1
|
Giannino G, Nocera L, Andolfatto M, Braia V, Giacobbe F, Bruno F, Saglietto A, Angelini F, De Filippo O, D'Ascenzo F, De Ferrari GM, Dusi V. Vagal nerve stimulation in myocardial ischemia/reperfusion injury: from bench to bedside. Bioelectron Med 2024; 10:22. [PMID: 39267134 PMCID: PMC11395864 DOI: 10.1186/s42234-024-00153-6] [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: 04/28/2024] [Accepted: 07/31/2024] [Indexed: 09/14/2024] Open
Abstract
The identification of acute cardioprotective strategies against myocardial ischemia/reperfusion (I/R) injury that can be applied in the catheterization room is currently an unmet clinical need and several interventions evaluated in the past at the pre-clinical level have failed in translation. Autonomic imbalance, sustained by an abnormal afferent signalling, is a key component of I/R injury. Accordingly, there is a strong rationale for neuromodulation strategies, aimed at reducing sympathetic activity and/or increasing vagal tone, in this setting. In this review we focus on cervical vagal nerve stimulation (cVNS) and on transcutaneous auricular vagus nerve stimulation (taVNS); the latest has the potential to overcome several of the issues of invasive cVNS, including the possibility of being used in an acute setting, while retaining its beneficial effects. First, we discuss the pathophysiology of I/R injury, that is mostly a consequence of the overproduction of reactive oxygen species. Second, we describe the functional anatomy of the parasympathetic branch of the autonomic nervous system and the most relevant principles of bioelectronic medicine applied to electrical vagal modulation, with a particular focus on taVNS. Then, we provide a detailed and comprehensive summary of the most relevant pre-clinical studies of invasive and non-invasive VNS that support its strong cardioprotective effect whenever there is an acute or chronic cardiac injury and specifically in the setting of myocardial I/R injury. The potential benefit in the emerging field of post cardiac arrest syndrome (PCAS) is also mentioned. Indeed, electrical cVNS has a strong anti-adrenergic, anti-inflammatory, antioxidants, anti-apoptotic and pro-angiogenic effect; most of the involved molecular pathways were already directly confirmed to take place at the cardiac level for taVNS. Pre-clinical data clearly show that the sooner VNS is applied, the better the outcome, with the possibility of a marked infarct size reduction and almost complete left ventricular reverse remodelling when VNS is applied immediately before and during reperfusion. Finally, we describe in detail the limited but very promising clinical experience of taVNS in I/R injury available so far.
Collapse
Affiliation(s)
- Giuseppe Giannino
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Corso Bramante 88, Turin, 10126, Italy
| | - Lorenzo Nocera
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Corso Bramante 88, Turin, 10126, Italy
| | - Maria Andolfatto
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Corso Bramante 88, Turin, 10126, Italy
| | - Valentina Braia
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Corso Bramante 88, Turin, 10126, Italy
| | - Federico Giacobbe
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Corso Bramante 88, Turin, 10126, Italy
| | - Francesco Bruno
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
| | - Andrea Saglietto
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
| | - Filippo Angelini
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
| | - Ovidio De Filippo
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
| | - Fabrizio D'Ascenzo
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Corso Bramante 88, Turin, 10126, Italy
| | - Gaetano Maria De Ferrari
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Corso Bramante 88, Turin, 10126, Italy
| | - Veronica Dusi
- Cardiology, Department of Medical Sciences, University of Turin, Torino, Italy.
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Corso Bramante 88, Turin, 10126, Italy.
| |
Collapse
|
2
|
Villeda-González JD, Gómez-Olivares JL, Baiza-Gutman LA. New paradigms in the study of the cholinergic system and metabolic diseases: Acetyl-and-butyrylcholinesterase. J Cell Physiol 2024; 239:e31274. [PMID: 38605655 DOI: 10.1002/jcp.31274] [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: 08/24/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024]
Abstract
Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are enzymes that belong to the neuromuscular cholinergic system, their main function is to hydrolyze the neurotransmitter acetylcholine (ACh), through their hydrolysis these enzymes regulate the neuronal and neuromuscular cholinergic system. They have recently attracted considerable attention due to the discovery of new enzymatic and nonenzymatic functions. These discoveries have aroused the interest of numerous scientists, consolidating the relevance of this group of enzymes. Recent investigations have revealed a positive correlation between several risk factors for metabolic syndrome (MetS) and the expression of cholinesterases (ChE's), which underscore the impact of high ChE's activity on the pro-inflammatory state associated with MetS. In addition, the excessive hydrolysis of ACh and other choline esters (succinylcholine, propionylcholine, butyrylcholine, etc.) by both ChE's results in the overproduction of fatty acid precursor metabolites, which facilitate the synthesis of very low-density lipoproteins and triacylglycerols. Participation in these processes may represent the link between ChE's and metabolic disorders. However, further scientific research is required to fully elucidate the involvement of ChE's in metabolic diseases. This review aims to collect recent research studies that contribute to understanding the association between the cholinergic system and metabolic diseases.
Collapse
Affiliation(s)
- Juan David Villeda-González
- Estancia Posdoctoral CONAHCYT, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - José Luis Gómez-Olivares
- Laboratorio de Biomembranas, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, México City, México
| | - Luis Arturo Baiza-Gutman
- Laboratorio en Biología del Desarrollo, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Estado de México, México
| |
Collapse
|
3
|
Kim JT, Song K, Han SW, Youn DH, Jung H, Kim KS, Lee HJ, Hong JY, Cho YJ, Kang SM, Jeon JP. Modeling of the brain-lung axis using organoids in traumatic brain injury: an updated review. Cell Biosci 2024; 14:83. [PMID: 38909262 PMCID: PMC11193205 DOI: 10.1186/s13578-024-01252-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
Clinical outcome after traumatic brain injury (TBI) is closely associated conditions of other organs, especially lungs as well as degree of brain injury. Even if there is no direct lung damage, severe brain injury can enhance sympathetic tones on blood vessels and vascular resistance, resulting in neurogenic pulmonary edema. Conversely, lung damage can worsen brain damage by dysregulating immunity. These findings suggest the importance of brain-lung axis interactions in TBI. However, little research has been conducted on the topic. An advanced disease model using stem cell technology may be an alternative for investigating the brain and lungs simultaneously but separately, as they can be potential candidates for improving the clinical outcomes of TBI.In this review, we describe the importance of brain-lung axis interactions in TBI by focusing on the concepts and reproducibility of brain and lung organoids in vitro. We also summarize recent research using pluripotent stem cell-derived brain organoids and their preclinical applications in various brain disease conditions and explore how they mimic the brain-lung axis. Reviewing the current status and discussing the limitations and potential perspectives in organoid research may offer a better understanding of pathophysiological interactions between the brain and lung after TBI.
Collapse
Affiliation(s)
- Jong-Tae Kim
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, 24252, Republic of Korea
| | - Kang Song
- Department of Green Chemical Engineering, Sangmyung University, Cheonan, 31066, Republic of Korea
| | - Sung Woo Han
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, 24252, Republic of Korea
| | - Dong Hyuk Youn
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, 24252, Republic of Korea
| | - Harry Jung
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, 24252, Republic of Korea
| | - Keun-Suh Kim
- Department of Periodontology, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Hyo-Jung Lee
- Department of Periodontology, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Ji Young Hong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Hallym University College of Medicine, Chuncheon, 24252, Republic of Korea
| | - Yong-Jun Cho
- Department of Neurosurgery, Hallym University College of Medicine, Chuncheon, 24252, Republic of Korea
| | - Sung-Min Kang
- Department of Green Chemical Engineering, Sangmyung University, Cheonan, 31066, Republic of Korea.
| | - Jin Pyeong Jeon
- Department of Neurosurgery, Hallym University College of Medicine, Chuncheon, 24252, Republic of Korea.
| |
Collapse
|
4
|
Mather M. The emotion paradox in the aging body and brain. Ann N Y Acad Sci 2024; 1536:13-41. [PMID: 38676452 DOI: 10.1111/nyas.15138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
With age, parasympathetic activity decreases, while sympathetic activity increases. Thus, the typical older adult has low heart rate variability (HRV) and high noradrenaline levels. Younger adults with this physiological profile tend to be unhappy and stressed. Yet, with age, emotional experience tends to improve. Why does older adults' emotional well-being not suffer as their HRV decreases? To address this apparent paradox, I present the autonomic compensation model. In this model, failing organs, the initial phases of Alzheimer's pathology, and other age-related diseases trigger noradrenergic hyperactivity. To compensate, older brains increase autonomic regulatory activity in the pregenual prefrontal cortex (PFC). Age-related declines in nerve conduction reduce the ability of the pregenual PFC to reduce hyperactive noradrenergic activity and increase peripheral HRV. But these pregenual PFC autonomic compensation efforts have a significant impact in the brain, where they bias processing in favor of stimuli that tend to increase parasympathetic activity (e.g., stimuli that increase feelings of safety) and against stimuli that tend to increase sympathetic activity (e.g., threatening stimuli). In summary, the autonomic compensation model posits that age-related chronic sympathetic/noradrenergic hyperactivity stimulates regulatory attempts that have the side effect of enhancing emotional well-being.
Collapse
Affiliation(s)
- Mara Mather
- Leonard Davis School of Gerontology, Department of Psychology, and Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
5
|
Yang M, Pan L, Tian H, Zhou T, Xin H, Feng Y, Zou X, Lv Z, Xu Y, Jin X, Gui S, Lu X. pH- and Matrix Metalloproteinase-Responsive Multifunctional Bilayer Microneedles Platform for Treatment of Tinea Pedis. ACS Biomater Sci Eng 2024; 10:3108-3119. [PMID: 38659287 PMCID: PMC11094678 DOI: 10.1021/acsbiomaterials.4c00305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Persistent foot odor and itchiness are common symptoms of tinea pedis, significantly disrupting the daily life of those affected. The cuticular barrier at the site of the tinea pedis is thickened, which impedes the effective penetration of antifungal agents. Additionally, fungi can migrate from the skin surface to deeper tissues, posing challenges in the current clinical treatment for tinea pedis. To effectively treat tinea pedis, we developed a platform of bilayer gelatin methacrylate (GelMA) microneedles (MNs) loaded with salicylic acid (SA) and FK13-a1 (SA/FK13-a1@GelMA MNs). SA/FK13-a1@GelMA MNs exhibit pH- and matrix metalloproteinase (MMP)-responsive properties for efficient drug delivery. The MNs are designed to deliver salicylic acid (SA) deep into the stratum corneum, softening the cuticle and creating microchannels. This process enables the antibacterial peptide FK13-a1 to penetrate through the stratum corneum barrier, facilitating intradermal diffusion and exerting antifungal and anti-inflammatory effects. In severe cases of tinea pedis, heightened local pH levels and MMP activity further accelerate drug release. Our research demonstrates that SA/FK13-a1@GelMA MNs are highly effective against Trichophyton mentagrophytes, Trichophyton rubrum, and Candida albicans. They also reduced stratum corneum thickness, fungal burden, and inflammation in a guinea pig model of tinea pedis induced by T. mentagrophytes. Furthermore, it was discovered that SA/FK13-a1@GelMA MNs exhibit excellent biocompatibility. These findings suggest that SA/FK13-a1@GelMA MNs have significant potential for the clinical treatment of tinea pedis as well as other fungal skin disorders.
