1
|
Chu XL, Zhao XX, Liu SY, Li YJ, Ding N, Liu MQ, Li QW, Li Q. Research progress in different physical therapies for treating peripheral nerve injuries. Front Neurol 2025; 16:1508604. [PMID: 40260135 PMCID: PMC12009707 DOI: 10.3389/fneur.2025.1508604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/28/2025] [Indexed: 04/23/2025] Open
Abstract
Physical therapy is gaining recognition as an effective therapeutic approach in the realm of peripheral nerve injury (PNI) research. This article seeks to provide a comprehensive review of the latest advancements, applications, and mechanisms of action of four physical therapy modalities-ultrasound, electrical stimulation, photobiomodulation, and aerobic exercise-in the context of PNI. Ultrasound, characterized by its mechanical and thermal effects, is widely regarded as an effective non-invasive or minimally invasive method for neural modulation. Electrical stimulation therapy, a prevalent technique in PNI treatment, entails the application of electric currents to stimulate nerve and muscle tissues, thereby facilitating nerve regeneration and mitigating muscle atrophy. Photobiomodulation, a process that influences cell metabolism through the absorption of photon energy, is closely associated with neural regeneration in the field of rehabilitation medicine. Additionally, aerobic exercise, a popular form of physical activity, serves to enhance blood circulation and improve neuronal function. The article discusses various physical therapy methods for peripheral nerve injuries, including hyperbaric oxygen therapy, magnetic therapy, and biofeedback therapy, in addition to traditional approaches. Despite advancements, challenges in nerve injury treatment persist, such as the need for standardized treatment protocols, consideration of individual variations, and assessment of long-term effectiveness. Future research is needed to address these issues. In summary, this article offers theoretical and empirical evidence supporting the utilization of physical therapy in the management of PNI. This research aims to promote further research and clinical practice in this field, contributing to enhancing patient quality of life and recovery outcomes.
Collapse
Affiliation(s)
- Xiao-Lei Chu
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| | - Xiao-Xuan Zhao
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Shuai-Yi Liu
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Ya-Jie Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Ning Ding
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Min-Qi Liu
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Qing-Wen Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Qi Li
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| |
Collapse
|
2
|
Alzu'bi A, Abu-El-Rub E, Almahasneh F, Tahat L, Athamneh RY, Khasawneh R, Alzoubi H, Ghorab DS, Almazari R, Zoubi MSA, Al-Zoubi RM. Delineating the molecular mechanisms of hippocampal neurotoxicity induced by chronic administration of synthetic cannabinoid AB-FUBINACA in mice. Neurotoxicology 2024; 103:50-59. [PMID: 38823587 DOI: 10.1016/j.neuro.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
Chronic use of synthetic cannabinoids (SCs) has been associated with cognitive and behavioural deficits and an increased risk of neuropsychiatric disorders. The underlying molecular and cellular mechanisms of the neurotoxic effects of long-term use of SCs have not been well investigated in the literature. Herein, we evaluated the in vivo effects of chronic administration of AB-FUBINACA on the hippocampus in mice. Our results revealed that the administration of AB-FUBINACA induced a significant impairment in recognition memory associated with histopathological changes in the hippocampus. These findings were found to be correlated with increased level of oxidative stress, neuroinflammation, and apoptosis markers, and reduced expression of brain-derived neurotrophic factor (BDNF), which plays an essential role in modulating synaptic plasticity integral for promoting learning and memory in the hippocampus. Additionally, we showed that AB-FUBINACA significantly decreased the expression of NR1, an important functional subunit of glutamate/NMDA receptors and closely implicated in the development of toxic psychosis. These findings shed light on the long-term neurotoxic effects of SCs on hippocampus and the underlying mechanisms of these effects. This study provided new targets for possible medical interventions to improve the treatment guidelines for SCs addiction.
