1
|
Pavlov VA. Novel opportunities for treating complex neuropsychiatric and neurocognitive conditions based on recent developments with xanomeline. Front Psychiatry 2025; 16:1593341. [PMID: 40443751 PMCID: PMC12119645 DOI: 10.3389/fpsyt.2025.1593341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/24/2025] [Indexed: 06/02/2025] Open
Affiliation(s)
- Valentin A. Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
| |
Collapse
|
2
|
Cembellin-Prieto A, Luo Z, Kulaga H, Baumgarth N. B cells modulate lung antiviral inflammatory responses via the neurotransmitter acetylcholine. Nat Immunol 2025; 26:775-789. [PMID: 40263611 PMCID: PMC12043518 DOI: 10.1038/s41590-025-02124-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 03/11/2025] [Indexed: 04/24/2025]
Abstract
The rapid onset of innate immune defenses is critical for early control of viral replication in an infected host and yet it can also lead to irreversible tissue damage, especially in the respiratory tract. Sensitive regulators must exist that modulate inflammation, while controlling the infection. In the present study, we identified acetylcholine (ACh)-producing B cells as such early regulators. B cells are the most prevalent ACh-producing leukocyte population in the respiratory tract demonstrated with choline acetyltransferase (ChAT)-green fluorescent protein (GFP) reporter mice, both before and after infection with influenza A virus. Mice lacking ChAT in B cells, disabling their ability to generate ACh (ChatBKO), but not those lacking ChAT in T cells, significantly, selectively and directly suppressed α7-nicotinic-ACh receptor-expressing interstitial, but not alveolar, macrophage activation and their ability to secrete tumor necrosis factor (TNF), while better controlling virus replication at 1 d postinfection. Conversely, TNF blockade via monoclonal antibody treatment increased viral loads at that time. By day 10 of infection, ChatBKO mice showed increased local and systemic inflammation and reduced signs of lung epithelial repair despite similar viral loads and viral clearance. Thus, B cells are key participants of an immediate early regulatory cascade that controls lung tissue damage after viral infection, shifting the balance toward reduced inflammation at the cost of enhanced early viral replication.
Collapse
Affiliation(s)
- Antonio Cembellin-Prieto
- Graduate Group in Immunology, University of California Davis, Davis, CA, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Lyme and Tickborne Diseases Research and Education Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Zheng Luo
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Heather Kulaga
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Lyme and Tickborne Diseases Research and Education Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Nicole Baumgarth
- Graduate Group in Immunology, University of California Davis, Davis, CA, USA.
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Lyme and Tickborne Diseases Research and Education Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA.
- Department of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
3
|
Zhai J, Li Y, Liu J, Dai C. Neuroimmune interactions: The bridge between inflammatory bowel disease and the gut microbiota. Clin Transl Med 2025; 15:e70329. [PMID: 40400119 PMCID: PMC12095209 DOI: 10.1002/ctm2.70329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The multidimensional regulatory mechanism of the gut-brain-immune axis in the context of inflammatory bowel disease (IBD) has garnered significant attention, particularly regarding how intestinal microbiota finely regulates immune responses through immune cells and sensory neurons. MAIN BODY Metabolites produced by intestinal microbiota influence the phenotype switching of immune cells via complex signalling pathways, thereby modulating their anti-inflammatory and pro-inflammatory functions during intestinal inflammation. Furthermore, sensory neurons exhibit heightened sensitivity to microbial-derived signals, which is essential for preserving intestinal balance and controlling pathological inflammation by integrating peripheral environmental signals with local immune responses. The dynamic equilibrium between immune cells and the neuroimmunoregulation mediated by sensory neurons collectively sustains immune homeostasis within the intestine. However, this coordination mechanism is markedly disrupted under the pathological conditions associated with IBD. CONCLUSION An in-depth exploration of the interactions among immune cells, gut microbiota and sensory neurons may yield significant insights into the pathological mechanisms underlying IBD and guide the creation of new treatment approaches. KEY POINTS The gut microbiota regulates the gut-brain-immune axis, modulating neuroimmune interactions in IBD. Microbiota-derived metabolites influence immune cells, thereby affecting neurons. Neurons secrete mediators, enabling bidirectional neuroimmune communication essential for intestinal homeostasis. Disruptions contribute to IBD, offering therapeutic targets.
Collapse
Affiliation(s)
- Jinxia Zhai
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| | - Yingjie Li
- Department of GastroenterologyFirst Affiliated Hospital, Jinzhou Medical UniversityJinzhou CityLiaoning ProvinceChina
| | - Jiameng Liu
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| | - Cong Dai
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| |
Collapse
|
4
|
Larsson JW, Olofsson PS, Sundman E. The innervated gut and critical illness. Curr Opin Crit Care 2025; 31:198-203. [PMID: 40047233 DOI: 10.1097/mcc.0000000000001260] [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: 05/13/2025]
Abstract
PURPOSE OF REVIEW This review highlights brain-gut neuroimmune interactions in the context of critical illness. Neural regulation of inflammation, gut innervation, and the brain-gut axis in critical illness are discussed. RECENT FINDINGS Recent studies indicate that the brain-gut axis and the enteric nervous system are integral to the regulation of local and systemic inflammation. Experimental evidence suggests that neural reflexes control immune responses, and specific neural signals promote gastrointestinal homeostasis. The understanding of these interactions in the clinical context remains limited, necessitating further investigation. Notably, therapeutic interventions targeting neuro-immune pathways have shown promise in preclinical models, suggesting that a better understanding of the neuro-immune crosstalk in the critically ill may potentially identify novel therapeutic targets. SUMMARY Critical illness involves complex organ dysfunction, not least in the gastrointestinal system. A multitude of neuroimmune interactions between the intestinal wall, immune cells, peripheral nerves and the central nervous system regulate inflammation. While experimental evidence supports the role of neural reflexes in controlling immune responses, clinical validation is lacking in the context of critical care. Future research needs to explore whether specific neural signals or mechanisms of neuro-immune crosstalk can be harnessed to restore and support gastrointestinal homeostasis in the critically ill.
Collapse
Affiliation(s)
- Jacob W Larsson
- Function Perioperative Medicine and Intensive Care, Karolinska University Hospital
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Peder S Olofsson
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- The Feinstein Institutes for Medical Research, Manhasset, NY, New York, USA
| | - Eva Sundman
- Function Perioperative Medicine and Intensive Care, Karolinska University Hospital
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Baldwin A, States G, Pikov V, Gunalan P, Elyahoodayan S, Kilgore K, Meng E. Recent advances in facilitating the translation of bioelectronic medicine therapies. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2025; 33:100575. [PMID: 39896232 PMCID: PMC11781353 DOI: 10.1016/j.cobme.2024.100575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Bioelectronic medicine is a growing field which involves directly interfacing with the vagus, sacral, enteric, and other autonomic nerves to treat conditions. Therapies based on bioelectronic medicine could address previously intractable diseases and provide an alternative to pharmaceuticals. However, translating a bioelectronic medicine therapy to the clinic requires overcoming several challenges, including titrating stimulation parameters to an individual's physiology, selectively stimulating target nerves without inducing off-target activation or block, and improving accessibility to clinically approved devices. This review describes recent progress towards solving these problems, including advances in mapping and characterizing the human autonomic nervous system, new sensor technology and signal processing techniques to enable closed-loop therapies, new methods for selectively stimulating autonomic nerves without inducing off-target effects, and efforts to develop open-source implantable devices. Recent commercial successes in bringing bioelectronic medicine therapies to the clinic are highlighted showing how addressing these challenges can lead to novel therapies.
Collapse
Affiliation(s)
- Alex Baldwin
- Alfred E. Mann Department of Biomedical Engineering,
University of Southern California, USA
| | - Gregory States
- Department of Physical Medicine & Rehabilitation, Case
Western Reserve University and The MetroHealth System, Cleveland, OH, USA
| | | | - Pallavi Gunalan
- Alfred E. Mann Department of Biomedical Engineering,
University of Southern California, USA
| | - Sahar Elyahoodayan
- Alfred E. Mann Department of Biomedical Engineering,
University of Southern California, USA
| | - Kevin Kilgore
- Department of Physical Medicine & Rehabilitation, Case
Western Reserve University and The MetroHealth System, Cleveland, OH, USA
| | - Ellis Meng
- Alfred E. Mann Department of Biomedical Engineering,
University of Southern California, USA
| |
Collapse
|
6
|
Wang Q, Yang M, Sun R, Liu W, Li W, Xu B, Yang S, Chen K, Xiao J, Chen X, Meng X, Feng J, Yu C, Luo Z. A biodegradable capacitive-coupling neurostimulator for wireless electroceutical treatment of inflammatory bowel diseases. SCIENCE ADVANCES 2025; 11:eadu5887. [PMID: 39951521 PMCID: PMC11827631 DOI: 10.1126/sciadv.adu5887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025]
Abstract
Electroceuticals based on peripheral nerve stimulation offer promising treatment for refractory inflammatory diseases such as inflammatory bowel diseases (IBDs). For pediatric IBD (PIBD) patients, wireless, biodegradable miniaturized bioelectronic devices are crucial to prevent neural damage and support neural development during and after therapy. Here we demonstrate a battery-free, miniaturized neurostimulator based on biodegradable materials and capacitive-coupling wireless power transfer. The biodegradable capacitive-coupling (BCC) neurostimulator consists of molybdenum (Mo) electronic components and self-healing biodegradable polyurethane elastomer (SBPUE) encapsulations. The self-healing property of SBPUE enables a stable neural interface. Capacitive coupling wirelessly transfers high-frequency electric fields through a single capacitor between wearable transmitters and implanted BCC neurostimulators. Programmed electrical stimulation of the vagus nerve alleviates PIBD symptoms by restoring CD4+ T cell balance, enhancing anti-inflammatory effects and suppressing pro-inflammatory effects in the intestines.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ming Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Materials Research Laboratory, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Renyuan Sun
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Wenliang Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Wenlong Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Baochun Xu
- Materials Research Laboratory, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Shiming Yang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ke Chen
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jun Xiao
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xuyong Chen
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xinyao Meng
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jiexiong Feng
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's Disease and Allied Disorders, Wuhan, Hubei 430030, China
| | - Cunjiang Yu
- Materials Research Laboratory, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, Department of Materials Science and Engineering, Department of Mechanical Science and Engineering, and Department of Bioengineering, Beckman Institute for Advanced Science and Technology, Nick Holonyak Micro and Nanotechnology Laboratory, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Zhiqiang Luo
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
7
|
Sic A, Bogicevic M, Brezic N, Nemr C, Knezevic NN. Chronic Stress and Headaches: The Role of the HPA Axis and Autonomic Nervous System. Biomedicines 2025; 13:463. [PMID: 40002876 PMCID: PMC11852498 DOI: 10.3390/biomedicines13020463] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/27/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Chronic stress significantly influences the pathogenesis of headache disorders, affecting millions worldwide. This review explores the intricate relationship between stress and headaches, focusing on the dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and autonomic nervous system (ANS). Persistent stress could lead to neuroinflammation, increased pain sensitivity, and vascular changes that could contribute to headache development and progression. The bidirectional nature of this relationship creates a vicious cycle, with recurrent headaches becoming a source of additional stress. Dysregulation of the HPA axis and ANS imbalance could amplify susceptibility to headaches, intensifying their frequency and severity. While pharmacological interventions remain common, non-pharmacological approaches targeting stress reduction, such as cognitive-behavioral therapy, biofeedback, and relaxation techniques, offer promising avenues for comprehensive headache management. By addressing the underlying stress-related mechanisms, these approaches provide a sustainable strategy to reduce headache frequency and improve patients' quality of life.