Collapse
Affiliation(s)
- Musheng Yang
- Guangdong
Provincial Key Laboratory of Pharmaceutical Bioactive Substances,
School of Basic Medical Sciences, Guangdong
Pharmaceutical University, Guangzhou 510006, China
- Shenzhen
Center for Disease Control and Prevention, Shenzhen 518055, China
- Intensive
Care Unit, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518031, China
| | - Lingling Pan
- Guangdong
Provincial Key Laboratory of Pharmaceutical Bioactive Substances,
School of Basic Medical Sciences, Guangdong
Pharmaceutical University, Guangzhou 510006, China
- Shenzhen
Center for Disease Control and Prevention, Shenzhen 518055, China
- Intensive
Care Unit, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518031, China
| | - Hongmei Tian
- Guangdong
Provincial Key Laboratory of Pharmaceutical Bioactive Substances,
School of Basic Medical Sciences, Guangdong
Pharmaceutical University, Guangzhou 510006, China
- Shenzhen
Center for Disease Control and Prevention, Shenzhen 518055, China
- Intensive
Care Unit, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518031, China
| | - Tao Zhou
- Guangdong
Provincial Key Laboratory of Pharmaceutical Bioactive Substances,
School of Basic Medical Sciences, Guangdong
Pharmaceutical University, Guangzhou 510006, China
- Shenzhen
Center for Disease Control and Prevention, Shenzhen 518055, China
- Intensive
Care Unit, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518031, China
| | - Hui Xin
- Guangdong
Provincial Key Laboratory of Pharmaceutical Bioactive Substances,
School of Basic Medical Sciences, Guangdong
Pharmaceutical University, Guangzhou 510006, China
- Shenzhen
Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Yonglin Feng
- Guangdong
Provincial Key Laboratory of Pharmaceutical Bioactive Substances,
School of Basic Medical Sciences, Guangdong
Pharmaceutical University, Guangzhou 510006, China
- Shenzhen
Center for Disease Control and Prevention, Shenzhen 518055, China
- Intensive
Care Unit, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518031, China
| | - Xuan Zou
- Shenzhen
Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Ziquan Lv
- Shenzhen
Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Yinghua Xu
- Key
Laboratory of the Ministry of Health for Research on Quality and Standardization
of Biotechnology Products, National Institutes
for Food and Drug Control, Beijing 102629, China
| | - Xiaobao Jin
- Guangdong
Provincial Key Laboratory of Pharmaceutical Bioactive Substances,
School of Basic Medical Sciences, Guangdong
Pharmaceutical University, Guangzhou 510006, China
| | - Shuiqing Gui
- Intensive
Care Unit, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518031, China
| | - Xuemei Lu
- Guangdong
Provincial Key Laboratory of Pharmaceutical Bioactive Substances,
School of Basic Medical Sciences, Guangdong
Pharmaceutical University, Guangzhou 510006, China
- Shenzhen
Center for Disease Control and Prevention, Shenzhen 518055, China
| |
Collapse
|
6
|
Yang K, Huang Z, Wang S, Zhao Z, Yi P, Chen Y, Xiao M, Quan J, Hu X. The Hepatic Nerves Regulated Inflammatory Effect in the Process of Liver Injury: Is Nerve the Key Treating Target for Liver Inflammation? Inflammation 2023; 46:1602-1611. [PMID: 37490221 DOI: 10.1007/s10753-023-01854-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/09/2023] [Accepted: 06/05/2023] [Indexed: 07/26/2023]
Abstract
Liver injury is a common pathological basis for various liver diseases. Chronic liver injury is often an important initiating factor in liver fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Currently, hepatitis A and E infections are the most common causes of acute liver injury worldwide, whereas drug toxicity (paracetamol overdose) in the USA and part of Western Europe. In recent years, chronic liver injury has become a common disease that harms human health. Meanwhile, the main causes of chronic liver injury are viral hepatitis (B, C) and long-term alcohol consumption worldwide. During the process of liver injury, massive inflammatory cytokines are stimulated by these hazardous factors, leading to a systemic inflammatory response syndrome, followed by a compensatory anti-inflammatory response, which causes immune cell dysfunction and sepsis, subsequent multi-organ failure. Cytokine release and immune cell infiltration-mediated aseptic inflammation are the most important features of the pathobiology of liver failure. From this perspective, diminishing the onset and progression of liver inflammation is of clinical importance in the treatment of liver injury. Although many studies have hinted at the critical role of nerves in regulating inflammation, there largely remains undetermined how hepatic nerves mediate immune inflammation and how the inflammatory factors released by these nerves are involved in the process of liver injury. Therefore, the purpose of this article is to summarize previous studies in the field related to hepatic nerve and inflammation as well as future perspectives on the aforementioned questions. Our findings were presented in three aspects: types of nerve distribution in the liver, how these nerves regulate immunity, and the role of liver nerves in hepatitis and liver failure.
Collapse
Affiliation(s)
- Kaili Yang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zebing Huang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shuyi Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhihong Zhao
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Panpan Yi
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yayu Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Meifang Xiao
- Department of Health Management Center, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Jun Quan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Xingwang Hu
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, 87Th of Xiangya Road, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
7
|
Kobori N, Moore AN, Redell JB, Dash PK. Caudal DMN neurons innervate the spleen and release CART peptide to regulate neuroimmune function. J Neuroinflammation 2023; 20:158. [PMID: 37403174 PMCID: PMC10318820 DOI: 10.1186/s12974-023-02838-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/19/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Inflammation is a fundamental biological response to injury and infection, which if unregulated can contribute to the pathophysiology of many diseases. The vagus nerve, which primarily originates from the dorsal motor nucleus (DMN), plays an important role in rapidly dampening inflammation by regulating splenic function. However, direct vagal innervation of the spleen, which houses the majority of immune and inflammatory cells, has not been established. As an alternative to direct innervation, an anti-inflammatory reflex pathway has been proposed which involves the vagus nerve, the sympathetic celiac ganglion, and the neurotransmitter norepinephrine. Although sympathetic regulation of inflammation has been shown, the interaction of the vagus nerve and the celiac ganglia requires a unique interaction of parasympathetic and sympathetic inputs, making this putative mechanism of brain-spleen interaction controversial. BODY: As neuropeptides can be expressed at relatively high levels in neurons, we reasoned that DMN neuropeptide immunoreactivity could be used to determine their target innervation. Employing immunohistochemistry, subdiaphragmatic vagotomy, viral tract tracing, CRISPR-mediated knock-down, and functional assays, we show that cocaine and amphetamine-regulated transcript (CART) peptide-expressing projection neurons in the caudal DMN directly innervate the spleen. In response to lipopolysaccharide (LPS) stimulation, CART acts to reduce inflammation, an effect that can be augmented by intrasplenic administration of a synthetic CART peptide. These in vivo effects could be recapitulated in cultured splenocytes, suggesting that these cells express the as yet unidentified CART receptor(s). CONCLUSION Our results provide evidence for direct connections between the caudal DMN and spleen. In addition to acetylcholine, these neurons express the neuropeptide CART that, once released, acts to suppress inflammation by acting directly upon splenocytes.
Collapse
Affiliation(s)
- Nobuhide Kobori
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - John B Redell
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA.
| |
Collapse
|
8
|
Functional Two-Way Crosstalk Between Brain and Lung: The Brain-Lung Axis. Cell Mol Neurobiol 2023; 43:991-1003. [PMID: 35678887 PMCID: PMC9178545 DOI: 10.1007/s10571-022-01238-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/25/2022] [Indexed: 11/03/2022]
Abstract
The brain has many connections with various organs. Recent advances have demonstrated the existence of a bidirectional central nervous system (CNS) and intestinal tract, that is, the brain-gut axis. Although studies have suggested that the brain and lung can communicate with each other through many pathways, whether there is a brain-lung axis remains still unknown. Based on previous findings, we put forward a hypothesis: there is a cross-talk between the central nervous system and the lung via neuroanatomical pathway, endocrine pathway, immune pathway, metabolites and microorganism pathway, gas pathway, that is, the brain-lung axis. Beyond the regulation of the physiological state in the body, bi-directional communication between the lung and the brain is associated with a variety of disease states, including lung diseases and CNS diseases. Exploring the brain-lung axis not only helps us to understand the development of the disease from different aspects, but also provides an important target for treatment strategies.
Collapse
|
9
|
Xu X, Xu H, Zhang Z. Cerebral amyloid angiopathy-related cardiac injury: Focus on cardiac cell death. Front Cell Dev Biol 2023; 11:1156970. [PMID: 36910141 PMCID: PMC9998697 DOI: 10.3389/fcell.2023.1156970] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 02/16/2023] [Indexed: 03/14/2023] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a kind of disease in which amyloid β (Aβ) and other amyloid protein deposits in the cerebral cortex and the small blood vessels of the brain, causing cerebrovascular and brain parenchymal damage. CAA patients are often accompanied by cardiac injury, involving Aβ, tau and transthyroxine amyloid (ATTR). Aβ is the main injury factor of CAA, which can accelerate the formation of coronary artery atherosclerosis, aortic valve osteogenesis calcification and cardiomyocytes basophilic degeneration. In the early stage of CAA (pre-stroke), the accompanying locus coeruleus (LC) amyloidosis, vasculitis and circulating Aβ will induce first hit to the heart. When the CAA progresses to an advanced stage and causes a cerebral hemorrhage, the hemorrhage leads to autonomic nervous function disturbance, catecholamine surges, and systemic inflammation reaction, which can deal the second hit to the heart. Based on the brain-heart axis, CAA and its associated cardiac injury can create a vicious cycle that accelerates the progression of each other.
Collapse
Affiliation(s)
- Xiaofang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huikang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhaocai Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of the Diagnosis and Treatment for Severe Trauma and Burn of Zhejiang Province, Hangzhou, China.,Zhejiang Province Clinical Research Center for Emergency and Critical care medicine, Hangzhou, China
| |
Collapse
|
10
|
Espeter F, Künne D, Garczarek L, Kuhlmann H, Skarabis A, Zivkovic AR, Brenner T, Schmidt K. Critically Ill COVID-19 Patients Show Reduced Point of Care-Measured Butyrylcholinesterase Activity—A Prospective, Monocentric Observational Study. Diagnostics (Basel) 2022; 12:diagnostics12092150. [PMID: 36140551 PMCID: PMC9498245 DOI: 10.3390/diagnostics12092150] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 01/08/2023] Open
Abstract
A biomarker for risk stratification and disease severity assessment in SARS-CoV-2 infections has not yet been established. Point of care testing (POCT) of butyrylcholinesterase (BChE) enables early detection of systemic inflammatory responses and correlates with disease severity in sepsis and burns. In acute care or resource-limited settings, POCT facilitates rapid clinical decision making, a particularly beneficial aspect in the management of pandemic situations. In this prospective observational study, POCT-measured BChE activity was assessed in 52 critically ill COVID-19 patients within 24 h of ICU admission and on the third and seventh day after ICU admission. Forty (77%) of these patients required venovenous extracorporeal membrane oxygenation (vvECMO). In critically ill COVID-19 patients, BChE activity is significantly decreased compared with healthy subjects, but also compared with other inflammatory conditions such as sepsis, burns, or trauma. POCT BChE activity reflects the severity of organ dysfunction and allows prediction of 28-day mortality in critically ill COVID-19 patients. Implementing early POCT BChE measurement could facilitate risk stratification and support admission and transfer decisions in resource-limited settings.
Collapse
Affiliation(s)
- Florian Espeter
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- Correspondence: ; Tel.: +49-201-723-84485
| | - David Künne
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Lena Garczarek
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Henning Kuhlmann
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Annabell Skarabis
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | | | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Karsten Schmidt
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
11
|
Time course of beat-to-beat blood pressure variability and outcome in patients with spontaneous intracerebral haemorrhage. J Hypertens 2022; 40:1744-1750. [PMID: 35943102 DOI: 10.1097/hjh.0000000000003206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Increased blood pressure variability (BPV) over 24 h or longer was associated with poor clinical outcomes in patients with intracerebral haemorrhage (ICH). However, the characteristics of beat-to-beat BPV, a rapid assessment of BPV and its association with outcome in ICH patients remain unknown. METHODS We consecutively and prospectively recruited patients with ICH between June 2014 and December 2020. Five-minute noninvasive beat-to-beat recordings were measured serially at three time points, 1-2, 4-6 and 10-12 days after ICH onset. BPV was calculated using standard deviation (SD) and variation independent of mean (VIM). Favourable outcome was defined as modified Rankin Scale score of less than 2 at 90 days. RESULTS The analysis included 66 participants (54.12 ± 10.79 years; 71.2% men) and 66 age and sex-matched healthy controls. Compared with that in healthy adults, beat-to-beat BPV was significantly increased 1-2 days after ICH and was completely recovered 10-12 days later. BPV recorded 1-2 days after ICH onset was higher among patients with unfavourable outcomes than among those with favourable outcomes (all P < 0.05) and higher BPV on days 1-2 was independently associated with a 3-month unfavourable outcome after adjustment for major covariates. CONCLUSION Beat-to-beat BPV was significantly increased among patients with ICH and could be completely recovered 10-12 days later. In addition, beat-to-beat BPV 1-2 days after ICH was independently associated with prognosis and could be regarded as a potential prognostic predictor and effective therapeutic target in the future.
Collapse
|
12
|
Kobrzycka AT, Stankiewicz AM, Goscik J, Gora M, Burzynska B, Iwanicka-Nowicka R, Pierzchala-Koziec K, Wieczorek M. Hypothalamic Neurochemical Changes in Long-Term Recovered Bilateral Subdiaphragmatic Vagotomized Rats. Front Behav Neurosci 2022; 16:869526. [PMID: 35874650 PMCID: PMC9304976 DOI: 10.3389/fnbeh.2022.869526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/10/2022] [Indexed: 11/26/2022] Open
Abstract
Background Vagus nerve is one of the crucial routes in communication between the immune and central nervous systems. The impaired vagal nerve function may intensify peripheral inflammatory processes. This effect subsides along with prolonged recovery after permanent nerve injury. One of the results of such compensation is a normalized plasma concentration of stress hormone corticosterone – a marker of hypothalamic-pituitary-adrenal (HPA) axis activity. In this work, we strive to explain this corticosterone normalization by studying the mechanisms responsible for compensation-related neurochemical alterations in the hypothalamus. Materials and Methods Using microarrays and high performance liquid chromatography (HPLC), we measured genome-wide gene expression and major amino acid neurotransmitters content in the hypothalamus of bilaterally vagotomized rats, 1 month after surgery. Results Our results show that, in the long term, vagotomy affects hypothalamic amino acids concentration but not mRNA expression of tested genes. Discussion We propose an alternative pathway of immune to CNS communication after vagotomy, leading to activation of the HPA axis, by influencing central amino acids and subsequent monoaminergic neurotransmission.
Collapse
Affiliation(s)
- Anna Teresa Kobrzycka
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Łodz, Łodz, Poland
- *Correspondence: Anna Teresa Kobrzycka,
| | - Adrian Mateusz Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Poland
| | - Joanna Goscik
- Software Department, Faculty of Computer Science, Bialystok University of Technology, Bialystok, Poland
| | - Monika Gora
- Department of Genetics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Burzynska
- Department of Genetics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Roksana Iwanicka-Nowicka
- Department of Genetics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
- Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | | - Marek Wieczorek
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Łodz, Łodz, Poland
- Marek Wieczorek,
| |
Collapse
|
13
|
Diamond LM, Alley J. Rethinking minority stress: A social safety perspective on the health effects of stigma in sexually-diverse and gender-diverse populations. Neurosci Biobehav Rev 2022; 138:104720. [PMID: 35662651 DOI: 10.1016/j.neubiorev.2022.104720] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/25/2022] [Accepted: 05/30/2022] [Indexed: 12/13/2022]
Abstract
For over two decades, the minority stress model has guided research on the health of sexually-diverse individuals (those who are not exclusively heterosexual) and gender-diverse individuals (those whose gender identity/expression differs from their birth-assigned sex/gender). According to this model, the cumulative stress caused by stigma and social marginalization fosters stress-related health problems. Yet studies linking minority stress to physical health outcomes have yielded mixed results, suggesting that something is missing from our understanding of stigma and health. Social safety may be the missing piece. Social safety refers to reliable social connection, inclusion, and protection, which are core human needs that are imperiled by stigma. The absence of social safety is just as health-consequential for stigmatized individuals as the presence of minority stress, because the chronic threat-vigilance fostered by insufficient safety has negative long-term effects on cognitive, emotional, and immunological functioning, even when exposure to minority stress is low. We argue that insufficient social safety is a primary cause of stigma-related health disparities and a key target for intervention.