Collapse
Affiliation(s)
- Ayman Alzu'bi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan.
| | - Ejlal Abu-El-Rub
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Fatimah Almahasneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Lena Tahat
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 211-63, Jordan
| | - Rabaa Y Athamneh
- Department of Medical Laboratory Sciences, Faculty of Allied Science, Zarqa University, Zarqa 13110, Jordan
| | - Ramada Khasawneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Hiba Alzoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Doaa S Ghorab
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan; Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rawan Almazari
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Mazhar Salim Al Zoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Raed M Al-Zoubi
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha, Qatar; Department of Biomedical Sciences, QU-Health, College of Health Sciences, Qatar University, Doha 2713, Qatar; Department of Chemistry, Jordan University of Science and Technology, P.O.Box 3030, Irbid 22110, Jordan.
| |
Collapse
|
3
|
Bellver-Sanchis A, Geng Q, Navarro G, Ávila-López PA, Companys-Alemany J, Marsal-García L, Larramona-Arcas R, Miró L, Perez-Bosque A, Ortuño-Sahagún D, Banerjee DR, Choudhary BS, Soriano FX, Poulard C, Pallàs M, Du HN, Griñán-Ferré C. G9a Inhibition Promotes Neuroprotection through GMFB Regulation in Alzheimer's Disease. Aging Dis 2024; 15:311-337. [PMID: 37307824 PMCID: PMC10796087 DOI: 10.14336/ad.2023.0424-2] [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: 01/22/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023] Open
Abstract
Epigenetic alterations are a fundamental pathological hallmark of Alzheimer's disease (AD). Herein, we show the upregulation of G9a and H3K9me2 in the brains of AD patients. Interestingly, treatment with a G9a inhibitor (G9ai) in SAMP8 mice reversed the high levels of H3K9me2 and rescued cognitive decline. A transcriptional profile analysis after G9ai treatment revealed increased gene expression of glia maturation factor β (GMFB) in SAMP8 mice. Besides, a H3K9me2 ChIP-seq analysis after G9a inhibition treatment showed the enrichment of gene promoters associated with neural functions. We observed the induction of neuronal plasticity and a reduction of neuroinflammation after G9ai treatment, and more strikingly, these neuroprotective effects were reverted by the pharmacological inhibition of GMFB in mice and cell cultures; this was also validated by the RNAi approach generating the knockdown of GMFB/Y507A.10 in Caenorhabditis elegans. Importantly, we present evidence that GMFB activity is controlled by G9a-mediated lysine methylation as well as we identified that G9a directly bound GMFB and catalyzed the methylation at lysine (K) 20 and K25 in vitro. Furthermore, we found that the neurodegenerative role of G9a as a GMFB suppressor would mainly rely on methylation of the K25 position of GMFB, and thus G9a pharmacological inhibition removes this methylation promoting neuroprotective effects. Then, our findings confirm an undescribed mechanism by which G9a inhibition acts at two levels, increasing GMFB and regulating its function to promote neuroprotective effects in age-related cognitive decline.
Collapse
Affiliation(s)
- Aina Bellver-Sanchis
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Qizhi Geng
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, RNA Institute, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Department Biochemistry and Physiology, Faculty of Pharmacy. Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Pedro A. Ávila-López
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| | - Júlia Companys-Alemany
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Laura Marsal-García
- Department of Biochemistry, McGill University, Montréal, Québec, Canada.
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Québec, Canada.
| | - Raquel Larramona-Arcas
- Department of Cell Biology, Physiology, and Immunology, Celltec-UB, University of Barcelona, Barcelona, Spain, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Lluisa Miró
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Anna Perez-Bosque
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunología Molecular, Instituto de Investigación de Ciencias Biomédicas (IICB) CUCS, Universidad de Guadalajara, Jalisco 44340, México.
| | | | - Bhanwar Singh Choudhary
- Department of Pharmacy, Central University of Rajasthan, Ajmer, Rajasthan, India.
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana, Gujarat, India.
| | - Francesc X Soriano
- Department of Cell Biology, Physiology, and Immunology, Celltec-UB, University of Barcelona, Barcelona, Spain, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Coralie Poulard
- Cancer Research Cancer Lyon, Université de Lyon, F-69000 Lyon, France.
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.