Collapse
Affiliation(s)
- Aleksandar Sic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (A.S.); (M.B.); (N.B.); (C.N.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Marko Bogicevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (A.S.); (M.B.); (N.B.); (C.N.)
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Nebojsa Brezic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (A.S.); (M.B.); (N.B.); (C.N.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Clara Nemr
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (A.S.); (M.B.); (N.B.); (C.N.)
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (A.S.); (M.B.); (N.B.); (C.N.)
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
- Department of Surgery, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
8
|
Wakefield C, Courchesne M, Nygard K, Frasch MG. Microglial and Macrophage Plasticity and Regional Cerebral Blood Flow in the Prenatal Brain and Gut Under Vagus Nerve Stimulation. Methods Mol Biol 2025; 2868:285-301. [PMID: 39546236 DOI: 10.1007/978-1-0716-4200-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
An intricate relationship exists between the vagus nerve and systemic immune cell regulation, specifically during fetal development. Little is known about the connection between the vagus nerve and the brain's regional circulatory control. In this chapter, we present a methodology for studying the impact of vagus nerve signaling on these connections in the developing fetus using the sheep model for human fetal physiology. First, we present the protocol to study the connection between the vagus nerve physiology and the regional cerebral blood flow (rCBF). Next, we detail the protocol for measuring how vagal signaling alters microglial cell plasticity in gut and brain. In previous work, our team showed that vagotomy results in amplified redistribution of rCBF toward subcortical structures in the fetal brain. Conversely, efferent VNS reduces rCBF to cortical structures while afferent VNS diminishes the rise of rCBF to subcortical structures (independent of cortical rCBF) when compared to controls in the fetal brain. Additionally, our team showed that Iba-1 expression, a marker for microglial cellular signaling activation, rises in a dose-dependent relationship with systemic inflammatory activation in the setting of vagotomy. The findings support existing preclinical and clinical evidence in adult human physiology that vagotomy is neuroprotective for neurodegenerative diseases such as Parkinson's likely via a glial cell-mediated mechanism. Vagus nerve stimulation (VNS) has also been shown to alter rCBF patterns in adults with treatment-resistant depression, underscoring the importance of further investigation of the relationship between the vagus nerve and rCBF as early as in utero. Together, the body of evidence emphasizes that the vagal pathway is an important player in the programming of microglial cell phenotypes within the developing brain. Further study is needed to better understand the significance of these relationships for the development and treatment of early susceptibility to neuroinflammatory and neurodegenerative disorders in later life. Therefore, we present a methodology for assessing rCBF and morphometric features of microglial and macrophage cell activation to allow future teams to expand on the existing body of work and further examine these relationships at a cellular and systems' levels.
Collapse
Affiliation(s)
| | - Marc Courchesne
- Biotron Imaging Facility, Western University, London, ON, Canada
| | - Karen Nygard
- Biotron Imaging Facility, Western University, London, ON, Canada
| | - Martin G Frasch
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington, Seattle, WA, USA.
| |
Collapse
|
9
|
Cui X, Gao X, Zhu B. CGRP: Does A Novel Neuroimmune Modulator Facilitate Tissue Repair? Neurosci Bull 2024; 40:2019-2022. [PMID: 39096347 PMCID: PMC11625025 DOI: 10.1007/s12264-024-01275-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/09/2024] [Indexed: 08/05/2024] Open
Affiliation(s)
- Xiang Cui
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Xinyan Gao
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Bing Zhu
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| |
Collapse
|
10
|
Mravec B, Szantova M. The role of the nervous system in liver diseases. Hepatol Res 2024; 54:970-980. [PMID: 39392763 DOI: 10.1111/hepr.14125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/26/2024] [Accepted: 09/28/2024] [Indexed: 10/13/2024]
Abstract
The nervous system significantly participates in maintaining homeostasis, and modulating repair and regeneration processes in the liver. Moreover, the nervous system also plays an important role in the processes associated with the development and progression of liver disease, and can either potentiate or inhibit these processes. The aim of this review is to describe the mechanisms and pathways through which the nervous system influences the development and progression of liver diseases, such as alcohol-associated liver disease, nonalcoholic fatty liver disease, cholestatic liver disease, hepatitis, cirrhosis, and hepatocellular carcinoma. Possible therapeutic implications based on modulation of signals transduction between the nervous system and the liver are also discussed.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Maria Szantova
- 3rd Department of Internal Medicine, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| |
Collapse
|
11
|
Oldroyd P, Hadwe SE, Barone DG, Malliaras GG. Thin-film implants for bioelectronic medicine. MRS BULLETIN 2024; 49:1045-1058. [PMID: 39397879 PMCID: PMC11469980 DOI: 10.1557/s43577-024-00786-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/01/2024] [Indexed: 10/15/2024]
Abstract
This article is based on the MRS Mid-Career Researcher Award "for outstanding contributions to the fundamentals and development of organic electronic materials and their application in biology and medicine" presentation given by George G. Malliaras, University of Cambridge, at the 2023 MRS Spring Meeting in San Francisco, Calif.Bioelectronic medicine offers a revolutionary approach to treating disease by stimulating the body with electricity. While current devices show safety and efficacy, limitations, including bulkiness, invasiveness, and scalability, hinder their wider application. Thin-film implants promise to overcome these limitations. Made using microfabrication technologies, these implants conform better to neural tissues, reduce tissue damage and foreign body response, and provide high-density, multimodal interfaces with the body. This article explores how thin-film implants using organic materials and novel designs may contribute to disease management, intraoperative monitoring, and brain mapping applications. Additionally, the technical challenges to be addressed for this technology to succeed are discussed. Graphical abstract
Collapse
Affiliation(s)
- Poppy Oldroyd
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
| | - Salim El Hadwe
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Damiano G. Barone
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - George G. Malliaras
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
| |
Collapse
|
12
|
Bolonduro OA, Chen Z, Fucetola CP, Lai YR, Cote M, Kajola RO, Rao AA, Liu H, Tzanakakis ES, Timko BP. An Integrated Optogenetic and Bioelectronic Platform for Regulating Cardiomyocyte Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402236. [PMID: 39054679 PMCID: PMC11423186 DOI: 10.1002/advs.202402236] [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: 03/01/2024] [Revised: 06/24/2024] [Indexed: 07/27/2024]
Abstract
Bioelectronic medicine is emerging as a powerful approach for restoring lost endogenous functions and addressing life-altering maladies such as cardiac disorders. Systems that incorporate both modulation of cellular function and recording capabilities can enhance the utility of these approaches and their customization to the needs of each patient. Here we report an integrated optogenetic and bioelectronic platform for stable and long-term stimulation and monitoring of cardiomyocyte function in vitro. Optical inputs are achieved through the expression of a photoactivatable adenylyl cyclase, that when irradiated with blue light causes a dose-dependent and time-limited increase in the secondary messenger cyclic adenosine monophosphate with subsequent rise in autonomous cardiomyocyte beating rate. Bioelectronic readouts are obtained through a multi-electrode array that measures real-time electrophysiological responses at 32 spatially-distinct locations. Irradiation at 27 µW mm-2 results in a 14% elevation of the beating rate within 20-25 min, which remains stable for at least 2 h. The beating rate can be cycled through "on" and "off" light states, and its magnitude is a monotonic function of irradiation intensity. The integrated platform can be extended to stretchable and flexible substrates, and can open new avenues in bioelectronic medicine, including closed-loop systems for cardiac regulation and intervention, for example, in the context of arrythmias.
Collapse
Affiliation(s)
| | - Zijing Chen
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, 02155, USA
| | - Corey P Fucetola
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Yan-Ru Lai
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Megan Cote
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Rofiat O Kajola
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Akshita A Rao
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Haitao Liu
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, China
| | - Emmanuel S Tzanakakis
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, 02155, USA
- Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Brian P Timko
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| |
Collapse
|
13
|
Micera S, Menciassi A, Cianferotti L, Gruppioni E, Lionetti V. Organ Neuroprosthetics: Connecting Transplanted and Artificial Organs with the Nervous System. Adv Healthc Mater 2024; 13:e2302896. [PMID: 38656615 DOI: 10.1002/adhm.202302896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/01/2024] [Indexed: 04/26/2024]
Abstract
Implantable neural interfaces with the central and peripheral nervous systems are currently used to restore sensory, motor, and cognitive functions in disabled people with very promising results. They have also been used to modulate autonomic activities to treat diseases such as diabetes or hypertension. Here, this study proposes to extend the use of these technologies to (re-)establish the connection between new (transplanted or artificial) organs and the nervous system in order to increase the long-term efficacy and the effective biointegration of these solutions. In this perspective paper, some clinically relevant applications of this approach are briefly described. Then, the choices that neural engineers must implement about the type, implantation location, and closed-loop control algorithms to successfully realize this approach are highlighted. It is believed that these new "organ neuroprostheses" are going to become more and more valuable and very effective solutions in the years to come.
