1
|
Baker MR, Klassen BT, Jensen MA, Ojeda Valencia G, Heydari H, Ince NF, Müller KR, Miller KJ. Parameterization of intraoperative human microelectrode recordings: Linking action potential morphology to brain anatomy. PLoS Comput Biol 2025; 21:e1013184. [PMID: 40526776 DOI: 10.1371/journal.pcbi.1013184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 06/27/2025] [Accepted: 05/30/2025] [Indexed: 06/19/2025] Open
Abstract
Deep brain stimulation (DBS) is a targeted manipulation of brain circuitry to treat neurological and neuropsychiatric conditions. Optimal DBS lead placement is essential for treatment efficacy. Current targeting practice is based on preoperative and intraoperative brain imaging, intraoperative electrophysiology, and stimulation mapping. Electrophysiological mapping using extracellular microelectrode recordings aids in identifying functional subdomains, anatomical boundaries, and disease-correlated physiology. The shape of single-unit action potentials may differ due to different biophysical properties between cell-types and brain regions. Here, we describe a technique to parameterize the structure and duration of sorted spike units using a novel algorithmic approach based on canonical response parameterization, and illustrate how it may be used on DBS microelectrode recordings. Isolated spike shapes are parameterized then compared using a spike similarity metric and grouped by hierarchical clustering. When spike morphology is associated with anatomy, we find regional clustering in the human globus pallidus. This method is widely applicable for spike removal and single-unit characterization and could be integrated into intraoperative array-based technologies to enhance targeting and clinical outcomes in DBS lead placement.
Collapse
Affiliation(s)
- Matthew R Baker
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Bryan T Klassen
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Michael A Jensen
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Gabriela Ojeda Valencia
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Hossein Heydari
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Nuri F Ince
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Klaus-Robert Müller
- Department of Computer Science, TU Berlin, Berlin, Germany
- Dept of Artificial Intelligence, Korea University, Seoul, Republic of Korea
- Max Planck Institute for Informatics, Saarbrücken, Germany
| | - Kai J Miller
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Pediatrics, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
2
|
Tort ABL, Laplagne DA, Draguhn A, Gonzalez J. Global coordination of brain activity by the breathing cycle. Nat Rev Neurosci 2025; 26:333-353. [PMID: 40204908 DOI: 10.1038/s41583-025-00920-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
Neuronal activities that synchronize with the breathing rhythm have been found in humans and a host of mammalian species, not only in brain areas closely related to respiratory control or olfactory coding but also in areas linked to emotional and higher cognitive functions. In parallel, evidence is mounting for modulations of perception and action by the breathing cycle. In this Review, we discuss the extent to which brain activity locks to breathing across areas, levels of organization and brain states, and the physiological origins of this global synchrony. We describe how waves of sensory activity evoked by nasal airflow spread through brain circuits, synchronizing neuronal populations to the breathing cycle and modulating faster oscillations, cell assembly formation and cross-area communication, thereby providing a mechanistic link from breathing to neural coding, emotion and cognition. We argue that, through evolution, the breathing rhythm has come to shape network functions across species.
Collapse
Affiliation(s)
- Adriano B L Tort
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Diego A Laplagne
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Andreas Draguhn
- Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Joaquin Gonzalez
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Neuroscience Institute and Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
3
|
Wang H, Jiang J, Zhou G, Zhang Y. The Roles of Heart Rate Variability in Cerebral Stroke. Neuropsychiatr Dis Treat 2025; 21:1057-1065. [PMID: 40395407 PMCID: PMC12091237 DOI: 10.2147/ndt.s515306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025] Open
Abstract
Heart rate variability (HRV), as a safe and noninvasive marker of autonomic activity, is becoming a prescreening tool for the prediction of cardiovascular complications such as myocardial infarction and heart failure after cerebral stroke. With good reproducibility and quantitative assessment of autonomic nervous system function, the association of some HRV parameters with neurological functional prognosis and stroke recurrence has been widely reported. However, current studies on the relationship between HRV and the clinical outcomes of stroke remain controversial. Previous studies on HRV have mainly focused on stroke prognosis or its role as a clinical biomarker, whereas other roles have rarely been explored. In this article, we review recent advances in the role of HRV in cerebral stroke according to current progress. Specifically, we summarized the role of HRV in the outcomes, complications, treatments, and mechanisms of stroke, based on the latest research. Further, in-depth implications of HRV in stroke are discussed.
Collapse
Affiliation(s)
- Huijun Wang
- Department of Neurosurgery, Xishan People’s Hospital of Wuxi City, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, People’s Republic of China
| | - Jiayao Jiang
- Department of Orthopedics, The 904th Hospital of the Joint Logistics Support Force of the PLA, Wuxi, Jiangsu Province, People’s Republic of China
| | - Guochao Zhou
- Department of Orthopedics, The Army 947th Hospital, Kashgar, People’s Republic of China
| | - Yelei Zhang
- Department of Neurosurgery, Xishan People’s Hospital of Wuxi City, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, People’s Republic of China
| |
Collapse
|
4
|
Lyons SH, Gottfried JA. Predictive coding in the human olfactory system. Trends Cogn Sci 2025:S1364-6613(25)00084-1. [PMID: 40345946 DOI: 10.1016/j.tics.2025.04.005] [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: 11/21/2024] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/11/2025]
Abstract
The human olfactory system is unusual. It deviates from the classical structure and function of other sensory cortices, and many of its basic computations remain mysterious. These idiosyncrasies have challenged the development of a clear and comprehensive theoretical framework in olfactory neuroscience. To address this challenge, we develop a theory of olfactory predictive coding that aims to unify diverse olfactory phenomena. Under this scheme, the olfactory system is not merely passively processing sensory information. Instead, it is actively issuing predictions about sensory inputs before they even arrive. We map this conceptual framework onto the micro- and macroscale neurobiology of the human olfactory system and review a variety of neurobiological, computational, and behavioral evidence in support of this scheme.
Collapse
Affiliation(s)
- Sam H Lyons
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jay A Gottfried
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Menuet C, Ben-Tal A, Linossier A, Allen AM, Machado BH, Moraes DJA, Farmer DGS, Paterson DJ, Mendelowitz D, Lakatta EG, Taylor EW, Ackland GL, Zucker IH, Fisher JP, Schwaber JS, Shanks J, Paton JFR, Buron J, Spyer KM, Shivkumar K, Dutschmann M, Joyner MJ, Herring N, Grossman P, McAllen RM, Ramchandra R, Yao ST, Ritz T, Gourine AV. Redefining respiratory sinus arrhythmia as respiratory heart rate variability: an international Expert Recommendation for terminological clarity. Nat Rev Cardiol 2025:10.1038/s41569-025-01160-z. [PMID: 40328963 DOI: 10.1038/s41569-025-01160-z] [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: 04/07/2025] [Indexed: 05/08/2025]
Abstract
The variation of heart rate in phase with breathing, known as 'respiratory sinus arrhythmia' (RSA), is a physiological phenomenon present in all air-breathing vertebrates. RSA arises from the interaction of several physiological mechanisms but is primarily mediated by rhythmic changes in cardiac parasympathetic (vagal) activity, increasing heart rate during inspiration and decreasing heart rate during expiration. RSA amplitude is an indicator of autonomic and cardiac health; RSA is diminished or absent in common pathological conditions such as chronic heart failure and hypertension. In this Expert Recommendation, we argue that the term 'RSA', although historically important, is semantically inaccurate and carries misleading pathological connotations, contributing to misunderstanding and misinterpretation of the origin and the physiological importance of the phenomenon. We propose replacing 'RSA' with the term 'respiratory heart rate variability' (RespHRV), which avoids pathological connotations and emphasizes the specific respiratory contribution to heart rate variability. We clarify that RespHRV encompasses respiratory-related heart rate variations in both the low-frequency and high-frequency bands traditionally defined in heart rate variability analysis, and that its amplitude should not be misconstrued as a measure of vagal tone. Adopting the proposed term 'RespHRV' is expected to unify understanding and stimulate further experimental and clinical research into the physiological mechanisms and functional importance of this phenomenon.
Collapse
Affiliation(s)
- Clément Menuet
- INMED, INSERM, Aix-Marseille University, Marseille, France.
| | - Alona Ben-Tal
- Insightful Modelling, Auckland, New Zealand
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Auckland, New Zealand
| | | | - Andrew M Allen
- Department of Anatomy & Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Benedito H Machado
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Davi J A Moraes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - David G S Farmer
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - David J Paterson
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MA, USA
| | - Edwin W Taylor
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Gareth L Ackland
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - James P Fisher
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Auckland, New Zealand
| | - James S Schwaber
- Department of Pathology, Anatomy, and Cell Biology, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Julia Shanks
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Auckland, New Zealand
| | - Julian F R Paton
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Auckland, New Zealand
| | - Julie Buron
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - K Michael Spyer
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Kalyanam Shivkumar
- University of California Los Angeles (UCLA) Cardiac Arrhythmia Center, Los Angeles, CA, USA
| | - Mathias Dutschmann
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University Hospitals Cleveland Medical Center and Case Western Reserve University, Cleveland, OH, USA
- Center for Sleep Disorders Research, Louis Stokes Cleveland VA Medical Center and Case Western Reserve University, Cleveland, OH, USA
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Paul Grossman
- Department of Psychosomatic Medicine, University Hospital Basel, Basel, Switzerland
| | - Robin M McAllen
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Rohit Ramchandra
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Auckland, New Zealand
| | - Song T Yao
- Department of Anatomy & Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Thomas Ritz
- Department of Psychology, Southern Methodist University, Dallas, TX, USA
| | - Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology & Pharmacology, University College London, London, UK
| |
Collapse
|
6
|
Zuo X, Bai H, Qiu J, Li R, Kuang X, Zhao Y, Tuo J, Zhao Q, Zhao X, Feng X. 17β-Trenbolone modulates anxiety-related synaptic plasticity by affecting the interaction between hippocampal TACR3 and systemic testosterone in male mice. ENVIRONMENTAL RESEARCH 2025; 279:121796. [PMID: 40340005 DOI: 10.1016/j.envres.2025.121796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 05/01/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Anxiety Disorder is a common neurological disorder for which ubiquitous environmental endocrine disruptors may be risk factors. 17β-trenbolone (17-TB) has been recognized as a potential environmental endocrine disruptor and its neurotoxic effects have attracted attention. Tachykinin receptor 3 (TACR3), a G protein-coupled receptor involved in pubertal anxiety, and its underlying neural mechanisms remain enigmatic. Therefore, this study investigated the possible relationship between 17-TB and TACR3, testosterone (T), and synaptic plasticity. Our results showed that adolescent male Balb/c mice developed significant anxiety-like behavior after four weeks of exposure to environmentally relevant concentrations of 17-TB (100 μg/kg/day). Transcriptomic RNA-Seq results showed that 17-TB affected abnormal synaptic transmission signals in the hippocampus. Electrophysiological results showed that 17-TB reduced the activity of hippocampal dentate gyrus (DG) neurons and led to the downregulation of hippocampal TACR3 and T levels. In addition, Western blot, immunohistochemistry, ELISA, and qRT-PCR showed that 17-TB exposure led to the downregulation of key hippocampal synaptic proteins PSD95, Gephyrin, and Syn, and induced a metabolic imbalance in glutamate (Glu)/GABA signaling, affecting dopamine signaling. Interestingly, testosterone supplementation (10 mg/kg/day) was effective in ameliorating the above phenomena and alleviating anxiety-like behaviors. These results suggest that 17-TB modulates anxiety-related synaptic plasticity by regulating hippocampal TACR3 and T interactions in vivo. In conclusion, our study contributes to understanding the neurobehavioral and potential mechanistic effects of environmental 17-TB exposure in mammals. It alerts the public to the health risks of chemicals to mammals and suggests new research directions and potential therapeutic targets.
