, Gómez-Climent MÁ, Dogbevia GK, Bertocchi I, Sprengel R, Larkum ME, Vlachos A, Gruart A, Delgado-García JM, Hasan MT. Dentate gyrus is needed for memory retrieval. Mol Psychiatry 2024; 29:2939-2950. [PMID: 38609585 PMCID: PMC11449802 DOI: 10.1038/s41380-024-02546-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/03/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024]
Abstract
The hippocampus is crucial for acquiring and retrieving episodic and contextual memories. In previous studies, the inactivation of dentate gyrus (DG) neurons by chemogenetic- and optogenetic-mediated hyperpolarization led to opposing conclusions about DG's role in memory retrieval. One study used Designer Receptors Exclusively Activated by Designer Drugs (DREADD)-mediated clozapine N-oxide (CNO)-induced hyperpolarization and reported that the previously formed memory was erased, thus concluding that denate gyrus is needed for memory maintenance. The other study used optogenetic with halorhodopsin induced hyperpolarization and reported and dentate gyrus is needed for memory retrieval. We hypothesized that this apparent discrepancy could be due to the length of hyperpolarization in previous studies; minutes by optogenetics and several hours by DREADD/CNO. Since hyperpolarization interferes with anterograde and retrograde neuronal signaling, it is possible that the memory engram in the dentate gyrus and the entorhinal to hippocampus trisynaptic circuit was erased by long-term, but not with short-term hyperpolarization. We developed and applied an advanced chemogenetic technology to selectively silence synaptic output by blocking neurotransmitter release without hyperpolarizing DG neurons to explore this apparent discrepancy. We performed in vivo electrophysiology during trace eyeblink in a rabbit model of associative learning. Our work shows that the DG output is required for memory retrieval. Based on previous and recent findings, we propose that the actively functional anterograde and retrograde neuronal signaling is necessary to preserve synaptic memory engrams along the entorhinal cortex to the hippocampal trisynaptic circuit.
Collapse
Affiliation(s)
- Alejandro Carretero-Guillén
- Division of Neuroscience, University Pablo de Olavide, Seville, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Mario Treviño
- Max Planck Institute for Medical Research, Heidelberg, Germany
- Instituto de Neurociencias, Universidad de Guadalajara, Guadalajara, 44130, México
| | | | - Godwin K Dogbevia
- Max Planck Institute for Medical Research, Heidelberg, Germany
- Health Canada, Ottawa, ON, Canada
| | - Ilaria Bertocchi
- Max Planck Institute for Medical Research, Heidelberg, Germany
- Neuroscience Institute Cavalieri-Ottolenghi (NICO), University of Turin, Turin, Italy
| | - Rolf Sprengel
- Max Planck Institute for Medical Research, Heidelberg, Germany
| | | | | | - Agnès Gruart
- Division of Neuroscience, University Pablo de Olavide, Seville, Spain
| | | | - Mazahir T Hasan
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- Max Planck Institute for Medical Research, Heidelberg, Germany.
- NeuroCure, Charité - Universitätsmedizin, Berlin, Germany.
- Ikerbasque - Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
46
|
Niu X, Zhang Y, Wang Y. Co-culture models for investigating cellular crosstalk in the glioma microenvironment. CANCER PATHOGENESIS AND THERAPY 2024; 2:219-230. [PMID: 39371093 PMCID: PMC11447344 DOI: 10.1016/j.cpt.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 10/08/2024]
Abstract
Glioma is the most prevalent primary malignant tumor in the central nervous system (CNS). It represents a diverse group of brain malignancies characterized by the presence of various cancer cell types as well as an array of noncancerous cells, which together form the intricate glioma tumor microenvironment (TME). Understanding the interactions between glioma cells/glioma stem cells (GSCs) and these noncancerous cells is crucial for exploring the pathogenesis and development of glioma. To invesigate these interactions requires in vitro co-culture models that closely mirror the actual TME in vivo. In this review, we summarize the two- and three-dimensional in vitro co-culture model systems for glioma-TME interactions currently available. Furthermore, we explore common glioma-TME cell interactions based on these models, including interactions of glioma cells/GSCs with endothelial cells/pericytes, microglia/macrophages, T cells, astrocytes, neurons, or other multi-cellular interactions. Together, this review provides an update on the glioma-TME interactions, offering insights into glioma pathogenesis.