Collapse
Affiliation(s)
- Lisa M Diamond
- Department of Psychology, University of Utah, Salt Lake City, UT, USA.
| | - Jenna Alley
- Department of Psychology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
14
|
Zhu L, Huang L, Le A, Wang TJ, Zhang J, Chen X, Wang J, Wang J, Jiang C. Interactions between the Autonomic Nervous System and the Immune System after Stroke. Compr Physiol 2022; 12:3665-3704. [PMID: 35766834 DOI: 10.1002/cphy.c210047] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acute stroke is one of the leading causes of morbidity and mortality worldwide. Stroke-induced immune-inflammatory response occurs in the perilesion areas and the periphery. Although stroke-induced immunosuppression may alleviate brain injury, it hinders brain repair as the immune-inflammatory response plays a bidirectional role after acute stroke. Furthermore, suppression of the systemic immune-inflammatory response increases the risk of life-threatening systemic bacterial infections after acute stroke. Therefore, it is essential to explore the mechanisms that underlie the stroke-induced immune-inflammatory response. Autonomic nervous system (ANS) activation is critical for regulating the local and systemic immune-inflammatory responses and may influence the prognosis of acute stroke. We review the changes in the sympathetic and parasympathetic nervous systems and their influence on the immune-inflammatory response after stroke. Importantly, this article summarizes the mechanisms on how ANS regulates the immune-inflammatory response through neurotransmitters and their receptors in immunocytes and immune organs after stroke. To facilitate translational research, we also discuss the promising therapeutic approaches modulating the activation of the ANS or the immune-inflammatory response to promote neurologic recovery after stroke. © 2022 American Physiological Society. Compr Physiol 12:3665-3704, 2022.
Collapse
Affiliation(s)
- Li Zhu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Anh Le
- Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Tom J Wang
- Winston Churchill High School, Potomac, Maryland, USA
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Xuemei Chen
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Jian Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.,Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
15
|
Zhang Y, Zheng L, Deng H, Feng D, Hu S, Zhu L, Xu W, Zhou W, Wang Y, Min K, Zhou Q, Chen Y, Zhou H, Yang H, Lv X. Electroacupuncture Alleviates LPS-Induced ARDS Through α7 Nicotinic Acetylcholine Receptor-Mediated Inhibition of Ferroptosis. Front Immunol 2022; 13:832432. [PMID: 35222419 PMCID: PMC8866566 DOI: 10.3389/fimmu.2022.832432] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an uncontrollable, progressive pulmonary inflammatory disease, and as a common clinical critical disease, there is no effective treatment available. Electroacupuncture (EA) therapy is a type of traditional Chinese medicine physiotherapy that can alleviate the inflammatory response. However, the potential mechanism of EA in the treatment of ARDS is not yet clear. Ferroptosis is a new type of programmed cell death characterized by intracellular iron accumulation and lipid peroxidation. Recently, emerging evidence has shown that ferroptosis is closely related to the occurrence and development of ARDS caused by various pathological factors. Here, we further investigated whether EA-mediated inhibition of ferroptosis in lung tissue could attenuate lipopolysaccharide (LPS)-induced ARDS and explored its underlying mechanisms. In this study, mice were administered LPS intraperitoneally to establish a model of LPS-induced ARDS. We found that EA stimulation could not only reduce the exudation of inflammatory cells and proteins in the alveolar lumen but also significantly alleviate the pathological changes of lung tissue, inhibit the production of proinflammatory cytokines and improve the survival rate of mice. Concurrently, we also found that ferroptosis events occurred in the lung tissue of LPS-induced ARDS mice, manifested by elevated iron levels, ROS production and lipid peroxidation. Intriguingly, our results showed that EA stimulation at the Zusanli (ST36) acupoint activated α7 nicotinic acetylcholine receptor (α7nAchR) in lung tissue mainly through the sciatic nerve and cervical vagus nerve, thus exerting anti-ferroptosis and pulmonary protective effects. Additionally, these effects were eliminated by methyllycaconitine (MLA), a selective antagonist of α7nAchR. In vitro experiments, activation of α7nAchR protected alveolar epithelial cells from LPS-induced ferroptosis. Furthermore, our experiments showed that the pulmonary protective effects of EA stimulation were effectively reversed by erastin, a ferroptosis activator. Collectively, we demonstrated that EA stimulation could alleviate LPS-induced ARDS by activating α7nAchR to inhibit LPS-induced ferroptosis in alveolar epithelial cells. Targeting and regulating ferroptosis in alveolar epithelial cells may be a potential intervention approach for the treatment of LPS-induced ALI/ARDS in the future.
Collapse
Affiliation(s)
- Yiguo Zhang
- Graduate School, Wannan Medical College, Wuhu, China
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li Zheng
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huimin Deng
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Di Feng
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Song Hu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lina Zhu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenting Xu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenyu Zhou
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Wang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Keting Min
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qing Zhou
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanli Chen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huanping Zhou
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Yang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Goldstein Ferber S, Weller A, Ben-Shachar M, Klinger G, Geva R. Development of the Ontogenetic Self-Regulation Clock. Int J Mol Sci 2022; 23:993. [PMID: 35055184 PMCID: PMC8778416 DOI: 10.3390/ijms23020993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/07/2022] [Accepted: 01/15/2022] [Indexed: 01/27/2023] Open
Abstract
To date, there is no overarching proposition for the ontogenetic-neurobiological basis of self-regulation. This paper suggests that the balanced self-regulatory reaction of the fetus, newborn and infant is based on a complex mechanism starting from early brainstem development and continuing to progressive control of the cortex over the brainstem. It is suggested that this balance occurs through the synchronous reactivity between the sympathetic and parasympathetic systems, both which originate from the brainstem. The paper presents an evidence-based approach in which molecular excitation-inhibition balance, interchanges between excitatory and inhibitory roles of neurotransmitters as well as cardiovascular and white matter development across gestational ages, are shown to create sympathetic-parasympathetic synchrony, including the postnatal development of electroencephalogram waves and vagal tone. These occur in developmental milestones detectable in the same time windows (sensitive periods of development) within a convergent systematic progress. This ontogenetic stepwise process is termed "the self-regulation clock" and suggest that this clock is located in the largest connection between the brainstem and the cortex, the corticospinal tract. This novel evidence-based new theory paves the way towards more accurate hypotheses and complex studies of self-regulation and its biological basis, as well as pointing to time windows for interventions in preterm infants. The paper also describes the developing indirect signaling between the suprachiasmatic nucleus and the corticospinal tract. Finally, the paper proposes novel hypotheses for molecular, structural and functional investigation of the "clock" circuitry, including its associations with other biological clocks. This complex circuitry is suggested to be responsible for the developing self-regulatory functions and their neurobehavioral correlates.
Collapse
Affiliation(s)
- Sari Goldstein Ferber
- Department of Psychology, Bar Ilan University, Ramat Gan 5290002, Israel; (A.W.); (R.G.)
- The Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel;
| | - Aron Weller
- Department of Psychology, Bar Ilan University, Ramat Gan 5290002, Israel; (A.W.); (R.G.)
- The Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel;
| | - Michal Ben-Shachar
- The Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel;
| | - Gil Klinger
- Department of Neonatology, Schneider Children’s Medical Center, Sackler Medical School, Tel Aviv University, Petach Tikvah 4920235, Israel;
| | - Ronny Geva
- Department of Psychology, Bar Ilan University, Ramat Gan 5290002, Israel; (A.W.); (R.G.)
- The Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel;
| |
Collapse
|
17
|
Kiryachkov YY, Bosenko SA, Muslimov BG, Petrova MV. Dysfunction of the Autonomic Nervous System and its Role in the Pathogenesis of Septic Critical Illness (Review). Sovrem Tekhnologii Med 2021; 12:106-116. [PMID: 34795998 PMCID: PMC8596275 DOI: 10.17691/stm2020.12.4.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Indexed: 12/05/2022] Open
Abstract
Dysfunction of the autonomic nervous system (ANS) of the brain in sepsis can cause severe systemic inflammation and even death. Numerous data confirmed the role of ANS dysfunction in the occurrence, course, and outcome of systemic sepsis. The parasympathetic part of the ANS modifies the inflammation through cholinergic receptors of internal organs, macrophages, and lymphocytes (the cholinergic anti-inflammatory pathway). The sympathetic part of ANS controls the activity of macrophages and lymphocytes by influencing β2-adrenergic receptors, causing the activation of intracellular genes encoding the synthesis of cytokines (anti-inflammatory beta2-adrenergic receptor interleukin-10 pathway, β2AR–IL-10). The interaction of ANS with infectious agents and the immune system ensures the maintenance of homeostasis or the appearance of a critical generalized infection. During inflammation, the ANS participates in the inflammatory response by releasing sympathetic or parasympathetic neurotransmitters and neuropeptides. It is extremely important to determine the functional state of the ANS in critical conditions, since both cholinergic and sympathomimetic agents can act as either anti- or pro-inflammatory stimuli.
Collapse
Affiliation(s)
- Y Y Kiryachkov
- Head of the Department of Surgical and Resuscitation Technologies; Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25, Bldg 2, Petrovka St., Moscow, 107031, Russia
| | - S A Bosenko
- Anesthesiologist; Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25, Bldg 2, Petrovka St., Moscow, 107031, Russia
| | - B G Muslimov
- Deputy Chief Physician for Anesthesiology and Intensive Care; Konchalovsky Central City Hospital, 2, Bldg 1, Kashtanovaya Alley, Zelenograd, Moscow, 124489, Russia
| | - M V Petrova
- Professor, Deputy Director Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25, Bldg 2, Petrovka St., Moscow, 107031, Russia
| |
Collapse
|
18
|
Rangon CM, Barruet R, Mazouni A, Le Cossec C, Thevenin S, Guillaume J, Léguillier T, Huysman F, Luis D. Auricular Neuromodulation for Mass Vagus Nerve Stimulation: Insights From SOS COVID-19 a Multicentric, Randomized, Controlled, Double-Blind French Pilot Study. Front Physiol 2021; 12:704599. [PMID: 34408665 PMCID: PMC8365750 DOI: 10.3389/fphys.2021.704599] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022] Open
Abstract
Importance: An exacerbated inflammatory response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is believed to be one of the major causes of the morbidity and mortality of the coronavirus disease 2019 (COVID-19). Neuromodulation therapy, based on vagus nerve stimulation, was recently hypothesized to control both the SARS-CoV-2 replication and the ensuing inflammation likely through the inhibition of the nuclear factor kappa-light-chain-enhancer of activated B cells pathway and could improve the clinical outcomes as an adjunct treatment. We proposed to test it by the stimulation of the auricular branch of the vagus nerve, i.e., auricular neuromodulation (AN), a non-invasive procedure through the insertion of semipermanent needles on the ears. Objective: The aim of this study was to assess the effect of AN on the clinical outcomes in patients affected by COVID-19. Design, Setting, and Participants: A multicenter, randomized, placebo-controlled, double-blind clinical trial included 31 patients with respiratory failure due to COVID-19 requiring hospitalization. Within 72 h after admission, patients received either AN (n = 14) or sham neuromodulation (SN, n = 15) in addition to the conventional treatments. Main Outcome and Measures: The primary endpoint of the study was the rate of a clinical benefit conferred by AN at Day 14 (D14) as assessed by a 7-point Clinical Progression Scale. The secondary endpoint of the study was the impact of AN on the rate of transfer to the intensive care unit (ICU) and on the survival rate at D14. Results: The AN procedure was well-tolerated without any reported side effects but with no significant improvement for the measures of both primary (p > 0.3) and secondary (p > 0.05) endpoints at the interim analysis. None of the AN-treated patients died but one in the SN group did (81 years). Two AN-treated patients (73 and 79 years, respectively) and one SN-treated patient (59 years) were transferred to ICU. Remarkably, AN-treated patients were older with more representation by males than in the SN arm (i.e., the median age of 75 vs. 65 years, 79% male vs. 47%). Conclusion: The AN procedure, which was used within 72 h after the admission of patients with COVID-19, was safe and could be successfully implemented during the first two waves of COVID-19 in France. Nevertheless, AN did not significantly improve the outcome of the patients in our small preliminary study. It is pertinent to explore further to validate AN as the non-invasive mass vagal stimulation solution for the forthcoming pandemics. Clinical Trial Registration: [https://clinicaltrials.gov/], identifier [NCT04341415].