Collapse
Affiliation(s)
- Silvestro Micera
- The BioRobotics Institute and Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Pisa, 56127, Italy
- Interdisciplinary Research Center Health Science, Scuola Superiore Sant'Anna, Pisa, 56127, Italy
- Bertarelli Foundation Chair in Translational Neuroengineering, Neuro-X Institute, School of Engineering, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, 1015, Switzerland
| | - Arianna Menciassi
- The BioRobotics Institute and Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Pisa, 56127, Italy
- Interdisciplinary Research Center Health Science, Scuola Superiore Sant'Anna, Pisa, 56127, Italy
| | - Luisella Cianferotti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, 50121, Italy
| | | | - Vincenzo Lionetti
- Interdisciplinary Research Center Health Science, Scuola Superiore Sant'Anna, Pisa, 56127, Italy
- UOSVD Anesthesia and Resuscitation, Fondazione Toscana G. Monasterio, Pisa, 56127, Italy
| |
Collapse
|
14
|
Giannotti A, Santanché R, Zinno C, Carpaneto J, Micera S, Riva ER. Characterization of a conductive hydrogel@Carbon fibers electrode as a novel intraneural interface. Bioelectron Med 2024; 10:20. [PMID: 39187894 PMCID: PMC11348655 DOI: 10.1186/s42234-024-00154-5] [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: 06/05/2024] [Accepted: 08/02/2024] [Indexed: 08/28/2024] Open
Abstract
Peripheral neural interfaces facilitate bidirectional communication between the nervous system and external devices, enabling precise control for prosthetic limbs, sensory feedback systems, and therapeutic interventions in the field of Bioelectronic Medicine. Intraneural interfaces hold great promise since they ensure high selectivity in communicating only with the desired nerve fascicles. Despite significant advancements, challenges such as chronic immune response, signal degradation over time, and lack of long-term biocompatibility remain critical considerations in the development of such devices. Here we report on the development and benchtop characterization of a novel design of an intraneural interface based on carbon fiber bundles. Carbon fibers possess low impedance, enabling enhanced signal detection and stimulation efficacy compared to traditional metal electrodes. We provided a 3D-stabilizing structure for the carbon fiber bundles made of PEDOT:PSS hydrogel, to enhance the biocompatibility between the carbon fibers and the nervous tissue. We further coated the overall bundles with a thin layer of elastomeric material to provide electrical insulation. Taken together, our results demonstrated that our electrode possesses adequate structural and electrochemical properties to ensure proper stimulation and recording of peripheral nerve fibers and a biocompatible interface with the nervous tissue.
Collapse
Affiliation(s)
- Alice Giannotti
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Department of Excellence in Robotics&AI, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Ranieri Santanché
- Dipartimento Di Ingegneria Civile E Industriale (DICI), Università Di Pisa, Largo Lucio Lazzarino 1, 56122, Pisa, Italy
| | - Ciro Zinno
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Department of Excellence in Robotics&AI, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Jacopo Carpaneto
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Department of Excellence in Robotics&AI, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Silvestro Micera
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Centre for Neuroprosthetics and Institute of Bioengineering, School of Engineering, Bertarelli Foundation Chair in Translational Neuroengineering, ÉcolePolytechniqueFédérale de Lausanne (EPFL), 1007, Lausanne, Switzerland
| | - Eugenio Redolfi Riva
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy.
- Department of Excellence in Robotics&AI, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy.
| |
Collapse
|
15
|
Zhao Q. Navigating internal senses: A road map for the vagal interoceptive system. Science 2024; 385:507-508. [PMID: 39088631 DOI: 10.1126/science.adq8578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
A road map for the vagal interoceptive system.
Collapse
Affiliation(s)
- Qiancheng Zhao
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
16
|
Rodríguez‐Meana B, del Valle J, Viana D, Walston ST, Ria N, Masvidal‐Codina E, Garrido JA, Navarro X. Engineered Graphene Material Improves the Performance of Intraneural Peripheral Nerve Electrodes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308689. [PMID: 38863325 PMCID: PMC11304253 DOI: 10.1002/advs.202308689] [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: 11/14/2023] [Revised: 04/09/2024] [Indexed: 06/13/2024]
Abstract
Limb neuroprostheses aim to restore motor and sensory functions in amputated or severely nerve-injured patients. These devices use neural interfaces to record and stimulate nerve action potentials, creating a bidirectional connection with the nervous system. Most neural interfaces are based on standard metal microelectrodes. In this work, a new generation of neural interfaces which replaces metals with engineered graphene, called EGNITE, is tested. In vitro and in vivo experiments are conducted to assess EGNITE biocompatibility. In vitro tests show that EGNITE does not impact cell viability. In vivo, no significant functional decrease or harmful effects are observed. Furthermore, the foreign body reaction to the intraneural implant is similar compared to other materials previously used in neural interfaces. Regarding functionality, EGNITE devices are able to stimulate nerve fascicles, during two months of implant, producing selective muscle activation with about three times less current compared to larger microelectrodes of standard materials. CNAP elicited by electrical stimuli and ENG evoked by mechanical stimuli are recorded with high resolution but are more affected by decreased functionality over time. This work constitutes further proof that graphene-derived materials, and specifically EGNITE, is a promising conductive material of neural electrodes for advanced neuroprostheses.
Collapse
Affiliation(s)
- Bruno Rodríguez‐Meana
- Institute of NeurosciencesDepartment of Cell BiologyPhysiology and ImmunologyUniversitat Autònoma de BarcelonaBellaterra08193Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadrid28031Spain
| | - Jaume del Valle
- Institute of NeurosciencesDepartment of Cell BiologyPhysiology and ImmunologyUniversitat Autònoma de BarcelonaBellaterra08193Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadrid28031Spain
- Department de Bioquímica i FisiologiaUniversitat de BarcelonaBarcelona08028Spain
| | - Damià Viana
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)CSIC and BISTCampus UABBellaterra08193Spain
| | - Steven T. Walston
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)CSIC and BISTCampus UABBellaterra08193Spain
| | - Nicola Ria
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)CSIC and BISTCampus UABBellaterra08193Spain
| | - Eduard Masvidal‐Codina
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)CSIC and BISTCampus UABBellaterra08193Spain
| | - Jose A. Garrido
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)CSIC and BISTCampus UABBellaterra08193Spain
- ICREABarcelona08010Spain
| | - Xavier Navarro
- Institute of NeurosciencesDepartment of Cell BiologyPhysiology and ImmunologyUniversitat Autònoma de BarcelonaBellaterra08193Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadrid28031Spain
- Institut Guttmann of NeurorehabilitationBadalona08916Spain
| |
Collapse
|
17
|
Zou J, Li J, Wang X, Tang D, Chen R. Neuroimmune modulation in liver pathophysiology. J Neuroinflammation 2024; 21:188. [PMID: 39090741 PMCID: PMC11295927 DOI: 10.1186/s12974-024-03181-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
The liver, the largest organ in the human body, plays a multifaceted role in digestion, coagulation, synthesis, metabolism, detoxification, and immune defense. Changes in liver function often coincide with disruptions in both the central and peripheral nervous systems. The intricate interplay between the nervous and immune systems is vital for maintaining tissue balance and combating diseases. Signaling molecules and pathways, including cytokines, inflammatory mediators, neuropeptides, neurotransmitters, chemoreceptors, and neural pathways, facilitate this complex communication. They establish feedback loops among diverse immune cell populations and the central, peripheral, sympathetic, parasympathetic, and enteric nervous systems within the liver. In this concise review, we provide an overview of the structural and compositional aspects of the hepatic neural and immune systems. We further explore the molecular mechanisms and pathways that govern neuroimmune communication, highlighting their significance in liver pathology. Finally, we summarize the current clinical implications of therapeutic approaches targeting neuroimmune interactions and present prospects for future research in this area.
Collapse
Affiliation(s)
- Ju Zou
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jie Li
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoxu Wang
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ruochan Chen
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
18
|
Falvey A, Palandira SP, Chavan SS, Brines M, Dantzer R, Tracey KJ, Pavlov VA. Electrical stimulation of the dorsal motor nucleus of the vagus in male mice can regulate inflammation without affecting the heart rate. Brain Behav Immun 2024; 120:630-639. [PMID: 38670240 PMCID: PMC11957331 DOI: 10.1016/j.bbi.2024.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/01/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The vagus nerve plays an important role in neuroimmune interactions and in the regulation of inflammation. A major source of efferent vagus nerve fibers that contribute to the regulation of inflammation is the brainstem dorsal motor nucleus of the vagus (DMN), as recently shown using optogenetics. In contrast to optogenetics, electrical neuromodulation has broad therapeutic implications. However, the anti-inflammatory effectiveness of electrical stimulation of the DMN (eDMNS) and the possible heart rate (HR) alterations associated with this approach have not been investigated. Here, we examined the effects of eDMNS on HR and cytokine levels in mice administered with lipopolysaccharide (LPS, endotoxin) and in mice subjected to cecal ligation and puncture (CLP) sepsis. METHODS Anesthetized male 8-10-week-old C57BL/6 mice on a stereotaxic frame were subjected to eDMNS using a concentric bipolar electrode inserted into the left or right DMN or sham stimulation. eDMNS (500, 250 or 50 μA at 30 Hz, for 1 min) was performed and HR recorded. In endotoxemia experiments, sham or eDMNS utilizing 250 μA or 50 μA was performed for 5 mins and was followed by LPS (0.5 mg/kg) i.p. administration. eDMNS was also applied in mice with cervical unilateral vagotomy or sham operation. In CLP experiments sham or left eDMNS was performed immediately post CLP. Cytokines and corticosterone were analyzed 90 mins after LPS administration or 24 h after CLP. CLP survival was monitored for 14 days. RESULTS Either left or right eDMNS at 500 μA and 250 μA decreased HR, compared with baseline pre-stimulation. This effect was not observed at 50 μA. Left side eDMNS at 50 μA, compared with sham stimulation, significantly decreased serum and splenic levels of the pro-inflammatory cytokine TNF and increased serum levels of the anti-inflammatory cytokine IL-10 during endotoxemia. The anti-inflammatory effect of eDMNS was abrogated in mice with unilateral vagotomy and was not associated with serum corticosterone alterations. Right side eDMNS in endotoxemic mice suppressed serum TNF and increased serum IL-10 levels but had no effects on splenic cytokines. In mice with CLP, left side eDMNS suppressed serum IL-6, as well as splenic IL-6 and increased splenic IL-10 and significantly improved the survival rate of CLP mice. CONCLUSIONS For the first time we show that a regimen of eDMNS which does not cause bradycardia alleviates LPS-induced inflammation. These eDMNS anti-inflammatory effects require an intact vagus nerve and are not associated with corticosteroid alterations. eDMNS also decreases inflammation and improves survival in a model of polymicrobial sepsis. These findings are of interest for further studies exploring bioelectronic anti-inflammatory approaches targeting the brainstem DMN.
Collapse
Affiliation(s)
- Aidan Falvey
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Santhoshi P Palandira
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Sangeeta S Chavan
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, NY 11549, USA; Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Michael Brines
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Robert Dantzer
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - Kevin J Tracey
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, NY 11549, USA; Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Valentin A Pavlov
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, NY 11549, USA; Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA.
| |
Collapse
|
19
|
Zhang Z, Zhang D, Lin Q, Cui X. Therapeutically Fine-Tuning Autonomic Nervous System to Treat Sepsis: A New Perspective on the Immunomodulatory Effects of Acupuncture. J Inflamm Res 2024; 17:4373-4387. [PMID: 38988505 PMCID: PMC11233988 DOI: 10.2147/jir.s477181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
Recent studies have highlighted the immunomodulatory effects of acupuncture on sepsis and proposed novel non-pharmacological or bioelectronic approaches to managing inflammatory illnesses. Establishing rules for selectively activating sympathetic or vagal nerve-mediated anti-inflammatory pathways using acupuncture has valuable clinical applications. Over the years, studies have revealed the segmental modulatory role of acupuncture in regulating visceral function by targeting the autonomic nervous system (ANS). In this review, we aim to summarize recent findings on acupuncture in treating sepsis, focusing on the underlying ANS mechanism, as well as the rules of acupoint specificity, intensity, frequency, and other parameters utilized in these studies. Mechanistically, the immunomodulatory properties of the sympathetic nervous system have been highlighted. Furthermore, we explore the immunotherapeutic benefits of acupuncture in treating sepsis. A better understanding of the immunoregulatory mechanism of sympathetic nervous system may offer novel approaches for the development of therapeutics to treat or prevent a variety of inflammatory diseases.