Collapse
Affiliation(s)
- Xiaodong Niu
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
47
|
Myung J. Enhancing Bioluminescence Imaging of Cultured Tissue Explants Using Optical Telecompression. SENSORS (BASEL, SWITZERLAND) 2024; 24:6041. [PMID: 39338785 PMCID: PMC11436007 DOI: 10.3390/s24186041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
Long-term observation of single-cell oscillations within tissue networks is now possible by combining bioluminescence reporters with stable tissue explant culture techniques. This method is particularly effective in revealing the network dynamics in systems with slow oscillations, such as circadian clocks. However, the low intensity of luciferase-based bioluminescence requires signal amplification using specialized cameras (e.g., I-CCDs and EM-CCDs) and prolonged exposure times, increasing baseline noise and reducing temporal resolution. To address this limitation, we implemented a cost-effective optical enhancement technique called telecompression, first used in astrophotography and now commonly used in digital photography. By combining a high numerical aperture objective lens with a magnification-reducing relay lens, we significantly increased the collection efficiency of the bioluminescence signal without raising the baseline CCD noise. This method allows for shorter exposure times in time-lapse imaging, enhancing temporal resolution and enabling more precise period estimations. Our implementation demonstrates the feasibility of telecompression for enhancing bioluminescence imaging for the tissue-level network observation of circadian clocks.
Collapse
Affiliation(s)
- Jihwan Myung
- Braintime Laboratory, Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan;
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
48
|
Steiner K, Yilmaz SN, Gern A, Marksteiner J, Faserl K, Villunger M, Sarg B, Humpel C. From Organotypic Mouse Brain Slices to Human Alzheimer Plasma Biomarkers: A Focus on Microglia. Biomolecules 2024; 14:1109. [PMID: 39334874 PMCID: PMC11430359 DOI: 10.3390/biom14091109] [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: 08/08/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Alzheimer's disease is a severe neurodegenerative disorder, and the discovery of biomarkers is crucial for early diagnosis. While the analysis of biomarkers in cerebrospinal fluid is well accepted, there are currently no blood biomarkers available. Our research focuses on identifying novel plasma biomarkers for Alzheimer's disease. To achieve this, we employed a technique that involves coupling human plasma to mouse organotypic brain slices via microcontact prints. After culturing for two weeks, we assessed Iba1-immunopositive microglia on these microcontact prints. We hypothesized that plasma from Alzheimer's patients contains factors that affect microglial migration. Our data indicated that plasma from Alzheimer's patients significantly inhibited the migration of round Iba1-immunoreactive microglia (13 ± 3, n = 24, p = 0.01) compared to healthy controls (50 ± 16, n = 23). Based on these findings, we selected the most promising plasma samples and conducted mass spectrometry using a differential approach, and we identified four potential biomarkers: mannose-binding protein C, macrophage receptor MARCO, complement factor H-related protein-3, and C-reactive protein. Our method represents a novel and innovative approach to translate research findings from mouse models to human applications.
Collapse
Affiliation(s)
- Katharina Steiner
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.S.); (S.N.Y.); (A.G.)
| | - Sakir Necat Yilmaz
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.S.); (S.N.Y.); (A.G.)
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, Mersin 33110, Turkey
| | - Alessa Gern
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.S.); (S.N.Y.); (A.G.)
| | - Josef Marksteiner
- Department of Psychiatry and Psychotherapy A, Hall State Hospital, 6060 Hall in Tirol, Austria;
| | - Klaus Faserl
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Mathias Villunger
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Bettina Sarg
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.S.); (S.N.Y.); (A.G.)
