1
|
Claeys W, Geerts A, Van Hoecke L, Van Steenkiste C, Vandenbroucke RE. Role of astrocytes and microglia in hepatic encephalopathy associated with advanced chronic liver disease: lessons from animal studies. Neural Regen Res 2025; 20:3461-3475. [PMID: 39688562 PMCID: PMC11974659 DOI: 10.4103/nrr.nrr-d-24-00600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/05/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Hepatic encephalopathy, defined as neuropsychiatric dysfunction secondary to liver disease, is a frequent decompensating event in cirrhosis. Its clinical impact is highlighted by a notable increase in patient mortality rates and a concomitant reduction in overall quality of life. Systemically, liver disease, liver function failure, portosystemic shunting, and associated multi-organ dysfunction result in the increase of disease-causing neurotoxins in the circulation, which impairs cerebral homeostasis. Key circulating neurotoxins are ammonia and inflammatory mediators. In the brain, pathophysiology is less well understood, but is thought to be driven by glial cell dysfunction. Astrocytes are the only brain resident cells that have ammonia-metabolizing machinery and are therefore putatively most susceptible to ammonia elevation. Based on a large body of mostly in vitro evidence, ammonia-induced cellular and molecular disturbances include astrocyte swelling and oxidative stress. Microglia, the brain resident macrophages, have been linked to the translation of systemic inflammation to the brain microenvironment. Recent evidence from animal studies has provided novel insights into old and new downstream effects of astrocyte and microglial dysfunction such as toxin clearance disruption and myeloid cell attraction to the central nervous system parenchyma. Furthermore, state of the art research increasingly implicates neuronal dysfunction and possibly even irreversible neuronal cell death. Cell-type specific investigation in animal models highlights the need for critical revision of the contribution of astrocytes and microglia to well-established and novel cellular and molecular alterations in hepatic encephalopathy. In this review, we therefore give a current and comprehensive overview of causes, features, and consequences of astrocyte and microglial dysfunction in hepatic encephalopathy, including areas of interest for future investigation.
Collapse
Affiliation(s)
- Wouter Claeys
- Department of Internal Medicine and Paediatrics, Hepatology Research Unit, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Anja Geerts
- Department of Internal Medicine and Paediatrics, Hepatology Research Unit, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium
- Department of Gastroenterology and Hepatology, Ghent University Hospital, Ghent, Belgium
| | - Lien Van Hoecke
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Christophe Van Steenkiste
- Department of Gastroenterology and Hepatology, Antwerp University, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Maria Middelares Hospital, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
2
|
He Q, Yang Q, Wu L, He Y, Zeng N, Wang Z. Neurotoxic effects of per- and polyfluoroalkyl substances (PFAS) mixture exposure in mice: Accumulations in brain and associated changes of behaviors, metabolome, and transcriptome. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137699. [PMID: 39987740 DOI: 10.1016/j.jhazmat.2025.137699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/22/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Humans are exposed to complex per- and polyfluoroalkyl substances (PFAS) mixtures, yet their neurotoxicity and mechanisms remains unclear. This study exposed male mice to 17 PFAS mixtures at low levels (0.2-20 µg/L) for 49 days via drinking water. Perfluoropentanoic acid (PFPeA), perfluoroheptanoic acid (PFHpA), 6:2 fluorotelomer sulfonic acid (6:2 FTS), and perfluorooctane Sulfonate (PFOS) accumulated in brain tissues, with brain/plasma ratios of 2.03-5.87, 2.94-12.88, 1.90-3.19, and 0.62-0.93, respectively. Electroencephalogram (EEG) results showed significant alterations, including a reduction in beta spectral edge (21.47-13.85 Hz) and an increase in gamma spectral edge (57.64-79.07 Hz). Histopathological analysis revealed necrosis in the hippocampus, contributing to the observed anxiety-like behaviors and memory impairments in exposed mice. Plasma metabolomics highlighted disrupted osmoprotectants, impaired glutamatergic synapse function, and tryptophan metabolism. Brain metabolomics demonstrated suppression of purine metabolism and activation of arachidonic acid metabolism, suggesting involvement in neurotoxic effects. Transcriptomic profiling further identified dysregulation in neuroactive ligand-receptor interactions, cholinergic and GABAergic synapses, and calcium signaling pathways, with oxytocin signaling highlighted as a critical mechanism. This study, for the first time, links PFAS mixture to neurotoxicity via neurotransmitter-related pathways, underscoring the need for public health policies and preventive strategies to mitigate PFAS exposure risks.
Collapse
Affiliation(s)
- Qiurong He
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qingkun Yang
- West China School of Public Health, Sichuan University, Chengdu 610041, PR China
| | - Lin Wu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yuhang He
- West China School of Public Health, Sichuan University, Chengdu 610041, PR China
| | - Ni Zeng
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zhenglu Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
3
|
Orciani C, Foret MK, Cuello AC, Do Carmo S. Long-term nucleus basalis cholinergic lesions alter the structure of cortical vasculature, astrocytic density and microglial activity in Wistar rats. Neurobiol Aging 2025; 150:132-145. [PMID: 40121723 DOI: 10.1016/j.neurobiolaging.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Basal forebrain cholinergic neurons (BFCNs) are the sole source of cholinergic innervation to the cerebral cortex and hippocampus in humans and the primary source in rodents. This system undergoes early degeneration in Alzheimer's disease. BFCNs terminal synapses are involved in the regulation of the cerebral blood flow by making classical synaptic contacts with other neurons. Additionally, they are located in proximity to cortical cerebral blood vessels, forming connections with various cell types of the neurovascular unit (NVU), including vascular smooth muscle cells, endothelial cells, and astrocytic end-feet. However, the effects of the BFCNs input on NVU components remain unresolved. To address this issue, we immunolesioned the nucleus basalis by administering bilateral stereotaxic injections of the cholinergic immunotoxin 192-IgG-Saporin in 2.5-month-old Wistar rats. Seven months post-lesion, we observed a significant reduction in cortical vesicular acetylcholine transporter-immunoreactive synapses. This was accompanied by changes in the diameter of cortical capillaries and precapillary arterioles, as well as lower levels of vascular endothelial growth factor A (VEGF-A). Additionally, the cholinergic immunolesion increased the density of cortical astrocytes and microglia in the cortex. At these post-BFCN-lesion stages, astrocytic end-feet exhibited an increased co-localization with arterioles. The number of microglia in the parietal cortex correlated with cholinergic loss and exhibited morphological changes indicative of an intermediate activation state. This was supported by decreased levels of proinflammatory mediators IFN-γ, IL-1β, and KC/GRO (CXCL1), and by increased expression of M2 markers SOCS3, IL4Rα, YM1, ARG1, and Fizz1. Our findings offer a novel insight: that the loss of nucleus basalis cholinergic input negatively impacts cortical blood vessels, NVU components, and microglia phenotype.
Collapse
Affiliation(s)
- Chiara Orciani
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Morgan K Foret
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
| | - A Claudio Cuello
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada; Department of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada; Department of Anatomy & Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada; Department of Pharmacology, Oxford University, Oxford, UK.
| | - Sonia Do Carmo
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
4
|
Lavanco G, Castelli V, D'Amico C, Vaccaro F, Tringali G, Clementi ME, Bottoni P, Kuchar M, Palivec P, Engmann O, Brancato A, Cannizzaro C. Gestational THC exposure perturbates hippocampal mitochondrial respiration in the memory-impaired adolescent progeny: Is there a role for mitochondrial CB1 receptor? Biomed Pharmacother 2025; 187:118144. [PMID: 40339229 DOI: 10.1016/j.biopha.2025.118144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025] Open
Abstract
Mitochondria are central to cellular energy metabolism, contributing to synaptic transmission and plasticity. The mitochondrial membranes present the cannabinoid type-1 receptor (mito-CB1R), which has been functionally linked to neuronal energy supply and cognitive processing. Prenatal exposure to Δ9-tetrahydrocannabinol (pTHC) has been associated with cognitive impairments associated with molecular cellular and functional abnormalities in several brain regions, including the hippocampus. This study aims at assessing whether, besides the memory impairment, pTHC exposure may result in mitochondrial molecular and functional alterations in the hippocampus of the offspring. Moreover, the assessment of CB1R expression is also carried out as a proxy of CB1 signalling in pTHC-exposed offspring. THC (2 mg/Kg), or vehicle, was administered to the dams from gestational day (GD) 5 to GD20, and the offspring were tested for declarative memory using the Novel Object Recognition test in the L-maze. We also assessed: mitochondrial respiration by high-resolution respirometry; mitochondrial respiratory complex-I subunit NDUFS1 protein levels, and mito-CB1R expression by ELISA. Our results revealed: significant memory impairment in pTHC-exposed offspring; attenuated mitochondrial respiration in the hippocampus alongside a marked reduction in complex-I-subunit NDUFS1; a significant increase in mito-CB1R expression. This is the first evidence of pTHC exposure-induced impairment in memory processing in the offspring that suggests a functional link between an attenuation in mitochondrial bioenergetics and abnormal CB1R signalling in the hippocampus.
Collapse
Affiliation(s)
- Gianluca Lavanco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties of Excellence "G. D'Alessandro", University of Palermo, Palermo, Italy.
| | - Valentina Castelli
- University of Palermo, Dept. of Biomedicine, Neuroscience and Advanced Diagnostics, via del Vespro 129, Palermo 90127, Italy
| | - Cesare D'Amico
- University of Palermo, Dept. of Biomedicine, Neuroscience and Advanced Diagnostics, via del Vespro 129, Palermo 90127, Italy
| | - Francesca Vaccaro
- University of Palermo, Dept. of Biomedicine, Neuroscience and Advanced Diagnostics, via del Vespro 129, Palermo 90127, Italy
| | - Giuseppe Tringali
- Pharmacology Section, Department of Healthcare Surveillance and Bioethics, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome, Italy
| | - Maria Elisabetta Clementi
- CNR‑ICRM Institute of 'Chemistry of Molecular Recognition', Institute of Biochemistry and Clinical Biochemistry, Catholic University Medical School, Rome, Italy
| | - Patrizia Bottoni
- Department of Basic Biotechnology Sciences, Intensive Care and Perioperative Clinics, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Martin Kuchar
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague, Czechia; Psychedelics Research Centre, National Institute of Mental Health, Prague, Czechia
| | - Petr Palivec
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague, Czechia; Psychedelics Research Centre, National Institute of Mental Health, Prague, Czechia
| | - Olivia Engmann
- Institute for Biochemistry and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany; Institute of Human Genetics, Jena University Hospital, Am Klinikum 1, F2E20, Jena 07747, Germany
| | - Anna Brancato
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties of Excellence "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Carla Cannizzaro
- University of Palermo, Dept. of Biomedicine, Neuroscience and Advanced Diagnostics, via del Vespro 129, Palermo 90127, Italy
| |
Collapse
|
5
|
Yogesh B, Heindorf M, Jordan R, Keller GB. Quantification of the effect of hemodynamic occlusion in two-photon imaging of mouse cortex. eLife 2025; 14:RP104914. [PMID: 40434064 PMCID: PMC12119086 DOI: 10.7554/elife.104914] [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] [Indexed: 05/29/2025] Open
Abstract
The last few years have seen an explosion in the number of tools available to measure neuronal activity using fluorescence imaging (Chen et al., 2013; Feng et al., 2019; Jing et al., 2019; Sun et al., 2018; Wan et al., 2021). When performed in vivo, these measurements are invariably contaminated by hemodynamic occlusion artifacts. In widefield calcium imaging, this problem is well recognized. For two-photon imaging, however, the effects of hemodynamic occlusion have only been sparsely characterized. Here, we perform a quantification of hemodynamic occlusion effects using measurements of fluorescence changes observed with GFP expression using both widefield and two-photon imaging in mouse cortex. We find that in many instances the magnitude of signal changes attributable to hemodynamic occlusion is comparable to that observed with activity sensors. Moreover, we find that hemodynamic occlusion effects were spatially heterogeneous, both over cortical regions and across cortical depth, and exhibited a complex relationship with behavior. Thus, hemodynamic occlusion is an important caveat to consider when analyzing and interpreting not just widefield but also two-photon imaging data.
Collapse
Affiliation(s)
- Baba Yogesh
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Faculty of Natural Sciences, University of BaselBaselSwitzerland
| | - Matthias Heindorf
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Rebecca Jordan
- Simons Initiative for the Developing Brain, University of EdinburghEdinburghUnited Kingdom
| | - Georg B Keller
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Faculty of Natural Sciences, University of BaselBaselSwitzerland
| |
Collapse
|
6
|
Wang J, Zhou J, Zhu J, Sheng J, Jiang R, Zhang X. Brain remodeling in stroke patients: A comprehensive review of mechanistic and neuroimaging studies. Behav Brain Res 2025; 486:115548. [PMID: 40122286 DOI: 10.1016/j.bbr.2025.115548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Stroke-induced brain remodeling involves a complex interplay of neurovascular components, including endothelial cells, microglia, astrocytes, and pericytes, which collectively contribute to the restoration of brain function. These processes are crucial for repairing the blood-brain barrier, regulating inflammation, and promoting neurogenesis. This review examines the mechanisms underlying brain remodeling and the role of advanced neuroimaging techniques-such as functional MRI (fMRI), positron emission tomography (PET), functional near-infrared spectroscopy (fNIRS), and functional ultrasound (fUS)-in assessing these changes. We also discuss various therapeutic approaches aimed at enhancing brain remodeling, including pharmacological agents, stem cell therapy, and rehabilitation strategies that target neurovascular repair and functional recovery. Despite significant progress, challenges remain in translating imaging insights into effective treatments. Future research should focus on integrating multiple imaging modalities to provide a comprehensive view of neurovascular changes and refining therapeutic interventions to optimize recovery and functional outcomes in stroke patients.
