1
|
Feng L, Li B, Cai M, Zhang Z, Zhao Y, Yong SS, Tian Z. Resistance exercise alleviates the prefrontal lobe injury and dysfunction by activating SESN2/AMPK/PGC-1α signaling pathway and inhibiting oxidative stress and inflammation in mice with myocardial infarction. Exp Neurol 2023; 370:114559. [PMID: 37788754 DOI: 10.1016/j.expneurol.2023.114559] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/14/2023] [Accepted: 09/29/2023] [Indexed: 10/05/2023]
Abstract
OBJECTIVES Myocardial infarction (MI) induces inflammatory response and oxidative stress in the brain, which would be one of the causes of cardiac dysfunction. Exercise training is viewed as a feasible strategy to improve cardiac function of the infarcted heart. The aim of this study was to investigate whether exercise training could alleviate MI-induced prefrontal lobe injury via activating Sestrin2 (SESN2) signaling and inhibiting oxidative stress and inflammation. METHODS Male C57BL/6 mice were divided into five groups: control group (CON), aerobic exercise group (AE), resistance exercise group (RE), whole-body vibration group (WBV) and electrical stimulation group (ES); and three groups: sham-operated group (S), sedentary MI group (MI) and MI with resistance exercise group (MRE). After four weeks of training, sensorimotor function, spatial learning, long-term and spatial memory, and cardiac function were detected. Then, mice were euthanized, and the prefrontal areas were separated for HE, Nissl, SESN2, microtubule-associated protein 2 (MAP2), neuron-specific nucleoprotein (NeuN), and TUNEL staining. Activation of SESN2/adenosine monophosphate-activated protein kinase (AMPK)/peroxisome proliferator activated receptor γ coactivator-1α (PGC-1α) signaling pathway and expression of proteins related to oxidative stress, inflammation and apoptosis in the prefrontal lobe were detected by western blotting. RESULTS Different types of exercise training all activated the SESN2/AMPK/PGC-1α signaling pathway, and the effect of RE is the best. RE improved sensorimotor, learning, and memory impairments, increased the expressions of antioxidant, anti-inflammatory and anti-apoptotic proteins, reduced oxidative stress, inflammation and apoptosis, ultimately alleviated the prefrontal lobe injury and dysfunction in mice with MI. CONCLUSION RE alleviates MI-indued prefrontal lobe injury and dysfunction by inhibiting the levels of oxidative stress, inflammation and apoptosis, partially via activating SESN2/AMPK/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Lili Feng
- Department of Sport and Exercise Science, College of Education, Zhejiang University, Hangzhou 310058, China; Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China.
| | - Bowen Li
- Department of Sport and Exercise Science, College of Education, Zhejiang University, Hangzhou 310058, China.
| | - Mengxin Cai
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China
| | - Zezhou Zhang
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China
| | - Yifang Zhao
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China
| | - Su Sean Yong
- Department of Sport and Exercise Science, College of Education, Zhejiang University, Hangzhou 310058, China
| | - Zhenjun Tian
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
2
|
Bonsale R, Infantino R, Perrone M, Marabese I, Ricciardi F, Fusco A, Teweldemedhin MM, Boccella S, Guida F, Rinaldi B. The long-term exercise after traumatic brain injury: Reharmonizing brain by sound body. Brain Res 2023; 1816:148471. [PMID: 37356701 DOI: 10.1016/j.brainres.2023.148471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 06/27/2023]
Abstract
Traumatic brain injuries (TBI) refer to multiple acquired dysfunctions arising from damage to the brain caused by an external force, including rapid acceleration/deceleration and concussion. Among them, mild TBI (mTBI) accounts for most cases (up to 90%) of injuries. It is responsible for a variety of symptoms, including anxiety, depression, and cognitive impairments that remain difficult to be treated. It has been reported that regular physical activity, as well as, improving life quality, display a neuroprotective function, suggesting a possible role in post-traumatic rehabilitation. In this study, we investigated the effects of treadmill exercise in a mice mTBI model by behavioural, electrophysiological and neurochemical analysis. Daily exercise decreased anxiety, aggressive behavior, and depression in mTBI mice. Accordingly, electrophysiological and neurochemical maladaptive rearrangement occurring in the hippocampus of mTBI mice were prevented by the exercise.
Collapse
Affiliation(s)
- Roozbe Bonsale
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Michela Perrone
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Federica Ricciardi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Antimo Fusco
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Milena Melake Teweldemedhin
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Barbara Rinaldi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy.
| |
Collapse
|
3
|
Traub J, Frey A, Störk S. Chronic Neuroinflammation and Cognitive Decline in Patients with Cardiac Disease: Evidence, Relevance, and Therapeutic Implications. Life (Basel) 2023; 13:life13020329. [PMID: 36836686 PMCID: PMC9962280 DOI: 10.3390/life13020329] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Acute and chronic cardiac disorders predispose to alterations in cognitive performance, ranging from mild cognitive impairment to overt dementia. Although this association is well-established, the factors inducing and accelerating cognitive decline beyond ageing and the intricate causal pathways and multilateral interdependencies involved remain poorly understood. Dysregulated and persistent inflammatory processes have been implicated as potentially causal mediators of the adverse consequences on brain function in patients with cardiac disease. Recent advances in positron emission tomography disclosed an enhanced level of neuroinflammation of cortical and subcortical brain regions as an important correlate of altered cognition in these patients. In preclinical and clinical investigations, the thereby involved domains and cell types of the brain are gradually better characterized. Microglia, resident myeloid cells of the central nervous system, appear to be of particular importance, as they are extremely sensitive to even subtle pathological alterations affecting their complex interplay with neighboring astrocytes, oligodendrocytes, infiltrating myeloid cells, and lymphocytes. Here, we review the current evidence linking cognitive impairment and chronic neuroinflammation in patients with various selected cardiac disorders including the aspect of chronic neuroinflammation as a potentially druggable target.