Collapse
Affiliation(s)
- Xiang Zuo
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Huijuan Bai
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Jinyu Qiu
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China
| | - Ruimin Li
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China
| | - Xiaochen Kuang
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Yudi Zhao
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Jingyi Tuo
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Qili Zhao
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China
| | - Xin Zhao
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China.
| | - Xizeng Feng
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
7
|
Zhang Y, Fang M, Yu L, Liu X, Wang J, Li N, Li L, Zhou C. Enhanced cellular viability and osteogenic activity in oxygen-self-generating and magnetically responsive alginate microgels as advanced cell carriers. BIOMATERIALS ADVANCES 2025; 170:214198. [PMID: 39893887 DOI: 10.1016/j.bioadv.2025.214198] [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: 11/22/2024] [Revised: 01/09/2025] [Accepted: 01/19/2025] [Indexed: 02/04/2025]
Abstract
Improving the accuracy of in vitro three-dimensional (3D) cellular cultures more closely replicates the in vivo microenvironment by mimicking the complex tissue structures, enhancing cell-cell interactions, and increasing differentiation potential along with functional capabilities. Natural materials aid in cell adhesion and proliferation within the 3D matrix, providing a more realistic growth environment. Oxygen availability is also critical for cell survival in 3D cultures, as a lack of oxygen can impede proliferation, reduce functionality, and ultimately result in cell death. To address the issue of oxygen supply in such systems, a novel magnetic alginate-based microcarrier that generates oxygen autonomously has been developed. This microcarrier contains calcium peroxide encapsulated within polylactic acid microspheres (CP), which act as an internal oxygen reservoir. The release of calcium ions results in weak interactions with alginate, thus improving structural integrity while also supporting bone marrow stromal cells (BMSCs) and creating a more in vivo-like microenvironment. Notably, when exposed to an external magnetic field, BMSCs on these CP/Fe3O4/SA microcarriers show improved viability, a marked decrease in hypoxia-inducible factor-1α (HIF-1α), and increased osteogenic gene expression. Therefore, the CP/Fe3O4/SA microcarriers represent a promising approach for enhancing in vitro 3D culture methods and offer significant potential for tissue repair and regeneration.
Collapse
Affiliation(s)
- Yifan Zhang
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China; College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Min Fang
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Lanqin Yu
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Xinshuo Liu
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Jizhuang Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Na Li
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan 528225, China
| | - Lihua Li
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China; College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China.
| | - Changren Zhou
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| |
Collapse
|
8
|
Li YY, Li H, Zheng Y, Xu DD, Liu L, Liu A, Li T, Pang DW, Tang HW. Optical Tweezer-Driven Mechanotransduction: Probing pN-Scale Forces and Calcium-Mediated Redox Signaling in Single Endothelial Cells. ACS NANO 2025; 19:16084-16095. [PMID: 40232231 DOI: 10.1021/acsnano.5c03122] [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: 04/16/2025]
Abstract
Endothelial cells (ECs) regulate vascular function by converting mechanical forces into biochemical signals; however, the molecular mechanisms of pN-scale mechanotransduction remain elusive. Here, we develop an optical tweezer-integrated confocal microscopy system that allows precise, noninvasive manipulation of the cell membrane localization with mechanical stimuli within the 0-100 pN range while monitoring Ca2+-mediated NO/ROS redox signaling in situ in single ECs under varying force parameters. We show that pN-scale mechanical stimulation regulates extracellular Ca2+ influx, triggering downstream production of NO and ROS, which subsequently affects intracellular redox homeostasis. Key mechanosensitive ion channels (e.g., Piezo1 and TRPV4) and cytoskeletal components (e.g., F-actin) facilitate force-induced redox signaling. We further delineate the roles of membrane tension-dominant versus hybrid tension-tether models in mechanotransduction, revealing their differential engagement in force transmission pathways. This mechanistic framework establishes direct connections between pN-scale mechanical input characteristics and redox-regulated vascular homeostasis.
Collapse
Affiliation(s)
- Yu-Yao Li
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Haodong Li
- The Institute for Advanced Studies, Wuhan University, Wuhan 430072, P. R. China
| | - Yawen Zheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Da-Di Xu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Liu Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Ao Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Tianning Li
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Dai-Wen Pang
- College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Hong-Wu Tang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
9
|
Li Y, Zhao D, Xiang L, Jiang Q, Cui W. Revolutionizing cerebral monitoring: the innovative leap of ultrasound patch technology and its future horizons. Sci Bull (Beijing) 2025; 70:1007-1009. [PMID: 39956672 DOI: 10.1016/j.scib.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Affiliation(s)
- Yihan Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ding Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lei Xiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qinzhe Jiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
10
|
Liu X, Zhang Z, Cao Z, Yuan H, Xing C. Near-Infrared Light-Controlled Dynamic Hydrogel for Modulating Mechanosensitive Ion Channels in 3-Dimensional Environment. Biomater Res 2025; 29:0182. [PMID: 40207256 PMCID: PMC11979339 DOI: 10.34133/bmr.0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/26/2025] [Accepted: 03/15/2025] [Indexed: 04/11/2025] Open
Abstract
The extracellular matrix (ECM) creates a dynamic mechanical environment for cellular functions, continuously influencing cellular activities via the mechanotransduction pathway. Mechanosensitive ion channels, recently identified as key mechanotransducers, convert mechanical stimuli into electrical or chemical signals when they detect membrane deformation. This process facilitates extracellular Ca2+ influx, cytoskeletal reorganization, and transcriptional regulation, all of which are essential for cellular physiological functions. In this study, we developed a fibrous hydrogel composite (PIC/OEG-NPs) with near-infrared (NIR) light-controlled dynamic mechanical properties to modulate mechanosensitive ion channels in cells, by using oligo-ethylene glycol (OEG)-assembled polyisocyanide (PIC) polymer and OEG-grafted conjugated polymer nanoparticles (OEG-NPs). PIC and OEG-NPs assemble into PIC/OEG-NPs composites through OEG-mediated hydrophobic interactions when heated. Under NIR stimulation, the PIC/OEG-NPs composites exhibit increased mechanical tension and form tighter fibrous networks due to their thermoresponsive behavior. These changes are reversible and allow for the dynamic regulation of mechanosensitive ion channels, including Piezo1 in transfected HEK-293T cells and the endogenous TRPV4 in human umbilical vein endothelial cells (HUVECs), by switching NIR on and off. Furthermore, this process enhances the angiogenic potential of HUVECs. In summary, we present a simple and effective platform for in situ modulation of mechanosensitive ion channels in 3 dimensions.
Collapse
Affiliation(s)
- Xiaoning Liu
- School of Materials Science and Engineering,
Hebei University of Technology, Tianjin 300401, China
| | - Zimeng Zhang
- Key Laboratory of Molecular Biophysics of Hebei Province, School of Health Sciences and Biomedical Engineering,
Hebei University of Technology, Tianjin 300401, China
| | - Zhanshuo Cao
- School of Chemical Engineering,
Hebei University of Technology, Tianjin 300401, China.
| | - Hongbo Yuan
- Key Laboratory of Molecular Biophysics of Hebei Province, School of Health Sciences and Biomedical Engineering,
Hebei University of Technology, Tianjin 300401, China
- Molecular Imaging and Photonics, Chemistry Department, KU Leuven, 3001 Heverlee, Belgium
| | - Chengfen Xing
- School of Materials Science and Engineering,
Hebei University of Technology, Tianjin 300401, China
- Key Laboratory of Molecular Biophysics of Hebei Province, School of Health Sciences and Biomedical Engineering,
Hebei University of Technology, Tianjin 300401, China
| |
Collapse
|
11
|
Ni B, Ye L, Zhang Y, Hu S, Lei W. Advances in humanoid organoid-based research on inter-organ communications during cardiac organogenesis and cardiovascular diseases. J Transl Med 2025; 23:380. [PMID: 40156006 PMCID: PMC11951738 DOI: 10.1186/s12967-025-06381-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
The intimate correlation between cardiovascular diseases and other organ pathologies, such as metabolic and kidney diseases, underscores the intricate interactions among these organs. Understanding inter-organ communications is crucial for developing more precise drugs and effective treatments for systemic diseases. While animal models have traditionally been pivotal in studying these interactions, human-induced pluripotent stem cells (hiPSCs) offer distinct advantages when constructing in vitro models. Beyond the conventional two-dimensional co-culture model, hiPSC-derived humanoid organoids have emerged as a substantial advancement, capable of replicating essential structural and functional attributes of internal organs in vitro. This breakthrough has spurred the development of multilineage organoids, assembloids, and organoids-on-a-chip technologies, which allow for enhanced physiological relevance. These technologies have shown great potential for mimicking coordinated organogenesis, exploring disease pathogenesis, and facilitating drug discovery. As the central organ of the cardiovascular system, the heart serves as the focal point of an extensively studied network of interactions. This review focuses on the advancements and challenges of hiPSC-derived humanoid organoids in studying interactions between the heart and other organs, presenting a comprehensive exploration of this cutting-edge approach in systemic disease research.