Collapse
Affiliation(s)
- Jing Wang
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Jian Zhou
- Department of Radiology, No. 945 Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army, Yaan, Sichuan 625000, China.
| | - Jing Zhu
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Jinping Sheng
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Rui Jiang
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Xiao Zhang
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| |
Collapse
|
7
|
Artemiev D, Valmaggia C, Tschuppert S, Kotliar K, Türksever C, Todorova MG. Retinal Vessel Flicker Light Responsiveness and Its Relation to Analysis Protocols and Static and Metabolic Data in Healthy Subjects. Biomedicines 2025; 13:1201. [PMID: 40427028 PMCID: PMC12108832 DOI: 10.3390/biomedicines13051201] [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: 03/19/2025] [Revised: 04/15/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Background: The aim of this study was to assess the agreement between different analysis protocols for the determination of retinal vessel dilation response to flicker light (FL) and its relation to static and metabolic parameters of retinal vessels in healthy subjects. Methods: In total, 24 right eyes of 24 healthy controls (mean age: 36.04 ± SD 14.4 years) who underwent dynamic and static retinal diameter and oxygen saturation measurements on a Retinal Vessel Analyzer (RVA, Imedos, Jena, Germany) were included. Using repeated video analyses, responses to FL were measured with RVA. These measurements were conducted at three specific retinal locations: within the superotemporal area-within a distance of less than one optic disk (OD) diameter to optic nerve head (ONH) (group 1); greater than one OD diameter to ONH (group 2); and areas near the ONH within the VesselMap region (group 3). For comparability, the static and oxygen saturation parameters were also calculated in the superotemporal peripapillary area using the VesselMap tool of the RVA and were evaluated in relation to the corresponding dynamic area (group 3). Results: In all groups, the vascular FL response of arteries was less pronounced compared to venules (p = 0.0014). Even though FL responses (mean ± SD: FL-A; FL-V) in group 1 were more pronounced (3.36 ± 2.31; 4.42 ± 1.69) compared to those in group 2 (2.97 ± 2.40; 4.08 ± 1.55) and group 3 (2.84 ± 2.29; 4.21 ± 2.03), they did not reach statistically significant values. The mean flicker response of venules (VDil) in all groups showed negative correlations to the corresponding static parameter: central retinal venous equivalent (CRV) (r = -0.0437; p = 0.015). The mean flicker response of arteries (ADil) in all groups showed negative correlations to the corresponding metabolic parameter: arterio-venous oxygen extraction fraction (r = -0.101; p = 0.041). Conclusions: Our study confirms that the flicker light response, despite slight variations in its duration and location, allows for reliable measurements, proving the Retinal Vessel Analyzer to be a valuable diagnostic tool. Furthermore, we were able to highlight the relationship between the dynamic and metabolic components of retinal supply, which enables early diagnosis concerning the development of diseases within this spectrum.
Collapse
Affiliation(s)
- Dmitri Artemiev
- Department of Ophthalmology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland (M.G.T.)
| | - Christophe Valmaggia
- Department of Ophthalmology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland (M.G.T.)
- Medical Faculty, University of Zurich, CH-8008 Zurich, Switzerland
| | - Scott Tschuppert
- Department of Ophthalmology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland (M.G.T.)
| | - Konstantin Kotliar
- Department of Medical Engineering and Technomathematics, FH Aachen University of Applied Sciences, 52428 Juelich, Germany
| | - Cengiz Türksever
- Department of Ophthalmology, VISTA Clinic, 4102 Binningen, Switzerland
| | - Margarita G. Todorova
- Department of Ophthalmology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland (M.G.T.)
- Medical Faculty, University of Zurich, CH-8008 Zurich, Switzerland
| |
Collapse
|
8
|
Czpakowska J, Głąbiński A, Szpakowski P. The Potential Role of Exosomes in Communication Between Astrocytes and Endothelial Cells. Int J Mol Sci 2025; 26:4676. [PMID: 40429819 PMCID: PMC12111803 DOI: 10.3390/ijms26104676] [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: 04/05/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Exosomes are extracellular vesicles secreted by almost all types of cells. Their release allows for the transport of specific regulatory cargo into the recipient cells and the modulation of their activity. Vesicular communication has also been identified as an important mechanism for the regulation of numerous cellular activities in the brain tissue, contributing to proper neuronal functions and brain homeostasis. In this work, we focus on the role of exosomes and extracellular vesicles in the communication between astrocytes and brain endothelial cells, two major components of the blood-brain barrier. We perform a comprehensive review of the latest studies highlighting the role of exosomes in astrocyte-endothelial cell crosstalk within the blood-brain barrier. We have also described the role of particular exosomal miRNAs in the regulation of astrocytes and brain endothelial cell functions, and discuss some future implications.
Collapse
Affiliation(s)
| | - Andrzej Głąbiński
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland;
| | - Piotr Szpakowski
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland;
| |
Collapse
|
9
|
Tirado-García P, Ferreiro A, González-Alday R, Arias-Ramos N, Lizarbe B, López-Larrubia P. Aquaporin-4 inhibition alters cerebral glucose dynamics predominantly in obese animals: an MRI study. Sci Rep 2025; 15:15649. [PMID: 40325102 PMCID: PMC12052982 DOI: 10.1038/s41598-025-99641-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
Glucose uptake and metabolism are linked to microvascular blood flow and cellular swelling events, which are altered during obesity and can be quantified using magnetic resonance imaging (MRI). Aquaporin-4 (AQP4), the most abundant water-transporting transmembrane protein in the central nervous system, facilitates glucose transport and metabolism-derived water influx. However, its significance and regulatory capacity remain largely unknown. To better understand these processes, we acquired sequential diffusion tensor and T2*-weighted images of the brains of obese and non-obese mice, both before administering an AQP4 inhibitor and after a subsequent glucose challenge. We then subjected the resulting variables to principal component and linear mixed model analyses to assess the influence of diet, sex, administration of the inhibitor, and brain region on the data. Our findings indicate that AQP4-inhibited mice exhibit MRI values consistent with reduced microvascular blood flow and region-specific inhibition of glucose-induced cell swelling during obesity, highlighting a key role for AQP4 in glucose uptake and metabolism. Additionally, we observed that, prior to any experimental manipulation, obese mice displayed MRI signs of lower hippocampal blood flow and cerebral cellular anisotropy compared to controls, in agreement with vascular alterations and reactive gliosis processes.
Collapse
Affiliation(s)
- Pablo Tirado-García
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain
| | - Adriana Ferreiro
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain
| | - Raquel González-Alday
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain
| | - Nuria Arias-Ramos
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain
| | - Blanca Lizarbe
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain.
- Department of Biochemistry, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Pilar López-Larrubia
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain.
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
10
|
Lacoste B, Prat A, Freitas-Andrade M, Gu C. The Blood-Brain Barrier: Composition, Properties, and Roles in Brain Health. Cold Spring Harb Perspect Biol 2025; 17:a041422. [PMID: 38951020 PMCID: PMC12047665 DOI: 10.1101/cshperspect.a041422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Blood vessels are critical to deliver oxygen and nutrients to tissues and organs throughout the body. The blood vessels that vascularize the central nervous system (CNS) possess unique properties, termed the blood-brain barrier (BBB), which allow these vessels to tightly regulate the movement of ions, molecules, and cells between the blood and the brain. This precise control of CNS homeostasis allows for proper neuronal function and protects the neural tissue from toxins and pathogens, and alterations of this barrier are important components of the pathogenesis and progression of various neurological diseases. The physiological barrier is coordinated by a series of physical, transport, and metabolic properties possessed by the brain endothelial cells (ECs) that form the walls of the blood vessels. These properties are regulated by interactions between different vascular, perivascular, immune, and neural cells. Understanding how these cell populations interact to regulate barrier properties is essential for understanding how the brain functions in both health and disease contexts.
Collapse
Affiliation(s)
- Baptiste Lacoste
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, Ontario K1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada
| | - Alexandre Prat
- Department of Neuroscience, Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Moises Freitas-Andrade
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, Ontario K1H 8M5, Canada
| | - Chenghua Gu
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
11
|
Dokic I, Moustafa M, Tessonnier T, Meister S, Ciamarone F, Akbarpour M, Krunic D, Haberer T, Debus J, Mairani A, Abdollahi A. Ultrahigh Dose Rate Helium Ion Beams: Minimizing Brain Tissue Damage while Preserving Tumor Control. Mol Cancer Ther 2025; 24:763-771. [PMID: 39739545 PMCID: PMC12046314 DOI: 10.1158/1535-7163.mct-24-0536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/16/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
Ultrahigh dose rate radiotherapy (RT) with electrons and protons has shown potential for cancer treatment by effectively targeting tumors while sparing healthy tissues (FLASH effect). This study aimed to investigate the potential FLASH sparing effect of ultrahigh dose rate helium ion irradiation, focusing on acute brain injury and subcutaneous tumor response in a preclinical in vivo setting. Raster-scanned helium ion beams were used to compare the effects of standard dose rate (SDR; at 0.2 Gy/second) and FLASH (at 141 Gy/second) RT on healthy brain tissue. Irradiation-induced brain injury was studied in C57BL/6 mice via DNA damage response, using nuclear γH2AX as a marker for double-strand breaks. The integrity of neurovascular and immune compartments was assessed through CD31+ microvascular density and activation of microglia/macrophages. IBA1+ ramified and CD68+ phagocytic microglia/macrophages were quantified, along with the expression of inducible nitric oxide synthetase. Tumor response to SDR (0.2 Gy/second) and FLASH (250 Gy/second) RT was evaluated in an A549 carcinoma model, using tumor volume and Kaplan-Meier survival as endpoints. The results showed that helium FLASH RT significantly reduced acute brain tissue injury compared with SDR, evidenced by lower levels of double-strand breaks and preserved the neurovascular endothelium. Additionally, FLASH RT reduced neuroinflammatory signals compared with SDR, as indicated by fewer CD68+ inducible nitric oxide synthetase-positive microglia/macrophages. FLASH RT achieved tumor control comparable with that of SDR RT. To the best of our knowledge, this is the first study to report the FLASH sparing effect of raster scanning helium ion RT in vivo, highlighting its potential for neuroprotection and effective tumor control.
Collapse
Affiliation(s)
- Ivana Dokic
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Mahmoud Moustafa
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- Department of Clinical Pathology, Suez Canal University, Ismailia, Egypt
| | - Thomas Tessonnier
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Sarah Meister
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Federica Ciamarone
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Mahdi Akbarpour
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Damir Krunic
- Light Microscopy Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Haberer
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Jürgen Debus
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Andrea Mairani
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Amir Abdollahi
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| |
Collapse
|
12
|
Yoshikawa T, Tateno T. Localized Theta-Burst Magnetic Stimulation Induces Bidirectional Neural Modulation in the Mouse Auditory Cortex In Vivo. eNeuro 2025; 12:ENEURO.0577-24.2025. [PMID: 40246552 PMCID: PMC12077811 DOI: 10.1523/eneuro.0577-24.2025] [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: 12/18/2024] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a noninvasive method that has been used to treat various brain disorders. The modulatory effects of rTMS can be adjusted by changing the repetition patterns. Theta-burst magnetic stimulation (TBS) is a magnetic stimulation pattern that can induce long-lasting modulatory effects with a short stimulation period. However, its effects on auditory brain regions remain unclear because of a lack of animal studies in which invasive techniques allow for a detailed exploration of the underlying neural mechanisms. In the current study, we investigated the effects of TBS on the C57BL/6J mouse auditory cortex using a custom-built 7 mm magnetic stimulation coil. Extracellular recordings were made before, during, and after the application of intermittent TBS (iTBS), continuous TBS (cTBS), or sham stimulation. Local field potential amplitudes were increased for 5-20 min post-iTBS compared with the sham condition and were decreased at 10 min post-cTBS compared with the sham condition. The bidirectional modulatory effects observed in our study are consistent with previous findings from other brain regions. Additionally, multiunit activities were significantly altered in cortical layers 2/3 and 4 but not layer 5, indicating that the modulatory effects were localized to the surface region of the auditory cortex. Interestingly, in the iTBS group, the amplitude of average spike waveforms increased with a 15 min delay. Our findings provide physiological evidence of TBS modulation of the rodent auditory cortex and may guide future research seeking to optimize rTMS for modulating hearing abilities.