Collapse
Affiliation(s)
- Ziyi Zhang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People’s Republic of China
| | - Dingdan Zhang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People’s Republic of China
| | - Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, 21287, USA
| | - Xiang Cui
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People’s Republic of China
| |
Collapse
|
20
|
Margolis KG, Shea-Donohue T, Cummings DM, Greenwel P, Lunsford RD, Gulbransen BD, Chiu IM. 2023 Workshop: Neuroimmune Crosstalk in the Gut - Impact on Local, Autonomic and Gut-Brain Function. Gastroenterology 2024; 167:223-230. [PMID: 38518873 DOI: 10.1053/j.gastro.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/12/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024]
Affiliation(s)
- Kara G Margolis
- New York University Pain Research Center and Department of Molecular Pathobiology, New York University, College of Dentistry, New York, New York; Departments of Pediatrics and Cell Biology, Grossman School of Medicine, New York, New York.
| | - Terez Shea-Donohue
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Diana M Cummings
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Patricia Greenwel
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Robert D Lunsford
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
21
|
Baumgarth N, Prieto AC, Luo Z, Kulaga H. B cells modulate lung antiviral inflammatory responses via the neurotransmitter acetylcholine. RESEARCH SQUARE 2024:rs.3.rs-4421566. [PMID: 38978583 PMCID: PMC11230464 DOI: 10.21203/rs.3.rs-4421566/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The rapid onset of innate immune defenses is critical for early control of viral replication in an infected host, yet it can also lead to irreversible tissue damage, especially in the respiratory tract. Intricate regulatory mechanisms must exist that modulate inflammation, while controlling the infection. Here, B cells expressing choline acetyl transferase (ChAT), an enzyme required for production of the metabolite and neurotransmitter acetylcholine (ACh) are identified as such regulators of the immediate early response to influenza A virus. Lung tissue ChAT + B cells are shown to interact with a7 nicotinic Ach receptor-expressing lung interstitial macrophages in mice within 24h of infection to control their production of TNFa, shifting the balance towards reduced inflammation at the cost of enhanced viral replication. Thus, innate-stimulated B cells are key participants of an immediate-early regulatory cascade that controls lung tissue damage after viral infection.
Collapse
|
22
|
Sahasrabudhe A, Rupprecht LE, Orguc S, Khudiyev T, Tanaka T, Sands J, Zhu W, Tabet A, Manthey M, Allen H, Loke G, Antonini MJ, Rosenfeld D, Park J, Garwood IC, Yan W, Niroui F, Fink Y, Chandrakasan A, Bohórquez DV, Anikeeva P. Multifunctional microelectronic fibers enable wireless modulation of gut and brain neural circuits. Nat Biotechnol 2024; 42:892-904. [PMID: 37349522 PMCID: PMC11180606 DOI: 10.1038/s41587-023-01833-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/23/2023] [Indexed: 06/24/2023]
Abstract
Progress in understanding brain-viscera interoceptive signaling is hindered by a dearth of implantable devices suitable for probing both brain and peripheral organ neurophysiology during behavior. Here we describe multifunctional neural interfaces that combine the scalability and mechanical versatility of thermally drawn polymer-based fibers with the sophistication of microelectronic chips for organs as diverse as the brain and the gut. Our approach uses meters-long continuous fibers that can integrate light sources, electrodes, thermal sensors and microfluidic channels in a miniature footprint. Paired with custom-fabricated control modules, the fibers wirelessly deliver light for optogenetics and transfer data for physiological recording. We validate this technology by modulating the mesolimbic reward pathway in the mouse brain. We then apply the fibers in the anatomically challenging intestinal lumen and demonstrate wireless control of sensory epithelial cells that guide feeding behaviors. Finally, we show that optogenetic stimulation of vagal afferents from the intestinal lumen is sufficient to evoke a reward phenotype in untethered mice.
Collapse
Affiliation(s)
- Atharva Sahasrabudhe
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura E Rupprecht
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Sirma Orguc
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tural Khudiyev
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tomo Tanaka
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Secure System Platform Research Laboratories, NEC Corporation, Kawasaki, Japan
| | - Joanna Sands
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Weikun Zhu
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anthony Tabet
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie Manthey
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Harrison Allen
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gabriel Loke
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marc-Joseph Antonini
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT Health Sciences and Technology Graduate Program, Cambridge, MA, USA
| | - Dekel Rosenfeld
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jimin Park
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Indie C Garwood
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT Health Sciences and Technology Graduate Program, Cambridge, MA, USA
| | - Wei Yan
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Farnaz Niroui
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yoel Fink
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anantha Chandrakasan
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Diego V Bohórquez
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
23
|
Olofsson PS. Living bioelectronics resolve inflammation. Science 2024; 384:962-963. [PMID: 38815044 DOI: 10.1126/science.adp5201] [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: 06/01/2024]
Abstract
Coupling skin bacteria and electronics opens paths to adaptive treatment of inflammation.
Collapse
Affiliation(s)
- Peder S Olofsson
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Metz CN, Brines M, Pavlov VA. Bridging cholinergic signalling and inflammation in schizophrenia. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:51. [PMID: 38734678 PMCID: PMC11088617 DOI: 10.1038/s41537-024-00472-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Affiliation(s)
- Christine N Metz
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11550, USA
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Michael Brines
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Valentin A Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA.
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11550, USA.
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
25
|
Adkisson PW, Steinhardt CR, Fridman GY. Galvanic vs. pulsatile effects on decision-making networks: reshaping the neural activation landscape. J Neural Eng 2024; 21:10.1088/1741-2552/ad36e2. [PMID: 38518369 PMCID: PMC11877455 DOI: 10.1088/1741-2552/ad36e2] [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: 10/10/2023] [Accepted: 03/22/2024] [Indexed: 03/24/2024]
Abstract
Objective. Primarily due to safety concerns, biphasic pulsatile stimulation (PS) is the present standard for electrical excitation of neural tissue with a diverse set of applications. While pulses have been shown to be effective to achieve functional outcomes, they have well-known deficits. Due to recent technical advances, galvanic stimulation (GS), delivery of current for extended periods of time (>1 s), has re-emerged as an alternative to PS.Approach. In this paper, we use a winner-take-all decision-making cortical network model to investigate differences between pulsatile and GS in the context of a perceptual decision-making task.Main results. Based on previous work, we hypothesized that GS would produce more spatiotemporally distributed, network-sensitive neural responses, while PS would produce highly synchronized activation of a limited group of neurons. Our results in-silico support these hypotheses for low-amplitude GS but deviate when galvanic amplitudes are large enough to directly activate or block nearby neurons.Significance. We conclude that with careful parametrization, GS could overcome some limitations of PS to deliver more naturalistic firing patterns in the group of targeted neurons.
Collapse
Affiliation(s)
- Paul W Adkisson
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, United States of America
| | - Cynthia R Steinhardt
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, United States of America
- Center for Theoretical Neuroscience, Columbia University, New York, NY 10027, United States of America
- Simons Society of Fellows, Simons Foundation, New York, NY 10010, United States of America
| | - Gene Y Fridman
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, United States of America
- Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University, Baltimore, MD 21205, United States of America
| |
Collapse
|
26
|
Lloyd DA, Alejandra Gonzalez-Gonzalez M, Romero-Ortega MI. AxoDetect: an automated nerve image segmentation and quantification workflow for computational nerve modeling. J Neural Eng 2024; 21:026017. [PMID: 38457836 PMCID: PMC10976901 DOI: 10.1088/1741-2552/ad31c3] [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: 10/20/2023] [Revised: 02/11/2024] [Accepted: 03/08/2024] [Indexed: 03/10/2024]
Abstract
Objective.Bioelectronic treatments targeting near-organ innervation have unprecedented clinical applications. Particularly in the spleen, the inhibition of the cholinergic inflammatory response by near-organ nerve stimulation has potential to replace pharmacological treatments in chronic and autoimmune diseases. A caveat is that the optimization of therapeutic stimulation parameters relies onin vivoexperimentation, which becomes challenging due to the small nerve diameters (2 μm), complex anatomy, and mixed axon type composition of the autonomic nerves. Effective development ofin silicomodels requires tools which allow for fast and efficient quantification of axonal composition of specific nerves. Current approaches to generate such information rely on manual image segmentation and quantification.Approach.We developed a combined image-segmentation and model-generation software called AxoDetect: a target- and format-agnostic computer vision algorithm which can segment myelin, endo/epineurium, and both myelinated and unmyelinated fibers from a nerve image without training.Main results.AxoDetect is over 10 times faster on average when compared with current automatic methods while maintaining flexibility through the use of tunable pixel threshold filters to detect different types of tissue. When compared to a distribution-based and a manually segmented model of the splenic nerve terminal branch 1, the model generated with AxoDetect had comparable threshold prediction and was able to accurately detect an increase in activation threshold caused by the addition of surrounding fat tissue to the modeled nerve.Significance.AxoDetect contributes to the acceleration of neuromodulation treatment development through faster model design and iteration without requiring training. Furthermore, the computer vision approach and tunable nature of the filters in our method allow for its use in a variety of histological applications. Our approach will impact not only the study of nerves but also the design of implantable neural interfaces to enhance bioelectronic therapeutic options.