Collapse
Affiliation(s)
- Jan Traub
- Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
- Department of Clinical Research & Epidemiology, Comprehensive Heart Failure Center, University and University Hospital Würzburg, 97078 Würzburg, Germany
- Correspondence: ; Tel.: +4993120139216
| | - Anna Frey
- Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
- Department of Clinical Research & Epidemiology, Comprehensive Heart Failure Center, University and University Hospital Würzburg, 97078 Würzburg, Germany
| | - Stefan Störk
- Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
- Department of Clinical Research & Epidemiology, Comprehensive Heart Failure Center, University and University Hospital Würzburg, 97078 Würzburg, Germany
| |
Collapse
|
4
|
Wu C, Liu R, Luo Z, Sun M, Qile M, Xu S, Jin S, Zhang L, Gross ER, Zhang Y, He S. Spinal cord astrocytes regulate myocardial ischemia-reperfusion injury. Basic Res Cardiol 2022; 117:56. [PMID: 36367592 PMCID: PMC10139732 DOI: 10.1007/s00395-022-00968-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/13/2022]
Abstract
Astrocytes play a key role in the response to injury and noxious stimuli, but its role in myocardial ischemia-reperfusion (I/R) injury remains largely unknown. Here we determined whether manipulation of spinal astrocyte activity affected myocardial I/R injury and the underlying mechanisms. By ligating the left coronary artery to establish an in vivo I/R rat model, we observed a 1.7-fold rise in glial fibrillary acidic protein (GFAP) protein level in spinal cord following myocardial I/R injury. Inhibition of spinal astrocytes by intrathecal injection of fluoro-citrate, an astrocyte inhibitor, decreased GFAP immunostaining and reduced infarct size by 29% relative to the I/R group. Using a Designer Receptor Exclusively Activated by Designer Drugs (DREADD) chemogenetic approach, we bi-directionally manipulated astrocyte activity employing GFAP promoter-driven Gq- or Gi-coupled signaling. The Gq-DREADD-mediated activation of spinal astrocytes caused transient receptor potential vanilloid 1 (TRPV1) activation and neuropeptide release leading to a 1.3-fold increase in infarct size, 1.2-fold rise in serum norepinephrine level and higher arrhythmia score relative to I/R group. In contrast, Gi-DREADD-mediated inhibition of spinal astrocytes suppressed TRPV1-mediated nociceptive signaling, resulting in 35% reduction of infarct size and 51% reduction of arrhythmia score from I/R group, as well as lowering serum norepinephrine level from 3158 ± 108 to 2047 ± 95 pg/mL. Further, intrathecal administration of TRPV1 or neuropeptide antagonists reduced infarct size and serum norepinephrine level. These findings demonstrate a functional role of spinal astrocytes in myocardial I/R injury and provide a novel potential therapeutic approach targeting spinal cord astrocytes for the prevention of cardiac injury.
Collapse
Affiliation(s)
- Chao Wu
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230061, Anhui Province, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Rongrong Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230061, Anhui Province, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Zhaofei Luo
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230061, Anhui Province, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Meiyan Sun
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230061, Anhui Province, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Muge Qile
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230061, Anhui Province, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Shijin Xu
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230061, Anhui Province, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Shiyun Jin
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230061, Anhui Province, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Li Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230061, Anhui Province, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Eric R Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230061, Anhui Province, China. .,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China.
| | - Shufang He
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230061, Anhui Province, China. .,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China.
| |
Collapse
|
5
|
Gelosa P, Castiglioni L, Rzemieniec J, Muluhie M, Camera M, Sironi L. Cerebral derailment after myocardial infarct: mechanisms and effects of the signaling from the ischemic heart to brain. J Mol Med (Berl) 2022; 100:23-41. [PMID: 34674004 PMCID: PMC8724191 DOI: 10.1007/s00109-021-02154-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/07/2021] [Accepted: 10/14/2021] [Indexed: 12/04/2022]
Abstract
Myocardial infarction (MI) is the leading cause of death among ischemic heart diseases and is associated with several long-term cardiovascular complications, such as angina, re-infarction, arrhythmias, and heart failure. However, MI is frequently accompanied by non-cardiovascular multiple comorbidities, including brain disorders such as stroke, anxiety, depression, and cognitive impairment. Accumulating experimental and clinical evidence suggests a causal relationship between MI and stroke, but the precise underlying mechanisms have not yet been elucidated. Indeed, the risk of stroke remains a current challenge in patients with MI, in spite of the improvement of medical treatment among this patient population has reduced the risk of stroke. In this review, the effects of the signaling from the ischemic heart to the brain, such as neuroinflammation, neuronal apoptosis, and neurogenesis, and the possible actors mediating these effects, such as systemic inflammation, immunoresponse, extracellular vesicles, and microRNAs, are discussed.
Collapse
Affiliation(s)
- Paolo Gelosa
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Laura Castiglioni
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Joanna Rzemieniec
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Majeda Muluhie
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Marina Camera
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
- Centro Cardiologico Monzino, 20138, Milan, Italy
| | - Luigi Sironi
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy.
| |
Collapse
|
6
|
Tóth K, Oroszi T, van der Zee EA, Nyakas C, Schoemaker RG. Effects of exercise training on behavior and brain function after high dose isoproterenol-induced cardiac damage. Sci Rep 2021; 11:23576. [PMID: 34880374 PMCID: PMC8654950 DOI: 10.1038/s41598-021-03107-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/26/2021] [Indexed: 02/02/2023] Open
Abstract
Acute sympathetic stress can result in cardiac fibrosis, but may also lead to mental dysfunction. Exercise training after isoproterenol (ISO)-induced acute sympathetic stress was investigated regarding cardiac damage, neuroinflammation, brain function and behavior. Male Wistar rats (12 months) received ISO or saline. One week later, treadmill running or control handling (sedentary) started. After 4 weeks, cognitive- and exploratory behavior were evaluated, and heart and brain tissues were analyzed regarding cardiac damage, hippocampal neuroinflammation and neuronal function. ISO did not affect cognitive performance nor hippocampal function. However, ISO reduced anxiety, coinciding with locally reduced microglia (processes) size in the hippocampus. Exercise in ISO rats reversed anxiety, did not affect microglia morphology, but increased brain function. Thus, exercise after ISO did not affect cardiac damage, cognition or hippocampal neuroinflammation, but normalized anxiety. Increased localized BDNF expression may indicate improved brain function.