Collapse
Affiliation(s)
- Takahiro Yoshikawa
- Bioengineering and Bioinformatics, Graduate School of Information Science and Technology, Hokkaido University, Sapporo 060-0814, Japan
| | - Takashi Tateno
- Division of Bioengineering and Bioinformatics, Faculty of Information Science and Technology, Hokkaido University, Sapporo 060-0814, Japan
| |
Collapse
|
13
|
Divecha YA, Rampes S, Tromp S, Boyanova ST, Fleckney A, Fidanboylu M, Thomas SA. The microcirculation, the blood-brain barrier, and the neurovascular unit in health and Alzheimer disease: The aberrant pericyte is a central player. Pharmacol Rev 2025; 77:100052. [PMID: 40215558 DOI: 10.1016/j.pharmr.2025.100052] [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: 07/19/2024] [Accepted: 02/28/2025] [Indexed: 05/27/2025] Open
Abstract
High fidelity neuronal signaling is enabled by a stable local microenvironment. A high degree of homeostatic regulation of the brain microenvironment, and its separation from the variable and potentially neurotoxic contents of the blood, is brought about by the central nervous system barriers. Evidence from clinical and preclinical studies implicates brain microcirculation, cerebral hypoperfusion, blood-brain barrier dysfunction, and reduced amyloid clearance in Alzheimer pathophysiology. Studying this dysregulation is key to understanding Alzheimer disease (AD), identifying drug targets, developing treatment strategies, and improving prescribing to this vulnerable population. This review has 2 parts: part 1 describes the cerebral microcirculation, cerebral blood flow, extracellular fluid drainage, and the neurovascular unit components with an emphasis on the blood-brain barrier, and part 2 summarizes how each aspect is altered in AD. Discussing the neurovascular unit structures separately allows us to conclude that aberrant pericytes are an early contributor and central to understanding AD pathophysiology. Pericytes have multiple functions including maintenance of blood-brain barrier integrity and the control of capillary blood flow, capillary stalling, neurovascular coupling, intramural periarterial drainage, glia-lymphatic (glymphatic) drainage, and consequently amyloid and tau clearance. Pericytes are vasoactive, express cholinergic and adrenergic receptors, and exhibit apolipoprotein E isoform-specific transport pathways. Hypoperfusion in AD is linked to a pericyte-mediated response. Deficient endothelial cell-pericyte (PDGBB-PDGFRβ) signaling loops cause pericyte dysfunction, which contributes and even initiates AD degeneration. We conclude that pericytes are central to understanding AD pathophysiology, are an interesting therapeutic target in AD, and have an emerging role in regenerative therapy. SIGNIFICANCE STATEMENT: Dysregulation and dysfunction of the neurovascular unit and fluid circulation (including blood, cerebrospinal fluid, and interstitial fluid) occurs in Alzheimer disease. A central player is the aberrant pericyte. This has fundamental implications to understanding disease pathophysiology and the development of therapies.
Collapse
Affiliation(s)
- Yasmin Amy Divecha
- King's College London, Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, Waterloo, London, United Kingdom
| | - Sanketh Rampes
- King's College London, Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, Waterloo, London, United Kingdom
| | - Sabine Tromp
- King's College London, Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, Waterloo, London, United Kingdom
| | - Sevda T Boyanova
- King's College London, Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, Waterloo, London, United Kingdom
| | - Alice Fleckney
- King's College London, Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, Waterloo, London, United Kingdom
| | - Mehmet Fidanboylu
- King's College London, Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, Waterloo, London, United Kingdom
| | - Sarah Ann Thomas
- King's College London, Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, Waterloo, London, United Kingdom.
| |
Collapse
|
14
|
Valjakka JS, Paasonen J, Salo RA, Paasonen E, Stenroos P, Gureviciene I, Kettunen M, Idiyatullin D, Tanila H, Michaeli S, Mangia S, Gröhn O. Correlation of zero echo time functional MRI with neuronal activity in rats. J Cereb Blood Flow Metab 2025; 45:855-870. [PMID: 39846159 PMCID: PMC11758440 DOI: 10.1177/0271678x251314682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/20/2024] [Accepted: 01/03/2025] [Indexed: 01/24/2025]
Abstract
Zero echo time (zero-TE) pulse sequences provide a quiet and artifact-free alternative to conventional functional magnetic resonance imaging (fMRI) pulse sequences. The fast readouts (<1 ms) utilized in zero-TE fMRI produce an image contrast with negligible contributions from blood oxygenation level-dependent (BOLD) mechanisms, yet the zero-TE contrast is highly sensitive to brain function. However, the precise relationship between the zero-TE contrast and neuronal activity has not been determined. Therefore, we aimed to derive a function to model the temporal dynamics of the zero-TE fMRI signal in response to neuronal activity. Furthermore, we examined the correlation of zero-TE fMRI with neuronal activity across stimulation frequencies. To these ends, we performed simultaneous electrophysiological recordings and zero-TE fMRI in rats subjected to whisker stimulation. The presented impulse response function provides a basis for the statistical modeling of neuronal activity-induced changes in the zero-TE fMRI signal. The temporal characteristics of the zero-TE fMRI response were found to be consistent with the previously postulated non-BOLD hemodynamic origin of the functional contrast. The zero-TE fMRI signal was well predicted by electrophysiological recordings, although systematic stimulation-dependent residuals were also observed, suggesting nonlinearities in neurovascular coupling. We conclude that zero-TE fMRI provides a robust proxy for neuronal activity.
Collapse
Affiliation(s)
- Juha S Valjakka
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, USA
| | - Jaakko Paasonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Raimo A Salo
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ekaterina Paasonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neurocenter, Kuopio University Hospital, Kuopio, Finland
| | - Petteri Stenroos
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Irina Gureviciene
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mikko Kettunen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Djaudat Idiyatullin
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, USA
| | - Heikki Tanila
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Shalom Michaeli
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, USA
| | - Silvia Mangia
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, USA
| | - Olli Gröhn
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
15
|
Zheng J, Wang M, Wang S, Shao Z. Temperature Regulates Astroglia Morphogenesis Through Thermosensory Circuitry in Caenorhabditis elegans. Glia 2025; 73:985-1003. [PMID: 39780488 DOI: 10.1002/glia.24668] [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: 07/21/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
Astrocytes are the most abundant type of macroglia in the brain and play crucial roles in regulating neural development and functions. The diverse functions of astrocytes are largely determined by their morphology, which is regulated by genetic and environmental factors. However, whether and how the astrocyte morphology is affected by temperature remains largely unknown. Here we discovered that elevated cultivation temperature (26°C) stimulates Caenorhabditis elegans ventral CEPsh glia endfoot extension during early developmental stages. This extension depends on the activation of glutamate AWC neurons, which inhibit the postsynaptic cholinergic AIY interneurons through glutamate-gated chloride channels, GLC-3 and GLC-4. In responding to the thermosensory signal, the guanyl-nucleotide exchange factor EPHX-1 and Rho GTPase CDC-42/Cdc42 in the glia facilitate the endfoot extension via F-actin assembly. This study elucidates the significant role of thermosensory circuitry in glia morphogenesis and the underlying molecular mechanism.
Collapse
Affiliation(s)
- Junyu Zheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Mengqing Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Shaocheng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Zhiyong Shao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| |
Collapse
|
16
|
Jobling AI, Greferath U, Dixon MA, Quiriconi P, Eyar B, van Koeverden AK, Mills SA, Vessey KA, Bui BV, Fletcher EL. Microglial regulation of the retinal vasculature in health and during the pathology associated with diabetes. Prog Retin Eye Res 2025; 106:101349. [PMID: 40020909 DOI: 10.1016/j.preteyeres.2025.101349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The high metabolic demand of retinal neurons requires a precisely regulated vascular system that can deliver rapid changes in blood flow in response to neural need. In the retina, this is achieved via the action of a coordinated group of cells that form the neurovascular unit. While cells such as pericytes, Müller cells, and astrocytes have long been linked to neurovascular coupling, more recently the resident microglial population have also been implicated. In the healthy retina, microglia make extensive contact with blood vessels, as well as neuronal synapses, and are important in vascular patterning during development. Work in the brain and retina has recently indicated that microglia can directly regulate the local vasculature. In the retina, the fractalkine-Cx3cr1 signalling axis has been shown to induce local capillary constriction within the superficial vascular plexus via a mechanism involving components of the renin-angiotensin system. Furthermore, aberrant microglial induced vasoconstriction may be at the centre of early vascular reactivity changes observed in those with diabetes. This review summarizes the recent emerging evidence that microglia play multiple roles in retinal homeostasis especially in regulating the vasculature. We highlight what is known about the role of microglia under normal circumstances, and then build on this to discuss how microglia contribute to early vascular compromise during diabetes. Further understanding of the mechanisms of microglial-vascular regulation may allow alternate treatment strategies to be devised to reduce vascular pathology in diseases such as diabetic retinopathy.
Collapse
Affiliation(s)
- Andrew I Jobling
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Ursula Greferath
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Michael A Dixon
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Pialuisa Quiriconi
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Belinda Eyar
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Anna K van Koeverden
- Department of Optometry and Vision Sciences, The University of Melbourne, Victoria, Australia
| | - Samuel A Mills
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Kirstan A Vessey
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
17
|
Longden T, Isaacs D. Pericyte Electrical Signalling and Brain Haemodynamics. Basic Clin Pharmacol Toxicol 2025; 136:e70030. [PMID: 40159653 PMCID: PMC11955720 DOI: 10.1111/bcpt.70030] [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: 12/21/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
Dynamic control of membrane potential lies at the nexus of a wide spectrum of biological processes, ranging from the control of individual cell secretions to the orchestration of complex thought and behaviour. Electrical signals in all vascular cell types (smooth muscle cells, endothelial cells and pericytes) contribute to the control of haemodynamics and energy delivery across spatiotemporal scales and throughout all tissues. Here, our goal is to review and synthesize key studies of electrical signalling within the brain vasculature and integrate these with recent data illustrating an important electrical signalling role for pericytes, in doing so attempting to work towards a holistic description of blood flow control in the brain by vascular electrical signalling. We use this as a framework for generating further questions that we believe are important to pursue. Drawing parallels with electrical signal integration in the nervous system may facilitate deeper insights into how signalling is organized within the vasculature and how it controls blood flow at the network level.
Collapse
Affiliation(s)
- Thomas A. Longden
- Department of Pharmacology and PhysiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Dominic Isaacs
- Department of Pharmacology and PhysiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Program in NeuroscienceUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
18
|
Li W, Qiu X, Chen J, Chen K, Chen M, Wang Y, Sun W, Su J, Chen Y, Liu X, Chu C, Wang J. Disentangling the Switching Behavior in Functional Connectivity Dynamics in Autism Spectrum Disorder: Insights from Developmental Cohort Analysis and Molecular-Cellular Associations. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403801. [PMID: 40344520 PMCID: PMC12120798 DOI: 10.1002/advs.202403801] [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: 04/11/2024] [Revised: 04/21/2025] [Indexed: 05/11/2025]
Abstract
Characterizing the transition or switching behavior between multistable brain states in functional connectivity dynamics (FCD) holds promise for uncovering the underlying neuropathology of Autism Spectrum Disorder (ASD). However, whether and how switching behaviors in FCD change in patients with developmental ASD, as well as their cellular and molecular basis, remains unexplored. This study develops a region-wise FCD switching index (RFSI) to investigate the drivers of FCD. This work finds that brain regions within the salience, default mode, and frontoparietal networks serve as abnormal drivers of FCD in ASD across different developmental stages. Additionally, changes in RFSI at different developmental stages of ASD correlated with transcriptomic profiles and neurotransmitter density maps. Importantly, the abnormal RFSI identifies in humans has also been observed in genetically edited ASD monkeys. Finally, single-nucleus RNA sequencing data from patients with developmental ASD are analyzed and aberrant switching behaviors in FCD may be mediated by somatostatin-expressing interneurons and altered differentiation patterns in astrocyte State2. In conclusion, this study provides the first evidence of abnormal drivers of FCD across different stages of ASD and their associated cellular and molecular mechanisms. These findings deepen the understanding of ASD neuropathology and offer valuable insights into treatment strategies.
Collapse
Affiliation(s)
- Wei Li
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunming650500China
- Faculty of Mechanical and Electrical EngineeringKunming University of Science and TechnologyKunming650500China
| | - Xia Qiu
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunming650500China
| | - Jin Chen
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunming650500China
| | - Kexuan Chen
- Medical SchoolKunming University of Science and TechnologyKunming650500China
| | - Meiling Chen
- Department of Clinical Psychologythe First People's Hospital of Yunnan ProvinceThe Affiliated Hospital of Kunming University of Science and TechnologyKunming650500China
| | - Yinyan Wang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
| | - Wenjie Sun
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunming650500China
| | - Jing Su
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunming650500China
| | - Yongchang Chen
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunming650500China
| | - Xiaobao Liu
- Faculty of Mechanical and Electrical EngineeringKunming University of Science and TechnologyKunming650500China
| | - Congying Chu
- Brainnetome Center & National Laboratory of Pattern RecognitionInstitute of AutomationChinese Academy of SciencesBeijing100190China
| | - Jiaojian Wang
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunming650500China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunming650500China
| |
Collapse
|
19
|
Le Gac B, Tournissac M, Belzic E, Picaud S, Dusart I, Soula H, Li D, Charpak S, Cauli B. Elevated pyramidal cell firing orchestrates arteriolar vasoconstriction through COX-2-derived prostaglandin E2 signaling. eLife 2025; 13:RP102424. [PMID: 40266667 PMCID: PMC12017770 DOI: 10.7554/elife.102424] [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: 04/24/2025] Open
Abstract
Neurovascular coupling, linking neuronal activity to cerebral blood flow, is essential for brain function and underpins functional brain imaging. Whereas mechanisms involved in vasodilation are well-documented, those controlling vasoconstriction remain overlooked. This study unravels the mechanisms by which pyramidal cells elicit arteriole vasoconstriction. Using patch-clamp recording, vascular and Ca2+ imaging in mouse cortical slices, we show that strong optogenetic activation of layer II/III pyramidal cells induces vasoconstriction, correlating with firing frequency and somatic Ca2+ increase. Ex vivo and in vivo pharmacological investigations indicate that this vasoconstriction predominantly recruits prostaglandin E2 through the cyclooxygenase-2 pathway, and activation of EP1 and EP3 receptors. We also present evidence that specific interneurons releasing neuropeptide Y, and astrocytes, through 20-hydroxyeicosatetraenoic acid, contribute to this process. By revealing the mechanisms by which pyramidal cells lead to vasoconstriction, our findings shed light on the complex regulation of neurovascular coupling.