| |
Collapse
|
49
|
Carreira RB, Dos Santos CC, de Oliveira JVR, da Silva VDA, David JM, Butt AM, Costa SL. Neuroprotective Effect of Flavonoid Agathisflavone in the Ex Vivo Cerebellar Slice Neonatal Ischemia. Molecules 2024; 29:4159. [PMID: 39275007 PMCID: PMC11396859 DOI: 10.3390/molecules29174159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
Agathisflavone is a flavonoid that exhibits anti-inflammatory and anti-oxidative properties. Here, we investigated the neuroprotective effects of agathisflavone on central nervous system (CNS) neurons and glia in the cerebellar slice ex vivo model of neonatal ischemia. Cerebellar slices from neonatal mice, in which glial fibrillary acidic protein (GFAP) and SOX10 drive expression of enhanced green fluorescent protein (EGFP), were used to identify astrocytes and oligodendrocytes, respectively. Agathisflavone (10 μM) was administered preventively for 60 min before inducing ischemia by oxygen and glucose deprivation (OGD) for 60 min and compared to controls maintained in normal oxygen and glucose (OGN). The density of SOX-10+ oligodendrocyte lineage cells and NG2 immunopositive oligodendrocyte progenitor cells (OPCs) were not altered in OGD, but it resulted in significant oligodendroglial cell atrophy marked by the retraction of their processes, and this was prevented by agathisflavone. OGD caused marked axonal demyelination, determined by myelin basic protein (MBP) and neurofilament (NF70) immunofluorescence, and this was blocked by agathisflavone preventative treatment. OGD also resulted in astrocyte reactivity, exhibited by increased GFAP-EGFP fluorescence and decreased expression of glutamate synthetase (GS), and this was prevented by agathisflavone pretreatment. In addition, agathisflavone protected Purkinje neurons from ischemic damage, assessed by calbindin (CB) immunofluorescence. The results demonstrate that agathisflavone protects neuronal and myelin integrity in ischemia, which is associated with the modulation of glial responses in the face of ischemic damage.
Collapse
Affiliation(s)
- Rodrigo Barreto Carreira
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil
| | - Cleonice Creusa Dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil
| | - Juciele Valeria Ribeiro de Oliveira
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil
| | - Victor Diogenes Amaral da Silva
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil
| | - Jorge Maurício David
- Department of General and Inorganic Chemistry, Institute of Chemistry, University Federal da Bahia, Salvador 40170-110, BA, Brazil
| | - Arthur Morgan Butt
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil
- National Institute of Translational Neuroscience (INNT), Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
50
|
Yang JS, Jang HJ, Sung KW, Rhie DJ, Yoon SH. Roles of metabotropic glutamate receptor 5 in low [Mg 2+] o-induced interictal epileptiform activity in rat hippocampal slices. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:413-422. [PMID: 39198222 PMCID: PMC11362004 DOI: 10.4196/kjpp.2024.28.5.413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 09/01/2024]
Abstract
Group I metabotropic glutamate receptors (mGluRs) modulate postsynaptic neuronal excitability and epileptogenesis. We investigated roles of group I mGluRs on low extracellular Mg2+ concentration ([Mg2+]o)-induced epileptiform activity and neuronal cell death in the CA1 regions of isolated rat hippocampal slices without the entorhinal cortex using extracellular recording and propidium iodide staining. Exposure to Mg2+-free artificial cerebrospinal fluid can induce interictal epileptiform activity in the CA1 regions of rat hippocampal slices. MPEP, a mGluR 5 antagonist, significantly inhibited the spike firing of the low [Mg2+]o-induced epileptiform activity, whereas LY367385, a mGluR1 antagonist, did not. DHPG, a group 1 mGluR agonist, significantly increased the spike firing of the epileptiform activity. U73122, a PLC inhibitor, inhibited the spike firing. Thapsigargin, an ER Ca2+-ATPase antagonist, significantly inhibited the spike firing and amplitude of the epileptiform activity. Both the IP3 receptor antagonist 2-APB and the ryanodine receptor antagonist dantrolene significantly inhibited the spike firing. The PKC inhibitors such as chelerythrine and GF109203X, significantly increased the spike firing. Flufenamic acid, a relatively specific TRPC 1, 4, 5 channel antagonist, significantly inhibited the spike firing, whereas SKF96365, a relatively non-specific TRPC channel antagonist, did not. MPEP significantly decreased low [Mg2+]o DMEM-induced neuronal cell death in the CA1 regions, but LY367385 did not. We suggest that mGluR 5 is involved in low [Mg2+]oinduced interictal epileptiform activity in the CA1 regions of rat hippocampal slices through PLC, release of Ca2+ from intracellular stores and PKC and TRPC channels, which could be involved in neuronal cell death.
Collapse
Affiliation(s)
- Ji Seon Yang
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
| | - Hyun-Jong Jang
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
| | - Ki-Wug Sung
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Duck-Joo Rhie
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
| | - Shin Hee Yoon
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|