Collapse
Affiliation(s)
- David A Lloyd
- Departments of Biomedical Engineering and Biomedical Sciences, University of Houston, Houston, TX, United States of America
| | - Maria Alejandra Gonzalez-Gonzalez
- Departments of Biomedical Engineering and Biomedical Sciences, University of Houston, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States of America
- Department of Pediatric Neurology, Baylor College of Medicine, Houston, TX, United States of America
| | - Mario I Romero-Ortega
- Departments of Biomedical Engineering and Biomedical Sciences, University of Houston, Houston, TX, United States of America
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
27
|
González-González MA, Conde SV, Latorre R, Thébault SC, Pratelli M, Spitzer NC, Verkhratsky A, Tremblay MÈ, Akcora CG, Hernández-Reynoso AG, Ecker M, Coates J, Vincent KL, Ma B. Bioelectronic Medicine: a multidisciplinary roadmap from biophysics to precision therapies. Front Integr Neurosci 2024; 18:1321872. [PMID: 38440417 PMCID: PMC10911101 DOI: 10.3389/fnint.2024.1321872] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/10/2024] [Indexed: 03/06/2024] Open
Abstract
Bioelectronic Medicine stands as an emerging field that rapidly evolves and offers distinctive clinical benefits, alongside unique challenges. It consists of the modulation of the nervous system by precise delivery of electrical current for the treatment of clinical conditions, such as post-stroke movement recovery or drug-resistant disorders. The unquestionable clinical impact of Bioelectronic Medicine is underscored by the successful translation to humans in the last decades, and the long list of preclinical studies. Given the emergency of accelerating the progress in new neuromodulation treatments (i.e., drug-resistant hypertension, autoimmune and degenerative diseases), collaboration between multiple fields is imperative. This work intends to foster multidisciplinary work and bring together different fields to provide the fundamental basis underlying Bioelectronic Medicine. In this review we will go from the biophysics of the cell membrane, which we consider the inner core of neuromodulation, to patient care. We will discuss the recently discovered mechanism of neurotransmission switching and how it will impact neuromodulation design, and we will provide an update on neuronal and glial basis in health and disease. The advances in biomedical technology have facilitated the collection of large amounts of data, thereby introducing new challenges in data analysis. We will discuss the current approaches and challenges in high throughput data analysis, encompassing big data, networks, artificial intelligence, and internet of things. Emphasis will be placed on understanding the electrochemical properties of neural interfaces, along with the integration of biocompatible and reliable materials and compliance with biomedical regulations for translational applications. Preclinical validation is foundational to the translational process, and we will discuss the critical aspects of such animal studies. Finally, we will focus on the patient point-of-care and challenges in neuromodulation as the ultimate goal of bioelectronic medicine. This review is a call to scientists from different fields to work together with a common endeavor: accelerate the decoding and modulation of the nervous system in a new era of therapeutic possibilities.
Collapse
Affiliation(s)
- María Alejandra González-González
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatric Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Silvia V. Conde
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NOVA University, Lisbon, Portugal
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Stéphanie C. Thébault
- Laboratorio de Investigación Traslacional en salud visual (D-13), Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Marta Pratelli
- Neurobiology Department, Kavli Institute for Brain and Mind, UC San Diego, La Jolla, CA, United States
| | - Nicholas C. Spitzer
- Neurobiology Department, Kavli Institute for Brain and Mind, UC San Diego, La Jolla, CA, United States
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- International Collaborative Center on Big Science Plan for Purinergic Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Cuneyt G. Akcora
- Department of Computer Science, University of Central Florida, Orlando, FL, United States
| | | | - Melanie Ecker
- Department of Biomedical Engineering, University of North Texas, Denton, TX, United States
| | | | - Kathleen L. Vincent
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, United States
| | - Brandy Ma
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
28
|
Hong W. Advances and Opportunities of Mobile Health in the Postpandemic Era: Smartphonization of Wearable Devices and Wearable Deviceization of Smartphones. JMIR Mhealth Uhealth 2024; 12:e48803. [PMID: 38252596 PMCID: PMC10823426 DOI: 10.2196/48803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 11/08/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Mobile health (mHealth) with continuous real-time monitoring is leading the era of digital medical convergence. Wearable devices and smartphones optimized as personalized health management platforms enable disease prediction, prevention, diagnosis, and even treatment. Ubiquitous and accessible medical services offered through mHealth strengthen universal health coverage to facilitate service use without discrimination. This viewpoint investigates the latest trends in mHealth technology, which are comprehensive in terms of form factors and detection targets according to body attachment location and type. Insights and breakthroughs from the perspective of mHealth sensing through a new form factor and sensor-integrated display overcome the problems of existing mHealth by proposing a solution of smartphonization of wearable devices and the wearable deviceization of smartphones. This approach maximizes the infinite potential of stagnant mHealth technology and will present a new milestone leading to the popularization of mHealth. In the postpandemic era, innovative mHealth solutions through the smartphonization of wearable devices and the wearable deviceization of smartphones could become the standard for a new paradigm in the field of digital medicine.
Collapse
Affiliation(s)
- Wonki Hong
- Department of Digital Healthcare, Daejeon University, Daejeon, Republic of Korea
| |
Collapse
|
29
|
Keever KR, Cui K, Casteel JL, Singh S, Hoover DB, Williams DL, Pavlov VA, Yakubenko VP. Cholinergic signaling via the α7 nicotinic acetylcholine receptor regulates the migration of monocyte-derived macrophages during acute inflammation. J Neuroinflammation 2024; 21:3. [PMID: 38178134 PMCID: PMC10765732 DOI: 10.1186/s12974-023-03001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND The involvement of the autonomic nervous system in the regulation of inflammation is an emerging concept with significant potential for clinical applications. Recent studies demonstrate that stimulating the vagus nerve activates the cholinergic anti-inflammatory pathway that inhibits pro-inflammatory cytokines and controls inflammation. The α7 nicotinic acetylcholine receptor (α7nAChR) on macrophages plays a key role in mediating cholinergic anti-inflammatory effects through a downstream intracellular mechanism involving inhibition of NF-κB signaling, which results in suppression of pro-inflammatory cytokine production. However, the role of the α7nAChR in the regulation of other aspects of the immune response, including the recruitment of monocytes/macrophages to the site of inflammation remained poorly understood. RESULTS We observed an increased mortality in α7nAChR-deficient mice (compared with wild-type controls) in mice with endotoxemia, which was paralleled with a significant reduction in the number of monocyte-derived macrophages in the lungs. Corroborating these results, fluorescently labeled α7nAChR-deficient monocytes adoptively transferred to WT mice showed significantly diminished recruitment to the inflamed tissue. α7nAChR deficiency did not affect monocyte 2D transmigration across an endothelial monolayer, but it significantly decreased the migration of macrophages in a 3D fibrin matrix. In vitro analysis of major adhesive receptors (L-selectin, β1 and β2 integrins) and chemokine receptors (CCR2 and CCR5) revealed reduced expression of integrin αM and αX on α7nAChR-deficient macrophages. Decreased expression of αMβ2 was confirmed on fluorescently labeled, adoptively transferred α7nAChR-deficient macrophages in the lungs of endotoxemic mice, indicating a potential mechanism for α7nAChR-mediated migration. CONCLUSIONS We demonstrate a novel role for the α7nAChR in mediating macrophage recruitment to inflamed tissue, which indicates an important new aspect of the cholinergic regulation of immune responses and inflammation.
Collapse
Affiliation(s)
- Kasey R Keever
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson, TN, USA
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson, TN, USA
| | - Kui Cui
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson, TN, USA
| | - Jared L Casteel
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson, TN, USA
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson, TN, USA
| | - Sanjay Singh
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson, TN, USA
| | - Donald B Hoover
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson, TN, USA
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson, TN, USA
| | - David L Williams
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson, TN, USA
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson, TN, USA
| | - Valentin A Pavlov
- Center for Biomedical Science and Center for Bioelectronic Medicine, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11550, USA
| | - Valentin P Yakubenko
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson, TN, USA.
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson, TN, USA.
| |
Collapse
|
30
|
Courchesne M, Wakefield C, Nygard K, Burns P, Fecteau G, Desrochers A, Cao M, Frasch MG. Vagus Nerve Manipulation and Microglial Plasticity in the Prenatal Brain. NEUROMETHODS 2024:69-94. [DOI: 10.1007/978-1-0716-3465-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
31
|
Martínez-Meza S, Singh B, Nixon DF, Dopkins N, Gangcuangco LMA. The brain-liver cholinergic anti-inflammatory pathway and viral infections. Bioelectron Med 2023; 9:29. [PMID: 38115148 PMCID: PMC10731847 DOI: 10.1186/s42234-023-00132-3] [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: 07/19/2023] [Accepted: 11/06/2023] [Indexed: 12/21/2023] Open
Abstract
Efferent cholinergic signaling is a critical and targetable source of immunoregulation. The vagus nerve (VN) is the primary source of cholinergic signaling in the body, and partially innervates hepatic functionality through the liver-brain axis. Virus-induced disruption of cholinergic signaling may promote pathogenesis in hepatotropic and neurotropic viruses. Therefore, restoring VN functionality could be a novel therapeutic strategy to alleviate pathogenic inflammation in hepatotropic and neurotropic viral infections alike. In this minireview, we discuss the physiological importance of cholinergic signaling in maintaining liver-brain axis homeostasis. Next, we explore mechanisms by which the VN is perturbed by viral infections, and how non-invasive restoration of cholinergic signaling pathways with bioelectronic medicine (BEM) might ameliorate hepatic inflammation and neuroinflammation in certain viral infections.
Collapse
Affiliation(s)
- Samuel Martínez-Meza
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Bhavya Singh
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Douglas F Nixon
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Nicholas Dopkins
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Louie Mar A Gangcuangco
- Hawaii Center for AIDS, Department of Medicine, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
32
|
Bolonduro OA, Chen Z, Lai YR, Cote M, Rao AA, Liu H, Tzanakakis ES, Timko BP. An Integrated Optogenetic and Bioelectronic Platform for Regulating Cardiomyocyte Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.15.571704. [PMID: 38168441 PMCID: PMC10760153 DOI: 10.1101/2023.12.15.571704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
We report an integrated optogenetic and bioelectronic platform for stable and long-term modulation and monitoring of cardiomyocyte function in vitro. Optogenetic inputs were achieved through expression of a photoactivatable adenylyl cyclase (bPAC), that when activated by blue light caused a dose-dependent and time-limited increase in autonomous cardiomyocyte beat rate. Bioelectronic readouts were achieved through an integrated planar multi-electrode array (MEA) that provided real-time readouts of electrophysiological activity from 32 spatially-distinct locations. Irradiation at 27 μW/mm2 resulted in a ca. 14% increase in beat rate within 20-25 minutes, which remained stable for at least 2 hours. The beating rate could be cycled through repeated "on" and "off' states, and its magnitude was a monotonic function of irradiation intensity. Our integrated platform opens new avenues in bioelectronic medicine, including closed-loop feedback systems, with potential applications for cardiac regulation including arrhythmia diagnosis and intervention.