- CNRS UMR5286, Centre de Recherche en Cancérlogie de Lyon, F-69000 Lyon, France.
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - Hai-Ning Du
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, RNA Institute, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Sancho-Alonso M, Arenas YM, Izquierdo-Altarejos P, Martinez-Garcia M, Llansola M, Felipo V. Enhanced Activation of the S1PR2-IL-1β-Src-BDNF-TrkB Pathway Mediates Neuroinflammation in the Hippocampus and Cognitive Impairment in Hyperammonemic Rats. Int J Mol Sci 2023; 24:17251. [PMID: 38139078 PMCID: PMC10744193 DOI: 10.3390/ijms242417251] [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: 11/10/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Hyperammonemia contributes to hepatic encephalopathy. In hyperammonemic rats, cognitive function is impaired by altered glutamatergic neurotransmission induced by neuroinflammation. The underlying mechanisms remain unclear. Enhanced sphingosine-1-phosphate receptor 2 (S1PR2) activation in the cerebellum of hyperammonemic rats contributes to neuroinflammation. in In hyperammonemic rats, we assessed if blocking S1PR2 reduced hippocampal neuroinflammation and reversed cognitive impairment and if the signaling pathways were involved. S1PR2 was blocked with intracerebral JTE-013, and cognitive function was evaluated. The signaling pathways inducing neuroinflammation and altered glutamate receptors were analyzed in hippocampal slices. JTE-013 improved cognitive function in the hyperammonemic rats, and hyperammonemia increased S1P. This increased IL-1β, which enhanced Src activity, increased CCL2, activated microglia and increased the membrane expression of the NMDA receptor subunit GLUN2B. This increased p38-MAPK activity, which altered the membrane expression of AMPA receptor subunits and increased BDNF, which activated the TrkB → PI3K → Akt → CREB pathway, inducing sustained neuroinflammation. This report unveils key pathways involved in the induction and maintenance of neuroinflammation in the hippocampus of hyperammonemic rats and supports S1PR2 as a therapeutic target for cognitive impairment.
Collapse
Affiliation(s)
- María Sancho-Alonso
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
- Systems Neuropharmacology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Yaiza M. Arenas
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
| | - Paula Izquierdo-Altarejos
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
| | - Mar Martinez-Garcia
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
| | - Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
| |
Collapse
|
5
|
He Q, Li Z, Li T, Zhang Z, Zhao J. ATP Stimulation Promotes Functional Recovery after Intracerebral Haemorrhage by Increasing the mBDNF/proBDNF Ratio. Neuroscience 2021; 459:104-117. [PMID: 33421569 DOI: 10.1016/j.neuroscience.2020.12.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/20/2022]
Abstract
Brain-derived neurotrophic factor (BDNF), including mature BDNF (mBDNF) and precursor BDNF (proBDNF), plays a pivotal role in neuronal survival, synaptic plasticity and neurogenesis. However, the functional effect of the mBDNF/proBDNF ratio in haemorrhagic stroke remains unclear. ATP is a known mediator of BDNF production in neurons and glia. Therefore, we hypothesized that ATP could facilitate BDNF production, increase the mBDNF/proBDNF ratio and thereby alleviate cerebral haemorrhage-induced injury. In this experiment, a model of intracerebral haemorrhage (ICH) was produced by injecting 50 μL autologous blood into the right corpus striatum in healthy male rats. ATP was injected to promote BDNF production and increase the mBDNF/proBDNF ratio. After ATP pretreatment, P2X4R-shRNA and SB203580 were used to inhibit P2X4R and p38-MAPK, respectively. We provide direct evidence that ATP administration was successful in promoting mBDNF expression and increasing the mBDNF/proBDNF ratio after ICH injury. Additionally, ATP stimulation could significantly improve cerebral neurological function and alleviate neuronal damage. Furthermore, ATP injection was able to upregulate the expression of P2X4R and p-p38-MAPK. Moreover, both P2X4R-shRNA and SB203580 could effectively abolish the effect of ATP injection on the levels of P2X4R and p-p38-MAPK and the mBDNF/proBDNF ratio. Together, these findings show that ATP stimulation contributes to functional recovery after cerebral haemorrhage and that neuroprotection induced by ATP administration in ICH rats is accompanied by a strong increase in the mBDNF/proBDNF ratio. Here, we also show a significant role of P2X4R-p38-MAPK signalling in the ATP-induced increase in the mBDNF/proBDNF ratio in ICH.