Collapse
Affiliation(s)
- Kata Tóth
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands.,Research Center for Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Tamás Oroszi
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands.,Research Center for Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Eddy A van der Zee
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Csaba Nyakas
- Research Center for Molecular Exercise Science, University of Physical Education, Budapest, Hungary.,Behavioral Physiology Research Laboratory, Health Science Faculty, Semmelweis University, Budapest, Hungary
| | - Regien G Schoemaker
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands. .,University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
7
|
Surinkaew P, Apaijai N, Sawaddiruk P, Jaiwongkam T, Kerdphoo S, Chattipakorn N, Chattipakorn SC. Mitochondrial Fusion Promoter Alleviates Brain Damage in Rats with Cardiac Ischemia/Reperfusion Injury. J Alzheimers Dis 2021; 77:993-1003. [PMID: 32804148 DOI: 10.3233/jad-200495] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cardiac ischemia/reperfusion (I/R) injury induces brain damage through increased blood-brain barrier (BBB) breakdown, microglial hyperactivity, pro-inflammatory cytokines, amyloid-β deposition, loss of dendritic spines, brain mitochondrial dysfunction, and imbalanced mitochondrial dynamics. Previous studies demonstrated that mitochondrial fusion promoter reduced cardiac damage from cardiac I/R injury; however, following cardiac I/R injury, the roles of mitochondrial dynamics on the brain have not been investigated. OBJECTIVE To investigate the effects of pharmacological modulation using mitochondrial fusion promoter (M1) in the brain of rats following cardiac I/R injury. METHODS Twenty-four male Wistar rats were separated into two groups; 1) sham-operation (n = 8) and 2) cardiac I/R injury (n = 16). Rats in the cardiac I/R injury group were randomly received either normal saline solution as a vehicle or a mitochondrial fusion promoter (M1, 2 mg/kg) intravenously. Both treatments were given to the rats 15 minutes before cardiac I/R injury. At the end of the reperfusion protocol, the brain was rapidly removed to investigate brain mitochondrial function, mitochondrial dynamics proteins, microglial activity, and Alzheimer's disease (AD) related proteins. RESULTS Cardiac I/R injury induced brain mitochondrial dynamics imbalance as indicated by reduced mitochondrial fusion proteins expression without alteration in mitochondrial fission, brain mitochondrial dysfunction, BBB breakdown, increased macrophage infiltration, apoptosis, and AD-related proteins. Pretreatment with M1 effectively increased the expression of mitofusin 2, a mitochondrial outer membrane fusion protein, reduced brain mitochondrial dysfunction, BBB breakdown, macrophage infiltration, apoptosis, and AD-related proteins in rats following cardiac I/R injury. CONCLUSION This mitochondrial fusion promoter significantly protected rats with cardiac I/R injury against brain damage.
Collapse
Affiliation(s)
- Poomarin Surinkaew
- Department of Anesthesiology, Lamphun Hospital, Lamphun, Thailand.,Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Passakorn Sawaddiruk
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thidarat Jaiwongkam
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sasiwan Kerdphoo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
8
|
Sun N, Mei Y, Hu Z, Xing W, Lv K, Hu N, Zhang T, Wang D. Ghrelin attenuates depressive-like behavior, heart failure, and neuroinflammation in postmyocardial infarction rat model. Eur J Pharmacol 2021; 901:174096. [PMID: 33848542 DOI: 10.1016/j.ejphar.2021.174096] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 11/27/2022]
Abstract
Depression after myocardial infarction (MI) and chronic heart failure (CHF) is a common condition that is resistant to anti-depressive drugs. Ghrelin (a peptide hormone) shows dual protective effects on heart and brain. Whether ghrelin treatment attenuated depression after MI was investigated. Coronary artery occlusion was performed to induce MI and subsequent CHF in rats. Ghrelin (100 μg/kg in 0.5 ml of saline) or vehicle (0.5 ml of saline) was injected subcutaneously twice a day for 4 weeks. At week 5, all the animals underwent behavioral assessments including sucrose preference test (SPT), elevated plus maze test (EPM), and open field test (OFT). After cardiac function analysis, brain tissues were processed to determine inflammatory cytokines and microglial activations in hippocampus. Results showed that ghrelin substantially improved cardiac dysfunction, infarction size, and cardiac remodeling and modulated the release of inflammatory cytokines and the increase of Iba-1 positive microglia and glial fibrillary acidic protein-positive astrocytes in the CA1 area of hippocampus. Behavioral tests revealed that this treatment remarkably increased sucrose preference and mobile times and numbers. These findings provided evidence that peripheral ghrelin administration inhibits depression-like behavior and neuroinflammation and thus could be a new approach for the treatment of CHF-associated depression.
Collapse
Affiliation(s)
- Nan Sun
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241001, PR China; Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, 241001, PR China
| | - Yong Mei
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241001, PR China; Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, 241001, PR China
| | - Zhengtao Hu
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241001, PR China
| | - Wen Xing
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241001, PR China
| | - Kun Lv
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, 241001, PR China
| | - Nengwei Hu
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241001, PR China; Department of Physiology and Neurobiology, Zhengzhou University School of Medicine, Zhengzhou, 450001, China; Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Ting Zhang
- Department of Psychology, Wannan Medical College, Wuhu, 241001, PR China.
| | - Deguo Wang
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241001, PR China; Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, 241001, PR China.
| |
Collapse
|
9
|
Vitamin D Deficiency Induces Chronic Pain and Microglial Phenotypic Changes in Mice. Int J Mol Sci 2021; 22:ijms22073604. [PMID: 33808491 PMCID: PMC8036382 DOI: 10.3390/ijms22073604] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
The bioactive form of vitamin D, 1,25-dihydroxyvitamin D (1,25D3), exerts immunomodulatory actions resulting in neuroprotective effects potentially useful against neurodegenerative and autoimmune diseases. In fact, vitamin D deficiency status has been correlated with painful manifestations associated with different pathological conditions. In this study, we have investigated the effects of vitamin D deficiency on microglia cells, as they represent the main immune cells responsible for early defense at central nervous system (CNS), including chronic pain states. For this purpose, we have employed a model of low vitamin D intake during gestation to evaluate possible changes in primary microglia cells obtained from postnatal day(P)2-3 pups. Afterwards, pain measurement and microglia morphological analysis in the spinal cord level and in brain regions involved in the integration of pain perception were performed in the parents subjected to vitamin D restriction. In cultured microglia, we detected a reactive-activated and proliferative-phenotype associated with intracellular reactive oxygen species (ROS) generation. Oxidative stress was closely correlated with the extent of DNA damage and increased β-galactosidase (B-gal) activity. Interestingly, the incubation with 25D3 or 1,25D3 or palmitoylethanolamide, an endogenous ligand of peroxisome proliferator-activated-receptor-alpha (PPAR-α), reduced most of these effects. Morphological analysis of ex-vivo microglia obtained from vitamin-D-deficient adult mice revealed an increased number of activated microglia in the spinal cord, while in the brain microglia appeared in a dystrophic phenotype. Remarkably, activated (spinal) or dystrophic (brain) microglia were detected in a prominent manner in females. Our data indicate that vitamin D deficiency produces profound modifications in microglia, suggesting a possible role of these cells in the sensorial dysfunctions associated with hypovitaminosis D.