Collapse
Affiliation(s)
| | - Zijing Chen
- Department of Chemical and Biological Engineering, Tufts University
| | - Yan-Ru Lai
- Department of Biomedical Engineering, Tufts University
| | - Megan Cote
- Department of Biomedical Engineering, Tufts University
| | | | - Haitao Liu
- Department of Biomedical Engineering, Tufts University
- General Surgery Department, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Emmanuel S. Tzanakakis
- Department of Chemical and Biological Engineering, Tufts University
- Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University
- Clinical and Translational Science Institute, Tufts Medical Center
| | | |
Collapse
|
33
|
Seicol BJ, Guo Z, Garrity K, Xie R. Potential uses of auditory nerve stimulation to modulate immune responses in the inner ear and auditory brainstem. Front Integr Neurosci 2023; 17:1294525. [PMID: 38162822 PMCID: PMC10755874 DOI: 10.3389/fnint.2023.1294525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
Bioelectronic medicine uses electrical stimulation of the nervous system to improve health outcomes throughout the body primarily by regulating immune responses. This concept, however, has yet to be applied systematically to the auditory system. There is growing interest in how cochlear damage and associated neuroinflammation may contribute to hearing loss. In conjunction with recent findings, we propose here a new perspective, which could be applied alongside advancing technologies, to use auditory nerve (AN) stimulation to modulate immune responses in hearing health disorders and following surgeries for auditory implants. In this article we will: (1) review the mechanisms of inflammation in the auditory system in relation to various forms of hearing loss, (2) explore nerve stimulation to reduce inflammation throughout the body and how similar neural-immune circuits likely exist in the auditory system (3) summarize current methods for stimulating the auditory system, particularly the AN, and (4) propose future directions to use bioelectronic medicine to ameliorate harmful immune responses in the inner ear and auditory brainstem to treat refractory conditions. We will illustrate how current knowledge from bioelectronic medicine can be applied to AN stimulation to resolve inflammation associated with implantation and disease. Further, we suggest the necessary steps to get discoveries in this emerging field from bench to bedside. Our vision is a future for AN stimulation that includes additional protocols as well as advances in devices to target and engage neural-immune circuitry for therapeutic benefits.
Collapse
Affiliation(s)
- Benjamin J. Seicol
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Zixu Guo
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Katy Garrity
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Ruili Xie
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
34
|
Yu M, Sun P, Sun C, Jin WL. Bioelectronic medicine potentiates endogenous NSCs for neurodegenerative diseases. Trends Mol Med 2023; 29:886-896. [PMID: 37735022 DOI: 10.1016/j.molmed.2023.08.005] [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: 05/31/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/23/2023]
Abstract
Neurodegenerative diseases (NDs) are commonly observed and while no therapy is universally applicable, cell-based therapies are promising. Stem cell transplantation has been investigated, but endogenous neural stem cells (eNSCs), despite their potential, especially with the development of bioelectronic medicine and biomaterials, remain understudied. Here, we compare stem cell transplantation therapy with eNSC-based therapy and summarize the combined use of eNSCs and developing technologies. The rapid development of implantable biomaterials has resulted in electronic stimulation becoming increasingly effective and decreasingly invasive. Thus, the combination of bioelectronic medicine and eNSCs has substantial potential for the treatment of NDs.
Collapse
Affiliation(s)
- Maifu Yu
- School of Life Science, Lanzhou University, Lanzhou 730000, China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Pin Sun
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Changkai Sun
- Research & Educational Center for the Control Engineering of Translational Precision Medicine (R-ECCE-TPM), School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian 116024, China.
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
35
|
Bok I, Vareberg A, Gokhale Y, Bhatt S, Masterson E, Phillips J, Zhu T, Ren X, Hai A. Wireless agents for brain recording and stimulation modalities. Bioelectron Med 2023; 9:20. [PMID: 37726851 PMCID: PMC10510192 DOI: 10.1186/s42234-023-00122-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/19/2023] [Indexed: 09/21/2023] Open
Abstract
New sensors and modulators that interact wirelessly with medical modalities unlock uncharted avenues for in situ brain recording and stimulation. Ongoing miniaturization, material refinement, and sensitization to specific neurophysiological and neurochemical processes are spurring new capabilities that begin to transcend the constraints of traditional bulky and invasive wired probes. Here we survey current state-of-the-art agents across diverse realms of operation and evaluate possibilities depending on size, delivery, specificity and spatiotemporal resolution. We begin by describing implantable and injectable micro- and nano-scale electronic devices operating at or below the radio frequency (RF) regime with simple near field transmission, and continue with more sophisticated devices, nanoparticles and biochemical molecular conjugates acting as dynamic contrast agents in magnetic resonance imaging (MRI), ultrasound (US) transduction and other functional tomographic modalities. We assess the ability of some of these technologies to deliver stimulation and neuromodulation with emerging probes and materials that provide minimally invasive magnetic, electrical, thermal and optogenetic stimulation. These methodologies are transforming the repertoire of readily available technologies paired with compatible imaging systems and hold promise toward broadening the expanse of neurological and neuroscientific diagnostics and therapeutics.
Collapse
Affiliation(s)
- Ilhan Bok
- Department of Biomedical Engineering, University of WI - Madison, 1550 Engineering Dr, Madison, WI, Rm 2112, USA
- Department of Electrical and Computer Engineering, University of WI - Madison, Madison, WI, USA
- Wisconsin Institute for Translational Neuroengineering (WITNe), Madison, WI, USA
| | - Adam Vareberg
- Department of Biomedical Engineering, University of WI - Madison, 1550 Engineering Dr, Madison, WI, Rm 2112, USA
- Wisconsin Institute for Translational Neuroengineering (WITNe), Madison, WI, USA
| | - Yash Gokhale
- Department of Biomedical Engineering, University of WI - Madison, 1550 Engineering Dr, Madison, WI, Rm 2112, USA
- Wisconsin Institute for Translational Neuroengineering (WITNe), Madison, WI, USA
| | - Suyash Bhatt
- Department of Electrical and Computer Engineering, University of WI - Madison, Madison, WI, USA
- Wisconsin Institute for Translational Neuroengineering (WITNe), Madison, WI, USA
| | - Emily Masterson
- Department of Biomedical Engineering, University of WI - Madison, 1550 Engineering Dr, Madison, WI, Rm 2112, USA
- Wisconsin Institute for Translational Neuroengineering (WITNe), Madison, WI, USA
| | - Jack Phillips
- Department of Biomedical Engineering, University of WI - Madison, 1550 Engineering Dr, Madison, WI, Rm 2112, USA
| | - Tianxiang Zhu
- Department of Electrical and Computer Engineering, University of WI - Madison, Madison, WI, USA
- Wisconsin Institute for Translational Neuroengineering (WITNe), Madison, WI, USA
| | - Xiaoxuan Ren
- Department of Biomedical Engineering, University of WI - Madison, 1550 Engineering Dr, Madison, WI, Rm 2112, USA
- Department of Electrical and Computer Engineering, University of WI - Madison, Madison, WI, USA
| | - Aviad Hai
- Department of Biomedical Engineering, University of WI - Madison, 1550 Engineering Dr, Madison, WI, Rm 2112, USA.
- Department of Electrical and Computer Engineering, University of WI - Madison, Madison, WI, USA.
- Wisconsin Institute for Translational Neuroengineering (WITNe), Madison, WI, USA.
| |
Collapse
|
36
|
Kaya L, Karatum O, Balamur R, Kaleli HN, Önal A, Vanalakar SA, Hasanreisoğlu M, Nizamoglu S. MnO 2 Nanoflower Integrated Optoelectronic Biointerfaces for Photostimulation of Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301854. [PMID: 37386797 PMCID: PMC10477844 DOI: 10.1002/advs.202301854] [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: 03/22/2023] [Revised: 06/09/2023] [Indexed: 07/01/2023]
Abstract
Optoelectronic biointerfaces have gained significant interest for wireless and electrical control of neurons. Three-dimentional (3D) pseudocapacitive nanomaterials with large surface areas and interconnected porous structures have great potential for optoelectronic biointerfaces that can fulfill the requirement of high electrode-electrolyte capacitance to effectively transduce light into stimulating ionic currents. In this study, the integration of 3D manganese dioxide (MnO2 ) nanoflowers into flexible optoelectronic biointerfaces for safe and efficient photostimulation of neurons is demonstrated. MnO2 nanoflowers are grown via chemical bath deposition on the return electrode, which has a MnO2 seed layer deposited via cyclic voltammetry. They facilitate a high interfacial capacitance (larger than 10 mF cm-2 ) and photogenerated charge density (over 20 µC cm-2 ) under low light intensity (1 mW mm-2 ). MnO2 nanoflowers induce safe capacitive currents with reversible Faradaic reactions and do not cause any toxicity on hippocampal neurons in vitro, making them a promising material for biointerfacing with electrogenic cells. Patch-clamp electrophysiology is recorded in the whole-cell configuration of hippocampal neurons, and the optoelectronic biointerfaces trigger repetitive and rapid firing of action potentials in response to light pulse trains. This study points out the potential of electrochemically-deposited 3D pseudocapacitive nanomaterials as a robust building block for optoelectronic control of neurons.
Collapse
Affiliation(s)
- Lokman Kaya
- Department of Electrical and Electronics EngineeringKoc University34450IstanbulTurkey
| | - Onuralp Karatum
- Department of Electrical and Electronics EngineeringKoc University34450IstanbulTurkey
| | - Rıdvan Balamur
- Department of Electrical and Electronics EngineeringKoc University34450IstanbulTurkey
| | - Hümeyra Nur Kaleli
- Research Center for Translational MedicineKoc University34450IstanbulTurkey
| | - Asım Önal
- Department of Biomedical Science and EngineeringKoc University34450IstanbulTurkey
| | | | - Murat Hasanreisoğlu
- Research Center for Translational MedicineKoc University34450IstanbulTurkey
- Department of OphthalmologySchool of MedicineKoc University34450IstanbulTurkey
| | - Sedat Nizamoglu
- Department of Electrical and Electronics EngineeringKoc University34450IstanbulTurkey
- Department of Biomedical Science and EngineeringKoc University34450IstanbulTurkey
| |
Collapse
|
37
|
Falvey A, Palandira SP, Chavan SS, Brines M, Tracey KJ, Pavlov VA. Electrical stimulation of the dorsal motor nucleus of the vagus regulates inflammation without affecting the heart rate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541191. [PMID: 37292846 PMCID: PMC10245723 DOI: 10.1101/2023.05.17.541191] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background The vagus nerve plays an important role in neuroimmune interactions and in the regulation of inflammation. A major source of efferent vagus nerve fibers that contribute to the regulation of inflammation is the brainstem dorsal motor nucleus of the vagus (DMN) as recently shown using optogenetics. In contrast to optogenetics, electrical neuromodulation has broad therapeutic implications, but the anti-inflammatory efficacy of electrical DMN stimulation (eDMNS) was not previously investigated. Here, we examined the effects of eDMNS on heart rate (HR) and cytokine levels in murine endotoxemia as well as the cecal ligation and puncture (CLP) model of sepsis. Methods Anesthetized male 8-10-week-old C57BL/6 mice on a stereotaxic frame were subjected to eDMNS using a concentric bipolar electrode inserted into the left or right DMN or sham stimulation. eDMNS (50, 250 or 500 μA and 30 Hz, for 1 min) was performed and HR recorded. In endotoxemia experiments, sham or eDMNS utilizing 250 μA or 50 μA was performed for 5 mins and was followed by LPS (0.5 mg/kg) i.p. administration. eDMNS was also applied in mice with cervical unilateral vagotomy or sham operation. In CLP experiments sham or left eDMNS was performed immediately post CLP. Cytokines and corticosterone were analyzed 90 mins after LPS administration or 24h after CLP. CLP survival was monitored for 14 days. Results Either left or right eDMNS at 250 μA and 500 μA decreased HR, compared with pre- and post-stimulation. This effect was not observed at 50 μA. Left side eDMNS at 50 μA, compared with sham stimulation, significantly decreased serum and splenic levels of the pro-inflammatory cytokine TNF and increased serum levels of the anti-inflammatory cytokine IL-10 during endotoxemia. The anti-inflammatory effect of eDMNS was abrogated in mice with unilateral vagotomy and were not associated with serum corticosterone alterations. Right side eDMNS suppressed serum TNF levels but had no effects on serum IL-10 and on splenic cytokines. In mice with CLP, left side eDMNS suppressed serum TNF and IL-6, as well as splenic IL-6 and increased splenic IL-10 and significantly improved the survival rate of CLP mice. Conclusions For the first time we show that a regimen of eDMNS which does not cause bradycardia alleviates LPS-induced inflammation and these effects require an intact vagus nerve and are not associated with corticosteroid alterations. eDMNS also decreases inflammation and improves survival in a model of polymicrobial sepsis. These findings are of interest for further studies exploring bioelectronic anti-inflammatory approaches targeting the brainstem DMN.