Collapse
Affiliation(s)
- Qi He
- The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhenyu Li
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Tiegang Li
- Institute of Materia Medica, Peking Union Medical College Hospital, Peking, People's Republic of China
| | - Zhiqian Zhang
- The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jing Zhao
- Department of Pathophysiology, Chongqing Medical University, Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
6
|
Changes in Hippocampal Plasticity in Depression and Therapeutic Approaches Influencing These Changes. Neural Plast 2020; 2020:8861903. [PMID: 33293948 PMCID: PMC7718046 DOI: 10.1155/2020/8861903] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/30/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Depression is a common neurological disease that seriously affects human health. There are many hypotheses about the pathogenesis of depression, and the most widely recognized and applied is the monoamine hypothesis. However, no hypothesis can fully explain the pathogenesis of depression. At present, the brain-derived neurotrophic factor (BDNF) and neurogenesis hypotheses have highlighted the important role of plasticity in depression. The plasticity of neurons and glial cells plays a vital role in the transmission and integration of signals in the central nervous system. Plasticity is the adaptive change in the nervous system in response to changes in external signals. The hippocampus is an important anatomical area associated with depression. Studies have shown that some antidepressants can treat depression by changing the plasticity of the hippocampus. Furthermore, caloric restriction has also been shown to affect antidepressant and hippocampal plasticity changes. In this review, we summarize the latest research, focusing on changes in the plasticity of hippocampal neurons and glial cells in depression and the role of BDNF in the changes in hippocampal plasticity in depression, as well as caloric restriction and mitochondrial plasticity. This review may contribute to the development of antidepressant drugs and elucidating the mechanism of depression.
Collapse
|
7
|
Engel DF, Bobbo VCD, Solon CS, Nogueira GA, Moura-Assis A, Mendes NF, Zanesco AM, Papangelis A, Ulven T, Velloso LA. Activation of GPR40 induces hypothalamic neurogenesis through p38- and BDNF-dependent mechanisms. Sci Rep 2020; 10:11047. [PMID: 32632088 PMCID: PMC7338363 DOI: 10.1038/s41598-020-68110-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Hypothalamic adult neurogenesis provides the basis for renewal of neurons involved in the regulation of whole-body energy status. In addition to hormones, cytokines and growth factors, components of the diet, particularly fatty acids, have been shown to stimulate hypothalamic neurogenesis; however, the mechanisms behind this action are unknown. Here, we hypothesized that GPR40 (FFAR1), the receptor for medium and long chain unsaturated fatty acids, could mediate at least part of the neurogenic activity in the hypothalamus. We show that a GPR40 ligand increased hypothalamic cell proliferation and survival in adult mice. In postnatal generated neurospheres, acting in synergy with brain-derived neurotrophic factor (BDNF) and interleukin 6, GPR40 activation increased the expression of doublecortin during the early differentiation phase and of the mature neuronal marker, microtubule-associated protein 2 (MAP2), during the late differentiation phase. In Neuro-2a proliferative cell-line GPR40 activation increased BDNF expression and p38 activation. The chemical inhibition of p38 abolished GPR40 effect in inducing neurogenesis markers in neurospheres, whereas BDNF immunoneutralization inhibited GPR40-induced cell proliferation in the hypothalamus of adult mice. Thus, GPR40 acts through p38 and BDNF to induce hypothalamic neurogenesis. This study provides mechanistic advance in the understating of how a fatty acid receptor regulates adult hypothalamic neurogenesis.