Collapse
Affiliation(s)
- Claire-Marie Rangon
- Pain and Neuromodulation Unit, Neurosurgery Department, Hôpital Fondation Adolphe de Rothschild, Paris, France
| | - Régine Barruet
- Infectious Diseases Department, Centre Hospitalier Simone Veil, Beauvais, France
| | | | - Chloé Le Cossec
- Clinical Research Department, Hôpital Fondation Adolphe de Rothschild, Paris, France
| | - Sophie Thevenin
- Clinical Research Department, Hôpital Fondation Adolphe de Rothschild, Paris, France
| | - Jessica Guillaume
- Clinical Research Department, Hôpital Fondation Adolphe de Rothschild, Paris, France
| | - Teddy Léguillier
- Clinical Research Department, Hôpital Fondation Adolphe de Rothschild, Paris, France
| | - Fabienne Huysman
- Clinical Research Department, Centre Hospitalier Simone Veil, Beauvais, France
| | - David Luis
- Clinical Research Department, Centre Hospitalier Simone Veil, Beauvais, France.,Intensive Care Unit, Centre Hospitalier Simone Veil, Beauvais, France
| |
Collapse
|
19
|
De Ridder D, Adhia D, Vanneste S. The anatomy of pain and suffering in the brain and its clinical implications. Neurosci Biobehav Rev 2021; 130:125-146. [PMID: 34411559 DOI: 10.1016/j.neubiorev.2021.08.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/09/2021] [Accepted: 08/13/2021] [Indexed: 02/08/2023]
Abstract
Pain is an unpleasant sensory and emotional experience associated with actual or potential tissue damage. Chronic pain, with a prevalence of 20-30 % is the major cause of human suffering worldwide, because effective, specific and safe therapies have yet to be developed. It is unevenly distributed among sexes, with women experiencing more pain and suffering. Chronic pain can be anatomically and phenomenologically dissected into three separable but interacting pathways, a lateral 'painfulness' pathway, a medial 'suffering' pathway and a descending pain inhibitory pathway. One may have pain(fullness) without suffering and suffering without pain(fullness). Pain sensation leads to suffering via a cognitive, emotional and autonomic processing, and is expressed as anger, fear, frustration, anxiety and depression. The medial pathway overlaps with the salience and stress networks, explaining that behavioural relevance or meaning determines the suffering associated with painfulness. Genetic and epigenetic influences trigger chronic neuroinflammatory changes which are involved in transitioning from acute to chronic pain. Based on the concept of the Bayesian brain, pain (and suffering) can be regarded as the consequence of an imbalance between the two ascending and the descending pain inhibitory pathways under control of the reward system. The therapeutic clinical implications of this simple pain model are obvious. After categorizing the working mechanisms of each of the available treatments (pain killers, psychopharmacology, psychotherapy, neuromodulation, psychosurgery, spinal cord stimulation) to 1 or more of the 3 pathways, a rational combination can be proposed of activating the descending pain inhibitory pathway in combination with inhibition of the medial and lateral pathway, so as to rebalance the pain (and suffering) pathways.
Collapse
Affiliation(s)
- Dirk De Ridder
- Section of Neurosurgery, Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.
| | - Divya Adhia
- Section of Neurosurgery, Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Sven Vanneste
- Global Brain Health Institute, Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
20
|
Cao J, Zhang Y, Li H, Yan Z, Liu X, Hou X, Chen W, Hodges S, Kong J, Liu B. Different modulation effects of 1 Hz and 20 Hz transcutaneous auricular vagus nerve stimulation on the functional connectivity of the periaqueductal gray in patients with migraine. J Transl Med 2021; 19:354. [PMID: 34404427 PMCID: PMC8371886 DOI: 10.1186/s12967-021-03024-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/05/2021] [Indexed: 01/02/2023] Open
Abstract
Background A growing body of evidence suggests that transcutaneous auricular vagus nerve stimulation (taVNS) may relieve symptoms of migraineurs. Frequency is one of the key stimulation parameters. The aim of this study is to investigate the modulation effect of taVNS frequency on the descending pain modulation system (DPMS) in patients with migraine. Methods Twenty-four episodic migraineurs without aura (21 females) were recruited for the single-blind, crossover, functional magnetic resonance imaging (fMRI) study. Each participant attended two separate fMRI scan sessions, one for 1 Hz and another for 20 Hz taVNS, in a random order. Seed-based functional connectivity analysis was applied using the ventrolateral periaqueductal gray (PAG) as the region of interest. Results Compared with the pre-taVNS resting state, continuous 1 Hz taVNS (during) produced a significant increase in functional connectivity between the PAG and the bilateral middle cingulate cortex (MCC), right precuneus, left middle frontal gyrus (MFG), and left cuneus. Compared with 20 Hz taVNS, 1 Hz taVNS produced greater PAG connectivity increases with the MCC, right precuneus/posterior cingulate cortex, left insula, and anterior cingulate cortex (ACC). A significant negative correlation was observed between the number of migraine attacks in the previous 4 weeks and the PAG-MCC functional connectivity in the pre-taVNS resting-state before 1 Hz taVNS. Conclusions Our findings suggest that taVNS with different frequencies may produce different modulation effects on the descending pain modulation system, demonstrating the important role of stimulation frequency in taVNS treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03024-9.
Collapse
Affiliation(s)
- Jin Cao
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yue Zhang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hui Li
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhaoxian Yan
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xian Liu
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaoyan Hou
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weicui Chen
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Sierra Hodges
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Jian Kong
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Bo Liu
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
21
|
Long-term endogenous acetylcholine deficiency potentiates pulmonary inflammation in a murine model of elastase-induced emphysema. Sci Rep 2021; 11:15918. [PMID: 34354132 PMCID: PMC8342425 DOI: 10.1038/s41598-021-95211-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
Acetylcholine (ACh), the neurotransmitter of the cholinergic system, regulates inflammation in several diseases including pulmonary diseases. ACh is also involved in a non-neuronal mechanism that modulates the innate immune response. Because inflammation and release of pro-inflammatory cytokines are involved in pulmonary emphysema, we hypothesized that vesicular acetylcholine transport protein (VAChT) deficiency, which leads to reduction in ACh release, can modulate lung inflammation in an experimental model of emphysema. Mice with genetical reduced expression of VAChT (VAChT KDHOM 70%) and wild-type mice (WT) received nasal instillation of 50 uL of porcine pancreatic elastase (PPE) or saline on day 0. Twenty-eight days after, animals were evaluated. Elastase instilled VAChT KDHOM mice presented an increase in macrophages, lymphocytes, and neutrophils in bronchoalveolar lavage fluid and MAC2-positive macrophages in lung tissue and peribronchovascular area that was comparable to that observed in WT mice. Conversely, elastase instilled VAChT KDHOM mice showed significantly larger number of NF-κB-positive cells and isoprostane staining in the peribronchovascular area when compared to elastase-instilled WT-mice. Moreover, elastase-instilled VAChT-deficient mice showed increased MCP-1 levels in the lungs. Other cytokines, extracellular matrix remodeling, alveolar enlargement, and lung function were not worse in elastase-instilled VAChT deficiency than in elastase-instilled WT-controls. These data suggest that decreased VAChT expression may contribute to the pathogenesis of emphysema, at least in part, through NF-κB activation, MCP-1, and oxidative stress pathways. This study highlights novel pathways involved in lung inflammation that may contribute to the development of chronic obstrutive lung disease (COPD) in cholinergic deficient individuals such as Alzheimer's disease patients.
Collapse
|
22
|
Li G, LeiQian, Gu P, Fan D. Dexmedetomidine post-conditioning attenuates cerebral ischemia following asphyxia cardiac arrest through down-regulation of apoptosis and neuroinflammation in rats. BMC Anesthesiol 2021; 21:180. [PMID: 34182937 PMCID: PMC8236741 DOI: 10.1186/s12871-021-01394-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 06/10/2021] [Indexed: 12/17/2022] Open
Abstract
Background Neuroprotection strategies after cardiac arrest (CA)/cardiopulmonary resuscitation (CPR) remain key areas of basic and clinical research. This study was designed to investigate the neuroprotective effects of dexmedetomidine following resuscitation and potential mechanisms. Methods Anesthetized rats underwent 6-min asphyxia-based cardiac arrest and resuscitation, after which the experimental group received a single intravenous dose of dexmedetomidine (25 μg/kg). Neurological outcomes and ataxia were assessed after the return of spontaneous circulation. The serum levels and brain expression of inflammation markers was examined, and apoptotic cells were quantified by TUNEL staining. Results Neuroprotection was enhanced by dexmedetomidine post-conditioning after the return of spontaneous circulation. This enhancement was characterized by the promotion of neurological function scores and coordination. In addition, dexmedetomidine post-conditioning attenuated the serum levels of the pro-inflammatory cytokine tumor necrosis factor (TNF)-α at 2 h, as well as interleukin IL-1β at 2, 24, and 48 h. TUNEL staining showed that the number of apoptotic cells in the dexmedetomidine post-conditioning group was significantly reduced compared with the control group. Further western blot analysis indicated that dexmedetomidine markedly reduced the levels of caspase-3 and nuclear factor-kappa B (NF-κB) in the brain. Conclusions Dexmedetomidine post-conditioning had a neuroprotective effect against cerebral injury following asphyxia-induced cardiac arrest. The mechanism was associated with the downregulation of apoptosis and neuroinflammation.
Collapse
Affiliation(s)
- Guangqian Li
- School of Medicine, Universityof Electronic Science and Technology of China, Chengdu, China
| | - LeiQian
- School of Medicine, Universityof Electronic Science and Technology of China, Chengdu, China
| | - Pan Gu
- School of Medicine, Universityof Electronic Science and Technology of China, Chengdu, China
| | - Dan Fan
- School of Medicine, Universityof Electronic Science and Technology of China, Chengdu, China. .,Department of Anesthesiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, No. 32 West Second Section, First RingRoad, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
23
|
Passeron T, Zouboulis CC, Tan J, Andersen ML, Katta R, Lyu X, Aguilar L, Kerob D, Morita A, Krutmann J, Peters EMJ. Adult skin acute stress responses to short-term environmental and internal aggression from exposome factors. J Eur Acad Dermatol Venereol 2021; 35:1963-1975. [PMID: 34077579 PMCID: PMC8519049 DOI: 10.1111/jdv.17432] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/18/2021] [Indexed: 12/02/2022]
Abstract
Exposome factors that lead to stressed skin can be defined as any disturbance to homeostasis from environmental (meteorological factors, solar radiation, pollution or tobacco smoke) and/or internal exposure (unhealthy diet, hormonal variations, lack of sleep, psychosocial stress). The clinical and biological impact of chronic exposome effects on skin functions has been extensively reviewed, whereas there is a paucity of information on the impact of short‐term acute exposure. Acute stress, which would typically last minutes to hours (and generally no more than a week), provokes a transient but robust neuroendocrine‐immune and tissue remodelling response in the skin and can alter the skin barrier. Firstly, we provide an overview of the biological effects of various acute stressors on six key skin functions, namely the skin physical barrier, pigmentation, defences (antioxidant, immune cell‐mediated, microbial and microbiome maintenance), structure (extracellular matrix and appendages), neuroendocrine and thermoregulation functions. Secondly, we describe the biological and clinical effects on adult skin from individual exposome factors that elicit an acute stress response and their consequences in skin health maintenance. Clinical manifestations of acutely stressed skin may include dry skin that might accentuate fine lines, oily skin, sensitive skin, pruritus, erythema, pale skin, sweating, oedema and flares of inflammatory skin conditions such as acne, rosacea, atopic dermatitis, pigmentation disorders and skin superinfection such as viral reactivation. Acute stresses can also induce scalp sensitivity, telogen effluvium and worsen alopecia.
Collapse
Affiliation(s)
- T Passeron
- Department of Dermatology, University Hospital Centre Nice, Côte d'Azur University, Nice, France.,INSERM U1065, team 12, C3M, Nice, France
| | - C C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Brandenburg Medical School Theodor Fontane and Faculty of Health Sciences Brandenburg, Dessau, Germany
| | - J Tan
- Windsor Clinical Research Inc., Windsor, ON, Canada.,Department of Medicine, University of Western Ontario, London, Canada
| | - M L Andersen
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP)/Escola Paulista de Medicina, São Paulo, Brazil
| | - R Katta
- Volunteer Clinical Faculty, Baylor College of Medicine, Houston, Texas, USA.,McGovern Medical School at UT Health, Houston, Texas, USA
| | - X Lyu
- Department of Dermatology, Anzhen Hospital, Capital Medical University, Beijing, China
| | - L Aguilar
- L'Oréal Advanced Research, Aulnay-sous-bois, France
| | - D Kerob
- Laboratoires Vichy, Levallois Perret, France
| | - A Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - J Krutmann
- IUF Leibniz Research Institute for Environmental Medicine, Dusseldorf, Germany.,Medical faculty, Heinrich-Heine-University, Dusseldorf, Germany
| | - E M J Peters
- Department of Psychosomatic Medicine and Psychotherapy, Justus-Liebig University, Gießen, Germany.,Charité Center 12 (CC12) for Internal Medicine and Dermatology, Berlin, Germany
| |
Collapse
|
24
|
Charles EJ, Tian Y, Zhang A, Wu D, Mehaffey JH, Gigliotti JC, Klibanov AL, Kron IL, Yang Z. Pulsed ultrasound attenuates the hyperglycemic exacerbation of myocardial ischemia-reperfusion injury. J Thorac Cardiovasc Surg 2021; 161:e297-e306. [PMID: 31839230 PMCID: PMC7195241 DOI: 10.1016/j.jtcvs.2019.10.096] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Acute hyperglycemia during myocardial infarction worsens outcomes in part by inflammatory mechanisms. Pulsed ultrasound has anti-inflammatory potential in bone healing and neuromodulation. We hypothesized that pulsed ultrasound would attenuate the hyperglycemic exacerbation of myocardial ischemia-reperfusion injury via the cholinergic anti-inflammatory pathway. METHODS Acute hyperglycemia was induced in wild-type C57BL6 or acetylcholine-receptor knockout (α7nAChR-/-) mice by intraperitoneal injection of glucose. Pulsed ultrasound (frequency 7 MHz, bursting mechanical index 1.2, duration 1 second, repeated every 6 seconds for 2 minutes, 20-second total exposure) was performed at the spleen or neck after glucose injection. Separate mice underwent vagotomy before treatment. The left coronary artery was occluded for 20 minutes, followed by 60 minutes of reperfusion. The primary end point was infarct size in explanted hearts. RESULTS Splenic pulsed ultrasound significantly decreased infarct size in wild-type C57BL6 mice exposed to acute hyperglycemia and myocardial ischemia-reperfusion injury (5.2% ± 4.4% vs 16.9% ± 12.5% of risk region, P = .013). Knockout of α7nAChR abrogated the beneficial effect of splenic pulsed ultrasound (22.2% ± 12.1%, P = .79 vs control). Neck pulsed ultrasound attenuated the hyperglycemic exacerbation of myocardial infarct size (3.5% ± 4.8%, P = .004 vs control); however, the cardioprotective effect disappeared in mice that underwent vagotomy. Plasma acetylcholine, β2 adrenergic receptor, and phosphorylated Akt levels were increased after splenic pulsed ultrasound treatment. CONCLUSIONS Pulsed ultrasound treatment of the spleen or neck attenuated the hyperglycemic exacerbation of myocardial ischemia-reperfusion injury leading to a 3-fold decrease in infarct size. Pulsed ultrasound may provide cardioprotection via the cholinergic anti-inflammatory pathway and could be a promising new nonpharmacologic, noninvasive therapy to reduce infarct size during acute myocardial infarction and improve patient outcomes.