Collapse
Affiliation(s)
- Baoqiang Ni
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Lingqun Ye
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Yan Zhang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Shijun Hu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | - Wei Lei
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
12
|
Xie W, Lückemeyer DD, Qualls KA, Prudente AS, Berta T, Gu M, Strong JA, Dong X, Zhang JM. Vascular motion in the dorsal root ganglion sensed by Piezo2 in sensory neurons triggers episodic neuropathic pain. Neuron 2025:S0896-6273(25)00178-3. [PMID: 40154477 DOI: 10.1016/j.neuron.2025.03.006] [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: 09/12/2024] [Revised: 01/27/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Spontaneous pain, characterized by episodic shooting or stabbing sensations, is a major complaint among neuropathic pain patients, yet its mechanisms remain poorly understood. Recent research indicates a connection between this pain condition and "clustered firing," wherein adjacent sensory neurons fire simultaneously. This study presents evidence that the triggers of spontaneous pain and clustered firing are the dynamic movements of small blood vessels within the nerve-injured sensory ganglion, along with increased blood vessel density/angiogenesis and increased number of pericytes around blood vessels. Pharmacologically or mechanically evoked myogenic vascular responses increase both spontaneous pain and clustered firing in a mouse model of neuropathic pain. The mechanoreceptor Piezo2 in sensory neurons plays a critical role in detecting blood vessel movements. An anti-VEGF monoclonal antibody that inhibits angiogenesis effectively blocks spontaneous pain and clustered firing. These findings suggest targeting Piezo2, angiogenesis, or abnormal vascular dynamics as potential therapeutic strategies for neuropathic spontaneous pain.
Collapse
Affiliation(s)
- Wenrui Xie
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Debora Denardin Lückemeyer
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Katherine A Qualls
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mingxia Gu
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA.
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
13
|
Elek D, Tóth M, Sonkodi B, Ács P, Kovács GL, Tardi P, Melczer C. The Efficacy of Soleus Push-Up in Individuals with Prediabetes: A Pilot Study. Sports (Basel) 2025; 13:81. [PMID: 40137805 PMCID: PMC11946342 DOI: 10.3390/sports13030081] [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: 01/23/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Hamilton and colleagues invented the soleus push-up exercise and showed that this exercise method was successful in reducing postprandial blood glucose levels in sedentary individuals. The objective of the current pilot study was to assess the efficacy of the soleus push-up in individuals with prediabetes and to evaluate the feasibility of incorporating this exercise method into their daily routine. METHODS Ten participants (mean age: 53.3 ± 2.7 years; four females, six males) with prediabetes were included in the study. Initially, participants underwent an oral glucose tolerance test (OGTT) while being sedentary to establish baseline postprandial blood glucose measurements. During a subsequent OGTT, participants concurrently performed the soleus push-up (SPU) exercise either with or without electromyographic (EMG) feedback. Blood glucose levels were measured at 15 min intervals over the two-hour duration of both OGTTs. RESULTS We observed that performing the SPU in a sitting position during the oral glucose tolerance test resulted in approximately a 32% reduction in postprandial glucose excursion compared to the sedentary baseline results. This effect was also present in the absence of EMG feedback. CONCLUSIONS Our findings suggest that this repetitive, prolonged contractile muscle activity can improve metabolic regulation in prediabetic individuals without the need for a laboratory setting. SPU may be a viable and effective exercise to support metabolic health in home or work environments. However, further validation is needed with a larger sample size.
Collapse
Affiliation(s)
- Dávid Elek
- Musculoskeletal Rehabilitation Department, Fejér County Szent György University Teaching Hospital Csákvár, 8083 Csákvár, Hungary
- Faculty of Health Sciences, Doctoral School of Health Sciences, University of Pécs, 7624 Pécs, Hungary
| | - Miklós Tóth
- Faculty of Health Sciences, Institute of Physiotherapy and Sport Science, University of Pécs, 7624 Pécs, Hungary
- Physical Activity Research Group, Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Health and Sports Medicine, Hungarian University of Sports Sciences, 1123 Budapest, Hungary
- Institute of Laboratory Medicine, Semmelweis University, 1089 Budapest, Hungary
| | - Balázs Sonkodi
- Faculty of Health Sciences, Institute of Physiotherapy and Sport Science, University of Pécs, 7624 Pécs, Hungary
- Physical Activity Research Group, Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Health and Sports Medicine, Hungarian University of Sports Sciences, 1123 Budapest, Hungary
- Department of Sports Medicine, Semmelweis University, 1122 Budapest, Hungary
| | - Pongrác Ács
- Faculty of Health Sciences, Institute of Physiotherapy and Sport Science, University of Pécs, 7624 Pécs, Hungary
- Physical Activity Research Group, Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
| | - Gábor L. Kovács
- National Laboratory on Human Reproduction, University of Pécs, 7624 Pécs, Hungary
- Molecular Medicine Research Group, Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
| | - Péter Tardi
- Faculty of Health Sciences, Institute of Physiotherapy and Sport Science, University of Pécs, 7624 Pécs, Hungary
| | - Csaba Melczer
- Faculty of Health Sciences, Institute of Physiotherapy and Sport Science, University of Pécs, 7624 Pécs, Hungary
- Physical Activity Research Group, Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Faculty of Health Sciences, Complex Sport Performance Diagnostic and Physiotherapy Research Institute, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
14
|
Sonkodi B. Delayed-Onset Muscle Soreness Begins with a Transient Neural Switch. Int J Mol Sci 2025; 26:2319. [PMID: 40076941 PMCID: PMC11901069 DOI: 10.3390/ijms26052319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Unaccustomed and/or strenuous eccentric contractions are known to cause delayed-onset muscle soreness. In spite of this fact, their exact cause and mechanism have been unknown for more than 120 years. The exploration of the diverse functionality of the Piezo2 ion channel, as the principal proprioceptive component, and its autonomously acquired channelopathy may bring light to this apparently simple but mysterious pain condition. Correspondingly, the neurocentric non-contact acute compression axonopathy theory of delayed-onset muscle soreness suggests two damage phases affecting two muscle compartments, including the intrafusal (within the muscle spindle) and the extrafusal (outside the muscle spindle) ones. The secondary damage phase in the extrafusal muscle space is relatively well explored. However, the suggested primary damage phase within the muscle spindle is far from being entirely known. The current manuscript describes how the proposed autonomously acquired Piezo2 channelopathy-induced primary damage could be the initiating transient neural switch in the unfolding of delayed-onset muscle soreness. This primary damage results in a transient proprioceptive neural switch and in a switch from quantum mechanical free energy-stimulated ultrafast proton-coupled signaling to rapid glutamate-based signaling along the muscle-brain axis. In addition, it induces a transient metabolic switch or, even more importantly, an energy generation switch in Type Ia proprioceptive terminals that eventually leads to a transient glutaminolysis deficit and mitochondrial deficiency, not to mention a force generation switch. In summary, the primary damage or switch is likely an inward unidirectional proton pathway reversal between Piezo2 and its auxiliary ligands, leading to acquired Piezo2 channelopathy.
Collapse
Affiliation(s)
- Balázs Sonkodi
- Department of Health Sciences and Sport Medicine, Hungarian University of Sports Science, 1123 Budapest, Hungary;
- Department of Sports Medicine, Semmelweis University, 1122 Budapest, Hungary
| |
Collapse
|
15
|
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
|
16
|
Candia-Rivera D, Chavez M. A method for dyadic cardiac rhythmicity analysis: Preliminary evidence on bilateral interactions in fetal-maternal cardiac dynamics. Exp Physiol 2025. [PMID: 39985150 DOI: 10.1113/ep092532] [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: 12/23/2024] [Accepted: 02/05/2025] [Indexed: 02/24/2025]
Abstract
Cardiac activity responds dynamically to metabolic demands and neural regulation. However, little is known about this process during pregnancy. Reports show occasional fetal-maternal heart rate couplings, but it has remained unclear whether these couplings extend to more complex oscillatory patterns of the heart rhythm. We developed a framework of time-varying measures of heart rate and rhythm, to test the presence of co-varying patterns in concurrent maternal and fetal measures (late pregnancy dataset, n = 10, and labour dataset, n = 12). These measures were derived from first and second-order Poincaré plots, with the aim to describe changes in short- and long-term rhythmicity, but also the dynamic shifts in acceleration and deceleration of heart rate. We found episodes of maternal-fetal co-varying patterns of cardiac rhythm in all the measures explored, in both datasets (at least 90% of the dataset presented a significant maternal-fetal correlation in each measure, with P < 0.001), with dynamic delays suggesting bilateral interactions at different time scales. We also found that these couplings intensify during labour (test between late pregnancy vs. labour datasets, P < 0.0015 in all second-order Poincaré plot-derived measures). While most literature suggests that the fetal heart responds to maternal breathing patterns or contractions, we propose the possibility that the fetal heart may also have a signalling function in the context of co-regulatory mechanisms and maternal inter-organ interactions. Understanding these complex visceral oscillations in utero may enhance the assessment of a healthy fetal development.
Collapse
Affiliation(s)
- Diego Candia-Rivera
- Sorbonne Université, Paris Brain Institute (ICM), Inria Paris, CNRS UMR7225, INSERM U1127, AP-HP Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Mario Chavez
- Sorbonne Université, Paris Brain Institute (ICM), Inria Paris, CNRS UMR7225, INSERM U1127, AP-HP Hôpital de la Pitié-Salpêtrière, Paris, France
| |
Collapse
|
17
|
Candia-Rivera D, Carrion-Falgarona S, de Vico Fallani F, Chavez M. Modelling the time-resolved modulations of cardiac activity in rats: A study on pharmacological autonomic stimulation. J Physiol 2025. [PMID: 39964815 DOI: 10.1113/jp288400] [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: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
Assessing cardiac dynamics over time is essential for understanding cardiovascular health and its parallel patterns of activity with the brain. We present a methodology to estimate the time-resolved sympathetic and parasympathetic modulations of cardiac dynamics, specifically tailored for the rat heart. To evaluate the performance of our method, we study a dataset comprising spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats. These rats were administered dobutamine to elicit autonomic dynamics. The results obtained from our method demonstrated accurate time-resolved depiction of sympathetic reactivity induced by dobutamine administration. These responses closely resembled the expected autonomic alterations observed during physical exercise conditions, albeit emulated pharmacologically. We further compared our method with standard measures of low-frequency (LF) and high-frequency (HF) components, which are commonly used, although debated, for sympathetic and parasympathetic activity estimation. The comparisons with LF and HF measures further confirmed the effectiveness of our method in better capturing autonomic changes in rat cardiac dynamics. Our findings highlight the potential of our adapted method for time-resolved analysis in future clinical and translational studies involving rodent models. The validation of our approach in animal models opens new avenues for investigating the relationship between ongoing changes in cardiac activity and parallel changes in brain dynamics. Such investigations are crucial for advancing our understanding of the brain-heart connection, particularly in cases involving neurodegeneration, brain injuries and cardiovascular conditions. KEY POINTS: We developed a method for time-resolved estimation of sympathetic and parasympathetic modulations in rat cardiac dynamics, validated against standard low-frequency and high-frequency measures. We used a cohort of spontaneously hypertensive rats and Wistar-Kyoto rats, with dobutamine administration to induce autonomic responses. Our method accurately depicted time-resolved sympathetic reactivity similar to autonomic changes during physical exercise. Our findings suggest potential for future clinical and translational studies on the brain-heart connection, particularly in cardiovascular conditions.