Collapse
|
10
|
Transcranial Magnetic Stimulation as a Tool to Investigate Motor Cortex Excitability in Sport. Brain Sci 2021; 11:brainsci11040432. [PMID: 33800662 PMCID: PMC8065474 DOI: 10.3390/brainsci11040432] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/17/2021] [Accepted: 03/24/2021] [Indexed: 11/28/2022] Open
Abstract
Transcranial magnetic stimulation, since its introduction in 1985, has brought important innovations to the study of cortical excitability as it is a non-invasive method and, therefore, can be used both in healthy and sick subjects. Since the introduction of this cortical stimulation technique, it has been possible to deepen the neurophysiological aspects of motor activation and control. In this narrative review, we want to provide a brief overview regarding TMS as a tool to investigate changes in cortex excitability in athletes and highlight how this tool can be used to investigate the acute and chronic responses of the motor cortex in sport science. The parameters that could be used for the evaluation of cortical excitability and the relative relationship with motor coordination and muscle fatigue, will be also analyzed. Repetitive physical training is generally considered as a principal strategy for acquiring a motor skill, and this process can elicit cortical motor representational changes referred to as use-dependent plasticity. In training settings, physical practice combined with the observation of target movements can enhance cortical excitability and facilitate the process of learning. The data to date suggest that TMS is a valid technique to investigate the changes in motor cortex excitability in trained and untrained subjects. Recently, interest in the possible ergogenic effect of non-invasive brain stimulation in sport is growing and therefore in the future it could be useful to conduct new experiments to evaluate the impact on learning and motor performance of these techniques.
Collapse
|
11
|
Gouweleeuw L, Wajant H, Maier O, Eisel ULM, Blankesteijn WM, Schoemaker RG. Effects of selective TNFR1 inhibition or TNFR2 stimulation, compared to non-selective TNF inhibition, on (neuro)inflammation and behavior after myocardial infarction in male mice. Brain Behav Immun 2021; 93:156-171. [PMID: 33444731 DOI: 10.1016/j.bbi.2021.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Myocardial infarction (MI) coinciding with depression worsens prognosis. Although Tumor Necrosis Factor alpha (TNF) is recognized to play a role in both conditions, the therapeutic potential of TNF inhibition is disappointing. TNF activates two receptors, TNFR1 and TNFR2, associated with opposite effects. Therefore, anti-inflammatory treatment with specific TNF receptor interference was compared to non-specific TNF inhibition regarding effects on heart, (neuro)inflammation, brain and behavior in mice with MI. METHODS Male C57BL/6 mice were subjected to MI or sham surgery. One hour later, MI mice were randomized to either non-specific TNF inhibition by Enbrel, specific TNFR1 antagonist-, or specific TNFR2 agonist treatment until the end of the protocol. Control sham and MI mice received saline. Behavioral evaluation was obtained day 10-14 after surgery. Eighteen days post-surgery, cardiac function was measured and mice were sacrificed. Blood and tissue samples were collected for analyses of (neuro)inflammation. RESULTS MI mice displayed left ventricular dysfunction, without heart failure, (neuro) inflammation or depressive-like behavior. Both receptor-specific interventions, but not Enbrel, doubled early post-MI mortality. TNFR2 agonist treatment improved left ventricular function and caused hyper-ramification of microglia, with no effect on depressive-like behavior. In contrast, TNFR1 antagonist treatment was associated with enhanced (neuro)inflammation: more plasma eosinophils and monocytes; increased plasma Lcn2 and hippocampal microglia and astrocyte activation. Moreover, increased baseline heart rate, with reduced beta-adrenergic responsiveness indicated sympathetic activation, and coincided with reduced exploratory behavior in the open field. Enbrel did not affect neuroinflammation nor behavior. CONCLUSION Early receptor interventions, but not non-specific TNF inhibition, increased mortality. Apart from this undesired effect, the general beneficial profile after TNFR2 stimulation, rather than the unfavourable effects of TNFR1 inhibition, would render TNFR2 stimulation preferable over non-specific TNF inhibition in MI with comorbid depression. However, follow-up studies regarding optimal timing and dosing are needed.
Collapse
Affiliation(s)
- L Gouweleeuw
- Department of Neurobiology, GELIFES, University of Groningen, the Netherlands
| | - H Wajant
- Department of Internal Medicine II, Division of Molecular Internal Medicine, University Hospital Wurzburg, Germany
| | - O Maier
- Institute of Cell Biology and Immunology, University of Stuttgart, Germany
| | - U L M Eisel
- Department of Neurobiology, GELIFES, University of Groningen, the Netherlands
| | - W M Blankesteijn
- Department of Pharmacology & Toxicology, CARIM, University of Maastricht, the Netherlands
| | - R G Schoemaker
- Department of Neurobiology, GELIFES, University of Groningen, the Netherlands; Department of Cardiology, University Medical Center Groningen, the Netherlands.