Collapse
Affiliation(s)
- Eric J Charles
- Department of Surgery, University of Virginia, Charlottesville, Va
| | - Yikui Tian
- Department of Surgery, University of Virginia, Charlottesville, Va; Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Aimee Zhang
- Department of Surgery, University of Virginia, Charlottesville, Va
| | - Di Wu
- Department of Surgery, University of Virginia, Charlottesville, Va
| | | | - Joseph C Gigliotti
- Department of Integrative Physiology and Pharmacology, Liberty University, Lynchburg, Va
| | | | - Irving L Kron
- Department of Surgery, University of Virginia, Charlottesville, Va; Department of Surgery, University of Arizona, Tucson, Ariz
| | - Zequan Yang
- Department of Surgery, University of Virginia, Charlottesville, Va.
| |
Collapse
|
25
|
Ertle CM, Rommel FR, Tumala S, Moriwaki Y, Klein J, Kruse J, Gieler U, Peters EMJ. New Pathways for the Skin's Stress Response: The Cholinergic Neuropeptide SLURP-1 Can Activate Mast Cells and Alter Cytokine Production in Mice. Front Immunol 2021; 12:631881. [PMID: 33815383 PMCID: PMC8012551 DOI: 10.3389/fimmu.2021.631881] [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: 11/21/2020] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Background: The alpha7 nicotinic acetylcholine receptor (Chrna7) plays an essential anti-inflammatory role in immune homeostasis and was recently found on mast cells (MC). Psychosocial stress can trigger MC hyperactivation and increases pro-inflammatory cytokines in target tissues such as the skin. If the cholinergic system (CS) and Chrna7 ligands play a role in these cascades is largely unknown. Objective: To elucidate the role of the CS in the response to psychosocial stress using a mouse-model for stress-triggered cutaneous inflammatory circuits. Methods: Key CS markers (ACh, Ch, SLURP-1, SLURP-2, Lynx1, Chrm3, Chrna7, Chrna9, ChAT, VAChT, Oct3, AChE, and BChE) in skin and its MC (sMC), MC activation, immune parameters (TNFα, IL1β, IL10, TGFβ, HIF1α, and STAT3) and oxidative stress were analyzed in skin from 24 h noise-stressed mice and in cultured MC (cMC) from C57BL/6 or Chrna7-Knockout mice. Results: First, Chrna7 and SLURP-1 mRNA were exclusively upregulated in stressed skin. Second, histomorphometry located Chrna7 and SLURP-1 in nerves and sMC and demonstrated upregulated contacts and increased Chrna7+ sMC in stressed skin, while 5 ng/mL SLURP-1 degranulated cMC. Third, IL1β+ sMC were high in stressed skin, and while SLURP-1 alone had no significant effect on cMC cytokines, it upregulated IL1β in cMC from Chrna7-KO and in IL1β-treated wildtype cMC. In addition, HIF1α+ sMC were high in stressed skin and Chrna7-agonist AR-R 17779 induced ROS in cMC while SLURP-1 upregulated TNFα and IL1β in cMC when HIF1α was blocked. Conclusions: These data infer that the CS plays a role in the regulation of stress-sensitive inflammatory responses but may have a surprising pro-inflammatory effect in healthy skin, driving IL1β expression if SLURP-1 is involved.
Collapse
Affiliation(s)
- Christoph M Ertle
- Psychoneuroimmunology Laboratory, Clinic for Psychosomatic Medicine and Psychotherapy, Justus-Liebig-University Giessen, Giessen, Germany
| | - Frank R Rommel
- Psychoneuroimmunology Laboratory, Clinic for Psychosomatic Medicine and Psychotherapy, Justus-Liebig-University Giessen, Giessen, Germany
| | - Susanne Tumala
- Psychoneuroimmunology Laboratory, Clinic for Psychosomatic Medicine and Psychotherapy, Justus-Liebig-University Giessen, Giessen, Germany
| | - Yasuhiro Moriwaki
- Department of Pharmacology, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Jochen Klein
- Department of Pharmacology, Biocenter N260, Goethe University Frankfurt, Frankfurt, Germany
| | - Johannes Kruse
- Clinic for Psychosomatic Medicine and Psychotherapy, Justus-Liebig-University Giessen, Giessen, Germany.,Clinic for Psychosomatic Medicine and Psychotherapy, Philipps University of Marburg, Marburg, Germany
| | - Uwe Gieler
- Department of Dermatology, University Hospital Giessen, Giessen, Germany
| | - Eva M J Peters
- Psychoneuroimmunology Laboratory, Clinic for Psychosomatic Medicine and Psychotherapy, Justus-Liebig-University Giessen, Giessen, Germany.,Charité Center 12 for Internal Medicine and Dermatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
26
|
Ruberti OM, Rodrigues B. Estrogen Deprivation and Myocardial Infarction: Role of Aerobic Exercise Training, Inflammation and Metabolomics. Curr Cardiol Rev 2021; 16:292-305. [PMID: 31362678 PMCID: PMC7903506 DOI: 10.2174/1573403x15666190729153026] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/01/2019] [Accepted: 07/09/2019] [Indexed: 12/15/2022] Open
Abstract
In general, postmenopausal women present higher mortality, and worse prognosis after myocardial infarction (MI) compared to men, due to estrogen deficiency. After MI, cardiovascular alterations occur such as the autonomic imbalance and the pro-inflammatory cytokines increase. In this sense, therapies that aim to minimize deleterious effects caused by myocardial ischemia are important. Aerobic training has been proposed as a promising intervention in the prevention of cardiovascular diseases. On the other hand, some studies have attempted to identify potential biomarkers for cardiovascular diseases or specifically for MI. For this purpose, metabolomics has been used as a tool in the discovery of cardiovascular biomarkers. Therefore, the objective of this work is to discuss the changes involved in ovariectomy, myocardial infarction, and aerobic training, with emphasis on inflammation and metabolism.
Collapse
Affiliation(s)
- Olívia M Ruberti
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Bruno Rodrigues
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| |
Collapse
|
27
|
D’Haese PF, Finomore V, Lesnik D, Kornhauser L, Schaefer T, Konrad PE, Hodder S, Marsh C, Rezai AR. Prediction of viral symptoms using wearable technology and artificial intelligence: A pilot study in healthcare workers. PLoS One 2021; 16:e0257997. [PMID: 34648513 PMCID: PMC8516235 DOI: 10.1371/journal.pone.0257997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 09/15/2021] [Indexed: 01/12/2023] Open
Abstract
Conventional testing and diagnostic methods for infections like SARS-CoV-2 have limitations for population health management and public policy. We hypothesize that daily changes in autonomic activity, measured through off-the-shelf technologies together with app-based cognitive assessments, may be used to forecast the onset of symptoms consistent with a viral illness. We describe our strategy using an AI model that can predict, with 82% accuracy (negative predictive value 97%, specificity 83%, sensitivity 79%, precision 34%), the likelihood of developing symptoms consistent with a viral infection three days before symptom onset. The model correctly predicts, almost all of the time (97%), individuals who will not develop viral-like illness symptoms in the next three days. Conversely, the model correctly predicts as positive 34% of the time, individuals who will develop viral-like illness symptoms in the next three days. This model uses a conservative framework, warning potentially pre-symptomatic individuals to socially isolate while minimizing warnings to individuals with a low likelihood of developing viral-like symptoms in the next three days. To our knowledge, this is the first study using wearables and apps with machine learning to predict the occurrence of viral illness-like symptoms. The demonstrated approach to forecasting the onset of viral illness-like symptoms offers a novel, digital decision-making tool for public health safety by potentially limiting viral transmission.
Collapse
Affiliation(s)
- Pierre-François D’Haese
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown, West Virginia, United States of America
- Health Sciences Center, West Virginia University, Morgantown, West Virginia, United States of America
- * E-mail:
| | - Victor Finomore
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown, West Virginia, United States of America
- Health Sciences Center, West Virginia University, Morgantown, West Virginia, United States of America
| | - Dmitry Lesnik
- Stratyfy, Inc, New York, New York, United States of America
| | | | | | - Peter E. Konrad
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown, West Virginia, United States of America
- Health Sciences Center, West Virginia University, Morgantown, West Virginia, United States of America
| | - Sally Hodder
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown, West Virginia, United States of America
- Health Sciences Center, West Virginia University, Morgantown, West Virginia, United States of America
| | - Clay Marsh
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown, West Virginia, United States of America
- Health Sciences Center, West Virginia University, Morgantown, West Virginia, United States of America
| | - Ali R. Rezai
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown, West Virginia, United States of America
- Health Sciences Center, West Virginia University, Morgantown, West Virginia, United States of America
| |
Collapse
|
28
|
Martins CC, Bagatini MD, Simões JLB, Cardoso AM, Baldissarelli J, Dalenogare DP, Dos Santos DL, Schetinger MRC, Morsch VM. Increased oxidative stress and inflammatory markers contrasting with the activation of the cholinergic anti-inflammatory pathway in patients with metabolic syndrome. Clin Biochem 2020; 89:63-69. [PMID: 33333061 DOI: 10.1016/j.clinbiochem.2020.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Metabolic syndrome (MetS) is a disorder that is closely associated with risk factors that increase the chance of atherosclerosis and cardiovascular diseases. We demonstrate the presence of inflammation and oxidative stress in patients with MetS through levels of antioxidants and oxidative and inflammatory markers, in order to determine influential variables in therapy. METHODS In this study, lipid peroxidation, carbonylated protein content and enzymatic and non-enzymatic antioxidants were evaluated in samples obtained from 30 patients with MetS and 30 control patients. In addition, acetylcholinesterase (AChE) activity, C-reactive protein (CRP) and uric acid (UA) levels were determined to investigate the inflammatory process in patients with MetS. RESULTS Our results demonstrated an increase in the levels of oxidative markers, such as substances reactive to thiobarbituric acid (TBARS) and carbonyl protein. In addition, a decrease in the defense of non-enzymatic antioxidants, such as levels of vitamin C and glutathione (GSH) in patients with MetS. As for inflammatory markers, CRP and UA were increased in patients with MetS. Finally, activation of the cholinergic anti-inflammatory pathway was observed due to decreased AchE activity in patients with MetS. CONCLUSION The analyzes indicated oxidative stress, together with a reduction in the levels of antioxidant enzymes, corroborating the high consumption of these proteins. In addition, inflammation and activation of the cholinergic anti-inflammatory pathway was observed by the AChE analysis. Thus, the activation of this pathway can be studied as a possible route to a potential therapy. In addition, the markers AChE, CRP and UA may be used as a focus for the treatment of MetS.
Collapse
Affiliation(s)
- Caroline Curry Martins
- Post-Graduation Program in Biological Sciences: Toxicological Biochemistry, Center of Natural and Exact Sciences of the Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Margarete Dulce Bagatini
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| | | | - Andreia Machado Cardoso
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Jucimara Baldissarelli
- Post-Graduation Program in Biological Sciences: Toxicological Biochemistry, Center of Natural and Exact Sciences of the Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Diéssica Padilha Dalenogare
- Post-Graduation Program in Biological Sciences: Toxicological Biochemistry, Center of Natural and Exact Sciences of the Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Daniela Lopes Dos Santos
- Physical Activity Group, Physical Education Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Maria Rosa Chitolina Schetinger
- Post-Graduation Program in Biological Sciences: Toxicological Biochemistry, Center of Natural and Exact Sciences of the Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Vera Maria Morsch
- Post-Graduation Program in Biological Sciences: Toxicological Biochemistry, Center of Natural and Exact Sciences of the Federal University of Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
29
|
Bursi F, Santangelo G, Sansalone D, Valli F, Vella AM, Toriello F, Barbieri A, Carugo S. Prognostic utility of quantitative offline 2D-echocardiography in hospitalized patients with COVID-19 disease. Echocardiography 2020; 37:2029-2039. [PMID: 32964483 PMCID: PMC7646664 DOI: 10.1111/echo.14869] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/04/2020] [Accepted: 09/05/2020] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To assess the prognostic utility of quantitative 2D-echocardiography, including strain, in patients with COVID-19 disease. METHODS COVID-19-infected patients admitted to the San Paolo University Hospital of Milan that underwent a clinically indicated echocardiographic examination were included in the study. To limit contamination, all measurements were performed offline. Quantitative measurements were obtained by an operator blinded to the clinical data. RESULTS Among the 49 patients, nonsurvivors (33%) had worse respiratory parameters, index of multiorgan failure, and worse markers of lung involvement. Right ventricular (RV) dysfunction (as assessed by conventional and 2-dimensional speckle tracking) was a common finding and a powerful independent predictor of mortality. At the ROC curve analyses, RV free wall longitudinal strain (LS) showed an AUC 0.77 ± 0.08 in predicting death, P = .008, and global RV LS (RV-GLS) showed an AUC 0.79 ± 0.04, P = .004. This association remained significant after correction for age (OR = 1.16, 95%CI 1.01-1.34, P = .029 for RV free wall LS and OR = 1.20, 95%CI 1.01-1.42, P = .033 for RV-GLS), for oxygen partial pressure at arterial gas analysis/fraction of inspired oxygen (OR = 1.28, 95%CI 1.04-1.57, P = .021 for RV free wall-LS and OR = 1.30, 95%CI 1.04-1.62, P = .020 for RV-GLS) and for the severity of pulmonary involvement measured by a computed tomography lung score (OR = 1.27, 95%CI 1.02-1.19, P = .034 for RV free wall LS and OR = 1.30, 95%CI 1.04-1.63, P = .022 for RV-GLS). CONCLUSIONS In patients hospitalized with COVID-19, offline quantitative 2D-echocardiographic assessment of cardiac function is feasible. Parameters of RV function are frequently abnormal and have an independent prognostic value over markers of lung involvement.