Collapse
Affiliation(s)
- Diego Candia-Rivera
- Paris Brain Institute (ICM), CNRS UMR7225, INRIA Paris, INSERM U1127, Hôpital de la Pitié Salpêtrière AP-HP, Sorbonne Université, Paris, France
| | - Sofia Carrion-Falgarona
- Paris Brain Institute (ICM), CNRS UMR7225, INRIA Paris, INSERM U1127, Hôpital de la Pitié Salpêtrière AP-HP, Sorbonne Université, Paris, France
| | - Fabrizio de Vico Fallani
- Paris Brain Institute (ICM), CNRS UMR7225, INRIA Paris, INSERM U1127, Hôpital de la Pitié Salpêtrière AP-HP, Sorbonne Université, Paris, France
| | - Mario Chavez
- Paris Brain Institute (ICM), CNRS UMR7225, INRIA Paris, INSERM U1127, Hôpital de la Pitié Salpêtrière AP-HP, Sorbonne Université, Paris, France
| |
Collapse
|
18
|
Galazis C, Chiu CE, Arichi T, Bharath AA, Varela M. PINNing cerebral blood flow: analysis of perfusion MRI in infants using physics-informed neural networks. FRONTIERS IN NETWORK PHYSIOLOGY 2025; 5:1488349. [PMID: 40028512 PMCID: PMC11868054 DOI: 10.3389/fnetp.2025.1488349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025]
Abstract
Arterial spin labelling (ASL) magnetic resonance imaging (MRI) enables cerebral perfusion measurement, which is crucial in detecting and managing neurological issues in infants born prematurely or after perinatal complications. However, cerebral blood flow (CBF) estimation in infants using ASL remains challenging due to the complex interplay of network physiology, involving dynamic interactions between cardiac output and cerebral perfusion, as well as issues with parameter uncertainty and data noise. We propose a new spatial uncertainty-based physics-informed neural network (PINN), SUPINN, to estimate CBF and other parameters from infant ASL data. SUPINN employs a multi-branch architecture to concurrently estimate regional and global model parameters across multiple voxels. It computes regional spatial uncertainties to weigh the signal. SUPINN can reliably estimate CBF (relative error - 0.3 ± 71.7 ), bolus arrival time (AT) ( 30.5 ± 257.8 ) , and blood longitudinal relaxation time ( T 1 b ) (-4.4 ± 28.9), surpassing parameter estimates performed using least squares or standard PINNs. Furthermore, SUPINN produces physiologically plausible spatially smooth CBF and AT maps. Our study demonstrates the successful modification of PINNs for accurate multi-parameter perfusion estimation from noisy and limited ASL data in infants. Frameworks like SUPINN have the potential to advance our understanding of the complex cardio-brain network physiology, aiding in the detection and management of diseases. Source code is provided at: https://github.com/cgalaz01/supinn.
Collapse
Affiliation(s)
- Christoforos Galazis
- Department of Computing, Imperial College London, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ching-En Chiu
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Electrical Engineering, Imperial College London, London, United Kingdom
| | - Tomoki Arichi
- Centre for the Developing Brain, King’s College London, London, United Kingdom
| | - Anil A. Bharath
- Imperial Global Singapore, CREATE Tower, Singapore, Singapore
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Marta Varela
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Cardiovascular and Genomics Research Institute, City St George’s University of London, London, United Kingdom
| |
Collapse
|
19
|
Garborg CS, Ghitti B, Zhang Q, Ricotta JM, Frank N, Mueller SJ, Greenawalt DI, Turner KL, Kedarasetti RT, Mostafa M, Lee H, Costanzo F, Drew PJ. Gut-Brain Hydraulics: Brain motion and CSF circulation is driven by mechanical coupling with the abdomen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635779. [PMID: 39974937 PMCID: PMC11838368 DOI: 10.1101/2025.01.30.635779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The brain moves within the skull, but the drivers and function of this motion are not understood. We visualized brain motion relative to the skull in awake head-fixed mice using high-speed, multi-plane two-photon microscopy. Brain motion was primarily rostrally and laterally directed, and was tightly correlated with locomotion, but not with respiration or the cardiac cycle. Electromyography recordings in abdominal muscles and microCT reconstructions of the trunk and spinal vasculature showed that brain motion was driven by abdominal muscle contractions that activate a hydraulic-like vascular connection between the nervous system and the abdominal cavity. Externally-applied abdominal pressure generated brain motion similar to those seen during abdominal muscle contractions. Simulations showed that brain motion drives substantial volumes of interstitial fluid through and out of the brain (at volumetric rates several times higher than production) into the subarachnoid space, in the opposite direction of fluid flow seen during sleep. The brain is hydraulically linked to the abdominal compartment, and fluid flow in the brain is coupled to body movements, providing a mechanism by which the mechanics of the viscera directly impact brain health.
Collapse
|
20
|
Zhang Y, Ma K, Fang X, Zhang Y, Miao R, Guan H, Tian J. Targeting ion homeostasis in metabolic diseases: Molecular mechanisms and targeted therapies. Pharmacol Res 2025; 212:107579. [PMID: 39756557 DOI: 10.1016/j.phrs.2025.107579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/13/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
The incidence of metabolic diseases-hypertension, diabetes, obesity, metabolic dysfunction-associated steatotic liver disease (MASLD), and atherosclerosis-is increasing annually, imposing a significant burden on both human health and the social economy. The occurrence and development of these diseases are closely related to the disruption of ion homeostasis, which is crucial for maintaining cellular functions and metabolic equilibrium. However, the specific mechanism of ion homeostasis in metabolic diseases is still unclear. This article reviews the role of ion homeostasis in the pathogenesis of metabolic diseases and assesses its potential as a therapeutic target. Furthermore, the article explores pharmacological strategies that target ion channels and transporters, including existing drugs and emerging drugs under development. Lastly, the article discusses the development direction of future therapeutic strategies, including the possibility of gene therapy targeting specific ion channels and personalized therapy using novel biomarkers. In summary, targeting ion homeostasis provides a new perspective and potential therapeutic approach for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Yanjiao Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Kaile Ma
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xinyi Fang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuxin Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Huifang Guan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
21
|
Baker MR, Klassen BT, Jensen MA, Valencia GO, Heydari H, Ince NF, Müller KR, Miller KJ. Parameterization of intraoperative human microelectrode recordings: Linking action potential morphology to brain anatomy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.633934. [PMID: 39896506 PMCID: PMC11785027 DOI: 10.1101/2025.01.20.633934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Deep brain stimulation (DBS) is a targeted manipulation of brain circuitry to treat neurological and neuropsychiatric conditions. Optimal DBS lead placement is essential for treatment efficacy. Current targeting practice is based on preoperative and intraoperative brain imaging, intraoperative electrophysiology, and stimulation mapping. Electrophysiological mapping using extracellular microelectrode recordings aids in identifying functional subdomains, anatomical boundaries, and disease-correlated physiology. The shape of single-unit action potentials may differ due to different biophysical properties between cell-types and brain regions. Here, we describe a technique to parameterize the structure and duration of sorted spike units using a novel algorithmic approach based on canonical response parameterization, and illustrate how it may be used on DBS microelectrode recordings. Isolated spike shapes are parameterized then compared using a spike similarity metric and grouped by hierarchical clustering. When spike morphology is associated with anatomy, we find regional clustering in the human globus pallidus. This method is widely applicable for spike removal and single-unit characterization and could be integrated into intraoperative array-based technologies to enhance targeting and clinical outcomes in DBS lead placement.
Collapse
Affiliation(s)
| | | | | | | | - Hossein Heydari
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Nuri F. Ince
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, USA
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Klaus-Robert Müller
- BIFOLD and Machine Learning Group, Department of Computer Science, TU Berlin, Berlin, Germany
- Dept of Artificial Intelligence, Korea University, Seoul, Republic of Korea
- Max Planck Institute for Informatics, Saarbrücken, Germany
| | - Kai J. Miller
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, USA
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Pediatrics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
22
|
Jacob MS, Roach BJ, Mathalon DH, Ford JM. Noncanonical EEG-BOLD coupling by default and in schizophrenia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.14.25320216. [PMID: 39867401 PMCID: PMC11759611 DOI: 10.1101/2025.01.14.25320216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Neuroimaging methods rely on models of neurovascular coupling that assume hemodynamic responses evolve seconds after changes in neural activity. However, emerging evidence reveals noncanonical BOLD (blood oxygen level dependent) responses that are delayed under stress and aberrant in neuropsychiatric conditions. To investigate BOLD coupling to resting-state fluctuations in neural activity, we simultaneously recorded EEG and fMRI in people with schizophrenia and psychiatrically unaffected participants. We focus on alpha band power to examine voxelwise, time-lagged BOLD correlations. Principally, we find diversity in the temporal profile of alpha-BOLD coupling within regions of the default mode network (DMN). This includes early coupling (0-2 seconds BOLD lag) for more posterior regions, thalamus and brainstem. Anterior regions of the DMN show coupling at canonical lags (4-6 seconds), with greater lag scores associated with self-reported measures of stress and greater lag scores in participants with schizophrenia. Overall, noncanonical alpha-BOLD coupling is widespread across the DMN and other non-cortical regions, and is delayed in people with schizophrenia. These findings are consistent with a "hemo-neural" hypothesis, that blood flow and/or metabolism can regulate ongoing neural activity, and further, that the hemo-neural lag may be associated with subjective arousal or stress. Our work highlights the need for more studies of neurovascular coupling in psychiatric conditions.