Collapse
Affiliation(s)
- Benjamin Le Gac
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| | - Marine Tournissac
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012ParisFrance
| | - Esther Belzic
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| | - Sandrine Picaud
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| | - Isabelle Dusart
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| | - Hédi Soula
- Sorbonne Université, INSERM, Nutrition and Obesities: Systemic Approaches, NutriOmics, Research UnitParisFrance
| | - Dongdong Li
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| | - Serge Charpak
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012ParisFrance
| | - Bruno Cauli
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| |
Collapse
|
20
|
Pan Y, Bi C, Ye Z, Lee H, Yu J, Yammine L, Ma T, Kochunov P, Hong LE, Chen S. Tobacco Smoking Functional Networks: A Whole-Brain Connectome Analysis in 24 539 Individuals. Nicotine Tob Res 2025; 27:917-925. [PMID: 39468718 DOI: 10.1093/ntr/ntae256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/04/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
INTRODUCTION Nicotine addiction, a multifaceted neuropsychiatric disorder, profoundly impacts brain functions through interactions with neural pathways. Despite its significance, the impact of tobacco smoking on the whole-brain functional connectome remains largely unexplored. AIMS AND METHODS We conducted a whole-brain analysis on 24 539 adults aged 40 and above from the United Kingdom Biobank cohort. Subjects were categorized into individuals who use nicotine and those who do not use nicotine based on current and chronic tobacco smoking information. Functional connectivity was assessed using resting-state functional magnetic resonance imaging. We employed a network analysis method to assess the systematic effects of tobacco smoking on brain connectome by identifying subnetworks that show nicotine-use-related differences. RESULTS Our analyses revealed two nicotine-use-related subnetworks with distinct network structure (permutation p < .001). In the first network, there is a significant decrease in resting-state functional connectivity (rsFC) between the basal ganglia regions (eg, nucleus accumbens) and 73% of the remaining brain regions, emphasizing the central hub role of basal ganglia in addictive smoking behaviors. Additionally, a data-driven subnetwork, mainly involving regions from frontal and occipital lobes, showed reduced rsFC among individuals who use nicotine. CONCLUSIONS The results suggest significant alterations in the communication and coordination among the basal ganglia and the broader network of brain regions. The observed changes in rsFC indicate a widespread disruption in the connectivity patterns associated with nicotine use. IMPLICATIONS This study identifies rsFC subnetworks related to chronic nicotine use through whole-brain connectome analysis. The findings confirm that widespread alterations in rsFC are centered around hub nodes within the basal ganglia, including bilateral nucleus accumbens, putamen, caudate, and globus pallidus. In addition, our analysis found a clique-forming subnetwork vulnerable to tobacco smoking consisting of regions from the visual, dorsal/ventral attention, and frontoparietal networks.
Collapse
Affiliation(s)
- Yezhi Pan
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
- Institute for Health Computing, University of Maryland, North Bethesda, MD, USA
| | - Chuan Bi
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Zhenyao Ye
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Hwiyoung Lee
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Jiaao Yu
- Department of Mathematics, University of Maryland, College Park, MD, USA
| | - Luba Yammine
- Department of Psychiatry and Behavioral Science, University of Texas Health Science Center Houston, Houston, TX, USA
| | - Tianzhou Ma
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD, USA
| | - Peter Kochunov
- Department of Psychiatry and Behavioral Science, University of Texas Health Science Center Houston, Houston, TX, USA
| | - L Elliot Hong
- Department of Psychiatry and Behavioral Science, University of Texas Health Science Center Houston, Houston, TX, USA
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
- Institute for Health Computing, University of Maryland, North Bethesda, MD, USA
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
21
|
Jordão J, Figueira J, Morgado M, Guimarães P, Serranho P, Castro-Farías D, DeBuc DC, Castelo-Branco M, Paques M, Bernardes R. Direct crosstalk between adult human retinas as suggested by interocular transfer of neurovascular coupling through photic stimulation. Sci Rep 2025; 15:13684. [PMID: 40258943 PMCID: PMC12012064 DOI: 10.1038/s41598-025-98631-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/14/2025] [Indexed: 04/23/2025] Open
Abstract
Crosstalk mechanisms between retinas were never documented in humans despite being documented for several other species, including non-human primates. Results of the first-in-human study that documents the crosstalk between retinas by measuring the vascular response in one retina to the photic stimulation of the contralateral eye in health and disease are reported herein. A stimulation apparatus was developed and integrated into an adaptive-optics fundus camera to image 32 healthy control (HC) subjects and 20 type 1 diabetes mellitus (DM) patients. Ipsilateral and contralateral neurovascular coupling effects were documented, and criteria were established to consider an actual response and find positive and negative responses. Ten (31.2%) and two (6.2%) subjects of the HC group presented contralateral positive and negative responses, respectively, and three (15.0%) positive and four (20.0%) negative responses were found for the DM group. Also, statistically significant differences in the ipsilateral (p < 0.001) and contralateral (p = 0.027) responses were found for the HC group, rejecting the null (non-response) hypothesis. This finding raises the need to revisit the current knowledge of neurovascular coupling mechanisms and the association between its dysregulation and neurological disorders. Further studies involving distinct populations and imaging centers are necessary to validate the findings herein.
Collapse
Affiliation(s)
- João Jordão
- University of Coimbra, Institute for Nuclear Sciences Applied to Health (ICNAS), Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Coimbra, Portugal
| | - João Figueira
- University of Coimbra, Faculty of Medicine, Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Faculty of Medicine, Department of Ophthalmology, Coimbra Hospital and University Centre (CHUC), Coimbra, Portugal
| | - Miguel Morgado
- University of Coimbra, Institute for Nuclear Sciences Applied to Health (ICNAS), Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Coimbra, Portugal
- University of Coimbra, Faculty of Science and Technology, Department of Physics, Coimbra, Portugal
| | - Pedro Guimarães
- University of Coimbra, Institute for Nuclear Sciences Applied to Health (ICNAS), Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Coimbra, Portugal
- University of Coimbra, Faculty of Medicine, Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Pedro Serranho
- University of Coimbra, Institute for Nuclear Sciences Applied to Health (ICNAS), Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Coimbra, Portugal
- Department of Sciences and Technology, Universidade Aberta, Lisboa, Portugal
| | | | - Delia Cabrera DeBuc
- Bascom Palmer Eye Institute, University Miami Miller School of Medicine, Miami, FL, USA
| | - Miguel Castelo-Branco
- University of Coimbra, Institute for Nuclear Sciences Applied to Health (ICNAS), Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Coimbra, Portugal
- University of Coimbra, Faculty of Medicine, Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Michel Paques
- Centre Hospitalier National des Quinze-Vingts, Paris Eye Imaging, Paris, France
| | - Rui Bernardes
- University of Coimbra, Institute for Nuclear Sciences Applied to Health (ICNAS), Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Coimbra, Portugal.
- University of Coimbra, Faculty of Medicine, Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal.
| |
Collapse
|
22
|
Sun B, Liu K, Zhang H, Gao M, Cai T, Sun M, Ding Z, Li P. Vessel skeleton-based vasodynamic optical flow for video deshaking in functional OCT angiography. OPTICS LETTERS 2025; 50:2659-2662. [PMID: 40232464 DOI: 10.1364/ol.554161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025]
Abstract
Functional OCT angiography (fOCTA) enables non-invasive detection of retinal neurovascular coupling (rNVC) function. However, involuntary eye movements during dynamic imaging can displace the field of view, thereby challenging the accurate quantification of rNVC function at the single-capillary level. In this Letter, we develop a vessel skeleton-based vasodynamic optical flow (VDO flow) algorithm designed for fOCTA video stabilization and vasodynamic preservation. This method not only registers dynamic OCTA images with high speed (∼0.39 s per frame) and high accuracy (63.4% vascular skeleton recombination rate) but also effectively preserves critical vasodynamic responses. By applying VDO flow, we could precisely detect rNVC dysfunction at the single-capillary level in early diabetic retinopathy, even in the absence of significant changes in retinal vascular morphology. This advancement facilitates the clinical application of fOCTA.
Collapse
|
23
|
Marowsky A, Wyss MT, Kindler D, Khalid NUA, Rudin M, Weber B, Arand M. Deletion of microsomal epoxide hydrolase gene leads to increased density in cerebral vasculature and enhances cerebral blood flow in mice. J Cereb Blood Flow Metab 2025:271678X251333234. [PMID: 40219924 PMCID: PMC11994649 DOI: 10.1177/0271678x251333234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 03/03/2025] [Accepted: 03/22/2025] [Indexed: 04/14/2025]
Abstract
Microsomal epoxide hydrolase (mEH), first identified as detoxifying enzyme, can hydrolyze epoxyeicosatrienoic acids (EETs) to less active diols (DHETs). EETs are potent vasodilatory and pro-angiogenic lipids, also implicated in neurovascular coupling. In mouse brain, mEH is strongly expressed in vascular and perivascular cells in contrast to the related soluble epoxide hydrolase (sEH), predominantly found in astrocytes. While sEH inhibition in stroke has demonstrated neuroprotective effects and increases cerebral blood flow (CBF), data regarding the role of mEH in brain are scarce. Here, we explored the function of mEH in cerebral vasculature by comparing mEH-KO, sEH-KO and WT mice. Basal cerebral volume (CBV0) was significantly higher in various mEH-KO brain areas compared to WT and sEH-KO. In line, quantification of cerebral vasculature in cortex and thalamus revealed a higher capillary density in mEH-KO, but not in sEH-KO brain. Whisker-stimulated CBF changes were by factor two higher in both mEH-KO and sEH-KO. In acutely isolated cerebral endothelial cells the loss of mEH, but not of sEH, augmented total EET levels and decreased the DHET:EET ratio. Collectively, these data suggest an important function of mEH in the regulation of cerebral vasculature and activity-modulated CBF, presumably by controlling local levels of endothelial-derived EETs.
Collapse
Affiliation(s)
- Anne Marowsky
- Institute of Pharmacology and Toxicology, University Zürich, Zürich, Switzerland
| | - Matthias T Wyss
- Institute of Pharmacology and Toxicology, University Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Diana Kindler
- Institute of Biomedical Engineering, University Zürich/Eidgenössisch-Technische Hochschule (ETH), Zürich, Switzerland
| | - Noor-Ul-Ain Khalid
- Institute of Pharmacology and Toxicology, University Zürich, Zürich, Switzerland
| | - Markus Rudin
- Loop Zurich, Medical Research Center, Zürich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Michael Arand
- Institute of Pharmacology and Toxicology, University Zürich, Zürich, Switzerland
| |
Collapse
|
24
|
Dalkara T, Østergaard L, Heusch G, Attwell D. Pericytes in the brain and heart: functional roles and response to ischaemia and reperfusion. Cardiovasc Res 2025; 120:2336-2348. [PMID: 39074200 PMCID: PMC11976724 DOI: 10.1093/cvr/cvae147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/28/2024] [Accepted: 05/03/2024] [Indexed: 07/31/2024] Open
Abstract
In the last 20 years, there has been a revolution in our understanding of how blood flow is regulated in many tissues. Whereas it used to be thought that essentially all blood flow control occurred at the arteriole level, it is now recognized that control of capillary blood flow by contractile pericytes plays a key role both in regulating blood flow physiologically and in reducing it in clinically relevant pathological conditions. In this article, we compare and contrast how brain and cardiac pericytes regulate cerebral and coronary blood flow, focusing mainly on the pathological events of cerebral and cardiac ischaemia. The cerebral and coronary capillary beds differ dramatically in morphology, yet in both cases, pericyte-mediated capillary constriction plays a key role in restricting blood flow after ischaemia and possibly in other pathological conditions. We conclude with suggestions for therapeutic approaches to relaxing pericytes, which may prove useful in the long-term for reducing pericyte-induced ischaemia.