Collapse
Affiliation(s)
- Francesca Bursi
- Division of Cardiology, Heart and Lung DepartmentSan Paolo HospitalASST Santi Paolo and CarloUniversity of MilanMilanItaly
| | - Gloria Santangelo
- Division of Cardiology, Heart and Lung DepartmentSan Paolo HospitalASST Santi Paolo and CarloUniversity of MilanMilanItaly
| | - Dario Sansalone
- Division of Cardiology, Heart and Lung DepartmentSan Paolo HospitalASST Santi Paolo and CarloUniversity of MilanMilanItaly
| | - Federica Valli
- Division of Cardiology, Heart and Lung DepartmentSan Paolo HospitalASST Santi Paolo and CarloUniversity of MilanMilanItaly
| | - Anna Maria Vella
- Division of Cardiology, Heart and Lung DepartmentSan Paolo HospitalASST Santi Paolo and CarloUniversity of MilanMilanItaly
| | - Filippo Toriello
- Division of Cardiology, Heart and Lung DepartmentSan Paolo HospitalASST Santi Paolo and CarloUniversity of MilanMilanItaly
| | - Andrea Barbieri
- Division of CardiologyDepartment of Diagnostics, Clinical and Public Health MedicinePoliclinico University Hospital of ModenaUniversity of Modena and Reggio EmiliaModenaItaly
| | - Stefano Carugo
- Division of Cardiology, Heart and Lung DepartmentSan Paolo HospitalASST Santi Paolo and CarloUniversity of MilanMilanItaly
| |
Collapse
|
30
|
Schmidt K, Zivkovic AR, Thiele M, Horter J, Brenner T, Weigand MA, Kleinschmidt S, Hofer S. Point-of-care measured serum cholinesterase activity predicts patient outcome following severe burns. Burns 2020; 47:863-872. [PMID: 33248805 DOI: 10.1016/j.burns.2020.10.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/04/2020] [Accepted: 10/19/2020] [Indexed: 10/23/2022]
Abstract
Risk stratification is of utmost importance in burn therapy. However, suitable bedside biomarkers to evaluate the emerging inflammatory response following burn injuries are missing. Serum cholinesterase (butyrylcholinesterase, BChE) has been shown to be a clinically relevant biomarker in acute inflammatory diseases including burns. In this observational cohort study BChE activity was measured by using point-of-care testing (POCT), a novel method in acute burn care. POCT measurements were performed at emergency room admission (ERA) of 35 patients and repeated 12, 24 and 48 h later. All patients or their legal designees gave informed consent. Patients with burn injuries showed sustained BChE activity reduction following hospital admission. BChE activity correlated negatively with burn injury severity, organ failure severity and intensive care unit resource requirements. BChE activity measured at ERA and 12 h later identified survivors and predicted 28-day patient outcome with noninferior efficacy compared to the abbreviated burn severity index (ABSI) scoring. Finally, POCT-measured BChE activity might complement ABSI scoring and possibly improve early risk stratification in acute burn care therapy.
Collapse
Affiliation(s)
- Karsten Schmidt
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Aleksandar R Zivkovic
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Martin Thiele
- Department of Anaesthesia, Intensive Care Medicine and Pain Therapy, BG Trauma Center Ludwigshafen/Rhine, Ludwig-Guttmann-Str. 13, 67071 Ludwigshafen, Germany.
| | - Johannes Horter
- Department of Hand, Plastic and Reconstructive Surgery-Burn Center, BG Trauma Center Ludwigshafen/Rhine, Ludwig-Guttmann-Str. 13, 67071 Ludwigshafen, Germany.
| | - Thorsten Brenner
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Stefan Kleinschmidt
- Department of Anaesthesia, Intensive Care Medicine and Pain Therapy, BG Trauma Center Ludwigshafen/Rhine, Ludwig-Guttmann-Str. 13, 67071 Ludwigshafen, Germany.
| | - Stefan Hofer
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany; Clinic for Anesthesiology, Intensive Care, Emergency Medicine I and Pain Therapy, Westpfalz Hospital, Hellmut-Hartert-Str. 1, 67655 Kaiserslautern, Germany.
| |
Collapse
|
31
|
Effect of Electroacupuncture at Zusanli (ST36) on Sepsis Induced by Cecal Ligation Puncture and Its Relevance to Spleen. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1914031. [PMID: 33082818 PMCID: PMC7563055 DOI: 10.1155/2020/1914031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/18/2020] [Accepted: 09/23/2020] [Indexed: 12/29/2022]
Abstract
Background Acupuncture at Zusanli (ST36), Quchi (LI11), and Tianshu (ST25) is commonly used in septic patients by traditional Chinese physicians. The protective effect of acupuncture at ST36 on the intestinal barrier is associated with Cholinergic Anti-Inflammatory Pathway (CAIP). However, its detailed mechanism and whether acupuncture at LI11 and ST25 have similar effects to ST36 remain unclear. Aim To explore the effects of electroacupuncture (EA) at ST36, LI11, and ST25 on septic rats and investigate the role of the spleen in the treatment of EA at ST36. Methods A septic rat model caused by cecal ligation and puncture (CLP) and a postsplenectomy (SPX) CLP rat model were established. Rats were divided into nine groups depending on different treatments. Serum levels of TNF-α, IL-10, D-lactic acidosis (D-LA), double amine oxidase (DAO), and T-lymphocyte subgroup level in intestinal lymph nodes were compared. Results EA could not improve the 2-day survival of CLP rats. For CLP rats, EA at ST36 and LI11 significantly decreased the levels of TNF-α, IL-10, DAO, and D-LA in serum and normalized intestinal T-cell immunity. For SPX CLP rats, EA at ST36 failed to reduce serum concentrations of TNF-α, IL-10, and D-LA but increased the values of CD3+CD4+/CD3+CD8+ cells and Treg/Th17 cells. Conclusions EA at ST36 and LI11, respectively, could alleviate inflammation reaction, protect the intestinal barrier, and maintain intestinal T-cell function in septic rats. Spleen participated in the protective effect of EA at ST36 in sepsis.
Collapse
|
32
|
Hutchings C, Phillips JA, Djamgoz MBA. Nerve input to tumours: Pathophysiological consequences of a dynamic relationship. Biochim Biophys Acta Rev Cancer 2020; 1874:188411. [PMID: 32828885 DOI: 10.1016/j.bbcan.2020.188411] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022]
Abstract
It is well known that tumours arising in different organs are innervated and that 'perineural invasion' (cancer cells escaping from the tumour by following the nerve trunk) is a negative prognostic factor. More surprisingly, increasing evidence suggests that the nerves can provide active inputs to tumours and there is two-way communication between nerves and cancer cells within the tumour microenvironment. Cells of the immune system also interact with the nerves and cancer cells. Thus, the nerve connections can exert significant control over cancer progression and modulating these (physically or chemically) can affect significantly the cancer process. Nerve inputs to tumours are derived mainly from the sympathetic (adrenergic) and the parasympathetic (cholinergic) systems, which are interactive. An important component of the latter is the vagus nerve, the largest of the cranial nerves. Here, we present a two-part review of the nerve inputs to tumours and their effects on tumorigenesis. First, we review briefly some relevant general issues including ultrastructural aspects, stemness, interactions between neurones and primary tumours, and communication between neurones and metastasizing tumour cells. Ultrastructural characteristics include synaptic vesicles, tumour microtubes and gap junctions enabling formation of cellular networks. Second, we evaluate the pathophysiology of the nerve input to five major carcinomas: cancers of prostate, stomach, colon, lung and pancreas. For each cancer, we present (i) the nerve inputs normally present in the cancer organ and (ii) how these interact and influence the cancer process. The best clinical evidence for the role of nerves in promoting tumorigenesis comes from prostate cancer patients where metastatic progression has been shown to be suppressed significantly in cases of spinal cord injury. The balance of the sympathetic and parasympathetic contributions to early versus late tumorigenesis varies amongst the different cancers. Different branches of the vagus provide functional inputs to several of the carcinomas and, in two-way interaction with the sympathetic nervous system, affect different stages of the cancer process. Overall, the impact of the vagus nerve can be 'direct' or 'indirect'. Directly, the effect of the vagus is primarily to promote tumorigenesis and this is mediated through cholinergic receptor mechanisms. Indirectly, pro- and anti-tumour effects can occur by stimulation or inhibition of the sympathetic nervous system, respectively. Less well understood are the 'indirect' anti-tumour effect of the vagus nerve via immunomodulation/inflammation, and the role of sensory innervation. A frequent occurrence in the nerve-tumour interactions is the presence of positive feedback driven by agents like nerve growth factor. We conclude that the nerve inputs to tumours can actively and dynamically impact upon cancer progression and are open to clinical exploitation.
Collapse
Affiliation(s)
- Charlotte Hutchings
- Imperial College London, Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK
| | - Jade A Phillips
- Imperial College London, Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK
| | - Mustafa B A Djamgoz
- Imperial College London, Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK; Biotechnology Research Centre, Cyprus International University, Haspolat, Nicosia, TRNC, Mersin 10, Turkey.
| |
Collapse
|
33
|
Zager A. Modulating the immune response with the wake-promoting drug modafinil: A potential therapeutic approach for inflammatory disorders. Brain Behav Immun 2020; 88:878-886. [PMID: 32311496 DOI: 10.1016/j.bbi.2020.04.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/16/2020] [Accepted: 04/15/2020] [Indexed: 01/03/2023] Open
Abstract
Modafinil is a psychostimulant drug approved by the FDA primarily for the treatment of sleep disorders such as narcolepsy, excessive daytime sleepiness and sleep apnea. Several documented but not yet approved uses for modafinil have been described over the last 30 years, including alleviating fatigue in neurological and neurodegenerative disorders. Recent evidence has suggested that modafinil may have an immunomodulatory effect. Here, we review the different effects of modafinil treatment in animal models of brain inflammation and peripheral immune function. We conclude that there is unequivocal evidence of an anti-inflammatory effect of modafinil in experimental animal models of brain inflammation and neurodegenerative disorders, including systemic inflammation and methamphetamine-induced neuroinflammation, Parkinson's disease, brain ischemia, and multiple sclerosis. Modafinil acts on resident glial cells and infiltrating immune cells, negatively affecting both innate and adaptive immune responses in the brain. We also review the outcomes of modafinil treatment on peripheral immune function. The results of studies on this subject are still controversial and far from conclusive, but point to a new avenue of research in relation to peripheral inflammation. The data reviewed here raise the possibility of modafinil being used as adjuvant treatment for neurological disorders in which inflammation plays an important role.
Collapse
Affiliation(s)
- Adriano Zager
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
34
|
Duan J, Wu Y, Liu C, Yang C, Yang L. Deleterious effects of viral pneumonia on cardiovascular system. Eur Heart J 2020; 41:1833-1838. [PMID: 32383765 DOI: 10.1093/eurheartj/ehaa325] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/19/2020] [Accepted: 04/10/2020] [Indexed: 12/22/2022] Open
Abstract
Abstract
Viral pneumonia has a significant effect on the cardiovascular system through various mechanisms; even though it is traditionally regarded as a pulmonary disease characterized by dyspnoea and hypoxaemia. Recent research works have shown that cardiovascular events outweigh all other causes of death in various influenza pandemics. Therefore, the exploration of the effects of viral pneumonia on cardiovascular system becomes increasingly essential. The objective of this review is three-fold: first, to summarize the knowledge about the epidemiological characteristics and clinical manifestations of viral infections that are the recent causes of global pandemics; second, to explore the cardiovascular response to these infections; and third, to attempt in identifying the possible coping strategies of the Wuhan epidemic and the future viral infection pandemics.