Collapse
Affiliation(s)
- Michael S Jacob
- San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA, 94121, United States
- University of California, San Francisco, 505 Parnassus Ave, San Francisco, CA, 94143, United States
| | - Brian J Roach
- San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA, 94121, United States
| | - Daniel H Mathalon
- San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA, 94121, United States
- University of California, San Francisco, 505 Parnassus Ave, San Francisco, CA, 94143, United States
| | - Judith M Ford
- San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA, 94121, United States
- University of California, San Francisco, 505 Parnassus Ave, San Francisco, CA, 94143, United States
| |
Collapse
|
23
|
Candia-Rivera D, de Vico Fallani F, Boehme R, Salamone PC. Linking heartbeats with the cortical network dynamics involved in self-social touch distinction. Commun Biol 2025; 8:52. [PMID: 39809818 PMCID: PMC11733256 DOI: 10.1038/s42003-024-07448-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: 06/01/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Research on interoception has revealed the role of heartbeats in shaping our perceptual awareness and embodying a first-person perspective. These heartbeat dynamics exhibit distinct responses to various types of touch. We advanced that those dynamics are directly associated to the brain activity that allows self-other distinction. In our study encompassing self and social touch, we employed a method to quantify the distinct couplings of temporal patterns in cardiac sympathetic and parasympathetic activities with brain connectivity. Our findings revealed that social touch led to an increase in the coupling between frontoparietal networks and parasympathetic/vagal activity, particularly in alpha and gamma bands. Conversely, as social touch progressed, we observed a decrease in the coupling between brain networks and sympathetic dynamics across a broad frequency range. These results show how heartbeat dynamics are intertwined with brain organization and provide fresh evidence on the neurophysiological mechanisms of self-social touch distinction.
Collapse
Affiliation(s)
- Diego Candia-Rivera
- Sorbonne Université, Paris Brain Institute (ICM), CNRS UMR7225, INRIA Paris, INSERM U1127, AP-HP Hôpital Pitié-Salpêtrière, Paris, France.
| | - Fabrizio de Vico Fallani
- Sorbonne Université, Paris Brain Institute (ICM), CNRS UMR7225, INRIA Paris, INSERM U1127, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Rebecca Boehme
- Center for Social and Affective Neuroscience, Linköping University Hospital, Linköping, Sweden
| | - Paula C Salamone
- Center for Social and Affective Neuroscience, Linköping University Hospital, Linköping, Sweden
| |
Collapse
|
24
|
Cao Q, Zhang P, Liu F, Jin M, Wang Y, Zeng H, Weng X, Xu F. The effect of deep magnetic stimulation on the cardiac-brain axis post-sleep deprivation: a pilot study. Front Neurosci 2025; 18:1464299. [PMID: 39867450 PMCID: PMC11757894 DOI: 10.3389/fnins.2024.1464299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction Sleep deprivation (SD) significantly disrupts the homeostasis of the cardiac-brain axis, yet the neuromodulation effects of deep magnetic stimulation (DMS), a non-invasive and safe method, remain poorly understood. Methods Sixty healthy adult males were recruited for a 36-h SD study, they were assigned to the DMS group or the control group according to their individual willing. All individuals underwent heart sound measurements and functional magnetic resonance imaging scans at the experiment's onset and terminal points. During the recovery sleep phase, DMS was applied twice for 30 min before sleep onset and upon awakening to the individuals in the DMS group. Two-factor analysis was used to disclose the changes in two status and intervention effect in groups, along with Spearman rank correlation analysis to assess the correlation between brain activity and heart activity, the linear regression analysis was performed to explore the effect of DMS on brain regions to regulated the heart activity. Additionally, bootstrapping analysis was employed to verify the mediation effect. Results The results indicated that the DMS group cardiac cycle duration was 0.81 ± 0.04 s, CON group was 0.80 ± 0.03 s, DMS presented a prolong effect (F = 0.32, p = 0.02), and all heart frequency and intensity indexes value were lower than CON group (p < 0.01). Two-factor analysis demonstrated the significant differences in the left insula and orbitofrontal inferior gyrus, which DC_Weight (0.25) value were lower 0.50 (p < 0.01), 0.42 (p < 0.01) after DMS. Furthermore, the correlation analysis confirmed that the negative association between the left orbital inferior frontal and left insula with the heart sound index (p < 0.05), such as Δ left orbital inferior frontal were negatively correlated with Δ Systolic_intensity (rho = -0.33, p < 0.05), Δ Diastolic_intensity (rho = -0.41, p < 0.05), Δ S1_intensity (rho = -0.36, p < 0.05), and Δ S2_intensity (rho = -0.43, p < 0.05). Δ Left insula was negatively correlated with Δ Diastolic_intensity (rho = -0.36, p < 0.05), Δ S1_intensity (rho = -0.33, p < 0.05), and Δ S2_intensity (rho = -0.36, p < 0.05). Mediated effect analysis showed that DMS affected S2_intensity by intervening in brain regions. Conclusion These findings suggest a causal effect on the cardiac-brain axis following 36 h of SD. The non-invasive intervention of DMS effectively regulates both brain and heart functions after SD, promoting homeostatic balance. The DMS can affect the cardiac-brain axis, offering a means to restore balance following extended periods of SD.
Collapse
Affiliation(s)
- Qiongfang Cao
- Department of Evidence-Based Medicine and Social Medicine, School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| | - Peng Zhang
- Department of Evidence-Based Medicine and Social Medicine, School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| | - Fangfang Liu
- Art College of Southwest Minzu University, Chengdu, Sichuan, China
| | - Mengyan Jin
- Department of Internal Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Yuhan Wang
- Department of Evidence-Based Medicine and Social Medicine, School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| | - Hanrui Zeng
- Department of Radiology, Sichuan Taikang Hospital, Chengdu, Sichuan, China
| | - Xiechuan Weng
- Department of Neuroscience, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Fan Xu
- Department of Evidence-Based Medicine and Social Medicine, School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
25
|
Beck C, Kunze A. Parallelized Mechanical Stimulation of Neuronal Calcium Through Cell-Internal Nanomagnetic Forces Provokes Lasting Shifts in the Network Activity State. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406678. [PMID: 39460486 PMCID: PMC11812431 DOI: 10.1002/smll.202406678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Neurons differentiate mechanical stimuli force and rate to elicit unique functional responses, driving the need for further tools to generate various mechanical stimuli. Here, cell-internal nanomagnetic forces (iNMF) are introduced by manipulating internalized magnetic nanoparticles with an external magnetic field across cortical neuron networks in vitro. Under iNMF, cortical neurons exhibit calcium (Ca2+) influx, leading to modulation of activity observed through Ca2+ event rates. Inhibiting particle uptake or altering nanoparticle exposure time reduced the neuronal response to nanomagnetic forces, exposing the requirement of nanoparticle uptake to induce the Ca2+ response. In highly active cortical networks, iNMF robustly modulates synchronous network activity, which is lasting and repeatable. Using pharmacological blockers, it is shown that iNMF activates mechanosensitive ion channels to induce the Ca2+ influx. Then, in contrast to transient mechanically evoked neuronal activity, iNMF activates Ca2+-activated potassium (KCa) channels to stabilize the neuronal membrane potential and induce network activity shifts. The findings reveal the potential of magnetic nanoparticle-mediated mechanical stimulation to modulate neuronal circuit dynamics, providing insights into the biophysics of neuronal computation.
Collapse
Affiliation(s)
- Connor Beck
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, USA
| | - Anja Kunze
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, USA
- Montana Nanotechnology Facility, Montana State University, Bozeman, Montana 59717, USA
- Optical Technology Center, Montana State University, Bozeman, Montana, 59717, USA
| |
Collapse
|
26
|
Greenwood BM, Garfinkel SN. Interoceptive Mechanisms and Emotional Processing. Annu Rev Psychol 2025; 76:59-86. [PMID: 39423429 DOI: 10.1146/annurev-psych-020924-125202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Interoception, the sensing of internal bodily signals, is intricately linked with the experience of emotions. Various theoretical models of emotion incorporate aspects of interoception as a fundamental component alongside higher-order processes such as the appraisal of internal signals guided by external context. Interoception can be delineated into different dimensions, which include the nature of afferent signals, the accuracy with which they can be sensed, their neural processing, and the higher-order interpretation of these signals. This review methodically evaluates these interoceptive dimensions through empirical research to illustrate their role in shaping emotions. Clinical and neurodevelopmental conditions characterized by altered emotional profiles, such as anxiety, depression, schizophrenia, posttraumatic stress disorder, emotionally unstable personality disorder, and autism, exhibit distinct changes in interoception. Various therapeutic approaches, including behavioral, pharmacological, and psychological strategies, may be efficacious for treating conditions associated with emotional alterations by targeting interoceptive mechanisms.
Collapse
Affiliation(s)
- Benedict M Greenwood
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom;
| | - Sarah N Garfinkel
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom;
| |
Collapse
|
27
|
Beck C, Kirby AM, Roberts S, Kunze A. Multimodal Characterization of Cortical Neuron Response to Permanent Magnetic Field Induced Nanomagnetic Force Maps. ACS NANO 2024; 18:34630-34645. [PMID: 39654337 PMCID: PMC11674720 DOI: 10.1021/acsnano.4c09542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024]
Abstract
Nanomagnetic forces deliver precise mechanical cues to biological systems through the remote pulling of magnetic nanoparticles under a permanent magnetic field. Cortical neurons respond to nanomagnetic forces with cytosolic calcium influx and event rate shifts. However, the underlying consequences of nanomagnetic force modulation on cortical neurons remain to be elucidated. Here, we integrate electrophysiological and optical recording modalities with nanomagnetic forces to characterize the in vitro functional response to mechanical cues. Neurons exposed to chitosan functionalized magnetic nanoparticles for 24 h and then exposed to magnetic fields capable of generating forces of 2-160 pN present elevated cytosolic calcium in neurons and a time-dynamic electrophysiological spike rate and magnitude response. Extracellular recordings with microelectrode arrays revealed a 2-8 pN force-specific increase in electrophysiological spiking with a trend in reduced activity following 2 min of continuous force exposure. Nanomagnetic forces in the 16-160 pN range produced increased electrophysiological activity and remained excited for up to 4 h under continuous stimulation before silencing. Furthermore, the neuronal response to nanomagnetic forces at 16-160 pN can be electrophysiologically mediated without calcium influx by altering the magnetic nanoparticle-neuron interactions. These results demonstrate that low pN nanomagnetic forces mediate neuronal function and suggest that magnetic nanoparticle interactions and force magnitudes can be harnessed to provoke different responses in cortical neurons.