Collapse
Affiliation(s)
- Daiane F Engel
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil.
| | - Vanessa C D Bobbo
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Carina S Solon
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Guilherme A Nogueira
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Alexandre Moura-Assis
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Natalia F Mendes
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Ariane M Zanesco
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Athanasios Papangelis
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Trond Ulven
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Licio A Velloso
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil.
| |
Collapse
|
8
|
Choudhury H, Chellappan DK, Sengupta P, Pandey M, Gorain B. Adenosine Receptors in Modulation of Central Nervous System Disorders. Curr Pharm Des 2020; 25:2808-2827. [PMID: 31309883 DOI: 10.2174/1381612825666190712181955] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022]
Abstract
The ubiquitous signaling nucleoside molecule, adenosine is found in different cells of the human body to provide its numerous pharmacological role. The associated actions of endogenous adenosine are largely dependent on conformational change of the widely expressed heterodimeric G-protein-coupled A1, A2A, A2B, and A3 adenosine receptors (ARs). These receptors are well conserved on the surface of specific cells, where potent neuromodulatory properties of this bioactive molecule reflected by its easy passage through the rigid blood-brainbarrier, to simultaneously act on the central nervous system (CNS). The minimal concentration of adenosine in body fluids (30-300 nM) is adequate to exert its neuromodulatory action in the CNS, whereas the modulatory effect of adenosine on ARs is the consequence of several neurodegenerative diseases. Modulatory action concerning the activation of such receptors in the CNS could be facilitated towards neuroprotective action against such CNS disorders. Our aim herein is to discuss briefly pathophysiological roles of adenosine on ARs in the modulation of different CNS disorders, which could be focused towards the identification of potential drug targets in recovering accompanying CNS disorders. Researches with active components with AR modulatory action have been extended and already reached to the bedside of the patients through clinical research in the improvement of CNS disorders. Therefore, this review consist of recent findings in literatures concerning the impact of ARs on diverse CNS disease pathways with the possible relevance to neurodegeneration.
Collapse
Affiliation(s)
- Hira Choudhury
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Dinesh K Chellappan
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Pallav Sengupta
- Department of Physiology, Faculty of Medicine, MA`HSA University, Kuala Lumpur, Malaysia
| | - Manisha Pandey
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Science, Taylor's University, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
9
|
Fukumoto M, Takeuchi T, Koubayashi E, Harada S, Ota K, Kojima Y, Higuchi K. Induction of brain-derived neurotrophic factor in enteric glial cells stimulated by interleukin-1β via a c-Jun N-terminal kinase pathway. J Clin Biochem Nutr 2019; 66:103-109. [PMID: 32231405 DOI: 10.3164/jcbn.19-55] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/29/2019] [Indexed: 12/12/2022] Open
Abstract
Brain-derived neurotrophic factor exhibits neurotropic and neuroprotective functions and is increased in the colonic mucosa of patients with irritable bowel syndrome in correlation with the severity and frequency of abdominal pain. However, there are no reports of brain-derived neurotrophic factor expression in enteric glial cells. We evaluated the mRNA and protein expressions of brain-derived neurotrophic factor in enteric glial cells and culture medium and levels of mitogen-activated protein kinase after stimulation with interleukin-1β. Brain-derived neurotrophic factor mRNA expression was increased by interleukin-1β (3.125-75 ng/ml) and time-dependently increased 3-fold (24 h) and 4-fold (48 h) by interleukin-1β (50 ng/ml). Pro- and mature brain-derived neurotrophic factor proteins were both significantly increased at 48 h by interleukin-1β. However, the mature form was predominant in the cultured medium. Interleukin-1β increased phosphorylated-p38 mitogen-activated protein kinase expressions 2-fold higher at 5 and 15 min, and also phosphorylated-c-Jun N-terminal kinase expression 5-fold at 5 min and 10-fold at 15 min. Prior treatment with phosphorylated-c-Jun N-terminal kinase inhibitors decreased interleukin-1β-induced brain-derived neurotrophic factor by 50%. Thus, brain-derived neurotrophic factor expression was induced by interleukin-1β in enteric glial cells via a phosphorylated-c-Jun N-terminal kinase pathway, which might affect the enteric nervous system during stress.