| |
Collapse
|
12
|
Kogel V, Trinh S, Gasterich N, Beyer C, Seitz J. Long-Term Glucose Starvation Induces Inflammatory Responses and Phenotype Switch in Primary Cortical Rat Astrocytes. J Mol Neurosci 2021; 71:2368-2382. [PMID: 33580474 PMCID: PMC8585803 DOI: 10.1007/s12031-021-01800-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/15/2021] [Indexed: 12/18/2022]
Abstract
Astrocytes are the most abundant cell type in the brain and crucial to ensure the metabolic supply of neurons and their synapse formation. Overnutrition as present in patients suffering from obesity causes astrogliosis in the hypothalamus. Other diseases accompanied by malnutrition appear to have an impact on the brain and astrocyte function. In the eating disorder anorexia nervosa (AN), patients suffer from undernutrition and develop volume reductions of the cerebral cortex, associated with reduced astrocyte proliferation and cell count. Although an effect on astrocytes and their function has already been shown for overnutrition, their role in long-term undernutrition remains unclear. The present study used primary rat cerebral cortex astrocytes to investigate their response to chronic glucose starvation. Cells were grown with a medium containing a reduced glucose concentration (2 mM) for 15 days. Long-term glucose starvation increased the expression of a subset of pro-inflammatory genes and shifted the primary astrocyte population to the pro-inflammatory A1-like phenotype. Moreover, genes encoding for proteins involved in the unfolded protein response were elevated. Our findings demonstrate that astrocytes under chronic glucose starvation respond with an inflammatory reaction. With respect to the multiple functions of astrocytes, an association between elevated inflammatory responses due to chronic starvation and alterations found in the brain of patients suffering from undernutrition seems possible.
Collapse
Affiliation(s)
- Vanessa Kogel
- Institute of Neuroanatomy, RWTH Aachen University, 52074, Aachen, Germany.
| | - Stefanie Trinh
- Institute of Neuroanatomy, RWTH Aachen University, 52074, Aachen, Germany
| | - Natalie Gasterich
- Institute of Neuroanatomy, RWTH Aachen University, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074, Aachen, Germany
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, RWTH Aachen University, 52074, Aachen, Germany
| |
Collapse
|
13
|
Sessa F, Messina G, Russo R, Salerno M, Castruccio Castracani C, Distefano A, Li Volti G, Calogero AE, Cannarella R, Mongioi' LM, Condorelli RA, La Vignera S. Consequences on aging process and human wellness of generation of nitrogen and oxygen species during strenuous exercise. Aging Male 2020; 23:14-22. [PMID: 29950140 DOI: 10.1080/13685538.2018.1482866] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Impairment of antioxidant defense system and increase in metabolic rate and production of reactive oxygen species have been demonstrated in strenuous exercise. Both at rest and during contractile activity, skeletal muscle generates a very complex set of reactive nitrogen and oxygen species; the main generated are superoxide and nitric oxide. The nature of the contractile activity influences the pattern and the magnitude of this reactive oxygen and nitrogen species (ROS) generation. The intracellular pro-oxidant/antioxidant homeostasis undergoes alteration owing to strenuous exercise and the major identified sources of intracellular free radical generation during physical activity are the mitochondrial electron transport chain, polymorphoneutrophil, and xanthine oxidase. Reactive oxygen species increased tissue susceptibility to oxidative damage and pose a serious threat to the cellular antioxidant defense system. The possible dangerous consequences of the aging process and human wellness are emphasized in this review.
Collapse
Affiliation(s)
- Francesco Sessa
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Raffaele Russo
- Department of Orthopaedic and Traumatology, Pellegrini Hospital, Naples, Italy
| | - Monica Salerno
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | | | - Alfio Distefano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Laura M Mongioi'
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
14
|
Donniacuo M, Urbanek K, Nebbioso A, Sodano L, Gallo L, Altucci L, Rinaldi B. Cardioprotective effect of a moderate and prolonged exercise training involves sirtuin pathway. Life Sci 2019; 222:140-147. [DOI: 10.1016/j.lfs.2019.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 12/21/2022]
|
15
|
Ragu-Varman D, Macedo-Mendoza M, Labrada-Moncada FE, Reyes-Ortega P, Morales T, Martínez-Torres A, Reyes-Haro D. Anorexia increases microglial density and cytokine expression in the hippocampus of young female rats. Behav Brain Res 2019; 363:118-125. [PMID: 30690107 DOI: 10.1016/j.bbr.2019.01.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 01/07/2019] [Accepted: 01/25/2019] [Indexed: 12/26/2022]
Abstract
Anorexia by osmotic dehydration is an adaptive response to hypernatremia and hyperosmolaemia induced by ingestion of a hypertonic solution. Dehydration-induced anorexia (DIA) reproduces weight loss and avoidance of food, despite its availability. By using this model, we previously showed increased reactive astrocyte density in the rat dorsal hippocampus, suggesting a pro-inflammatory environment where microglia may play an important role. However, whether such anorexic condition increases a pro-inflammatory response is unknown. The aim of this study was to test if DIA increases microglial density in the dorsal hippocampus, as well as the expression of pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6) and interleukin 1 beta (IL-1β) in the hippocampus of young female rats. Our results showed that DIA significantly increased microglial density in CA2-CA3 and dentate gyrus (DG) but not in CA1. However, forced food restriction (FFR) only increased microglial density in the DG. Accordingly, the activated/resting microglia ratio was significantly increased in CA2-CA3 and DG, in DIA and FFR groups. Finally, western blot analysis showed increased expression of IBA1, TNF-α, IL-6 and IL-1β in the hippocampus of both experimental groups. We conclude that anorexia triggers increased reactive microglial density and expression of TNF-α, IL-6 and IL-1β; this environment may result in hippocampal neuroinflammation.
Collapse
Affiliation(s)
- Durairaj Ragu-Varman
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla, Querétaro, CP76230, Mexico
| | - Mayra Macedo-Mendoza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla, Querétaro, CP76230, Mexico
| | - Francisco Emmanuel Labrada-Moncada
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla, Querétaro, CP76230, Mexico
| | - Pamela Reyes-Ortega
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla, Querétaro, CP76230, Mexico
| | - Teresa Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla, Querétaro, CP76230, Mexico
| | - Ataúlfo Martínez-Torres
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla, Querétaro, CP76230, Mexico
| | - Daniel Reyes-Haro
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla, Querétaro, CP76230, Mexico.