Collapse
Affiliation(s)
- Connor
L. Beck
- Department
of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, United States
| | - Andrew M. Kirby
- Department
of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, United States
| | - Samuel Roberts
- Department
of Chemical Engineering, Montana State University, Bozeman, Montana 59717, United States
| | - Anja Kunze
- Department
of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, United States
- Montana
Nanotechnology Facility, Montana State University, Bozeman, Montana 59717, United States
- Optical
Technology Center, Montana State University, Bozeman, Montana 59717, United States
| |
Collapse
|
28
|
Broillet-Olivier E, Wenger Y, Gilliand N, Cadas H, Sabatasso S, Broillet MC, Brechbühl J. Development of an rpS6-Based Ex Vivo Assay for the Analysis of Neuronal Activity in Mouse and Human Olfactory Systems. Int J Mol Sci 2024; 25:13173. [PMID: 39684883 DOI: 10.3390/ijms252313173] [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: 10/22/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Olfactory sensitivity to odorant molecules is a complex biological function influenced by both endogenous factors, such as genetic background and physiological state, and exogenous factors, such as environmental conditions. In animals, this vital ability is mediated by olfactory sensory neurons (OSNs), which are distributed across several specialized olfactory subsystems depending on the species. Using the phosphorylation of the ribosomal protein S6 (rpS6) in OSNs following sensory stimulation, we developed an ex vivo assay allowing the simultaneous conditioning and odorant stimulation of different mouse olfactory subsystems, including the main olfactory epithelium, the vomeronasal organ, and the Grueneberg ganglion. This approach enabled us to observe odorant-induced neuronal activity within the different olfactory subsystems and to demonstrate the impact of environmental conditioning, such as temperature variations, on olfactory sensitivity, specifically in the Grueneberg ganglion. We further applied our rpS6-based assay to the human olfactory system and demonstrated its feasibility. Our findings show that analyzing rpS6 signal intensity is a robust and highly reproducible indicator of neuronal activity across various olfactory systems, while avoiding stress and some experimental limitations associated with in vivo exposure. The potential extension of this assay to other conditioning paradigms and olfactory systems, as well as its application to other animal species, including human olfactory diagnostics, is also discussed.
Collapse
Affiliation(s)
- Emma Broillet-Olivier
- Faculty of Medicine Hradec Králové, Charles University, 500 00 Hradec Králové, Czech Republic
| | - Yaëlle Wenger
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Bugnon 27, CH-1011 Lausanne, Switzerland
| | - Noah Gilliand
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Bugnon 27, CH-1011 Lausanne, Switzerland
| | - Hugues Cadas
- Faculty of Biology and Medicine, University of Lausanne, Bugnon 9, CH-1005 Lausanne, Switzerland
- Faculty Unit of Anatomy and Morphology, University Center of Legal Medicine Lausanne-Geneva, Lausanne University Hospital and University of Lausanne, Vulliette 4, CH-1000 Lausanne, Switzerland
| | - Sara Sabatasso
- Faculty of Biology and Medicine, University of Lausanne, Bugnon 9, CH-1005 Lausanne, Switzerland
- Faculty Unit of Anatomy and Morphology, University Center of Legal Medicine Lausanne-Geneva, Lausanne University Hospital and University of Lausanne, Vulliette 4, CH-1000 Lausanne, Switzerland
| | - Marie-Christine Broillet
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Bugnon 27, CH-1011 Lausanne, Switzerland
| | - Julien Brechbühl
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Bugnon 27, CH-1011 Lausanne, Switzerland
| |
Collapse
|
29
|
Carvalho EM, Ding EA, Saha A, Garcia DC, Weldy A, Zushin PJH, Stahl A, Aghi MK, Kumar S. Viscoelastic High-Molecular-Weight Hyaluronic Acid Hydrogels Support Rapid Glioblastoma Cell Invasion with Leader-Follower Dynamics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404885. [PMID: 39508297 PMCID: PMC11637900 DOI: 10.1002/adma.202404885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/25/2024] [Indexed: 11/15/2024]
Abstract
Hyaluronic acid (HA), the primary component of brain extracellular matrix, is increasingly used to model neuropathological processes, including glioblastoma (GBM) tumor invasion. While elastic hydrogels based on crosslinked low-molecular-weight (LMW) HA are widely exploited for this purpose and have proven valuable for discovery and screening, brain tissue is both viscoelastic and rich in high-MW (HMW) HA, and it remains unclear how these differences influence invasion. To address this question, hydrogels comprised of either HMW (1.5 MDa) or LMW (60 kDa) HA are introduced, characterized, and applied in GBM invasion studies. Unlike LMW HA hydrogels, HMW HA hydrogels relax stresses quickly, to a similar extent as brain tissue, and to a greater extent than many conventional HA-based scaffolds. GBM cells implanted within HMW HA hydrogels invade much more rapidly than in their LMW HA counterparts and exhibit distinct leader-follower dynamics. Leader cells adopt dendritic morphologies similar to invasive GBM cells observed in vivo. Transcriptomic, pharmacologic, and imaging studies suggest that leader cells exploit hyaluronidase, an enzyme strongly enriched in human GBMs, to prime a path for followers. This study offers new insight into how HA viscoelastic properties drive invasion and argues for the use of highly stress-relaxing materials to model GBM.
Collapse
Affiliation(s)
- Emily M Carvalho
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Erika A Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Atul Saha
- Department of Neurosurgery, University of California, San Francisco, CA, 94158, USA
| | - Diana Cruz Garcia
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94158, USA
| | - Anna Weldy
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Peter-James H Zushin
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 94720, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 94720, USA
| | - Manish K Aghi
- Department of Neurosurgery, University of California, San Francisco, CA, 94158, USA
| | - Sanjay Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94158, USA
| |
Collapse
|
30
|
Sonkodi B. Proton-Mediated PIEZO2 Channelopathy: Linking Oxaliplatin Treatment to Impaired Proprioception and Cognitive Deficits. Cancers (Basel) 2024; 16:3898. [PMID: 39682086 DOI: 10.3390/cancers16233898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Oxaliplatin induces acute neuropathy within a few hours post-treatment, with symptoms persisting for several days. Delayed onset muscle soreness also causes the delayed onset of mechanical pain sensation starting at about 6-8 h and lasting up to a week after exercise. Both conditions come with impaired proprioception and could be chronic if these bouts are repeated frequently. The involvement of PIEZO2 ion channels, as the principal mechanosensory channels responsible for proprioception, is theorized in both conditions as well. The current opinion manuscript is meant to explain how the minor stretch-related microdamage of PIEZO2 on Type Ia proprioceptive terminals could explain the aforementioned symptoms of impaired proprioception. This includes a platinum-induced proton affinity 'switch' on these proprioceptive endings with PIEZO2 content, resulting in this being the likely initiating cause. Furthermore, it postulates how the proton-based ultrafast long-range oscillatory synchronization to the hippocampus could be impaired due to this microdamage on Type Ia proprioceptive terminals. Finally, the manuscript provides insight into how the impairment of the PIEZO2-initiated ultrafast muscle-brain axis may contribute to chemobrain and its associated cognitive and memory deficits.
Collapse
Affiliation(s)
- Balázs Sonkodi
- Department of Health Sciences and Sport Medicine, Hungarian University of Sports Science, 1123 Budapest, Hungary
- Department of Sports Medicine, Semmelweis University, 1122 Budapest, Hungary
| |
Collapse
|
31
|
Lovelace JW, Ma J, Augustine V. Defining cardioception: Heart-brain crosstalk. Neuron 2024; 112:3671-3674. [PMID: 39500326 DOI: 10.1016/j.neuron.2024.10.009] [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: 08/01/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/24/2024]
Abstract
Interoception, the sensation and perception of internal bodily states, should be conceptualized through specialized modalities like cardioception, pulmoception, gastroception, and uroception. This NeuroView emphasizes cardioception, exploring heart-brain interactions, cardiac reflexes, and their influence on mental states and behavior.
Collapse
Affiliation(s)
| | - Jingrui Ma
- Department of Neurobiology, University of California, San Diego, CA, USA
| | - Vineet Augustine
- Department of Neurobiology, University of California, San Diego, CA, USA.
| |
Collapse
|
32
|
Lai G, Landi D, Vidaurre C, Bhattacharya J, Herrojo Ruiz M. Cardiac cycle modulates alpha and beta suppression during motor imagery. Cereb Cortex 2024; 34:bhae442. [PMID: 39579013 PMCID: PMC11584698 DOI: 10.1093/cercor/bhae442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/13/2024] [Accepted: 10/23/2024] [Indexed: 11/25/2024] Open
Abstract
Previous interoception research has demonstrated that sensory processing is reduced during cardiac systole, an effect associated with diminished cortical excitability, possibly due to heightened baroreceptor activity. This study aims to determine how phases of the cardiac cycle-systole and diastole-modulate neural sensorimotor activity during motor imagery (MI) and motor execution (ME). We hypothesised that MI performance, indexed by enhanced suppression of contralateral sensorimotor alpha (8-13 Hz) and beta (14-30 Hz) activity, would be modulated by the cardiac phases, with improved performance during diastole due to enhanced sensory processing of movement cues. Additionally, we investigated whether movement cues during systole or diastole enhance muscle activity. To test these hypotheses, 29 participants were instructed to perform or imagine thumb abductions, while we recorded their electroencephalography, electrocardiogram, and electromyogram (EMG) activity. We show that imaginary movements instructed during diastole lead to more pronounced suppression of alpha and beta activity in contralateral sensorimotor cortices, with no significant cardiac timing effects observed during ME as confirmed by circular statistics. Additionally, diastole was associated with significantly increased EMG on the side of actual and, to a lesser degree, imagined movements. Our study identifies optimal cardiac phases for MI performance, suggesting potential pathways to enhance MI-based assistive technologies.