Collapse
Affiliation(s)
- Masanobu Fukumoto
- Internal Medicine (II), Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| | - Toshihisa Takeuchi
- Internal Medicine (II), Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| | - Eiko Koubayashi
- Internal Medicine (II), Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| | - Satoshi Harada
- Internal Medicine (II), Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| | - Kazuhiro Ota
- Internal Medicine (II), Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| | - Yuichi Kojima
- Internal Medicine (II), Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| | - Kazuhide Higuchi
- Internal Medicine (II), Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| |
Collapse
|
10
|
Electrical stimulation induces neurite outgrowth in PC12m3 cells via the p38 mitogen-activated protein kinase pathway. Neurosci Lett 2019; 698:81-84. [DOI: 10.1016/j.neulet.2019.01.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/29/2018] [Accepted: 01/07/2019] [Indexed: 11/20/2022]
|
11
|
Brain-Derived Neurotrophic Factor Is Required for the Neuroprotective Effect of Mifepristone on Immature Purkinje Cells in Cerebellar Slice Culture. Int J Mol Sci 2019; 20:ijms20020285. [PMID: 30642045 PMCID: PMC6359295 DOI: 10.3390/ijms20020285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/02/2022] Open
Abstract
Endogenous γ-aminobutyric acid (GABA)-dependent activity induces death of developing Purkinje neurons in mouse organotypic cerebellar cultures and the synthetic steroid mifepristone blocks this effect. Here, using brain-derived neurotrophic factor (BDNF) heterozygous mice, we show that BDNF plays no role in immature Purkinje cell death. However, interestingly, BDNF haploinsufficiency impairs neuronal survival induced by mifepristone and GABAA-receptors antagonist (bicuculline) treatments, indicating that the underlying neuroprotective mechanism requires the neurotrophin full expression.
Collapse
|
12
|
Chauhan G, Ray K, Sahu S, Roy K, Jain V, Wadhwa M, Panjwani U, Kishore K, Singh S. Adenosine A1 receptor antagonist mitigates deleterious effects of sleep deprivation on adult neurogenesis and spatial reference memory in rats. Neuroscience 2016; 337:107-116. [DOI: 10.1016/j.neuroscience.2016.09.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/30/2016] [Accepted: 09/05/2016] [Indexed: 01/19/2023]
|
13
|
Becker BK, Wang HJ, Tian C, Zucker IH. BDNF contributes to angiotensin II-mediated reductions in peak voltage-gated K+ current in cultured CATH.a cells. Physiol Rep 2015; 3:3/11/e12598. [PMID: 26537343 PMCID: PMC4673628 DOI: 10.14814/phy2.12598] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Increased central angiotensin II (Ang II) levels contribute to sympathoexcitation in cardiovascular disease states such as chronic heart failure and hypertension. One mechanism by which Ang II increases neuronal excitability is through a decrease in voltage-gated, rapidly inactivating K+ current (IA); however, little is known about how Ang II signaling results in reduced IA. Brain-derived neurotrophic factor (BDNF) has also been demonstrated to decrease IA and has signaling components common to Ang II. Therefore, we hypothesized that Ang II-mediated suppression of voltage-gated K+ currents is due, in part, to BDNF signaling. Differentiated CATH.a, catecholaminergic cell line treated with BDNF for 2 h exhibited a reduced IA in a manner similar to that of Ang II treatment as demonstrated by whole-cell patch-clamp analysis. Inhibiting BDNF signaling by pretreating neurons with an antibody against BDNF significantly attenuated the Ang II-induced reduction of IA. Inhibition of a common component of both BDNF and Ang II signaling, p38 MAPK, with SB-203580 attenuated the BDNF-mediated reductions in IA. These results implicate the involvement of BDNF signaling in Ang II-induced reductions of IA, which may cause increases in neuronal sensitivity and excitability. We therefore propose that BDNF may be a necessary component of the mechanism by which Ang II reduces IA in CATH.a cells.