| |
Collapse
|
16
|
Nijs J, Loggia ML, Polli A, Moens M, Huysmans E, Goudman L, Meeus M, Vanderweeën L, Ickmans K, Clauw D. Sleep disturbances and severe stress as glial activators: key targets for treating central sensitization in chronic pain patients? Expert Opin Ther Targets 2017; 21:817-826. [DOI: 10.1080/14728222.2017.1353603] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Jo Nijs
- Department of physiotherapy, human physiology and anatomy, Pain in Motion International Research Group, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Physical Medicine and Physiotherapy, University Hospital Brussels, Brussels, Belgium
| | - Marco L. Loggia
- MGH/HST A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Andrea Polli
- Department of physiotherapy, human physiology and anatomy, Pain in Motion International Research Group, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Maarten Moens
- Department of Neurosurgery and Radiology, University Hospital Brussels, Brussels, Belgium
- Department of Manual Therapy, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Huysmans
- Department of physiotherapy, human physiology and anatomy, Pain in Motion International Research Group, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lisa Goudman
- Department of physiotherapy, human physiology and anatomy, Pain in Motion International Research Group, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Neurosurgery and Radiology, University Hospital Brussels, Brussels, Belgium
| | - Mira Meeus
- Department of physiotherapy, human physiology and anatomy, Pain in Motion International Research Group, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Ghent, Belgium
- Department of Rehabilitation Sciences and Physiotherapy, University of Antwerp, Antwerp, Belgium
| | - Luc Vanderweeën
- Department of physiotherapy, human physiology and anatomy, Pain in Motion International Research Group, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
- Private Practice for Spinal Manual Therapy, Schepdaal-Dilbeek, Belgium
| | - Kelly Ickmans
- Department of physiotherapy, human physiology and anatomy, Pain in Motion International Research Group, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Physical Medicine and Physiotherapy, University Hospital Brussels, Brussels, Belgium
| | - Daniel Clauw
- Chronic Pain and Fatigue Research Center, University of Michigan, Ann Arbor, USA
| |
Collapse
|
17
|
Koo JH, Jang YC, Hwang DJ, Um HS, Lee NH, Jung JH, Cho JY. Treadmill exercise produces neuroprotective effects in a murine model of Parkinson's disease by regulating the TLR2/MyD88/NF-κB signaling pathway. Neuroscience 2017; 356:102-113. [PMID: 28527958 DOI: 10.1016/j.neuroscience.2017.05.016] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 05/02/2017] [Accepted: 05/09/2017] [Indexed: 12/30/2022]
Abstract
Parkinson's disease (PD) is characterized by progressive dopamine depletion and a loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Treadmill exercise is a promising non-pharmacological approach for reducing the risk of PD and other neuroinflammatory disorders, such as Alzheimer's disease. The goal of this study was to investigate the effects of treadmill exercise on α-synuclein-induced neuroinflammation and neuronal cell death in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. Eight weeks of treadmill exercise improved motor deficits and reduced α-synuclein expression, a major causative factor of PD-like symptoms, in MPTP mice. Treadmill exercise also down-regulated the expression of toll-like receptor 2 and its associated downstream signaling molecules, including myeloid differentiation factor-88, tumor necrosis factor receptor-associated factor 6, and transforming growth factor-β-activated protein kinase 1. These effects were associated with reduced ionized calcium-binding adapter molecule 1 expression, decreased IκBα and nuclear transcription factor-κB phosphorylation, decreased tumor necrosis factor α and interleukin-1β expression, and decreased NADPH oxidase subunit expression in the SNpc and striatum. Additionally, it promoted the expression of tyrosine hydroxylase and the dopamine transporter, as well as plasma dopamine levels, in MPTP mice; these effects were associated with decreased caspase-3 expression and cleavage, as well as increased Bcl-2 expression in the SNpc. Taken together, our data suggest that treadmill exercise improves MPTP-associated motor deficits by exerting neuroprotective effects in the SNpc and striatum, supporting the notion that treadmill exercise is useful as a non-pharmacological tool for the management of PD.
Collapse
Affiliation(s)
- Jung-Hoon Koo
- Exercise Biochemistry Laboratory, Korea National Sport University, 1239, Yangjae, Songpa-gu, Seoul 05541, Republic of Korea; Institute of Sport Science, Korea National Sport University, 1239, Yangjae, Songpa-gu, Seoul 05541, Republic of Korea
| | - Yong-Chul Jang
- Exercise Biochemistry Laboratory, Korea National Sport University, 1239, Yangjae, Songpa-gu, Seoul 05541, Republic of Korea
| | - Dong-Ju Hwang
- Exercise Biochemistry Laboratory, Korea National Sport University, 1239, Yangjae, Songpa-gu, Seoul 05541, Republic of Korea
| | - Hyun-Seob Um
- Department of Exercise Prescription, Kon-Yang University, 119 Daehangro, Nonsan City, Chungnam 320-711, Republic of Korea
| | - Nam-Hee Lee
- Exercise Biochemistry Laboratory, Dan Kook University, Cheonan 330-714, Republic of Korea
| | - Jae-Hoon Jung
- Department of Physical Education, Han-Yang University, 222 Wangsibri-ro, sungdong-gu, Seoul 04763, Republic of Korea
| | - Joon-Yong Cho
- Exercise Biochemistry Laboratory, Korea National Sport University, 1239, Yangjae, Songpa-gu, Seoul 05541, Republic of Korea.
| |
Collapse
|
18
|
Guida F, Luongo L, Boccella S, Giordano ME, Romano R, Bellini G, Manzo I, Furiano A, Rizzo A, Imperatore R, Iannotti FA, D'Aniello E, Piscitelli F, Sca Rossi F, Cristino L, Di Marzo V, de Novellis V, Maione S. Palmitoylethanolamide induces microglia changes associated with increased migration and phagocytic activity: involvement of the CB2 receptor. Sci Rep 2017; 7:375. [PMID: 28336953 PMCID: PMC5428303 DOI: 10.1038/s41598-017-00342-1] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 02/22/2017] [Indexed: 12/22/2022] Open
Abstract
The endogenous fatty acid amide palmitoylethanolamide (PEA) has been shown to exert anti-inflammatory actions mainly through inhibition of the release of pro-inflammatory molecules from mast cells, monocytes and macrophages. Indirect activation of the endocannabinoid (eCB) system is among the several mechanisms of action that have been proposed to underlie the different effects of PEA in vivo. In this study, we used cultured rat microglia and human macrophages to evaluate whether PEA affects eCB signaling. PEA was found to increase CB2 mRNA and protein expression through peroxisome proliferator-activated receptor-α (PPAR-α) activation. This novel gene regulation mechanism was demonstrated through: (i) pharmacological PPAR-α manipulation, (ii) PPAR-α mRNA silencing, (iii) chromatin immunoprecipitation. Moreover, exposure to PEA induced morphological changes associated with a reactive microglial phenotype, including increased phagocytosis and migratory activity. Our findings suggest indirect regulation of microglial CB2R expression as a new possible mechanism underlying the effects of PEA. PEA can be explored as a useful tool for preventing/treating the symptoms associated with neuroinflammation in CNS disorders.