Collapse
Affiliation(s)
- Giuseppe Lai
- Goldsmiths, University of London, New Cross London SE14 6NW, UK
| | | | - Carmen Vidaurre
- Basque Center on Cognition Brain and Language, Paseo Mikeletegi 69, 2°20009 Donostia San Sebastián, Gipuzkoa, Spain
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi, 548009 Bilbao, Spain
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Straße des 17, Juni 13510623, Berlin, Germany
| | | | | |
Collapse
|
33
|
Mou YK, Song XY, Wang HR, Wang Y, Liu WC, Yang T, Zhang MJ, Hu Y, Ren C, Song XC. Understanding the nose-brain axis and its role in related diseases: A conceptual review. Neurobiol Dis 2024; 202:106690. [PMID: 39389156 DOI: 10.1016/j.nbd.2024.106690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
The nose-brain axis (NBA), a critical component of the body-brain axis, not only serves as a drug transport route for the treatment of brain diseases but also mediates changes such as neuroimmune disorders, which may be an important mechanism in the occurrence and development of some nasal or brain diseases. Despite its importance, there are substantial gaps that remain in our understanding of the characteristics of NBA-mediated diseases and of the cellular and molecular mechanisms underlying the bidirectional NBA crosstalk. These gaps have limited the translational application of NBA-related research findings to some extent. Therefore, this review aims to address the conceptual framework of NBA and highlight its values in representative diseases by combining existing literature with new research results from our group. We hope that this paper will provide a basis for further in-depth research in this field, and facilitate the clinical translation of NBA.
Collapse
Affiliation(s)
- Ya-Kui Mou
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Xiao-Yu Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Han-Rui Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Yao Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Wan-Chen Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Ting Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Ming-Jun Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Yue Hu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Chao Ren
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Department of Neurology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
| | - Xi-Cheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
| |
Collapse
|
34
|
Zhao Y, Lian Y, Di H, Zhao W. Rapid coupling between vasculature and neurons through mechanosensitive channels in the olfactory lobe. Front Hum Neurosci 2024; 18:1435859. [PMID: 39435349 PMCID: PMC11491361 DOI: 10.3389/fnhum.2024.1435859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/20/2024] [Indexed: 10/23/2024] Open
Affiliation(s)
- Yilin Zhao
- International Vegetative State and Consciousness Science Institute, Hangzhou Normal University, Hangzhou, China
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yitong Lian
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Haibo Di
- International Vegetative State and Consciousness Science Institute, Hangzhou Normal University, Hangzhou, China
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Weiqiao Zhao
- International Vegetative State and Consciousness Science Institute, Hangzhou Normal University, Hangzhou, China
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
35
|
Candia-Rivera D, Engelen T, Babo-Rebelo M, Salamone PC. Interoception, network physiology and the emergence of bodily self-awareness. Neurosci Biobehav Rev 2024; 165:105864. [PMID: 39208877 DOI: 10.1016/j.neubiorev.2024.105864] [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: 06/28/2024] [Revised: 08/06/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The interplay between the brain and interoceptive signals is key in maintaining internal balance and orchestrating neural dynamics, encompassing influences on perceptual and self-awareness. Central to this interplay is the differentiation between the external world, others and the self, a cornerstone in the construction of bodily self-awareness. This review synthesizes physiological and behavioral evidence illustrating how interoceptive signals can mediate or influence bodily self-awareness, by encompassing interactions with various sensory modalities. To deepen our understanding of the basis of bodily self-awareness, we propose a network physiology perspective. This approach explores complex neural computations across multiple nodes, shifting the focus from localized areas to large-scale neural networks. It examines how these networks operate in parallel with and adapt to changes in visceral activities. Within this framework, we propose to investigate physiological factors that disrupt bodily self-awareness, emphasizing the impact of interoceptive pathway disruptions, offering insights across several clinical contexts. This integrative perspective not only can enhance the accuracy of mental health assessments but also paves the way for targeted interventions.
Collapse
Affiliation(s)
- Diego Candia-Rivera
- Sorbonne Université, Paris Brain Institute (ICM), CNRS UMR7225, INSERM U1127, Hôpital de la Pitié-Salpêtrière AP-HP, Inria Paris, 75013, Paris, France.
| | - Tahnée Engelen
- Department of Psychology and Centre for Interdisciplinary Brain Research, University of Jyväskylä, Mattilanniemi 6, Jyväskylä FI-40014, Finland
| | - Mariana Babo-Rebelo
- Laboratory of Cognitive Neuroscience, Neuro-X Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | - Paula C Salamone
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| |
Collapse
|
36
|
Guo F, Zhao C, Shou Q, Jin N, Jann K, Shao X, Wang DJJ. Assessing Cerebral Microvascular Volumetric Pulsatility with High-Resolution 4D CBV MRI at 7T. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.04.24313077. [PMID: 39281763 PMCID: PMC11398588 DOI: 10.1101/2024.09.04.24313077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Arterial pulsation is crucial for promoting fluid circulation and for influencing neuronal activity. Previous studies assessed the pulsatility index based on blood flow velocity pulsatility in relatively large cerebral arteries of human. Here, we introduce a novel method to quantify the volumetric pulsatility of cerebral microvasculature across cortical layers and in white matter (WM), using high-resolution 4D vascular space occupancy (VASO) MRI with simultaneous recording of pulse signals at 7T. Microvascular volumetric pulsatility index (mvPI) and cerebral blood volume (CBV) changes across cardiac cycles are assessed through retrospective sorting of VASO signals into cardiac phases and estimating mean CBV in resting state (CBV0) by arterial spin labeling (ASL) MRI at 7T. Using data from 11 young (28.4±5.8 years) and 7 older (61.3±6.2 years) healthy participants, we investigated the aging effect on mvPI and compared microvascular pulsatility with large arterial pulsatility assessed by 4D-flow MRI. We observed the highest mvPI in the cerebrospinal fluid (CSF) on the cortical surface (0.19±0.06), which decreased towards the cortical layers as well as in larger arteries. In the deep WM, a significantly increased mvPI (p = 0.029) was observed in the older participants compared to younger ones. Additionally, mvPI in deep WM is significantly associated with the velocity pulsatility index (vePI) of large arteries (r = 0.5997, p = 0.0181). We further performed test-retest scans, non-parametric reliability test and simulations to demonstrate the reproducibility and accuracy of our method. To the best of our knowledge, our method offers the first in vivo measurement of microvascular volumetric pulsatility in human brain which has implications for cerebral microvascular health and its relationship research with glymphatic system, aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Fanhua Guo
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Chenyang Zhao
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Qinyang Shou
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | | | - Kay Jann
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Danny JJ Wang
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| |
Collapse
|
37
|
Decoster L, Trova S, Zucca S, Bulk J, Gouveia A, Ternier G, Lhomme T, Legrand A, Gallet S, Boehm U, Wyatt A, Wahl V, Wartenberg P, Hrabovszky E, Rácz G, Luzzati F, Nato G, Fogli M, Peretto P, Schriever SC, Bernecker M, Pfluger PT, Steculorum SM, Bovetti S, Rasika S, Prevot V, Silva MSB, Giacobini P. A GnRH neuronal population in the olfactory bulb translates socially relevant odors into reproductive behavior in male mice. Nat Neurosci 2024; 27:1758-1773. [PMID: 39095587 DOI: 10.1038/s41593-024-01724-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Hypothalamic gonadotropin-releasing hormone (GnRH) neurons regulate fertility and integrate hormonal status with environmental cues to ensure reproductive success. Here we show that GnRH neurons in the olfactory bulb (GnRHOB) of adult mice can mediate social recognition. Specifically, we show that GnRHOB neurons extend neurites into the vomeronasal organ and olfactory epithelium and project to the median eminence. GnRHOB neurons in males express vomeronasal and olfactory receptors, are activated by female odors and mediate gonadotropin release in response to female urine. Male preference for female odors required the presence and activation of GnRHOB neurons, was impaired after genetic inhibition or ablation of these cells and relied on GnRH signaling in the posterodorsal medial amygdala. GnRH receptor expression in amygdala kisspeptin neurons appear to be required for GnRHOB neurons' actions on male mounting behavior. Taken together, these results establish GnRHOB neurons as regulating fertility, sex recognition and mating in male mice.
Collapse
Affiliation(s)
- Laurine Decoster
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Sara Trova
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
- Centro CMP3VdA, Istituto Italiano di Tecnologia (IIT), Aosta, Italy
| | - Stefano Zucca
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Janice Bulk
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
| | - Ayden Gouveia
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
| | - Gaetan Ternier
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Amandine Legrand
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Sarah Gallet
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Amanda Wyatt
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Vanessa Wahl
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Philipp Wartenberg
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Hun-Ren Institute of Experimental Medicine, Budapest, Hungary
| | - Gergely Rácz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Federico Luzzati
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Giulia Nato
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Marco Fogli
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Paolo Peretto
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Sonja C Schriever
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Miriam Bernecker
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of Neurobiology of Diabetes, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Paul T Pfluger
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of Neurobiology of Diabetes, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Sophie M Steculorum
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Serena Bovetti
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Sowmyalakshmi Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Mauro S B Silva
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France.
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France.
| |
Collapse
|
38
|
Horrocks EAB, Rodrigues FR, Saleem AB. Flexible neural population dynamics govern the speed and stability of sensory encoding in mouse visual cortex. Nat Commun 2024; 15:6415. [PMID: 39080254 PMCID: PMC11289260 DOI: 10.1038/s41467-024-50563-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: 12/11/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Time courses of neural responses underlie real-time sensory processing and perception. How these temporal dynamics change may be fundamental to how sensory systems adapt to different perceptual demands. By simultaneously recording from hundreds of neurons in mouse primary visual cortex, we examined neural population responses to visual stimuli at sub-second timescales, during different behavioural states. We discovered that during active behavioural states characterised by locomotion, single-neurons shift from transient to sustained response modes, facilitating rapid emergence of visual stimulus tuning. Differences in single-neuron response dynamics were associated with changes in temporal dynamics of neural correlations, including faster stabilisation of stimulus-evoked changes in the structure of correlations during locomotion. Using Factor Analysis, we examined temporal dynamics of latent population responses and discovered that trajectories of population activity make more direct transitions between baseline and stimulus-encoding neural states during locomotion. This could be partly explained by dampening of oscillatory dynamics present during stationary behavioural states. Functionally, changes in temporal response dynamics collectively enabled faster, more stable and more efficient encoding of new visual information during locomotion. These findings reveal a principle of how sensory systems adapt to perceptual demands, where flexible neural population dynamics govern the speed and stability of sensory encoding.