Collapse
Affiliation(s)
- Bryan K Becker
- Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Han-Jun Wang
- Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Changhai Tian
- Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Irving H Zucker
- Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
14
|
Huang W, Lv B, Zeng H, Shi D, Liu Y, Chen F, Li F, Liu X, Zhu R, Yu L, Jiang X. Paracrine Factors Secreted by MSCs Promote Astrocyte Survival Associated With GFAP Downregulation After Ischemic Stroke via p38 MAPK and JNK. J Cell Physiol 2015; 230:2461-75. [DOI: 10.1002/jcp.24981] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 02/23/2015] [Accepted: 03/02/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Weiyi Huang
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Bingke Lv
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Huijun Zeng
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Dandan Shi
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Yi Liu
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Fanfan Chen
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Feng Li
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Xinghui Liu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Rong Zhu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Lei Yu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Xiaodan Jiang
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| |
Collapse
|
15
|
Wang P, Chen FX, Du C, Li CQ, Yu YB, Zuo XL, Li YQ. Increased production of BDNF in colonic epithelial cells induced by fecal supernatants from diarrheic IBS patients. Sci Rep 2015; 5:10121. [PMID: 25998025 PMCID: PMC4441152 DOI: 10.1038/srep10121] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 03/30/2015] [Indexed: 12/27/2022] Open
Abstract
Colonic brain-derived neurotrophic factor (BDNF) plays an essential role in pathogenesis of abdominal pain in diarrhea-predominant irritable bowel syndrome (IBS-D), but regulation on its expression remains unclear. We investigated the role of fecal supernatants (FSN) from IBS-D patients on regulating BDNF expression in colonic epithelial cells of human and mice. Using human Caco-2 cells, we found that IBS-D FSN significantly increased BDNF mRNA and protein levels compared to control FSN, which were remarkably suppressed by the serine protease inhibitor. To further explore the potential mechanisms, we investigated the impact of protease-activated receptor-2 (PAR-2) on BDNF expression. We found a significant increase in PAR-2 expression in Caco-2 after IBS-D FSN stimulation. Knockdown of PAR-2 significantly inhibited IBS-D FSN-induced upregulation of BDNF. Moreover, we found that phosphorylation of p38 MAPK, not NF-κB p65, contributed to PAR-2-mediated BDNF overexpression. To confirm these results, we intracolonically infused IBS-D or control FSN in mice and found that IBS-D FSN significantly elevated colonic BDNF and visceral hypersensitivity in mice, which were both suppressed by the inhibitor of serine protease or antagonist of PAR-2. Together, our data indicate that activation of PAR-2 signaling by IBS-D FSN promotes expression of colonic BDNF, thereby contributing to IBS-like visceral hypersensitivity.
Collapse
Affiliation(s)
- Peng Wang
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Fei-Xue Chen
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Chao Du
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Chang-Qing Li
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Yan-Bo Yu
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Xiu-Li Zuo
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Yan-Qing Li
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| |
Collapse
|
16
|
Ittner AA, Gladbach A, Bertz J, Suh LS, Ittner LM. p38 MAP kinase-mediated NMDA receptor-dependent suppression of hippocampal hypersynchronicity in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2014; 2:149. [PMID: 25331068 PMCID: PMC4212118 DOI: 10.1186/s40478-014-0149-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/08/2014] [Indexed: 11/10/2022] Open
Abstract
Hypersynchronicity of neuronal brain circuits is a feature of Alzheimer's disease (AD). Mouse models of AD expressing mutated forms of the amyloid-β precursor protein (APP), a central protein involved in AD pathology, show cortical hypersynchronicity. We studied hippocampal circuitry in APP23 transgenic mice using telemetric electroencephalography (EEG), at the age of onset of memory deficits. APP23 mice display spontaneous hypersynchronicity in the hippocampus including epileptiform spike trains. Furthermore, spectral contributions of hippocampal theta and gamma oscillations are compromised in APP23 mice, compared to non-transgenic controls. Using cross-frequency coupling analysis, we show that hippocampal gamma amplitude modulation by theta phase is markedly impaired in APP23 mice. Hippocampal hypersynchronicity and waveforms are differentially modulated by injection of riluzole and the non-competitive N-methyl-D-aspartate (NMDA) receptor inhibitor MK801, suggesting specific involvement of voltage-gated sodium channels and NMDA receptors in hypersynchronicity thresholds in APP23 mice. Furthermore, APP23 mice show marked activation of p38 mitogen-activated protein (MAP) kinase in hippocampus, and injection of MK801 but not riluzole reduces activation of p38 in the hippocampus. A p38 inhibitor induces hypersynchronicity in APP23 mice to a similar extent as MK801, thus supporting suppression of hypersynchronicity involves NMDA receptors-mediated p38 activity. In summary, we characterize components of hippocampal hypersynchronicity, waveform patterns and cross-frequency coupling in the APP23 mouse model by pharmacological modulation, furthering the understanding of epileptiform brain activity in AD.