Collapse
Affiliation(s)
- Turgay Dalkara
- Department of Neuroscience, Bilkent University, Ankara 06800 Türkiye
- Department of Molecular Biology and Genetics, Bilkent University, Ankara 06800 Türkiye
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| |
Collapse
|
25
|
Shoemaker PA, Bekkouche BMB. Modeling traveling calcium waves in cellular structures. J Comput Neurosci 2025:10.1007/s10827-025-00898-2. [PMID: 40172607 DOI: 10.1007/s10827-025-00898-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 04/04/2025]
Abstract
We report a parametric simulation study of traveling calcium waves in two classes of cellular structures: dendrite-like processes and an idealized cell body. It is motivated by the hypothesis that calcium waves may participate in spatiotemporal sensory processing; accordingly, its objective is to elucidate the dependence of traveling wave characteristics (e.g., propagation speed and amplitude) on various anatomical and physiological parameters. The models include representations of inositol trisphosphate and ryanodine receptors (which mediate transient calcium entry into the cytoplasm from the endoplasmic reticulum), as well as other entities involved in calcium transport or reactions. These support traveling cytoplasmic calcium waves, which are fully regenerative for significant ranges of model parameters. We also observe Hopf bifurcations between stable and unstable regimes, the latter being characterized by periodic calcium spikes. Traveling waves are possible in unstable processes during phases with sufficiently high calcium levels in the endoplasmic reticulum. Damped and abortive waves are observed for some parameter values. When both receptor types are present and functional, we find wave speeds on the order of 100 to several hundred micrometers per second and cytosolic calcium transients with amplitudes of tens of micromolar; when ryanodine receptors are absent, these values are on the order of tens of micrometers per second and 1-6 micromolar. Even with significantly downgraded channel conductance, ryanodine receptors can significantly impact wave speeds and amplitudes. Receptor areal densities and the diffusion coefficient for cytoplasmic calcium are the parameters to which wave characteristics are most sensitive.
Collapse
Affiliation(s)
- Patrick A Shoemaker
- Computational Science Research Center, San Diego State University, San Diego, CA, USA.
| | - Bo M B Bekkouche
- , Nevrobo AB (5593306664), Stockholm, Sweden
- Department of Biology, Lund University, Lund, Sweden
| |
Collapse
|
26
|
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
|
27
|
Fang X, Border JJ, Zhang H, Challagundla L, Kaur J, Hwang SH, Hammock BD, Fan F, Roman RJ. A Soluble Epoxide Hydrolase Inhibitor Improves Cerebrovascular Dysfunction, Neuroinflammation, Amyloid Burden, and Cognitive Impairments in the hAPP/PS1 TgF344-AD Rat Model of Alzheimer's Disease. Int J Mol Sci 2025; 26:2433. [PMID: 40141075 PMCID: PMC11942141 DOI: 10.3390/ijms26062433] [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: 12/30/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Alzheimer's disease (AD) is an increasing global healthcare crisis with few effective treatments. The accumulation of amyloid plaques and hyper-phosphorylated tau are thought to underlie the pathogenesis of AD. However, current studies have recognized a prominent role of cerebrovascular dysfunction in AD. We recently reported that SNPs in soluble epoxide hydrolase (sEH) are linked to AD in human genetic studies and that long-term administration of an sEH inhibitor attenuated cerebral vascular and cognitive dysfunction in a rat model of AD. However, the mechanisms linking changes in cerebral vascular function and neuroprotective actions of sEH inhibitors in AD remain to be determined. This study investigated the effects of administration of an sEH inhibitor, 1-(1-Propanoylpiperidin-4-yl)-3-[4-(trifluoromethoxy)phenyl]urea (TPPU), on neurovascular coupling, blood-brain barrier (BBB) function, neuroinflammation, and cognitive dysfunction in an hAPP/PS1 TgF344-AD rat model of AD. We observed predominant β-amyloid accumulation in the brains of 9-10-month-old AD rats and that TPPU treatment for three months reduced amyloid burden. The functional hyperemic response to whisker stimulation was attenuated in AD rats, and TPPU normalized the response. The sEH inhibitor, TPPU, mitigated capillary rarefaction, BBB leakage, and activation of astrocytes and microglia in AD rats. TPPU increased the expression of pre- and post-synaptic proteins and reduced loss of hippocampal neurons and cognitive impairments in the AD rats, which was confirmed in a transcriptome and GO analysis. These results suggest that sEH inhibitors could be a novel therapeutic strategy for AD.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| | - Jane J. Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| | - Lavanya Challagundla
- Molecular and Genomics Facility, University of Mississippi Medical Center, Jackson, MS 39216, USA; (L.C.); (J.K.)
| | - Jasleen Kaur
- Molecular and Genomics Facility, University of Mississippi Medical Center, Jackson, MS 39216, USA; (L.C.); (J.K.)
| | - Sung Hee Hwang
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA; (S.H.H.); (B.D.H.)
| | - Bruce D. Hammock
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA; (S.H.H.); (B.D.H.)
| | - Fan Fan
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Richard J. Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| |
Collapse
|
28
|
Xu X, Su J, Zhu R, Li K, Zhao X, Fan J, Mao F. From morphology to single-cell molecules: high-resolution 3D histology in biomedicine. Mol Cancer 2025; 24:63. [PMID: 40033282 PMCID: PMC11874780 DOI: 10.1186/s12943-025-02240-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/18/2025] [Indexed: 03/05/2025] Open
Abstract
High-resolution three-dimensional (3D) tissue analysis has emerged as a transformative innovation in the life sciences, providing detailed insights into the spatial organization and molecular composition of biological tissues. This review begins by tracing the historical milestones that have shaped the development of high-resolution 3D histology, highlighting key breakthroughs that have facilitated the advancement of current technologies. We then systematically categorize the various families of high-resolution 3D histology techniques, discussing their core principles, capabilities, and inherent limitations. These 3D histology techniques include microscopy imaging, tomographic approaches, single-cell and spatial omics, computational methods and 3D tissue reconstruction (e.g. 3D cultures and spheroids). Additionally, we explore a wide range of applications for single-cell 3D histology, demonstrating how single-cell and spatial technologies are being utilized in the fields such as oncology, cardiology, neuroscience, immunology, developmental biology and regenerative medicine. Despite the remarkable progress made in recent years, the field still faces significant challenges, including high barriers to entry, issues with data robustness, ambiguous best practices for experimental design, and a lack of standardization across methodologies. This review offers a thorough analysis of these challenges and presents recommendations to surmount them, with the overarching goal of nurturing ongoing innovation and broader integration of cellular 3D tissue analysis in both biology research and clinical practice.
Collapse
Affiliation(s)
- Xintian Xu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Rongyi Zhu
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Kailong Li
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaolu Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and GynecologyNational Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital)Key Laboratory of Assisted Reproduction (Peking University), Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China.
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory for Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Beijing, China.
| |
Collapse
|
29
|
Valenza G, Matić Z, Catrambone V. The brain-heart axis: integrative cooperation of neural, mechanical and biochemical pathways. Nat Rev Cardiol 2025:10.1038/s41569-025-01140-3. [PMID: 40033035 DOI: 10.1038/s41569-025-01140-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 03/05/2025]
Abstract
The neural and cardiovascular systems are pivotal in regulating human physiological, cognitive and emotional states, constantly interacting through anatomical and functional connections referred to as the brain-heart axis. When this axis is dysfunctional, neurological conditions can lead to cardiovascular disorders and, conversely, cardiovascular dysfunction can substantially affect brain health. However, the mechanisms and fundamental physiological components of the brain-heart axis remain largely unknown. In this Review, we elucidate these components and identify three primary pathways: neural, mechanical and biochemical. The neural pathway involves the interaction between the autonomic nervous system and the central autonomic network in the brain. The mechanical pathway involves mechanoreceptors, particularly those expressing mechanosensitive Piezo protein channels, which relay crucial information about blood pressure through peripheral and cerebrovascular connections. The biochemical pathway comprises many endogenous compounds that are important mediators of neural and cardiovascular function. This multisystem perspective calls for the development of integrative approaches, leading to new clinical specialties in neurocardiology.
Collapse
Affiliation(s)
- Gaetano Valenza
- Neurocardiovascular Intelligence Lab, Department of Information Engineering & Research Center "E. Piaggio", University of Pisa, Pisa, Italy.
| | - Zoran Matić
- Neurocardiovascular Intelligence Lab, Department of Information Engineering & Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| | - Vincenzo Catrambone
- Neurocardiovascular Intelligence Lab, Department of Information Engineering & Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| |
Collapse
|
30
|
Kremer R, Williams A, Wardlaw J. Endothelial cells as key players in cerebral small vessel disease. Nat Rev Neurosci 2025; 26:179-188. [PMID: 39743557 DOI: 10.1038/s41583-024-00892-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 01/04/2025]
Abstract
Cerebral small vessel disease (SVD) is a vascular disorder that increases the risk of stroke and dementia and is diagnosed through brain MRI. Current primary prevention and secondary treatment of SVD are focused on lifestyle interventions and vascular risk factor control, including blood pressure reduction. However, these interventions have limited effects, a proportion of individuals with sporadic SVD do not have hypertension, and SVD shows strong familial and genetic underpinnings. Here, we describe the increasing evidence that cerebral endothelial cell dysfunction is a key mechanism of SVD. Dysfunctional endothelial cells can cause cerebral blood vessel dysfunction, alter blood-brain barrier integrity and interfere with cell-cell interactions in the neuro-glial-vascular unit, thereby causing damage to adjacent brain tissue. Endothelial cells in SVD may become dysfunctional through intrinsic mechanisms via genetic vulnerability to SVD and/or via extrinsic factors such as hypertension, smoking and diabetes. Drugs that act on endothelial pathways are already looking promising in clinical trials, and understanding their action on endothelial cells and the surrounding brain may lead to the development of other therapies to limit disease progression and improve outcomes for individuals with SVD.
Collapse
Affiliation(s)
- Ronja Kremer
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Joanna Wardlaw
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK.
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK.
- Edinburgh Imaging, The University of Edinburgh, Edinburgh, UK.
- Row Fogo Centre for Research into Ageing and the Brain, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
31
|
Van Eijgen J, Van Winckel L, Hanssen H, Kotliar K, Vanassche T, Van Craenenbroeck EM, Cornelissen V, Van Craenenbroeck AH, Jones E, Stalmans I. Retinal vessel analysis to assess microvascular function in the healthy eye: A systematic review on the response to acute physiological and pathological stressors. Surv Ophthalmol 2025; 70:200-214. [PMID: 39592075 DOI: 10.1016/j.survophthal.2024.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024]
Abstract
The retina allows noninvasive in vivo assessment of the microcirculation. Autoregulation of the retinal microvasculature meets the changing requirements of local metabolic demand and maintains adequate blood flow. Analysis of the retinal vascular reactivity contributes to the understanding of regulatory physiology and its relationship to the systemic microcirculation. We conducted a literature review on the effect of different acute stimuli onto the retinal vasculature was conducted in accordance with the PRISMA guidelines. A literature search between 1-1-2005 and 17-10-2022 was performed in Medline, Embase, Web of Science and the Cochrane Library. We report the retinal vascular behavior of healthy individuals in response to both physiological and pathological stressors in 106 included articles. We provide ables of methodological characteristics for each stressor. Hypoxia, hypercapnia, high altitude, flicker light stimulation, rise of core temperature, blood pressure lowering, and the condition immediately after endurance exercise associate with larger retinal vessels. Hyperoxia, hypocapnia, blood pressure rise (Bayliss effect), and the condition during isometric exercise associate with smaller retinal vessels. The retinal vasculature is highly reactive to physiological and pathological stressors. This autoregulatory capacity is hypothesized to be a source of biomarkers for vascular health. Dynamic and static retinal vessel analysis are noninvasive methods to assess this (micro)vascular function. Exploring its diagnostic potential and application into clinical practice requires the development of standardized assessment methods, for which some recommendations are made.
Collapse
Affiliation(s)
- Jan Van Eijgen
- Department of Ophthalmology, University Hospitals UZ Leuven, Leuven, Belgium; Research Group Ophthalmology, Department of Neurosciences, KU Leuven, Leuven, Belgium.
| | - Lien Van Winckel
- Department of Ophthalmology, University Hospitals UZ Leuven, Leuven, Belgium.
| | - Henner Hanssen
- Department of Sports, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland.
| | - Konstantin Kotliar
- Department of Medical Engineering and Technomathematics, FH Aachen University of Applied Sciences, Campus Jülich, Heinrich-Mussmann-Str. 1, Jülich 52428, Germany.
| | - Thomas Vanassche
- Centre for Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium.