Collapse
Affiliation(s)
- Edward A B Horrocks
- Institute of Behavioural Neuroscience, University College London, London, WC1V 0AP, UK.
| | - Fabio R Rodrigues
- Institute of Behavioural Neuroscience, University College London, London, WC1V 0AP, UK
| | - Aman B Saleem
- Institute of Behavioural Neuroscience, University College London, London, WC1V 0AP, UK.
| |
Collapse
|
39
|
Wang ZJ, Sun L, Heinbockel T. Firing Patterns of Mitral Cells and Their Transformation in the Main Olfactory Bulb. Brain Sci 2024; 14:678. [PMID: 39061419 PMCID: PMC11275187 DOI: 10.3390/brainsci14070678] [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: 05/31/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Mitral cells (MCs) in the main olfactory bulb relay odor information to higher-order olfactory centers by encoding the information in the form of action potentials. The firing patterns of these cells are influenced by both their intrinsic properties and their synaptic connections within the neural network. However, reports on MC firing patterns have been inconsistent, and the mechanisms underlying these patterns remain unclear. Using whole-cell patch-clamp recordings in mouse brain slices, we discovered that MCs exhibit two types of integrative behavior: regular/rhythmic firing and bursts of action potentials. These firing patterns could be transformed both spontaneously and chemically. MCs with regular firing maintained their pattern even in the presence of blockers of fast synaptic transmission, indicating this was an intrinsic property. However, regular firing could be transformed into bursting by applying GABAA receptor antagonists to block inhibitory synaptic transmission. Burst firing could be reverted to regular firing by blocking ionotropic glutamate receptors, rather than applying a GABAA receptor agonist, indicating that ionotropic glutamatergic transmission mediated this transformation. Further experiments on long-lasting currents (LLCs), which generated burst firing, also supported this mechanism. In addition, cytoplasmic Ca2+ in MCs was involved in the transformation of firing patterns mediated by glutamatergic transmission. Metabotropic glutamate receptors also played a role in LLCs in MCs. These pieces of evidence indicate that odor information can be encoded on a mitral cell (MC) platform, where it can be relayed to higher-order olfactory centers through intrinsic and dendrodendritic mechanisms in MCs.
Collapse
Affiliation(s)
- Ze-Jun Wang
- Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Liqin Sun
- Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA
| | - Thomas Heinbockel
- Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA
| |
Collapse
|
40
|
Molcan L. Short-term blood pressure variability and reduced cytosolic calcium levels. Hypertens Res 2024; 47:1425-1428. [PMID: 38467799 DOI: 10.1038/s41440-024-01640-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 02/24/2024] [Indexed: 03/13/2024]
Affiliation(s)
- Lubos Molcan
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, Bratislava, Slovakia.
| |
Collapse
|
41
|
Guo J, Wan X, Lian J, Ma H, Dong D, Liu Y, Zhao J. Electrophysiological Characteristics of Inhibitive Control for Adults with Different Physiological or Psychological Obesity. Nutrients 2024; 16:1252. [PMID: 38732499 PMCID: PMC11085209 DOI: 10.3390/nu16091252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 05/13/2024] Open
Abstract
Individuals exhibiting high scores on the fatness subscale of the negative-physical-self scale (NPSS-F) are characterized by heightened preoccupation with body fat accompanied by negative body image perceptions, often leading to excessive dieting behaviors. This demographic constitutes a considerable segment of the populace in China, even among those who are not obese. Nonetheless, scant empirical inquiries have delved into the behavioral and neurophysiological profiles of individuals possessing a healthy body mass index (BMI) alongside elevated NPSS-F scores. This study employed an experimental paradigm integrating go/no-go and one-back tasks to assess inhibitory control and working memory capacities concerning food-related stimuli across three adult cohorts: those with normal weight and low NPSS-F scores, those with normal weight and high NPSS-F scores, and individuals classified as obese. Experimental stimuli comprised high- and low-caloric-food pictures with concurrent electroencephalogram (EEG) and photoplethysmogram (PPG) recordings. Individuals characterized by high NPSS-F scores and normal weight exhibited distinctive electrophysiological responses compared to the other two cohorts, evident in event-related potential (ERP) components, theta and alpha band oscillations, and heart rate variability (HRV) patterns. In essence, the findings underscore alterations in electrophysiological reactivity among individuals possessing high NPSS-F scores and a healthy BMI in the context of food-related stimuli, underscoring the necessity for increased attention to this demographic alongside individuals affected by obesity.
Collapse
Affiliation(s)
- Jiaqi Guo
- Faculty of Psychology, Southwest University, Chongqing 400715, China; (J.G.); (X.W.); (J.L.); (H.M.); (D.D.)
| | - Xiaofang Wan
- Faculty of Psychology, Southwest University, Chongqing 400715, China; (J.G.); (X.W.); (J.L.); (H.M.); (D.D.)
| | - Junwei Lian
- Faculty of Psychology, Southwest University, Chongqing 400715, China; (J.G.); (X.W.); (J.L.); (H.M.); (D.D.)
| | - Hanqing Ma
- Faculty of Psychology, Southwest University, Chongqing 400715, China; (J.G.); (X.W.); (J.L.); (H.M.); (D.D.)
| | - Debo Dong
- Faculty of Psychology, Southwest University, Chongqing 400715, China; (J.G.); (X.W.); (J.L.); (H.M.); (D.D.)
- Key Laboratory of Cognition and Personality (Ministry of Education), Southwest University, Chongqing 400715, China
| | - Yong Liu
- Faculty of Psychology, Southwest University, Chongqing 400715, China; (J.G.); (X.W.); (J.L.); (H.M.); (D.D.)
- Key Laboratory of Cognition and Personality (Ministry of Education), Southwest University, Chongqing 400715, China
| | - Jia Zhao
- Faculty of Psychology, Southwest University, Chongqing 400715, China; (J.G.); (X.W.); (J.L.); (H.M.); (D.D.)
- Key Laboratory of Cognition and Personality (Ministry of Education), Southwest University, Chongqing 400715, China
| |
Collapse
|
42
|
Carvalho EM, Ding EA, Saha A, Weldy A, Zushin PJH, Stahl A, Aghi MK, Kumar S. Viscoelastic high-molecular-weight hyaluronic acid hydrogels support rapid glioblastoma cell invasion with leader-follower dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588167. [PMID: 38617333 PMCID: PMC11014578 DOI: 10.1101/2024.04.04.588167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Hyaluronic acid (HA), the primary component of brain extracellular matrix, is increasingly used to model neuropathological processes, including glioblastoma (GBM) tumor invasion. While elastic hydrogels based on crosslinked low-molecular-weight (LMW) HA are widely exploited for this purpose and have proven valuable for discovery and screening, brain tissue is both viscoelastic and rich in high-MW (HMW) HA, and it remains unclear how these differences influence invasion. To address this question, hydrogels comprised of either HMW (1.5 MDa) or LMW (60 kDa) HA are introduced, characterized, and applied in GBM invasion studies. Unlike LMW HA hydrogels, HMW HA hydrogels relax stresses quickly, to a similar extent as brain tissue, and to a greater extent than many conventional HA-based scaffolds. GBM cells implanted within HMW HA hydrogels invade much more rapidly than in their LMW HA counterparts and exhibit distinct leader-follower dynamics. Leader cells adopt dendritic morphologies, similar to invasive GBM cells observed in vivo. Transcriptomic, pharmacologic, and imaging studies suggest that leader cells exploit hyaluronidase, an enzyme strongly enriched in human GBMs, to prime a path for followers. This study offers new insight into how HA viscoelastic properties drive invasion and argues for the use of highly stress-relaxing materials to model GBM.
Collapse
Affiliation(s)
- Emily M Carvalho
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Erika A Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Atul Saha
- Department of Neurosurgery, University of California, San Francisco, CA 94158, USA
| | - Anna Weldy
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Peter-James H Zushin
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley 94720, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley 94720, USA
| | - Manish K Aghi
- Department of Neurosurgery, University of California, San Francisco, CA 94158, USA
| | - Sanjay Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
43
|
Whalley K. Olfactory neurons can feel the (heart) beat. Nat Rev Neurosci 2024; 25:210. [PMID: 38409297 DOI: 10.1038/s41583-024-00801-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
|
44
|
Huynh K. Heartbeat-induced pressure pulsations in cerebral arteries modulate neuronal activity. Nat Rev Cardiol 2024; 21:218. [PMID: 38374341 DOI: 10.1038/s41569-024-00999-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
|
45
|
Hamel R, Oyler R, Harms E, Bailey R, Rendeiro C, Jenkinson N. Dietary Cocoa Flavanols Do Not Alter Brain Excitability in Young Healthy Adults. Nutrients 2024; 16:969. [PMID: 38613003 PMCID: PMC11013095 DOI: 10.3390/nu16070969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
The ingestion of dietary cocoa flavanols acutely alters functions of the cerebral endothelium, but whether the effects of flavanols permeate beyond this to alter other brain functions remains unclear. Based on converging evidence, this work tested the hypothesis that cocoa flavanols would alter brain excitability in young healthy adults. In a randomised, cross-over, double-blinded, placebo-controlled design, transcranial magnetic stimulation was used to assess corticospinal and intracortical excitability before as well as 1 and 2 h post-ingestion of a beverage containing either high (695 mg flavanols, 150 mg (-)-epicatechin) or low levels (5 mg flavanols, 0 mg (-)-epicatechin) of cocoa flavanols. In addition to this acute intervention, the effects of a short-term chronic intervention where the same cocoa flavanol doses were ingested once a day for 5 consecutive days were also investigated. For both the acute and chronic interventions, the results revealed no robust alteration in corticospinal or intracortical excitability. One possibility is that cocoa flavanols yield no net effect on brain excitability, but predominantly alter functions of the cerebral endothelium in young healthy adults. Future studies should increase intervention durations to maximize the acute and chronic accumulation of flavanols in the brain, and further investigate if cocoa flavanols would be more effective at altering brain excitability in older adults and clinical populations than in younger adults.
Collapse
Affiliation(s)
- Raphael Hamel
- School of Sports, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Rebecca Oyler
- School of Sports, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Evie Harms
- School of Sports, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Rosamond Bailey
- School of Sports, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Catarina Rendeiro
- School of Sports, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Ned Jenkinson
- School of Sports, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
46
|
Hamill OP. Arterial pulses link heart-brain oscillations. Science 2024; 383:482-483. [PMID: 38301020 DOI: 10.1126/science.adn4942] [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: 02/03/2024]
Abstract
A central baroreceptor monitors arterial pressure to modulate brain activity.
Collapse
Affiliation(s)
- Owen P Hamill
- Department of Neurobiology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| |
Collapse
|