Collapse
Affiliation(s)
- Jiahao Duan
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Yeshun Wu
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| |
Collapse
|
35
|
Oshri A, Duprey EB, Liu S, Ehrlich KB. Harsh parenting and youth systemic inflammation: Modulation by the autonomic nervous system. Health Psychol 2020; 39:482-496. [PMID: 32202827 DOI: 10.1037/hea0000852] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The present study aimed to test the role of the autonomic nervous system (ANS) in modulating the impact of family stress induced by harsh parenting on youths' inflammation. First, we examined the direct effect of severity of adverse parenting behaviors on two serum biomarkers of systemic inflammation (C-reactive protein and interleukin-6) among youth. Second, we tested the moderating role of ANS reactivity in response to laboratory-induced stress in the association between harsh parenting and inflammation among these youth. METHOD The sample included 101 low-income children (75.2% African American) between 9 and 12 years of age (Mage = 10.9; SDage = 1.2) who participated in a conflict task with their primary caregiver in a laboratory setting. Heart rate variability reactivity (HRV-R), skin conductance level reactivity (SCL-R), and preejection period reactivity (PEPr-R) were used to index parasympathetic and sympathetic nervous system reactivity. Markers of low-grade inflammation (C-reactive protein and interleukin-6) were obtained from serum. RESULTS After adjusting for confounding variables, ANS activity moderated the associations between family stress and systemic inflammation. Specifically, elevated HRV-R buffered the effect of family stress on youths' inflammation, whereas elevated PEPr-R and SCL-R exacerbated the effect. CONCLUSION These findings show that self-regulatory capacity and threat sensitivity, as indicated by ANS function, may have an impact on the associations between family stress and systemic inflammation. (PsycInfo Database Record (c) 2020 APA, all rights reserved).
Collapse
|
36
|
Wessler I, Kirkpatrick CJ. Cholinergic signaling controls immune functions and promotes homeostasis. Int Immunopharmacol 2020; 83:106345. [PMID: 32203906 DOI: 10.1016/j.intimp.2020.106345] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/06/2020] [Accepted: 02/23/2020] [Indexed: 12/15/2022]
Abstract
Acetylcholine (ACh) was created by nature as one of the first signaling molecules, expressed already in procaryotes. Based on the positively charged nitrogen, ACh could initially mediate signaling in the absence of receptors. When evolution established more and more complex organisms the new emerging organs systems, like the smooth and skeletal muscle systems, energy-generating systems, sexual reproductive system, immune system and the nervous system have further optimized the cholinergic signaling machinery. Thus, it is not surprising that ACh and the cholinergic system are expressed in the vast majority of cells. Consequently, multiple common interfaces exist, for example, between the nervous and the immune system. Research of the last 20 years has unmasked these multiple regulating mechanisms mediated by cholinergic signaling and thus, the biological role of ACh has been revised. The present article summarizes new findings and describes the role of both non-neuronal and neuronal ACh in protecting the organism from external and internal health threats, in providing energy for the whole organism and for the individual cell, controling immune functions to prevent inflammatory dysbalance, and finally, the involvement in critical brain functions, such as learning and memory. All these capacities of ACh enable the organism to attain and maintain homeostasis under changing external conditions. However, the existence of identical interfaces between all these different organ systems complicates the research for new therapeutic interventions, making it essential that every effort should be undertaken to find out more specific targets to modulate cholinergic signaling in different diseases.
Collapse
Affiliation(s)
- Ignaz Wessler
- Institute of Pathology, University Medical Center, Johannes Gutenberg University, D-55101 Mainz, Germany.
| | - Charles James Kirkpatrick
- Institute of Pathology, University Medical Center, Johannes Gutenberg University, D-55101 Mainz, Germany
| |
Collapse
|
37
|
Maturo MG, Soligo M, Gibson G, Manni L, Nardini C. The greater inflammatory pathway-high clinical potential by innovative predictive, preventive, and personalized medical approach. EPMA J 2020; 11:1-16. [PMID: 32140182 PMCID: PMC7028895 DOI: 10.1007/s13167-019-00195-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND LIMITATIONS Impaired wound healing (WH) and chronic inflammation are hallmarks of non-communicable diseases (NCDs). However, despite WH being a recognized player in NCDs, mainstream therapies focus on (un)targeted damping of the inflammatory response, leaving WH largely unaddressed, owing to three main factors. The first is the complexity of the pathway that links inflammation and wound healing; the second is the dual nature, local and systemic, of WH; and the third is the limited acknowledgement of genetic and contingent causes that disrupt physiologic progression of WH. PROPOSED APPROACH Here, in the frame of Predictive, Preventive, and Personalized Medicine (PPPM), we integrate and revisit current literature to offer a novel systemic view on the cues that can impact on the fate (acute or chronic inflammation) of WH, beyond the compartmentalization of medical disciplines and with the support of advanced computational biology. CONCLUSIONS This shall open to a broader understanding of the causes for WH going awry, offering new operational criteria for patients' stratification (prediction and personalization). While this may also offer improved options for targeted prevention, we will envisage new therapeutic strategies to reboot and/or boost WH, to enable its progression across its physiological phases, the first of which is a transient acute inflammatory response versus the chronic low-grade inflammation characteristic of NCDs.
Collapse
Affiliation(s)
- Maria Giovanna Maturo
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Greg Gibson
- Center for Integrative Genomics, School of Biological Sciences, Georgia Tech, Atlanta, GA USA
| | - Luigi Manni
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Christine Nardini
- IAC Institute for Applied Computing, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
- Bio Unit, Scientific and Medical Direction, SOL Group, Monza, Italy
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Autonomic disorders sometimes occur in the context of systemic autoimmune disease or as a direct consequence of autoimmunity against the nervous system. This article provides an overview of autonomic disorders with potential autoimmune etiology. RECENT FINDINGS Recent evidence highlights a close association between the autonomic nervous system and inflammation. The autonomic nervous system regulates immune function, and autonomic manifestations may occur in a number of systemic autoimmune diseases. In a few instances, autoimmunity directly influences autonomic function. Autoimmune autonomic ganglionopathy is the prototypic antibody-mediated autonomic disorder. Over time, a better understanding of the clinical spectrum of autoimmune autonomic ganglionopathy, the significance of ganglionic nicotinic acetylcholine receptor antibodies, other immune-mediated autonomic neuropathies, and autonomic manifestations of other systemic or neurologic autoimmune disorders has emerged. SUMMARY Autoimmune autonomic disorders may be challenging, but correct identification of these conditions is important. In some cases, potential exists for effective immunomodulatory treatment.
Collapse
|
39
|
Fernandes P, Pereira D, Watkins PB, Bertrand D. Differentiating the Pharmacodynamics and Toxicology of Macrolide and Ketolide Antibiotics. J Med Chem 2019; 63:6462-6473. [PMID: 31644280 DOI: 10.1021/acs.jmedchem.9b01159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This is a review of the macrolide and ketolide field focusing on differentiating the pharmacodynamics and especially the toxicology of the macrolides and ketolides. We emphasize the diversity in pharmacodynamics and toxicity of the macrolides and ketolides, resulting from even small structural changes, which makes it important to consider the various different compounds separately, not necessarily as a class. The ketolide, telithromycin, was developed because of rising bacterial macrolide resistance but was withdrawn postapproval after visual disturbances, syncope, myasthenia gravis, and hepatotoxicity were noted. These diverse adverse effects could be attributed to inhibition of nicotinic acetylcholine receptors. Solithromycin, a later generation ketolide, was effective in treating bacterial pneumonia, but it was not approved by the U.S. Food and Drug Administration owing, in part, to its structural similarity to telithromycin. This Miniperspective describes that structurally similar macrolides/ketolides have clearly mechanistically distinct effects. Understanding these effects could help in developing and securing regulatory approval of a new macrolide/ketolide that is active against macrolide-resistant pathogenic bacteria.
Collapse
Affiliation(s)
| | - David Pereira
- Ponce De Leon Health, Fernandina Beach, Florida 32034, United States
| | - Paul B Watkins
- Schools of Pharmacy, Medicine and Public Health, Institute for Drug Safety Sciences, University of North Carolina, Chapel Hill, North Carolina 27514, United States
| | - Daniel Bertrand
- HiQScreen SÃrl, 6, Route de Compois, Vesenaz, 1222 Geneva, Switzerland
| |
Collapse
|
40
|
Saxton SN, Clark BJ, Withers SB, Eringa EC, Heagerty AM. Mechanistic Links Between Obesity, Diabetes, and Blood Pressure: Role of Perivascular Adipose Tissue. Physiol Rev 2019; 99:1701-1763. [PMID: 31339053 DOI: 10.1152/physrev.00034.2018] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Obesity is increasingly prevalent and is associated with substantial cardiovascular risk. Adipose tissue distribution and morphology play a key role in determining the degree of adverse effects, and a key factor in the disease process appears to be the inflammatory cell population in adipose tissue. Healthy adipose tissue secretes a number of vasoactive adipokines and anti-inflammatory cytokines, and changes to this secretory profile will contribute to pathogenesis in obesity. In this review, we discuss the links between adipokine dysregulation and the development of hypertension and diabetes and explore the potential for manipulating adipose tissue morphology and its immune cell population to improve cardiovascular health in obesity.
Collapse
Affiliation(s)
- Sophie N Saxton
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom; School of Environment and Life Sciences, University of Salford, Salford, United Kingdom; and Department of Physiology, VU University Medical Centre, Amsterdam, Netherlands
| | - Ben J Clark
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom; School of Environment and Life Sciences, University of Salford, Salford, United Kingdom; and Department of Physiology, VU University Medical Centre, Amsterdam, Netherlands
| | - Sarah B Withers
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom; School of Environment and Life Sciences, University of Salford, Salford, United Kingdom; and Department of Physiology, VU University Medical Centre, Amsterdam, Netherlands
| | - Etto C Eringa
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom; School of Environment and Life Sciences, University of Salford, Salford, United Kingdom; and Department of Physiology, VU University Medical Centre, Amsterdam, Netherlands
| | - Anthony M Heagerty
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom; School of Environment and Life Sciences, University of Salford, Salford, United Kingdom; and Department of Physiology, VU University Medical Centre, Amsterdam, Netherlands
| |
Collapse
|
41
|
Hajiasgharzadeh K, Sadigh-Eteghad S, Mansoori B, Mokhtarzadeh A, Shanehbandi D, Doustvandi MA, Asadzadeh Z, Baradaran B. Alpha7 nicotinic acetylcholine receptors in lung inflammation and carcinogenesis: Friends or foes? J Cell Physiol 2019; 234:14666-14679. [PMID: 30701535 DOI: 10.1002/jcp.28220] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 01/24/2023]
Abstract
The lung tissue expresses the cholinergic system including nicotinic acetylcholine receptors (nAChRs) which included in many physiologic and pathologic processes. Mounting evidence revealed that these receptors have important roles in lung carcinogenesis via modulating either stimulatory or inhibitory signaling pathways. Among different members of nicotinic receptors family, alpha7-subtype of nAChR (α7nAChR) is a critical mediator involved in both inflammatory responses and cancers. Several studies have shown that this receptor is the most powerful regulator of responses that stimulate lung cancer processes such as proliferation, angiogenesis, metastasis, and inhibition of apoptosis. Moreover, aside from its roles in the regulation of cancer pathways, there is growing evidence indicating that α7nAChR has profound impacts on lung inflammation through the cholinergic anti-inflammatory pathway. Regarding such diverse effects as well as the critical roles of nicotine as an activator of α7nAChR on lung cancer pathogenesis, its modulation has emerged as a promising target for drug developments. In this review, we aim to highlight the detrimental as well as the possible beneficial influences of α7nAChR downstream signaling cascades in the control of lung inflammation and cancer-associated properties. Consequently, by considering the significant global burden of lung cancer, delineating the complex influences of α7 receptors would be of great interest in designing novel anticancer and anti-inflammatory strategies for the patients suffering from lung cancer.
Collapse
Affiliation(s)
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
42
|
Nederhoff MGJ, Fransen DE, Verlinde SAMW, Brans MAD, Pasterkamp G, Bleys RLAW. Effect of vagus nerve stimulation on tissue damage and function loss in a mouse myocardial ischemia-reperfusion model. Auton Neurosci 2019; 221:102580. [PMID: 31491700 DOI: 10.1016/j.autneu.2019.102580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/14/2019] [Accepted: 08/14/2019] [Indexed: 11/25/2022]
Abstract
OBJECTIVES In cardiac ischemia, acute inflammatory responses further increase the detrimental effect on myocardial tissue. Since vagus nerve stimulation (VS) attenuates inflammatory responsiveness this study examines the effect of VS on myocardial damage development in a cardiac ischemia-reperfusion (IR) mouse model. METHODS 54 male C57Bl/6j mice were subjected to an IR procedure with or without prior VS. The effects on inflammatory responsiveness, infarct size, cardiac function, neutrophils, lymphocytes and vascular endothelial growth factor (VEGF) in the infarcted myocardium were measured at 48 h after intervention. Group results were compared with unpaired Mann-Whitney or Kruskall-Wallis test. RESULTS A significant decrease in inflammatory responsiveness was not verified by decreased TNFα levels in blood from VS and IR treated mice. The percentage infarct size over area at risk was smaller in the group with VS + IR compared with IR (22.4 ± 10.2% vs 37.6 ± 9.0%, p = 0.003). The degree of the reduction in cardiac function was not different between the IR groups with or without VS and no group differences were found in amounts of neutrophils, CD3+ lymphocytes and VEGF in the reperfused mouse heart. CONCLUSION The present study does not provide clear evidence of a reducing role for VS on cardiac function loss. This could mean that VS has a less inhibiting effect on myocardial inflammation than may be expected from the literature.