Collapse
Affiliation(s)
- Aidan Falvey
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Santhoshi P. Palandira
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Sangeeta S. Chavan
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, New York 11549, USA
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Michael Brines
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Kevin J. Tracey
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, New York 11549, USA
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Valentin A. Pavlov
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, New York 11549, USA
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| |
Collapse
|
38
|
Mylavarapu RV, Kanumuri VV, de Rivero Vaccari JP, Misra A, McMillan DW, Ganzer PD. Importance of timing optimization for closed-loop applications of vagus nerve stimulation. Bioelectron Med 2023; 9:8. [PMID: 37101239 PMCID: PMC10134677 DOI: 10.1186/s42234-023-00110-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/28/2023] Open
Abstract
In recent decades, vagus nerve stimulation (VNS) therapy has become widely used for clinical applications including epilepsy, depression, and enhancing the effects of rehabilitation. However, several questions remain regarding optimization of this therapy to maximize clinical outcomes. Although stimulation parameters such as pulse width, amplitude, and frequency are well studied, the timing of stimulation delivery both acutely (with respect to disease events) and chronically (over the timeline of a disease's progression) has generally received less attention. Leveraging such information would provide a framework for the implementation of next generation closed-loop VNS therapies. In this mini-review, we summarize a number of VNS therapies and discuss (1) general timing considerations for these applications and (2) open questions that could lead to further therapy optimization.
Collapse
Affiliation(s)
| | - Vivek V Kanumuri
- Department of Otolaryngology, University of Miami, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA
- Department of Neurological Surgery, University of Miami, Miami, FL, USA
| | - Amrit Misra
- Newton Wellesley Neurology Associates, Newton, MA, USA
| | - David W McMillan
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA
- Department of Neurological Surgery, University of Miami, Miami, FL, USA
| | - Patrick D Ganzer
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA.
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA.
| |
Collapse
|
39
|
Pio-Lopez L, Levin M. Morphoceuticals: perspectives for discovery of drugs targeting anatomical control mechanisms in regenerative medicine, cancer and aging. Drug Discov Today 2023; 28:103585. [PMID: 37059328 DOI: 10.1016/j.drudis.2023.103585] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/18/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
Morphoceuticals are a new class of interventions that target the setpoints of anatomical homeostasis for efficient, modular control of growth and form. Here, we focus on a subclass: electroceuticals, which specifically target the cellular bioelectrical interface. Cellular collectives in all tissues form bioelectrical networks via ion channels and gap junctions that process morphogenetic information, controlling gene expression and allowing cell networks to adaptively and dynamically control growth and pattern formation. Recent progress in understanding this physiological control system, including predictive computational models, suggests that targeting bioelectrical interfaces can control embryogenesis and maintain shape against injury, senescence and tumorigenesis. We propose a roadmap for drug discovery focused on manipulating endogenous bioelectric signaling for regenerative medicine, cancer suppression and antiaging therapeutics. Teaser: By taking advantage of the native problem-solving competencies of cells and tissues, a new kind of top-down approach to biomedicine becomes possible. Bioelectricity offers an especially tractable interface for interventions targeting the software of life for regenerative medicine applications.
Collapse
Affiliation(s)
- Léo Pio-Lopez
- Allen Discovery Center, Tufts University, Medford, MA, USA
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
40
|
de Moraes TL, Costa FO, Cabral DG, Fernandes DM, Sangaleti CT, Dalboni MA, Motta E Motta J, de Souza LA, Montano N, Irigoyen MC, Brines M, J Tracey K, Pavlov VA, Consolim Colombo FM. Brief periods of transcutaneous auricular vagus nerve stimulation improve autonomic balance and alter circulating monocytes and endothelial cells in patients with metabolic syndrome: a pilot study. Bioelectron Med 2023; 9:7. [PMID: 36998060 PMCID: PMC10064781 DOI: 10.1186/s42234-023-00109-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/11/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND There is emerging evidence that the nervous system regulates immune and metabolic alterations mediating Metabolic syndrome (MetS) pathogenesis via the vagus nerve. This study evaluated the effects of transcutaneous auricular vagus nerve stimulation (TAVNS) on key cardiovascular and inflammatory components of MetS. METHODS We conducted an open label, randomized (2:1), two-arm, parallel-group controlled trial in MetS patients. Subjects in the treatment group (n = 20) received 30 min of TAVNS with a NEMOS® device placed on the cymba conchae of the left ear, once weekly. Patients in the control group (n = 10) received no stimulation. Hemodynamic, heart rate variability (HRV), biochemical parameters, and monocytes, progenitor endothelial cells, circulating endothelial cells, and endothelial micro particles were evaluated at randomization, after the first TAVNS treatment, and again after 8 weeks of follow-up. RESULTS An improvement in sympathovagal balance (HRV analysis) was observed after the first TAVNS session. Only patients treated with TAVNS for 8 weeks had a significant decrease in office BP and HR, a further improvement in sympathovagal balance, with a shift of circulating monocytes towards an anti-inflammatory phenotype and endothelial cells to a reparative vascular profile. CONCLUSION These results are of interest for further study of TAVNS as treatment of MetS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Nicola Montano
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | - Michael Brines
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Kevin J Tracey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Valentin A Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Fernanda M Consolim Colombo
- Nove de Julho University - UNINOVE, São Paulo, Brazil.
- University of São Paulo, Hypertension Unit, São Paulo, Brazil.
| |
Collapse
|
41
|
Repeated use of 3,4-methylenedioxymethamphetamine is associated with the resilience in mice after chronic social defeat stress: A role of gut-microbiota-brain axis. Psychiatry Res 2023; 320:115020. [PMID: 36571897 DOI: 10.1016/j.psychres.2022.115020] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/29/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA), the most widely used illicit compound worldwide, is the most attractive therapeutic drug for post-traumatic stress disorder (PTSD). Recent observational studies of US adults demonstrated that lifetime MDMA use was associated with lower risk of depression. Here, we examined whether repeated administration of MDMA can affect resilience versus susceptibility in mice exposed to chronic social defeat stress (CSDS). CSDS produced splenomegaly, anhedonia-like phenotype, and higher plasma levels of interleukin-6 (IL-6) in the saline-treated mice. In contrast, CSDS did not cause these changes in the MDMA-treated mice. Analysis of gut microbiome found several microbes altered between saline + CSDS group and MDMA + CSDS group. Untargeted metabolomics analysis showed that plasma levels of N-epsilon-methyl-L-lysine in the saline + CSDS group were significantly higher than those in the control and MDMA + CSDS groups. Interestingly, there were positive correlations between plasma IL-6 levels and the abundance of several microbes (or plasma N-epsilon-methyl-L-lysine) in the three groups. Furthermore, there were also positive correlations between the abundance of several microbes and N-epsilon-methyl-L-lysine in the three groups. In conclusion, these data suggest that repeated administration of MDMA might contribute to stress resilience in mice subjected to CSDS through gut-microbiota-brain axis.
Collapse
|
42
|
Harikesh PC, Yang CY, Wu HY, Zhang S, Donahue MJ, Caravaca AS, Huang JD, Olofsson PS, Berggren M, Tu D, Fabiano S. Ion-tunable antiambipolarity in mixed ion-electron conducting polymers enables biorealistic organic electrochemical neurons. NATURE MATERIALS 2023; 22:242-248. [PMID: 36635590 PMCID: PMC9894750 DOI: 10.1038/s41563-022-01450-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/28/2022] [Indexed: 05/29/2023]
Abstract
Biointegrated neuromorphic hardware holds promise for new protocols to record/regulate signalling in biological systems. Making such artificial neural circuits successful requires minimal device/circuit complexity and ion-based operating mechanisms akin to those found in biology. Artificial spiking neurons, based on silicon-based complementary metal-oxide semiconductors or negative differential resistance device circuits, can emulate several neural features but are complicated to fabricate, not biocompatible and lack ion-/chemical-based modulation features. Here we report a biorealistic conductance-based organic electrochemical neuron (c-OECN) using a mixed ion-electron conducting ladder-type polymer with stable ion-tunable antiambipolarity. The latter is used to emulate the activation/inactivation of sodium channels and delayed activation of potassium channels of biological neurons. These c-OECNs can spike at bioplausible frequencies nearing 100 Hz, emulate most critical biological neural features, demonstrate stochastic spiking and enable neurotransmitter-/amino acid-/ion-based spiking modulation, which is then used to stimulate biological nerves in vivo. These combined features are impossible to achieve using previous technologies.
Collapse
Affiliation(s)
- Padinhare Cholakkal Harikesh
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Chi-Yuan Yang
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Han-Yan Wu
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Silan Zhang
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
- Wallenberg Wood Science Center, Linköping University, Norrköping, Sweden
| | - Mary J Donahue
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - April S Caravaca
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Jun-Da Huang
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Peder S Olofsson
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Berggren
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
- Wallenberg Wood Science Center, Linköping University, Norrköping, Sweden
- n-Ink AB, Norrköping, Sweden
| | - Deyu Tu
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Simone Fabiano
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden.
- Wallenberg Wood Science Center, Linköping University, Norrköping, Sweden.