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Campus Drie Eiken D.T.2.28, Universiteitsplein 1, Antwerp 2610, Belgium; Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem 2650, Belgium.
| | - Véronique Cornelissen
- Research Group of Rehabilitation of Internal Disorders, Department of Rehabilitation Sciences, Faculty of Movement and Rehabilitation Sciences, KU Leuven, Leuven, Belgium.
| | - Amaryllis H Van Craenenbroeck
- Division of Nephrology, University Hospitals UZ leuven, Leuven, Belgium; Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium.
| | - Elisabeth Jones
- Centre for Molecular and Vascular Biology, Herestraat 49, Bus 911, KU, Leuven 3000, Belgium; Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, Maastricht 6229 ER, Netherlands.
| | - Ingeborg Stalmans
- Department of Ophthalmology, University Hospitals UZ Leuven, Leuven, Belgium; Research Group Ophthalmology, Department of Neurosciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
32
|
Sacca V, Maleki N, Reddy S, Hodges S, Kong J. Assessing the Modulatory Effects of tDCS and Acupuncture on Cerebral Blood Flow in Chronic Low Back Pain Using Arterial Spin Labeling Perfusion Imaging. Brain Sci 2025; 15:261. [PMID: 40149782 PMCID: PMC11940449 DOI: 10.3390/brainsci15030261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Both transcranial direct current stimulation (tDCS) and acupuncture are promising methods for managing chronic low back pain (cLBP), however, their underlying mechanisms remain unclear. METHODS To explore the neural mechanisms of tDCS and acupuncture on cLBP, we examined how real and sham tDCS applied to the bilateral motor cortex (M1), combined with real or sham acupuncture, influenced cerebral blood flow (CBF) using pulsed continuous arterial spin labeling (pCASL) imaging. tDCS was administered over six sessions, combined with real or sham acupuncture, over one month. RESULTS Following real tDCS, we observed increased CBF in the bilateral occipital cortex, precuneus, left hippocampus, and parahippocampal gyrus/posterior cingulate cortex. After sham tDCS, CBF decreased in regions including the bilateral superior parietal lobule, precuneus, bilateral precentral and postcentral gyri, and left angular gyrus. Real acupuncture led to reduced CBF in the bilateral occipital cortex and hippocampus, and left posterior cingulate gyrus, and increased CBF in the right postcentral gyrus, superior parietal lobule, and frontal areas. Sham acupuncture was associated with decreased CBF in the bilateral hippocampus and anterior cingulate gyrus. CONCLUSIONS These results suggest both shared and distinct patterns of CBF changes between real and sham tDCS, as well as between real and sham acupuncture, reflecting mode-dependent effects on brain networks involved in pain processing and modulation. Our findings highlight the different neural circuits implicated in the therapeutic mechanisms of tDCS and acupuncture in the management of cLBP.
Collapse
Affiliation(s)
| | | | | | | | - Jian Kong
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA; (V.S.); (N.M.); (S.R.); (S.H.)
| |
Collapse
|
33
|
Jo MG, Hong J, Kim J, Kim SH, Lee B, Choi HN, Lee SE, Kim YJ, Park H, Park DH, Roh GS, Kim CS, Yun SP. Physiological change of striatum and ventral midbrain's glia cell in response to different exercise modalities. Behav Brain Res 2025; 479:115342. [PMID: 39571940 DOI: 10.1016/j.bbr.2024.115342] [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: 06/26/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
Exercise not only regulates neurotransmitters and synapse formation but also enhances the function of multiple brain regions, beyond cortical activation. Prolonged aerobic or resistance exercise modality has been widely applied to reveal the beneficial effects on the brain, but few studies have investigated the direct effects of different exercise modalities and variations in exercise intensity on the neuroinflammatory response in the brain and overall health. Therefore, in this study, we investigated changes in brain cells and the immune environment of the brain according to exercise modalities. This study was conducted to confirm whether different exercise modalities affect the location and function of dopaminergic neurons, which are responsible for regulating voluntary movement, before utilizing animal models of disease. The results showed that high-intensity interval exercise (HIE) increased the activity of A2-reactive astrocytes in the striatum (STR), which is directly involved in movement control, resulting in neuroprotective effects. Both HIE and combined exercises (CE) increased the expression of dopamine transporter (DAT) in the STR without damaging dopamine neurons in the ventral midbrain (VM). This means that exercise training can help improve and maintain exercise capacity. In conclusion, specific exercise modalities or intensity of exercise may contribute to preventing neurodegenerative diseases such as Parkinson's disease or enhancing therapeutic effects when combined with medication for patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Min Gi Jo
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Junyoung Hong
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jiyeon Kim
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Republic of Korea
| | - Seon-Hee Kim
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Bina Lee
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Ha Nyeoung Choi
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - So Eun Lee
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Young Jin Kim
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Heejung Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Dong-Ho Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea; Department of Kinesiology, Inha University, Incheon 22212, Republic of Korea
| | - Gu Seob Roh
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; Department of Anatomy, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Chang Sun Kim
- Department of Physical Education, Dongduk Women's University, Seoul 02748, Republic of Korea.
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea.
| |
Collapse
|
34
|
Zhao X, Yao M, Wang Y, Feng C, Yang Y, Tian L, Bao C, Li X, Zhu X, Zhang X. Neuroregulation during Bone Formation and Regeneration: Mechanisms and Strategies. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7223-7250. [PMID: 39869030 DOI: 10.1021/acsami.4c16786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The skeleton is highly innervated by numerous nerve fibers. These nerve fibers, in addition to transmitting information within the bone and mediating bone sensations, play a crucial role in regulating bone tissue formation and regeneration. Traditional bone tissue engineering (BTE) often fails to achieve satisfactory outcomes when dealing with large-scale bone defects, which is frequently related to the lack of effective reconstruction of the neurovascular network. In recent years, increasing research has revealed the critical role of nerves in bone metabolism. Nerve fibers regulate bone cells through neurotransmitters, neuropeptides, and peripheral glial cells. Furthermore, nerves also coordinate with the vascular and immune systems to jointly construct a microenvironment favorable for bone regeneration. As a signaling driver of bone formation, neuroregulation spans the entire process of bone physiological activities from the embryonic formation to postmaturity remodeling and repair. However, there is currently a lack of comprehensive summaries of these regulatory mechanisms. Therefore, this review sketches out the function of nerves during bone formation and regeneration. Then, we elaborate on the mechanisms of neurovascular coupling and neuromodulation of bone immunity. Finally, we discuss several novel strategies for neuro-bone tissue engineering (NBTE) based on neuroregulation of bone, focusing on the coordinated regeneration of nerve and bone tissue.
Collapse
Affiliation(s)
- Xiangrong Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meilin Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuyi Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cong Feng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuhan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Luoqiang Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
35
|
Csipo T, Lipecz A, Mukli P, Péterfi A, Szarvas Z, Ungvari A, Alaoui LE, Sándor M, Kállai A, Fekete M, Fülöp GÁ, Tarantini S, Csiszar A, Benyó Z, Sótonyi P, Tabak AG, Merkely B, Yabluchanskiy A, Ungvari Z. Advancing prediction of age-related vascular cognitive impairment based on peripheral and retinal vascular health in a pilot study: a novel comprehensive assessment developed for a prospective workplace-based cohort (The Semmelweis Study). GeroScience 2025; 47:1329-1344. [PMID: 39604626 PMCID: PMC11872852 DOI: 10.1007/s11357-024-01447-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: 08/04/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
With a growing elderly population in the European Union, age-related diseases associated with unhealthy aging pose increasing public health challenges, including a loss of independence and heightened societal burdens. The Semmelweis Study, a prospective occupational cohort study in Hungary, seeks to identify determinants of unhealthy aging, focusing on the complex relationship between lifestyle, environmental, occupational factors, and the development of chronic age-associated diseases, including age-related vascular cognitive impairment (VCI). The primary objective of this pilot study was to establish a robust, high-throughput assessment methodology to comprehensively evaluate both peripheral and cerebrovascular health to provide a solid foundation for the forthcoming Semmelweis Study framework. The study involved 49 participants aged 23 to 87 years, and it assessed multi-domain cognitive performance through an automated battery of tests (CANTAB). Vascular health was comprehensively evaluated using laser speckle contrast imaging (LSCI), flow-mediated dilation (FMD), static and dynamic retinal vessel analysis (SVA, DVA), and measurements of vascular stiffness. The retinal microvasculature, which closely mirrors the cerebral circulation in anatomy, physiology, and pathophysiology, provided a unique window for examination. Optical imaging through SVA and DVA enables the identification of structural and functional changes in the central nervous system's microcirculation, which are highly relevant to the pathogenesis of VCI. Subsequently, the collected measures were integrated into vascular health indices using principal component analysis (PCA) and the relationship to the age and cognitive status of study participants was explored. These comprehensive vascular health indices demonstrated a correlation not only with age but also with cognitive performance. This methodology holds promise for providing novel insights into the intricate interplay between vascular and cognitive health within the context of the Semmelweis Study.
Collapse
Affiliation(s)
- Tamas Csipo
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Agnes Lipecz
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Péterfi
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zsofia Szarvas
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Ungvari
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Lamyae El Alaoui
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Márton Sándor
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Attila Kállai
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Mónika Fekete
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Gábor Á Fülöp
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College, Health Sciences Program/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College, Health Sciences Program/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, H-1052, Hungary
| | - Péter Sótonyi
- Department of Vascular and Endovascular Surgery, Semmelweis University, Budapest, Hungary
| | - Adam G Tabak
- Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- UCL Brain Sciences, University College London, London, UK
- Department of Internal Medicine and Oncology, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College, Health Sciences Program/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College, Health Sciences Program/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| |
Collapse
|
36
|
Anderle S, Bonnar O, Henderson J, Shaw K, Chagas AM, McMullan L, Webber A, McGowan K, King SL, Hall CN. APOE4 and sedentary lifestyle synergistically impair neurovascular function in the visual cortex of awake mice. Commun Biol 2025; 8:144. [PMID: 39880935 PMCID: PMC11779976 DOI: 10.1038/s42003-025-07585-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025] Open
Abstract
Reduced cerebral blood flow occurs early in the development of Alzheimer's disease (AD), but the factors producing this reduction are unknown. Here, we ask whether genetic and lifestyle risk factors for AD-the ε4 allele of the Apolipoprotein (APOE) gene, and physical activity-can together produce this reduction in cerebral blood flow which leads eventually to AD. Using in vivo two-photon microscopy and haemodynamic measures, we record neurovascular function from the visual cortex of physically active or sedentary mice expressing APOE3 and APOE4 in place of murine APOE. Energy supply and demand are mismatched in APOE4 mice, with smaller increases in cerebral blood flow, blood volume and blood oxygenation occurring during neuronal activation as blood vessels frequently fail to dilate. Exercise dose-dependently overall improves neurovascular function, with an increased impact of exercise apparent after longer exposure times. Several haemodynamic measures show a larger beneficial effect of exercise in APOE4 vs. APOE3 mice. Thus, APOE4 genotype in conjunction with sedentary behaviour produces the worst neurovascular function. Promotion of physical activity may therefore be particularly important to improve cerebrovascular function and reduce dementia risk in APOE4 carriers.
Collapse
Affiliation(s)
- Silvia Anderle
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Orla Bonnar
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Joseph Henderson
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Kira Shaw
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Andre M Chagas
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Letitia McMullan
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Alexandra Webber
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Kirsty McGowan
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Sarah L King
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK
| | - Catherine N Hall
- School of Psychology and Sussex Neuroscience, University of Sussex, Brighton, UK.
| |
Collapse
|
37
|
Yoo J, Lee J, Ahn B, Han J, Lim MH. Multi-target-directed therapeutic strategies for Alzheimer's disease: controlling amyloid-β aggregation, metal ion homeostasis, and enzyme inhibition. Chem Sci 2025; 16:2105-2135. [PMID: 39810997 PMCID: PMC11726323 DOI: 10.1039/d4sc06762b] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia, marked by progressive cognitive decline and memory impairment. Despite advances in therapeutic research, single-target-directed treatments often fall short in addressing the complex, multifactorial nature of AD. This arises from various pathological features, including amyloid-β (Aβ) aggregate deposition, metal ion dysregulation, oxidative stress, impaired neurotransmission, neuroinflammation, mitochondrial dysfunction, and neuronal cell death. This review illustrates their interrelationships, with a particular emphasis on the interplay among Aβ, metal ions, and AD-related enzymes, such as β-site amyloid precursor protein cleaving enzyme 1 (BACE1), matrix metalloproteinase 9 (MMP9), lysyl oxidase-like 2 (LOXL2), acetylcholinesterase (AChE), and monoamine oxidase B (MAOB). We further underscore the potential of therapeutic strategies that simultaneously inhibit Aβ aggregation and address other pathogenic mechanisms. These approaches offer a more comprehensive and effective method for combating AD, overcoming the limitations of conventional therapies.
Collapse
Affiliation(s)
- Jeasang Yoo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jimin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Byeongha Ahn
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul Seoul 02504 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
38
|
Huang H, Hou J, Li M, Wei F, Liao Y, Xi B. Microplastics in the bloodstream can induce cerebral thrombosis by causing cell obstruction and lead to neurobehavioral abnormalities. SCIENCE ADVANCES 2025; 11:eadr8243. [PMID: 39841831 PMCID: PMC11753373 DOI: 10.1126/sciadv.adr8243] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025]
Abstract
Human health is being threatened by environmental microplastic (MP) pollution. MPs were detected in the bloodstream and multiple tissues of humans, disrupting the regular physiological processes of organs. Nanoscale plastics can breach the blood-brain barrier, leading to neurotoxic effects. How MPs cause brain functional irregularities remains unclear. This work uses high-depth imaging techniques to investigate the MPs within the brain in vivo. We show that circulating MPs are phagocytosed and lead these cells to obstruction in the capillaries of the brain cortex. These blockages as thrombus formation cause reduced blood flow and neurological abnormalities in mice. Our data reveal a mechanism by which MPs disrupt tissue function indirectly through regulation of cell obstruction and interference with local blood circulation, rather than direct tissue penetration. This revelation offers a lens through which to comprehend the toxicological implications of MPs that invade the bloodstream.