Collapse
Affiliation(s)
- M G J Nederhoff
- Department of Anatomy, Division Surgical Specialties, University Medical Center Utrecht, Universiteitsweg 100, room: Str. 0.305, 3584CG Utrecht, the Netherlands.
| | - D E Fransen
- Department of Anatomy, Division Surgical Specialties, University Medical Center Utrecht, Universiteitsweg 100, room: Str. 0.305, 3584CG Utrecht, the Netherlands
| | - S A M W Verlinde
- Department of Anatomy, Division Surgical Specialties, University Medical Center Utrecht, Universiteitsweg 100, room: Str. 0.305, 3584CG Utrecht, the Netherlands
| | - M A D Brans
- Experimental Cardiology Laboratory, Division Surgical Specialties, University Medical Center Utrecht, Universiteitsweg 100, room: Str. 0.305, 3584CG Utrecht, the Netherlands
| | - G Pasterkamp
- Experimental Cardiology Laboratory, Division Surgical Specialties, University Medical Center Utrecht, Universiteitsweg 100, room: Str. 0.305, 3584CG Utrecht, the Netherlands
| | - R L A W Bleys
- Department of Anatomy, Division Surgical Specialties, University Medical Center Utrecht, Universiteitsweg 100, room: Str. 0.305, 3584CG Utrecht, the Netherlands
| |
Collapse
|
43
|
Kobrzycka A, Napora P, Pearson BL, Pierzchała-Koziec K, Szewczyk R, Wieczorek M. Peripheral and central compensatory mechanisms for impaired vagus nerve function during peripheral immune activation. J Neuroinflammation 2019; 16:150. [PMID: 31324250 PMCID: PMC6642550 DOI: 10.1186/s12974-019-1544-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Determining the etiology and possible treatment strategies for numerous diseases requires a comprehensive understanding of compensatory mechanisms in physiological systems. The vagus nerve acts as a key interface between the brain and the peripheral internal organs. We set out to identify mechanisms compensating for a lack of neuronal communication between the immune and the central nervous system (CNS) during infection. METHODS We assessed biochemical and central neurotransmitter changes resulting from subdiaphragmatic vagotomy and whether they are modulated by intraperitoneal infection. We performed a series of subdiaphragmatic vagotomy or sham operations on male Wistar rats. Next, after full, 30-day recovery period, they were randomly assigned to receive an injection of Escherichia coli lipopolysaccharide or saline. Two hours later, animal were euthanized and we measured the plasma concentration of prostaglandin E2 (with HPLC-MS), interleukin-6 (ELISA), and corticosterone (RIA). We also had measured the concentration of monoaminergic neurotransmitters and their metabolites in the amygdala, brainstem, hippocampus, hypothalamus, motor cortex, periaqueductal gray, and prefrontal medial cortex using RP-HPLC-ED. A subset of the animals was evaluated in the elevated plus maze test immediately before euthanization. RESULTS The lack of immunosensory signaling of the vagus nerve stimulated increased activity of discrete inflammatory marker signals, which we confirmed by quantifying biochemical changes in blood plasma. Behavioral results, although preliminary, support the observed biochemical alterations. Many of the neurotransmitter changes observed after vagotomy indicated that the vagus nerve influences the activity of many brain areas involved in control of immune response and sickness behavior. Our studies show that these changes are largely eliminated during experimental infection. CONCLUSIONS Our results suggest that in vagotomized animals with blocked CNS, communication may transmit via a pathway independent of the vagus nerve to permit restoration of CNS activity for peripheral inflammation control.
Collapse
Affiliation(s)
- Anna Kobrzycka
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Paweł Napora
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Brandon L. Pearson
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, USA
| | | | - Rafał Szewczyk
- Department of Industrial Microbiology and Biotechnology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Marek Wieczorek
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| |
Collapse
|
44
|
Snow SJ, Henriquez AR, Costa DL, Kodavanti UP. Neuroendocrine Regulation of Air Pollution Health Effects: Emerging Insights. Toxicol Sci 2019; 164:9-20. [PMID: 29846720 DOI: 10.1093/toxsci/kfy129] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Air pollutant exposures are linked to cardiopulmonary diseases, diabetes, metabolic syndrome, neurobehavioral conditions, and reproductive abnormalities. Significant effort is invested in understanding how pollutants encountered by the lung might induce effects in distant organs. The role of circulating mediators has been predicted; however, their origin and identity have not been confirmed. New evidence has emerged which implicates the role of neuroendocrine sympathetic-adrenal-medullary (SAM) and hypothalamic-pituitary-adrenal (HPA) stress axes in mediating a wide array of systemic and pulmonary effects. Our recent studies using ozone exposure as a prototypical air pollutant demonstrate that increases in circulating adrenal-derived stress hormones (epinephrine and cortisol/corticosterone) contribute to lung injury/inflammation and metabolic effects in the liver, pancreas, adipose, and muscle tissues. When stress hormones are depleted by adrenalectomy in rats, most ozone effects including lung injury/inflammation are diminished. Animals treated with antagonists for adrenergic and glucocorticoid receptors show inhibition of the pulmonary and systemic effects of ozone, whereas treatment with agonists restore and exacerbate the ozone-induced injury/inflammation phenotype, implying the role of neuroendocrine activation. The neuroendocrine system is critical for normal homeostasis and allostatic activation; however, chronic exposure to stressors may lead to increases in allostatic load. The emerging mechanisms by which circulating mediators are released and are responsible for producing multiorgan effects of air pollutants insists upon a paradigm shift in the field of air pollution and health. Moreover, since these neuroendocrine responses are linked to both chemical and nonchemical stressors, the interactive influence of air pollutants, lifestyle, and environmental factors requires further study.
Collapse
Affiliation(s)
- Samantha J Snow
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, 27711
| | - Andres R Henriquez
- Oak Ridge Institute for Science and Education, Research Triangle Park, North Carolina, 27711
| | - Daniel L Costa
- Emeritus, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, 27711
| | - Urmila P Kodavanti
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, 27711
| |
Collapse
|
45
|
Abstract
Perivascular adipose tissue (PVAT) is no longer recognised as simply a structural support for the vasculature, and we now know that PVAT releases vasoactive factors which modulate vascular function. Since the discovery of this function in 1991, PVAT research is rapidly growing and the importance of PVAT function in disease is becoming increasingly clear. Obesity is associated with a plethora of vascular conditions; therefore, the study of adipocytes and their effects on the vasculature is vital. PVAT contains an adrenergic system including nerves, adrenoceptors and transporters. In obesity, the autonomic nervous system is dysfunctional; therefore, sympathetic innervation of PVAT may be the key mechanistic link between increased adiposity and vascular disease. In addition, not all obese people develop vascular disease, but a common feature amongst those that do appears to be the inflammatory cell population in PVAT. This review will discuss what is known about sympathetic innervation of PVAT, and the links between nerve activation and inflammation in obesity. In addition, we will examine the therapeutic potential of exercise in sympathetic stimulation of adipose tissue.
Collapse
Affiliation(s)
- Sophie N Saxton
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility (3rd floor), 46 Grafton Street, M13 9NT, Manchester, UK.
| | - Sarah B Withers
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility (3rd floor), 46 Grafton Street, M13 9NT, Manchester, UK
- School of Environment and Life Sciences, University of Salford, Manchester, UK
| | - Anthony M Heagerty
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility (3rd floor), 46 Grafton Street, M13 9NT, Manchester, UK
| |
Collapse
|
46
|
Abstract
Surgery and other invasive procedures, which are routinely performed during general anesthesia, may induce an inflammatory response in the patient. This inflammatory response is an inherent answer of the body to the intervention and can be both beneficial and potentially harmful. The immune system represents a unique evolutionary achievement equipping higher organisms with an effective defense mechanism against exogenous pathogens. However, not only bacteria might evoke an immune response but also other noninfectious stimuli like the surgical trauma or mechanical ventilation may induce an inflammatory response of varying degree. In these cases, the immune system activation is not always beneficial for the patients and might carry the risk of concomitant, harmful effects on host cells, tissues, or even whole organ systems. Research over the past decades has contributed substantial information in which ways surgical patients may be affected by inflammatory reactions. Modulations of the patient's immune system may be evoked by the use of anesthetic agents, the nature of surgical trauma and the use of any supportive therapy during the perioperative period. The effects on the patient may be manifold, including various proinflammatory effects. This review focuses on the causes and effects of inflammation in the perioperative period. In addition, we also highlight possible approaches by which inflammation in the perioperative may be modulated in the future.
Collapse
Affiliation(s)
- Jan Rossaint
- From the Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | | |
Collapse
|
47
|
Fornai M, van den Wijngaard RM, Antonioli L, Pellegrini C, Blandizzi C, de Jonge WJ. Neuronal regulation of intestinal immune functions in health and disease. Neurogastroenterol Motil 2018; 30:e13406. [PMID: 30058092 DOI: 10.1111/nmo.13406] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Nerve-mucosa interactions control various elements of gastrointestinal functions, including mucosal host defense, gut barrier function, and epithelial cell growth and differentiation. In both intestinal and extra-intestinal diseases, alterations of autonomic nerve activity have been observed to be concurrent with the disease course, such as in inflammatory and functional bowel diseases, and neurodegenerative diseases. This is relevant as the extrinsic autonomic nervous system is increasingly recognized to modulate gut inflammatory responses. The molecular and cellular mechanisms through which the extrinsic and intrinsic nerve pathways may regulate digestive mucosal functions have been investigated in several pre-clinical and clinical studies. PURPOSE The present review focuses on the involvement of neural pathways in gastrointestinal disease, and addresses the current strategies to intervene with neuronal pathway as a means of treatment.
Collapse
Affiliation(s)
- M Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - R M van den Wijngaard
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - L Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - C Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - C Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - W J de Jonge
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
48
|
A new hypothesis for the pathophysiology of complex regional pain syndrome. Med Hypotheses 2018; 119:41-53. [DOI: 10.1016/j.mehy.2018.07.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/18/2018] [Accepted: 07/27/2018] [Indexed: 12/21/2022]
|
49
|
Peng ZL, Huang LW, Yin J, Zhang KN, Xiao K, Qing GZ. Association between early serum cholinesterase activity and 30-day mortality in sepsis-3 patients: A retrospective cohort study. PLoS One 2018; 13:e0203128. [PMID: 30161257 PMCID: PMC6117034 DOI: 10.1371/journal.pone.0203128] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 08/15/2018] [Indexed: 12/29/2022] Open
Abstract
Low serum cholinesterase (SCHE) activity has been associated with poor prognoses in a variety of conditions, including sepsis. However, such an association has not been well characterized since the Third International Consensus Definitions Task Force modified the definition of sepsis to "life-threatening organ dysfunction due to a dysregulated host response to infection" (known as sepsis-3) in 2016. In the current retrospective cohort study, we examined whether 30-day mortality in sepsis-3 patients is associated with SCHE activity. A total of 166 sepsis-3 patients receiving treatment at an emergency intensive care unit (EICU) were included. The 30-day death rate was 33.1% (55/166). SCHE activity upon EICU admission was lower in nonsurvivors (3.3 vs. 4.5 KU/L in survivors, p = 0.0002). Subjects with low SCHE activity (defined as <4 KU/L) had higher 30-day mortality rates than subjects with normal SCHE activity (45.5%, 40/88 vs. 19.2%, 15/78; p<0.001). A multivariate logistic regression analysis revealed an association between 30-day mortality and lower SCHE activity after adjustments for relevant factors, such as acute multiple organ dysfunction. The odds ratio (OR) for every unit decrease in SCHE activity was 2.11 (95% confidence interval (CI), 1.37-3.27; p = 0.0008). The area under the curve (AUC) of SCHE activity for predicting 30-day mortality was 0.67 (95% CI 0.59-0.74), and the AUC of lactate for predicting 30-day mortality was 0.64 (95% CI 0.57-0.70). Using a combination of SCHE and lactate, the AUC was 0.74 (95% CI 0.69-0.83). These data suggest that lower SCHE activity is an independent risk factor for 30-day mortality in sepsis-3 patients.
Collapse
Affiliation(s)
- Zheng-Liang Peng
- EICU, First Affiliated Hospital of the University of South China, Hengyang, Hunan, China
- * E-mail: (ZLP); (GZQ)
| | - Liang-Wei Huang
- EICU, First Affiliated Hospital of the University of South China, Hengyang, Hunan, China
| | - Jian Yin
- EICU, First Affiliated Hospital of the University of South China, Hengyang, Hunan, China
| | - Ke-Na Zhang
- EICU, First Affiliated Hospital of the University of South China, Hengyang, Hunan, China
| | - Kang Xiao
- EICU, First Affiliated Hospital of the University of South China, Hengyang, Hunan, China
| | - Guo-Zhong Qing
- EICU, First Affiliated Hospital of the University of South China, Hengyang, Hunan, China
- * E-mail: (ZLP); (GZQ)
| |
Collapse
|
50
|
Bordoni B, Morabito B. Symptomatology Correlations Between the Diaphragm and Irritable Bowel Syndrome. Cureus 2018; 10:e3036. [PMID: 30258735 PMCID: PMC6153095 DOI: 10.7759/cureus.3036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022] Open
Abstract
Irritable bowel syndrome (IBS) is one of the most debilitating and common gastrointestinal disorders; nevertheless, its pathophysiology is still unclear. It affects 11% of the world's population, and is characterized by alternating periods of pain and/or motility disorders with periods of remission and without any evidence of any structural and functional organic variation. It has been recently proposed that an altered contractile ability of the diaphragm muscle might adversely influence intestinal motility. The text reviews the diaphragm's functions, anatomy, and neurological links in correlation with the presence of chronic symptoms associated to IBS, like chronic low back pain, chronic pelvic pain, chronic headache, and temporomandibular joint dysfunction, vagus nerve inflammation, and depression and anxiety. The interplay between an individual's breath dynamic and intestinal behaviour is still an unaddressed point in the physiopathology of IBS, and the paucity of scientific studies should recommend further research to better understand the importance of breathing in this syndrome.
Collapse
Affiliation(s)
- Bruno Bordoni
- Cardiology, Foundation Don Carlo Gnocchi (IRCCS)/Institute of Hospitalization and Care, Milano, ITA
| | - Bruno Morabito
- Osteopathy, School of Osteopathic Centre for Research and Studies, Rome, ITA
| |
Collapse
|