- n-Ink AB, Norrköping, Sweden.
| |
Collapse
|
43
|
Leitzke M. Is the post-COVID-19 syndrome a severe impairment of acetylcholine-orchestrated neuromodulation that responds to nicotine administration? Bioelectron Med 2023; 9:2. [PMID: 36650574 PMCID: PMC9845100 DOI: 10.1186/s42234-023-00104-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
Following a SARS-CoV-2 infection, many individuals suffer from post-COVID-19 syndrome. It makes them unable to proceed with common everyday activities due to weakness, memory lapses, pain, dyspnea and other unspecific physical complaints. Several investigators could demonstrate that the SARS-CoV-2 related spike glycoprotein (SGP) attaches not only to ACE-2 receptors but also shows DNA sections highly affine to nicotinic acetylcholine receptors (nAChRs). The nAChR is the principal structure of cholinergic neuromodulation and is responsible for coordinated neuronal network interaction. Non-intrinsic viral nAChR attachment compromises integrative interneuronal communication substantially. This explains the cognitive, neuromuscular and mood impairment, as well as the vegetative symptoms, characterizing post-COVID-19 syndrome. The agonist ligand nicotine shows an up to 30-fold higher affinity to nACHRs than acetylcholine (ACh). We therefore hypothesize that this molecule could displace the virus from nAChR attachment and pave the way for unimpaired cholinergic signal transmission. Treating several individuals suffering from post-COVID-19 syndrome with a nicotine patch application, we witnessed improvements ranging from immediate and substantial to complete remission in a matter of days.
Collapse
Affiliation(s)
- Marco Leitzke
- Department of Anesthesiology, Helios Clinics, Colditzer Straße 48, 04703, Leisnig, Germany.
| |
Collapse
|
44
|
Hashimoto K. Neuroinflammation through the vagus nerve-dependent gut–microbiota–brain axis in treatment-resistant depression. PROGRESS IN BRAIN RESEARCH 2023. [DOI: 10.1016/bs.pbr.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
45
|
Zhao Z, Li L, Xin C, Yin Y, Zhang R, Guo J. A bibliometric analysis of 100 top-cited journal articles related to acupuncture regulation of the autonomic nervous system. Front Neurosci 2022; 16:1086087. [PMID: 36620457 PMCID: PMC9813952 DOI: 10.3389/fnins.2022.1086087] [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: 11/01/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose Research on the effects of acupuncture on autonomic function has been conducted for several decades, and a few notable studies have emerged in recent years. This study used bibliometric analysis to assess 100 top-cited articles to characterize the current status and research trends over the last three decades. Methods The 100 top-cited publications were identified from the Web of Science Core Collection database. The bibliometrix package in R was used for quantitative and qualitative analyses of the publication patterns and the country/region, institution, and author contributions. VOSviewer was used to construct networks based on co-citation analysis of the journals and the keyword co-occurrence. Results The 100 top-cited articles were identified with a total of 8,123 citations (range: 37-345). The majority of the articles came from the USA (n = 42), followed by Japan (n = 14) and mainland China (n = 13). Articles from the USA exhibited the largest number of citations (3,582 citations), followed by articles from Japan (1,189 citations), then articles from mainland China (755 citations). Neurosciences/Neurology was the most studied research area (n = 41). The Autonomic Neuroscience: Basic and Clinical published the largest number of papers (n = 14), while Brain Research received the largest number of citations (205 citations). Longhurst JC was the most productive author (10 publications), and Sato A was first among the cited authors (87 citations). The most frequently cited articles that focused on gastrointestinal, cardiovascular, or gynecologic responses to acupuncture regulation of the autonomic nervous system first appeared in the 1990s, peaked in the 2000s, then decreased after 2010. Publication of articles focused on the anti-inflammatory effects of acupuncture associated with autonomic function demonstrated an increasing trend over the last three decades. Conclusion From the initial studies focusing on the autonomic mechanism of visceral responses to acupuncture, researchers concentrated on exploring the autonomic mechanism of acupuncture in the control of systemic inflammation. Non-invasive electrical methods that activate somato-autonomic reflexes are current translational directions in clinical practice. Additional investigation of the underlying neuroanatomical basis of somato-autonomic reflexes also is needed.
Collapse
Affiliation(s)
- Zhanhao Zhao
- Danyang Hospital of Traditional Chinese Medicine, Teaching Hospital of Nanjing University of Chinese Medicine, Danyang, China
| | - Li Li
- Acupuncture and Massage College, Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing, China,Department of Acupuncture Rehabilitation, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen Xin
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yaqun Yin
- Acupuncture and Massage College, Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Rong Zhang
- School of Health Sciences, Wuhan University, Wuhan, China
| | - Jing Guo
- Acupuncture and Massage College, Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing, China,*Correspondence: Jing Guo,
| |
Collapse
|
46
|
Kirkland LG, Garbe CG, Hadaya J, Benson PV, Wagener BM, Tankovic S, Hoover DB. Sympathetic innervation of human and porcine spleens: implications for between species variation in function. Bioelectron Med 2022; 8:20. [PMID: 36536461 PMCID: PMC9762010 DOI: 10.1186/s42234-022-00102-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The vagus nerve affects innate immune responses by activating spleen-projecting sympathetic neurons, which modulate leukocyte function. Recent basic and clinical research investigating vagus nerve stimulation to engage the cholinergic anti-inflammatory pathway (CAP) has shown promising therapeutic results for a variety of inflammatory diseases. Abundant sympathetic innervation occurs in rodent spleens, and use of these species has dominated mechanistic research investigating the CAP. However, previous neuroanatomical studies of human spleen found a more restricted pattern of innervation compared to rodents. Therefore, our primary goal was to establish the full extent of sympathetic innervation of human spleens using donor tissue with the shortest procurement to fixation time. Parallel studies of porcine spleen, a large animal model, were performed as a positive control and for comparison. METHODS Human and porcine spleen tissue were fixed immediately after harvest and prepared for immunohistochemistry. Human heart and porcine spleen were stained in conjunction as positive controls. Several immunohistochemical protocols were compared for best results. Tissue was stained for tyrosine hydroxylase (TH), a noradrenergic marker, using VIP purple chromogen. Consecutive tissue slices were stained for neuropeptide Y (NPY), which often co-localizes with TH, or double-labelled for TH and CD3, a T cell marker. High-magnification images and full scans of the tissue were obtained and analyzed for qualitative differences between species. RESULTS TH had dominant perivascular localization in human spleen, with negligible innervation of parenchyma, but such nerves were abundant throughout ventricular myocardium. In marked contrast, noradrenergic innervation was abundant in all regions of porcine spleen, with red pulp having more nerves than white pulp. NPY stain results were consistent with this pattern. In human spleen, noradrenergic nerves only ran close to T cells at the boundary of the periarterial lymphatic sheath and arteries. In porcine spleen, noradrenergic nerves were closely associated with T cells in both white and red pulp as well as other leukocytes in red pulp. CONCLUSION Sympathetic innervation of the spleen varies between species in both distribution and abundance, with humans and pigs being at opposite extremes. This has important implications for sympathetic regulation of neuroimmune interactions in the spleen of different species and focused targeting of the CAP in humans.
Collapse
Affiliation(s)
- Logan G. Kirkland
- grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Chloe G. Garbe
- grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Joseph Hadaya
- grid.19006.3e0000 0000 9632 6718UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095 USA ,grid.19006.3e0000 0000 9632 6718Molecular, Cellular, and Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA USA
| | - Paul V. Benson
- grid.265892.20000000106344187Department of Pathology, The University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL 35249 USA
| | - Brant M. Wagener
- grid.265892.20000000106344187Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL 35249 USA
| | - Sanjin Tankovic
- grid.265892.20000000106344187Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL 35249 USA
| | - Donald B. Hoover
- grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA ,grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine and Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN 37614 USA
| |
Collapse
|
47
|
Abstract
Approximately 20 years ago it was discovered that the vagus nerve regulates pro-inflammatory cytokine levels and inflammation. Subsequent research using several preclinical models revealed that vagus nerve stimulation evokes a protective decrease in pro-inflammatory cytokines in multiple inflammatory disorders. Consequently, the pro- and anti- inflammatory cytokine balance has become the predominant readout for indicating a positive outcome of vagus nerve stimulation. However, cytokine levels are just a single aspect of an effective immune response. It is conceivable that vagus nerve stimulation regulates inflammation through additional mechanisms. In this letter, I discuss a manuscript that describes how vagus nerve stimulation promotes resolution of inflammation via regulating the balance of specialised pro-resolving mediator levels and neutrophil activity.
Collapse
Affiliation(s)
- Aidan Falvey
- grid.416477.70000 0001 2168 3646Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, 11030 Manhasset, NY USA
| |
Collapse
|
48
|
Palandira SP, Carrion J, Turecki L, Falvey A, Zeng Q, Liu H, Tsaava T, Herschberg D, Brines M, Chavan SS, Chang EH, Vo A, Ma Y, Metz CN, Al-Abed Y, Tracey KJ, Pavlov VA. A dual tracer [ 11C]PBR28 and [ 18F]FDG microPET evaluation of neuroinflammation and brain energy metabolism in murine endotoxemia. Bioelectron Med 2022; 8:18. [PMID: 36451231 PMCID: PMC9710165 DOI: 10.1186/s42234-022-00101-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/09/2022] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Brain metabolic alterations and neuroinflammation have been reported in several peripheral inflammatory conditions and present significant potential for targeting with new diagnostic approaches and treatments. However, non-invasive evaluation of these alterations remains a challenge. METHODS Here, we studied the utility of a micro positron emission tomography (microPET) dual tracer ([11C]PBR28 - for microglial activation and [18F]FDG for energy metabolism) approach to assess brain dysfunction, including neuroinflammation in murine endotoxemia. MicroPET imaging data were subjected to advanced conjunction and individual analyses, followed by post-hoc analysis. RESULTS There were significant increases in [11C]PBR28 and [18F]FDG uptake in the hippocampus of C57BL/6 J mice 6 h following LPS (2 mg/kg) intraperitoneal (i.p.) administration compared with saline administration. These results confirmed previous postmortem observations. In addition, patterns of significant simultaneous activation were demonstrated in the hippocampus, the thalamus, and the hypothalamus in parallel with other tracer-specific and region-specific alterations. These changes were observed in the presence of robust systemic inflammatory responses manifested by significantly increased serum cytokine levels. CONCLUSIONS Together, these findings demonstrate the applicability of [11C]PBR28 - [18F]FDG dual tracer microPET imaging for assessing neuroinflammation and brain metabolic alterations in conditions "classically" characterized by peripheral inflammatory and metabolic pathogenesis.
Collapse
Affiliation(s)
| | - Joseph Carrion
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Lauren Turecki
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Aidan Falvey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Qiong Zeng
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Hui Liu
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Tea Tsaava
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Dov Herschberg
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Michael Brines
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Sangeeta S Chavan
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Eric H Chang
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - An Vo
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Yilong Ma
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Christine N Metz
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Yousef Al-Abed
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Kevin J Tracey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Valentin A Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA.
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|