Collapse
Affiliation(s)
- F Guida
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - L Luongo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - S Boccella
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - M E Giordano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - R Romano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - G Bellini
- Department of Women, Child and General and Specialistic Surgery, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - I Manzo
- Department of Women, Child and General and Specialistic Surgery, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - A Furiano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - A Rizzo
- Department of Experimental Medicine, Section of Microbiology and Clinical Microbiology, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - R Imperatore
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Department of Science and Technology, University of Sannio, Benevento, Italy
| | - F A Iannotti
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - E D'Aniello
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - F Piscitelli
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - F Sca Rossi
- Department of Women, Child and General and Specialistic Surgery, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy
| | - L Cristino
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - V Di Marzo
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - V de Novellis
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy.,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy
| | - S Maione
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN), 80138, Naples, Italy. .,Endocannabinoid Research Group, Institute of Biomolecular Chemistry, C.N.R., Pozzuoli, Italy.
| |
Collapse
|
19
|
Messina G, Valenzano A, Moscatelli F, Salerno M, Lonigro A, Esposito T, Monda V, Corso G, Messina A, Viggiano A, Triggiani AI, Chieffi S, Guglielmi G, Monda M, Cibelli G. Role of Autonomic Nervous System and Orexinergic System on Adipose Tissue. Front Physiol 2017; 8:137. [PMID: 28344558 PMCID: PMC5344930 DOI: 10.3389/fphys.2017.00137] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 02/23/2017] [Indexed: 01/16/2023] Open
Abstract
Adipose tissue, defined as white adipose tissue (WAT) and brown adipose tissue (BAT), is a biological caloric reservoir; in response to over-nutrition it expands and, in response to energy deficit, it releases lipids. The WAT primarily stores energy as triglycerides, whereas BAT dissipates chemical energy as heat. In mammals, the BAT is a key site for heat production and an attractive target to promote weight loss. The autonomic nervous system (ANS) exerts a direct control at the cellular and molecular levels in adiposity. The sympathetic nervous system (SNS) provides a complex homeostatic control to specifically coordinate function and crosstalk of both fat pads, as indicated by the increase of the sympathetic outflow to BAT, in response to cold and high-fat diet, but also by the increase or decrease of the sympathetic outflow to selected WAT depots, in response to different lipolytic requirements of these two conditions. More recently, a role has been attributed to the parasympathetic nervous system (PNS) in modulating both adipose tissue insulin-mediated glucose uptake and fatty free acid (FFA) metabolism in an anabolic way and its endocrine function. The regulation of adipose tissue is unlikely to be limited to the autonomic control, since a number of signaling cytokines and neuropeptides play an important role, as well. In this review, we report some experimental evidences about the role played by both the ANS and orexins into different fat pads, related to food intake and energy expenditure, with a special emphasis on body weight status and fat mass (FM) content.
Collapse
Affiliation(s)
- Giovanni Messina
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy; Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Monica Salerno
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Antonio Lonigro
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Teresa Esposito
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Vincenzo Monda
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Gaetano Corso
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Antonietta Messina
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Andrea Viggiano
- Department of Medicine and Surgery, University of Salerno Salerno, Italy
| | - Antonio I Triggiani
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Sergio Chieffi
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Giuseppe Guglielmi
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| |
Collapse
|
20
|
Osteopontin: Relation between Adipose Tissue and Bone Homeostasis. Stem Cells Int 2017; 2017:4045238. [PMID: 28194185 PMCID: PMC5282444 DOI: 10.1155/2017/4045238] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 11/19/2016] [Accepted: 12/18/2016] [Indexed: 12/20/2022] Open
Abstract
Osteopontin (OPN) is a multifunctional protein mainly associated with bone metabolism and remodeling. Besides its physiological functions, OPN is implicated in the pathogenesis of a variety of disease states, such as obesity and osteoporosis. Importantly, during the last decades obesity and osteoporosis have become among the main threats to health worldwide. Because OPN is a protein principally expressed in cells with multifaceted effects on bone morphogenesis and remodeling and because it seems to be one of the most overexpressed genes in the adipose tissue of the obese contributing to osteoporosis, this mini review will highlight recent insights about relation between adipose tissue and bone homeostasis.
Collapse
|
21
|
Esposito T, Lobaccaro JM, Esposito MG, Monda V, Messina A, Paolisso G, Varriale B, Monda M, Messina G. Effects of low-carbohydrate diet therapy in overweight subjects with autoimmune thyroiditis: possible synergism with ChREBP. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2939-2946. [PMID: 27695291 PMCID: PMC5028075 DOI: 10.2147/dddt.s106440] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The thyroid is one of the metabolism regulating glands. Its function is to determine the amount of calories that the body has to burn to maintain normal weight. Thyroiditides are inflammatory processes that mainly result in autoimmune diseases. We have conducted the present study in order to have a clear picture of both autoimmune status and the control of body weight. We have evaluated the amount of either thyroid hormones, or antithyroid, or anti-microsomal, or anti-peroxidase antibodies (Abs) in patients with high amounts of Abs. In a diet devoid of carbohydrates (bread, pasta, fruit, and rice), free from goitrogenic food, and based on body mass index, the distribution of body mass and intracellular and extracellular water conducted for 3 weeks gives the following results: patients treated as above showed a significant reduction of antithyroid (−40%, P<0.013), anti-microsomal (−57%, P<0.003), and anti-peroxidase (−44%, P<0,029) Abs. Untreated patients had a significant increase in antithyroid (+9%, P<0.017) and anti-microsomal (+30%, P<0.028) Abs. Even the level of anti-peroxidase Abs increased without reaching statistical significance (+16%, P>0064). With regard to the body parameters measured in patients who followed this diet, reduction in body weight (−5%, P<0.000) and body mass index (−4%, P<0.000) were observed. Since 83% of patients with high levels of autoantibodies are breath test positive to lactase with a lactase deficit higher than 50%, this fact led us to hypothesize a correlation with carbohydrate-responsive element-binding protein and therefore a possible role of carbohydrate metabolism in the development and maintenance of autoimmune thyroiditis associated with body weight increase and slower basic metabolism.