Collapse
Affiliation(s)
- Haipeng Huang
- State Key Laboratory of Environment Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing Raygen Health, Nanjing, China
| | - Jiaqi Hou
- State Key Laboratory of Environment Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Mingxiao Li
- State Key Laboratory of Environment Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Fangchao Wei
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Yilie Liao
- National University of Singapore, Lower Kent Ridge Road, Singapore, Singapore
| | - Beidou Xi
- State Key Laboratory of Environment Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China
| |
Collapse
|
39
|
Fu Z, Ganesana M, Hwang P, Tan X, Kinkaid MM, Sun YY, Bian E, Weybright A, Chen HR, Sol-Church K, Eyo UB, Pridans C, Quintana FJ, Robson SC, Kumar P, Venton BJ, Schaefer A, Kuan CY. Microglia modulate the cerebrovascular reactivity through ectonucleotidase CD39. Nat Commun 2025; 16:956. [PMID: 39843911 PMCID: PMC11754601 DOI: 10.1038/s41467-025-56093-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025] Open
Abstract
Microglia and the border-associated macrophages contribute to the modulation of cerebral blood flow, but the mechanisms have remained uncertain. Here, we show that microglia regulate the cerebral blood flow baseline and the responses to whisker stimulation or intra-cisternal magna injection of adenosine triphosphate, but not intra-cisternal magna injection of adenosine in mice model. Notably, microglia repopulation corrects these cerebral blood flow anomalies. The microglial-dependent regulation of cerebral blood flow requires the adenosine triphosphate-sensing P2RY12 receptor and ectonucleotidase CD39 that initiates the dephosphorylation of extracellular adenosine triphosphate into adenosine in both male and female mice. Pharmacological inhibition or CX3CR1-CreER-mediated deletion of CD39 mimics the cerebral blood flow anomalies in microglia-deficient mice and reduces the upsurges of extracellular adenosine following whisker stimulation. Together, these results suggest that the microglial CD39-initiated breakdown of extracellular adenosine triphosphate co-transmitter is an important step in neurovascular coupling and the regulation of cerebrovascular reactivity.
Collapse
Affiliation(s)
- Zhongxiao Fu
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | | | - Philip Hwang
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xiao Tan
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Melissa Marie Kinkaid
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Yu-Yo Sun
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Emily Bian
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Aden Weybright
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Hong-Ru Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Katia Sol-Church
- Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Ukpong B Eyo
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Clare Pridans
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Departments of Anesthesia and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Pankaj Kumar
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - B Jill Venton
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA
| | - Anne Schaefer
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- MPI Biology of Ageing, Cologne, Germany
| | - Chia-Yi Kuan
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
40
|
Suarez A, Fernandez L, Riera J. Characterizing astrocyte-mediated neurovascular coupling by combining optogenetics and biophysical modeling. J Cereb Blood Flow Metab 2025:271678X241311010. [PMID: 39791314 PMCID: PMC11719438 DOI: 10.1177/0271678x241311010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025]
Abstract
Vasoactive signaling from astrocytes is an important contributor to the neurovascular coupling (NVC), which aims at providing energy to neurons during brain activation by increasing blood perfusion in the surrounding vasculature. Pharmacological manipulations have been previously combined with experimental techniques (e.g., transgenic mice, uncaging, and multiphoton microscopy) and stimulation paradigms to isolate in vivo individual pathways of the astrocyte-mediated NVC. Unfortunately, these pathways are highly nonlinear and non-additive. To separate these pathways in a unified framework, we combine a comprehensive biophysical model of vasoactive signaling from astrocytes with a unique optogenetic stimulation method that selectively induces astrocytic Ca2+ signaling in a large population of astrocytes. We also use a sensitivity analysis and an optimization technique to estimate key model parameters. Optogenetically-induced Ca2+ signals in astrocytes cause a cerebral blood flow (CBF) response with two major components. Component-1 was rapid and smaller (ΔCBF∼13%, 18 seconds), while component-2 was slowest and highest (ΔCBF ∼18%, 45 seconds). The proposed biophysical model was adequate in reproducing component-2, which was validated with a pharmacological manipulation. Model's predictions were not in contradiction with previous studies. Finally, we discussed scenarios accounting for the existence of component-1, which once validated might be included in our model.
Collapse
Affiliation(s)
- Alejandro Suarez
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| | - Lazaro Fernandez
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| | - Jorge Riera
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| |
Collapse
|
41
|
Golf SR, Trotter JH, Wang J, Nakahara G, Han X, Wernig M, Südhof TC. Deletion of Neuroligins from Astrocytes Does Not Detectably Alter Synapse Numbers or Astrocyte Cytoarchitecture by Maturity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.04.10.536254. [PMID: 37090508 PMCID: PMC10120619 DOI: 10.1101/2023.04.10.536254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Astrocytes perform multifarious roles in the formation, regulation, and function of synapses in the brain, but the mechanisms involved are incompletely understood. Interestingly, astrocytes abundantly express neuroligins, postsynaptic adhesion molecules that function as synaptic organizers by binding to presynaptic neurexins. Here we examined the function of neuroligins in astrocytes with a rigorous genetic approach that uses the conditional deletion of all major neuroligins (Nlgn1-3) in astrocytes in vivo and complemented this approach by a genetic deletion of neuroligins in glia cells that are co-cultured with human neurons. Our results show that early postnatal deletion of neuroligins from astrocytes in vivo has no detectable effect on cortical or hippocampal synapses and does not alter the cytoarchitecture of astrocytes when evaluated in young adult mice. Moreover, deletion of astrocytic neuroligins in co-cultures of human neurons produced no detectable consequences for the formation and function of synapses. Thus, astrocytic neuroligins are unlikely to fundamentally shape synapse formation or astrocyte morphogenesis but likely perform other important roles that remain to be discovered.
Collapse
Affiliation(s)
- Samantha R. Golf
- Dept. of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Justin H. Trotter
- Dept. of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Dept. of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
- Dept. of Neurobiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Jinzhao Wang
- Dept. of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pathology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - George Nakahara
- Dept. of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xiao Han
- Dept. of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Marius Wernig
- Department of Pathology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Thomas C. Südhof
- Dept. of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
42
|
Wang W, Lutsey PL, Inciardi RM, Reyes JL, Mosley TH, Johansen MC, Gottesman RF, Alonso A, Jack CR, Solomon SD, Shah AM, Wasserman BA, Chen LY. Association of left atrial function with vascular brain injury: The Atherosclerosis Risk in Communities study. Eur J Neurol 2025; 32:e16549. [PMID: 39569699 PMCID: PMC11625916 DOI: 10.1111/ene.16549] [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: 08/16/2024] [Revised: 10/18/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND AND PURPOSE Lower left atrial (LA) function is associated with higher dementia risk and may be mechanistically linked through vascular brain injury, an established correlate for higher dementia risk. Using data from the Atherosclerosis Risk in Communities study, we assessed the cross-sectional association between LA function and brain magnetic resonance imaging (MRI) markers of vascular brain injury. METHODS We included 1488 participants who were free of prevalent dementia, stroke, or atrial fibrillation and who underwent a two-dimensional echocardiogram and brain MRI in 2011-2013 (mean [± standard deviation] age 76 [± 5] years, 60% female, 27% Black). LA function measures (reservoir, conduit, contractile strain) were assessed in quartiles. Brain MRI measures included cerebral microbleeds, brain infarcts, and white matter hyperintensity (WMH) volume. Logistic regression was used for dichotomous outcomes. Linear regression was used for WMH volume. RESULTS Overall, 343 (23%) and 344 participants (23%) had ≥1 cerebral microbleed or brain infarct. After multivariable adjustments, the lowest LA reservoir and conduit strain quartiles (vs. highest quartile) were associated with higher odds of the presence of ≥1 cerebral microbleed (odds ratios [95% confidence intervals] 1.78 [1.42-2.22] and 1.52 [1.22-1.90]). Compared to the highest quartile, participants in the lowest LA conduit strain quartile had 1.51 (95% confidence interval 1.22-1.88) times higher odds of having ≥1 brain infarct. Lower LA contractile strain was associated with lower odds of brain infarcts. No association with WMH volume was noted. CONCLUSIONS We found that LA reservoir and conduit strain were associated with cerebral microbleeds and brain infarcts. Lower LA function may be linked to dementia risk via vascular brain injury. Prospective studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Wendy Wang
- Division of Epidemiology and Community Health, School of Public HealthUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public HealthUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Riccardo M. Inciardi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Institute of CardiologyUniversity of BresciaBresciaItaly
| | - Jorge L. Reyes
- Lillehei Heart Institute and Department of Medicine (Cardiovascular Division)University of Minnesota Medical SchoolMinneapolisMinnesotaUSA
| | - Thomas H. Mosley
- The MIND CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | | | - Rebecca F. Gottesman
- Stroke BranchNational Institute of Neurological Disorders and Stroke Intramural Research ProgramBethesdaMarylandUSA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public HealthEmory UniversityAtlantaGeorgiaUSA
| | | | - Scott D. Solomon
- Cardiovascular DivisionBrigham and Women's HospitalBostonMassachusettsUSA
| | - Amil M. Shah
- Division of CardiologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Bruce A. Wasserman
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Lin Yee Chen
- Lillehei Heart Institute and Department of Medicine (Cardiovascular Division)University of Minnesota Medical SchoolMinneapolisMinnesotaUSA
| |
Collapse
|
43
|
Eyre B, Shaw K, Francis S, Howarth C, Berwick J. Voluntary locomotion induces an early and remote hemodynamic decrease in the large cerebral veins. NEUROPHOTONICS 2025; 12:S14609. [PMID: 40130194 PMCID: PMC11931294 DOI: 10.1117/1.nph.12.s1.s14609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/26/2025]
Abstract
Significance Behavior regulates dural and cerebral vessels, with spontaneous locomotion inducing dural vessel constriction and increasing stimulus-evoked cerebral hemodynamic responses. It is vital to investigate the function of different vascular network components, surrounding and within the brain, to better understand the role of the neurovascular unit in health and neurodegeneration. Aim We characterized locomotion-induced hemodynamic responses across vascular compartments of the whisker barrel cortex: artery, vein, parenchyma, draining, and meningeal vein. Approach Using 2D-OIS, hemodynamic responses during locomotion were recorded in 9- to 12-month-old awake mice: wild-type, Alzheimer's disease (AD), atherosclerosis, or mixed (atherosclerosis/AD) models. Within the somatosensory cortex, responses were taken from pial vessels inside the whisker barrel region [(WBR): "whisker artery" and "whisker vein"], a large vein from the sagittal sinus adjacent to the WBR (draining vein), and meningeal vessels from the dura mater (which do not penetrate cortical tissue). Results We demonstrate that locomotion evokes an initial decrease in total hemoglobin (HbT) within the draining vein before the increase in HbT within WBR vessels. The locomotion event size influences the magnitude of the HbT increase in the pial vessels of the WBR but not of the early HbT decrease within the draining veins. Following locomotion onset, an early HbT decrease was also observed in the overlying meningeal vessels, which unlike within the cortex did not go on to exceed baseline HbT levels during the remainder of the locomotion response. We show that locomotion-induced hemodynamic responses are altered in disease in the draining vein and whisker artery, suggesting this could be an important neurodegeneration biomarker. Conclusions This initial reduction in HbT within the draining and meningeal veins potentially serves as a "space-saving" mechanism, allowing for large increases in cortical HbT associated with locomotion. Given this mechanism is impacted by disease, it may provide an important target for vascular-based therapeutic interventions.
Collapse
Affiliation(s)
- Beth Eyre
- University of Sheffield, Sheffield Neurovascular Group, Department of Psychology, Sheffield, United Kingdom
- University of Sheffield, Neuroscience Institute, Sheffield, United Kingdom
- University of Sheffield, Healthy Lifespan Institute, Sheffield, United Kingdom
- Massachusetts General Hospital, Harvard Medical School, Department of Neurology, Boston, Massachusetts, United States
| | - Kira Shaw
- University of Sheffield, Sheffield Neurovascular Group, Department of Psychology, Sheffield, United Kingdom
- University of Sheffield, Neuroscience Institute, Sheffield, United Kingdom
- University of Sheffield, Healthy Lifespan Institute, Sheffield, United Kingdom
| | - Sheila Francis
- University of Sheffield, Neuroscience Institute, Sheffield, United Kingdom
- University of Sheffield, Healthy Lifespan Institute, Sheffield, United Kingdom
- University of Sheffield, School of Medicine and Population Health, Sheffield, United Kingdom
| | - Clare Howarth
- University of Sheffield, Sheffield Neurovascular Group, Department of Psychology, Sheffield, United Kingdom
- University of Sheffield, Neuroscience Institute, Sheffield, United Kingdom
- University of Sheffield, Healthy Lifespan Institute, Sheffield, United Kingdom
| | - Jason Berwick
- University of Sheffield, Sheffield Neurovascular Group, Department of Psychology, Sheffield, United Kingdom
- University of Sheffield, Neuroscience Institute, Sheffield, United Kingdom
- University of Sheffield, Healthy Lifespan Institute, Sheffield, United Kingdom
| |
Collapse
|
44
|
Zielińska M, Popek M, Albrecht J. Neuroglia in hepatic encephalopathy. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:191-212. [PMID: 40148045 DOI: 10.1016/b978-0-443-19102-2.00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Neuroglia contribute to the pathophysiology of hepatic encephalopathy (HE) either beneficially or detrimentally. Pathogenesis of HE is linked to damage triggered by blood-derived toxins, with ammonia being the main causative factor. Neuroglial cells, especially astrocytes and microglia, respond to HE-associated systemic and central signals and undergo complex and variable changes in their metabolism, morphology, and function, which include ion and water dyshomeostasis in conjunction with neurotransmission imbalance and neuroinflammation. HE-induced alterations of astrocytes are defined as astrocytopathy, with aberrant astrocytes resulting in either gain or loss of functions. In the chronic HE, the presence of Alzheimer type II cells is a histologic hallmark, with asthenic astrocytes emerging as a newcomer. In acute HE, rapid swelling of astrocytes is a primary cause of cerebral edema and mortality. This chapter reviews the dominant role of astrocytes in the pathogenesis of HE resulting from acute and chronic liver failure, mainly in experimental models. The focus is on the loss of homeostatic function bearing upon the functioning of the glymphatic system, aberrant neurotransmission as a consequence of astrocyte-neuron miscommunication, and the concordant neuroinflammatory response of astrocytes and microglia. The chapter concludes with a delineation of concepts for future research.