Collapse
Affiliation(s)
- Teresa Esposito
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine; Laboratory of Molecular Biology and Genetics, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Jean Marc Lobaccaro
- UMR, Clermont Université, Centre de Recherche en Nutrition Humaine d'Auvergne, Aubière Cedex, France
| | | | - Vincenzo Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine
| | - Antonietta Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine
| | - Giuseppe Paolisso
- Department of Scienze Mediche, Chirurgiche, Neurologiche, Metaboliche e dell'Invecchiamento, Second University of Naples, Naples
| | - Bruno Varriale
- Laboratory of Molecular Biology and Genetics, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine
| | - Giovanni Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine; Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
22
|
Anorexia Reduces GFAP+ Cell Density in the Rat Hippocampus. Neural Plast 2016; 2016:2426413. [PMID: 27579183 PMCID: PMC4992534 DOI: 10.1155/2016/2426413] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 05/18/2016] [Accepted: 06/16/2016] [Indexed: 11/17/2022] Open
Abstract
Anorexia nervosa is an eating disorder observed primarily in young women. The neurobiology of the disorder is unknown but recently magnetic resonance imaging showed a volume reduction of the hippocampus in anorexic patients. Dehydration-induced anorexia (DIA) is a murine model that mimics core features of this disorder, including severe weight loss due to voluntary reduction in food intake. The energy supply to the brain is mediated by astrocytes, but whether their density is compromised by anorexia is unknown. Thus, the aim of this study was to estimate GFAP+ cell density in the main regions of the hippocampus (CA1, CA2, CA3, and dentate gyrus) in the DIA model. Our results showed that GFAP+ cell density was significantly reduced (~20%) in all regions of the hippocampus, except in CA1. Interestingly, DIA significantly reduced the GFAP+ cells/nuclei ratio in CA2 (−23%) and dentate gyrus (−48%). The reduction of GFAP+ cell density was in agreement with a lower expression of GFAP protein. Additionally, anorexia increased the expression of the intermediate filaments vimentin and nestin. Accordingly, anorexia increased the number of reactive astrocytes in CA2 and dentate gyrus more than twofold. We conclude that anorexia reduces the hippocampal GFAP+ cell density and increases vimentin and nestin expression.
Collapse
|
23
|
Functional Assessment of Corticospinal System Excitability in Karate Athletes. PLoS One 2016; 11:e0155998. [PMID: 27218465 PMCID: PMC4878742 DOI: 10.1371/journal.pone.0155998] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/06/2016] [Indexed: 12/04/2022] Open
Abstract
Objectives To investigate the involvement of the primary motor cortex (M1) in the coordination performance of karate athletes through transcranial magnetic stimulation (TMS). Methods Thirteen right-handed male karate athletes (25.0±5.0 years) and 13 matched non-athlete controls (26.7±6.2 years) were enrolled. A single-pulse TMS was applied using a figure-eight coil stimulator. Resting motor threshold (rMT) was determined. Surface electromyography was recorded from the first dorsal interosseous muscle. Motor evoked potential (MEP) latencies and amplitudes at rMT, 110%, and 120% of rMT were considered. Functional assessment of the coordination performance was assessed by in-phase (IP) and anti-phase (AP) homolateral hand and foot coordination tasks performed at 80, 120, and 180 bpm. Results Compared to controls, athletes showed lower rMT (p<0.01), shorter MEP latency (p<0.01) and higher MEP amplitude (p<0.01), with a significant correlation (r = 0.50, p<0.01) between rMT and MEP latency. Coordination decreased with increasing velocity, and better IP performances emerged compared to AP ones (p<0.001). In general, a high correlation between rMT and coordination tasks was found for both IP and AP conditions. Conclusion With respect to controls, karate athletes present a higher corticospinal excitability indicating the presence of an activity-dependent alteration in the balance and interactions between inhibitory and facilitatory circuits determining the final output from the M1. Furthermore, the high correlation between corticospinal excitability and coordination performance could support sport-specific neurophysiological arrangements.
Collapse
|
24
|
Viggiano E, Monda V, Messina A, Moscatelli F, Valenzano A, Tafuri D, Cibelli G, De Luca B, Messina G, Monda M. Cortical spreading depression produces a neuroprotective effect activating mitochondrial uncoupling protein-5. Neuropsychiatr Dis Treat 2016; 12:1705-10. [PMID: 27468234 PMCID: PMC4946829 DOI: 10.2147/ndt.s107074] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Depression of electrocorticogram propagating over the cortex surface results in cortical spreading depression (CSD), which is probably related to the pathophysiology of stroke, epilepsy, and migraine. However, preconditioning with CSD produces neuroprotection to subsequent ischemic episodes. Such effects require the expression or activation of several genes, including neuroprotective ones. Recently, it has been demonstrated that the expression of the uncoupling proteins (UCPs) 2 and 5 is amplified during brain ischemia and their expression exerts a long-term effect upon neuron protection. To evaluate the neuroprotective consequence of CSD, the expression of UCP-5 in the brain cortex was measured following CSD induction. CSD was evoked in four samples of rats, which were sacrificed after 2 hours, 4 hours, 6 hours, and 24 hours. Western blot analyses were carried out to measure UCP-5 concentrations in the prefrontal cortices of both hemispheres, and immunohistochemistry was performed to determine the localization of UCP-5 in the brain cortex. The results showed a significant elevation in UCP-5 expression at 24 hours in all cortical strata. Moreover, UCP-5 was triggered by CSD, indicating that UCP-5 production can have a neuroprotective effect.
Collapse
Affiliation(s)
- Emanuela Viggiano
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Second University of Naples, Naples; Department of Medicine, University of Padua, Padua
| | - Vincenzo Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Second University of Naples, Naples
| | - Antonietta Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Second University of Naples, Naples
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia
| | - Domenico Tafuri
- Department of Motor Sciences and Wellness, University of Naples "Parthenope", Naples, Italy
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia
| | - Bruno De Luca
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Second University of Naples, Naples
| | - Giovanni Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Second University of Naples, Naples; Department of Clinical and Experimental Medicine, University of Foggia, Foggia
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Second University of Naples, Naples
| |
Collapse
|