Collapse
Affiliation(s)
- Magdalena Zielińska
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland.
| | - Mariusz Popek
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Jan Albrecht
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
45
|
Demmings MD, da Silva Chagas L, Traetta ME, Rodrigues RS, Acutain MF, Barykin E, Datusalia AK, German-Castelan L, Mattera VS, Mazengenya P, Skoug C, Umemori H. (Re)building the nervous system: A review of neuron-glia interactions from development to disease. J Neurochem 2025; 169:e16258. [PMID: 39680483 DOI: 10.1111/jnc.16258] [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: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 12/18/2024]
Abstract
Neuron-glia interactions are fundamental to the development and function of the nervous system. During development, glia, including astrocytes, microglia, and oligodendrocytes, influence neuronal differentiation and migration, synapse formation and refinement, and myelination. In the mature brain, glia are crucial for maintaining neural homeostasis, modulating synaptic activity, and supporting metabolic functions. Neurons, inherently vulnerable to various stressors, rely on glia for protection and repair. However, glia, in their reactive state, can also promote neuronal damage, which contributes to neurodegenerative and neuropsychiatric diseases. Understanding the dual role of glia-as both protectors and potential aggressors-sheds light on their complex contributions to disease etiology and pathology. By appropriately modulating glial activity, it may be possible to mitigate neurodegeneration and restore neuronal function. In this review, which originated from the International Society for Neurochemistry (ISN) Advanced School in 2019 held in Montreal, Canada, we first describe the critical importance of glia in the development and maintenance of a healthy nervous system as well as their contributions to neuronal damage and neurological disorders. We then discuss potential strategies to modulate glial activity during disease to protect and promote a properly functioning nervous system. We propose that targeting glial cells presents a promising therapeutic avenue for rebuilding the nervous system.
Collapse
Affiliation(s)
- Matthew D Demmings
- Neuroscience Program, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Luana da Silva Chagas
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Marianela E Traetta
- Instituto de Biología Celular y Neurociencia (IBCN), Facultad de Medicina, Conicet, Buenos Aires, Argentina
| | - Rui S Rodrigues
- University of Bordeaux, INSERM, Neurocentre Magendie U1215, Bordeaux, France
| | - Maria Florencia Acutain
- Instituto de Biología Celular y Neurociencia (IBCN), Facultad de Medicina, Conicet, Buenos Aires, Argentina
| | - Evgeny Barykin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER Raebareli), Raebareli, UP, India
| | - Liliana German-Castelan
- Neuroscience Program, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Vanesa S Mattera
- Instituto de Química y Fisicoquímica Biológica (IQUIFIB-FFyB-UBA), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pedzisai Mazengenya
- Center of Medical and bio-Allied Health Sciences Research, College of Medicine, Ajman University, Ajman, United Arab Emirates
| | - Cecilia Skoug
- Department of Neuroscience, Physiology & Pharmacology, Centre for Cardiovascular and Metabolic Neuroscience, University College London, London, UK
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Al-Eitan LN, Alahmad SZ, Khair IY. The Impact of Potent Addictive Substances on Angiogenic Behavior: A Comprehensive Review. Curr Neuropharmacol 2025; 23:511-523. [PMID: 39248059 DOI: 10.2174/1570159x23666240905125037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 09/10/2024] Open
Abstract
Angiogenesis, the formation of new vasculature from preexisting vasculature, is involved in the development of several diseases as well as various physiological processes. Strict cooperation of proangiogenic and antiangiogenic factors mediates the control of angiogenesis. The fundamental steps in angiogenesis include endothelial cell proliferation, migration, and invasion. Addictive substances, which are considered therapeutic candidates in research and medicine, are classified as natural substances, such as nicotine, or synthetic substances, such as synthetic cannabinoids. Addictive substances have been shown to either enhance or suppress angiogenesis. This review article provides an overview of recent studies concerning the effects of several addictive substances on the process of angiogenesis. Google Scholar and PubMed were used to collect the scientific literature used in this review. The addictive substances addressed in this review are nicotine, opioids such as morphine and heroin, alcohol, cocaine, methamphetamine, and cannabinoids. An accurate assessment of the influence of these substances on the angiogenic process may help to construct a potentially effective therapeutic protocol to control and treat several angiogenesis-related diseases.
Collapse
Affiliation(s)
- Laith Naser Al-Eitan
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Saif Zuhair Alahmad
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Iliya Yacoub Khair
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
47
|
Battini S, Cantarutti N, Kotsalos C, Roussel Y, Cattabiani A, Arnaudon A, Favreau C, Antonel S, Markram H, Keller D. Modeling of Blood Flow Dynamics in Rat Somatosensory Cortex. Biomedicines 2024; 13:72. [PMID: 39857656 PMCID: PMC11761867 DOI: 10.3390/biomedicines13010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/11/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Background: The cerebral microvasculature forms a dense network of interconnected blood vessels where flow is modulated partly by astrocytes. Increased neuronal activity stimulates astrocytes to release vasoactive substances at the endfeet, altering the diameters of connected vessels. Methods: Our study simulated the coupling between blood flow variations and vessel diameter changes driven by astrocytic activity in the rat somatosensory cortex. We developed a framework with three key components: coupling between the vasculature and synthesized astrocytic morphologies, a fluid dynamics model to compute flow in each vascular segment, and a stochastic process replicating the effect of astrocytic endfeet on vessel radii. Results: The model was validated against experimental flow values from the literature across cortical depths. We found that local vasodilation from astrocyte activity increased blood flow, especially in capillaries, exhibiting a layer-specific response in deeper cortical layers. Additionally, the highest blood flow variability occurred in capillaries, emphasizing their role in cerebral perfusion regulation. We discovered that astrocytic activity impacted blood flow dynamics in a localized, clustered manner, with most vascular segments influenced by two to three neighboring endfeet. Conclusions: These insights enhance our understanding of neurovascular coupling and guide future research on blood flow-related diseases.
Collapse
Affiliation(s)
- Stéphanie Battini
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Yang L, Zhao W, Kan Y, Ren C, Ji X. From Mechanisms to Medicine: Neurovascular Coupling in the Diagnosis and Treatment of Cerebrovascular Disorders: A Narrative Review. Cells 2024; 14:16. [PMID: 39791717 PMCID: PMC11719775 DOI: 10.3390/cells14010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
Neurovascular coupling (NVC) refers to the process of local changes in cerebral blood flow (CBF) after neuronal activity, which ensures the timely and adequate supply of oxygen, glucose, and substrates to the active regions of the brain. Recent clinical imaging and experimental technology advancements have deepened our understanding of the cellular mechanisms underlying NVC. Pathological conditions such as stroke, subarachnoid hemorrhage, cerebral small vascular disease, and vascular cognitive impairment can disrupt NVC even before clinical symptoms appear. However, the complexity of the underlying mechanism remains unclear. This review discusses basic and clinical experimental evidence on how neural activity sensitively communicates with the vasculature to cause spatial changes in blood flow in cerebrovascular diseases. A deeper understanding of how neurovascular unit-related cells participate in NVC regulation is necessary to better understand blood flow and nerve activity recovery in cerebrovascular diseases.
Collapse
Affiliation(s)
- Lu Yang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (L.Y.); (W.Z.); (Y.K.)
| | - Wenbo Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (L.Y.); (W.Z.); (Y.K.)
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100054, China
| | - Yuan Kan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (L.Y.); (W.Z.); (Y.K.)
| | - Changhong Ren
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100054, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100054, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
49
|
Sun J, Zelaya F, Sendt KV, McQueen G, Gillespie AL, Lally J, Howes OD, Barker GJ, McGuire P, MacCabe JH, Egerton A. Response to clozapine in treatment resistant schizophrenia is related to alterations in regional cerebral blood flow. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:122. [PMID: 39715777 DOI: 10.1038/s41537-024-00544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024]
Abstract
PET and SPECT studies in treatment-resistant schizophrenia (TRS) have revealed significant alterations in regional cerebral blood flow (CBF) during clozapine treatment, which may vary according to the clinical response. Here, we used the more recent MRI approach of arterial spin labelling (ASL) to evaluate regional CBF in participants with TRS (N = 36) before starting treatment with clozapine compared to in healthy volunteers (N = 16). We then compared CBF in the TRS group, before and after 12 weeks of treatment with clozapine (N = 24); and examined the relationship of those differences against changes in Positive and Negative Syndrome Scale for Schizophrenia (PANSS) scores over the treatment period. We observed widespread reductions in CBF in TRS compared to in healthy volunteers (p < 0.05). After covarying for global CBF and age, lower CBF in frontal and parietal regions was still evident (p < 0.05, FWE corrected). Clozapine treatment was associated with longitudinal decreases in CBF in the anterior cingulate cortex (ACC) (p < 0.05). Higher striatal CBF at baseline was associated with greater improvement in total and general symptoms following clozapine, and higher hippocampal CBF was associated with greater improvement in total and positive symptoms. Longitudinal reductions in CBF in the ACC and thalamus were associated with less improvement in negative (ACC), positive (thalamus), and total (thalamus) symptoms. These findings suggest that changes in CBF on clozapine administration in TRS may accompany symptomatic improvement, and that CBF prior to clozapine initiation may determine the degree of clinical response.
Collapse
Affiliation(s)
- Junyu Sun
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Fernando Zelaya
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Kyra-Verena Sendt
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Grant McQueen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Amy L Gillespie
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
| | - John Lally
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Department of Psychiatry, University College Dublin, Dublin, Ireland
- Department of Psychiatry, St Vincent's Hospital Fairview, Dublin, Ireland
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Gareth J Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Philip McGuire
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
| | - James H MacCabe
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- National Psychosis Unit, South London and Maudsley NHS Foundation Trust, London, UK
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| |
Collapse
|
50
|
Wang S, Zou M, Zhu Z, Wang Z, Li K, Ruan J, Zhao B, Pan C, Lan X, Zhang S, Foulkes NS, Zhao H. Oseltamivir phosphate (Tamiflu) alters neurobehavior of zebrafish larvae by inducing mitochondrial dysfunction. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:177077. [PMID: 39461536 DOI: 10.1016/j.scitotenv.2024.177077] [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: 08/25/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
Antiviral drugs are widely used, yet their potential risks during early development, particularly within the central nervous system, remain contentious. Oseltamivir phosphate (OSE), a commonly prescribed antiviral, is increasingly detected in various environments. However, its toxicity to organisms and the underlying mechanisms are not well understood. In this study, we employed the zebrafish model to evaluate the developmental neurotoxic effects of OSE at environmentally and therapeutically relevant doses, through high-throughput behavioral analysis, in vivo two-photon imaging, transcriptomic sequencing, pharmacological intervention, and biochemical and molecular assays. Our results indicated that OSE exposure increased heart rate and induced pericardial edema in zebrafish larvae. Additionally, OSE-exposed larvae exhibited hyperactive behavior, impaired social interactions, and reduced habitual learning capacity. Although OSE at our selected levels did not significantly affect neuron count in the brain, it activated neuroinflammatory responses, altered blood vessel morphology, modulated neurotransmitter levels and the expression of neurodevelopment-related genes. Transcriptomic analysis revealed upregulation of mitochondria-related genes associated with oxidative phosphorylation. Further assessments of mitochondrial function demonstrated altered activities of respiratory chain complexes, reduced mitochondrial membrane potential (MMP), and decreased ATP content. Notably, co-treatment with mitochondrial protectants acetyl-l-carnitine-hydrochloride (ALC) or nicotinamide riboside (NR) effectively mitigated OSE-induced neurobehavioral disorders. These findings suggest that overuse of OSE can pose neurodevelopmental risks for both humans and animals, potentially attributable to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Shuang Wang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou 730000, Gansu Province, China.
| | - Minjian Zou
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou 730000, Gansu Province, China.
| | - Zhirui Zhu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou 730000, Gansu Province, China.
| | - Zuo Wang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou 730000, Gansu Province, China.
| | - Kemin Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou 730000, Gansu Province, China.
| | - Jiayi Ruan
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou 730000, Gansu Province, China.
| | - Bixi Zhao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou 730000, Gansu Province, China.
| | - Chuanyin Pan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling 712100, Shaanxi Province, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling 712100, Shaanxi Province, China
| | - Shengxiang Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou 730000, Gansu Province, China.
| | - Nicholas S Foulkes
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany.
| | - Haiyu Zhao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|