Collapse
|
17
|
Neu3 sialidase-mediated ganglioside conversion is necessary for axon regeneration and is blocked in CNS axons. J Neurosci 2014; 34:2477-92. [PMID: 24523539 DOI: 10.1523/jneurosci.4432-13.2014] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PNS axons have a high intrinsic regenerative ability, whereas most CNS axons show little regenerative response. We show that activation of Neu3 sialidase, also known as Neuraminidase-3, causing conversion of GD1a and GT1b to GM1 ganglioside, is an essential step in regeneration occurring in PNS (sensory) but not CNS (retinal) axons in adult rat. In PNS axons, axotomy activates Neu3 sialidase, increasing the ratio of GM1/GD1a and GM1/GT1b gangliosides immediately after injury in vitro and in vivo. No change in the GM1/GD1a ratio after axotomy was observed in retinal axons (in vitro and in vivo), despite the presence of Neu3 sialidase. Externally applied sialidase converted GD1a ganglioside to GM1 and rescued axon regeneration in CNS axons and in PNS axons after Neu3 sialidase blockade. Neu3 sialidase activation in DRGs is initiated by an influx of extracellular calcium, activating P38MAPK and then Neu3 sialidase. Ganglioside conversion by Neu3 sialidase further activates the ERK pathway. In CNS axons, P38MAPK and Neu3 sialidase were not activated by axotomy.
Collapse
|
18
|
Corona G, Vauzour D, Hercelin J, Williams CM, Spencer JPE. Phenolic acid intake, delivered via moderate champagne wine consumption, improves spatial working memory via the modulation of hippocampal and cortical protein expression/activation. Antioxid Redox Signal 2013; 19:1676-89. [PMID: 23458470 DOI: 10.1089/ars.2012.5142] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
AIMS While much data exist for the effects of flavonoid-rich foods on spatial memory in rodents, there are no such data for foods/beverages predominantly containing hydroxycinnamates and phenolic acids. To address this, we investigated the effects of moderate Champagne wine intake, which is rich in these components, on spatial memory and related mechanisms relative to the alcohol- and energy-matched controls. RESULTS In contrast to the isocaloric and alcohol-matched controls, supplementation with Champagne wine (1.78 ml/kg BW, alcohol 12.5% vol.) for 6 weeks led to an improvement in spatial working memory in aged rodents. Targeted protein arrays indicated that these behavioral effects were paralleled by the differential expression of a number of hippocampal and cortical proteins (relative to the isocaloric control group), including those involved in signal transduction, neuroplasticity, apoptosis, and cell cycle regulation. Western immunoblotting confirmed the differential modulation of brain-derived neurotrophic factor, cAMP response-element-binding protein (CREB), p38, dystrophin, 2',3'-cyclic-nucleotide 3'-phosphodiesterase, mammalian target of rapamycin (mTOR), and Bcl-xL in response to Champagne supplementation compared to the control drink, and the modulation of mTOR, Bcl-xL, and CREB in response to alcohol supplementation. INNOVATION Our data suggest that smaller phenolics such as gallic acid, protocatechuic acid, tyrosol, caftaric acid, and caffeic acid, in addition to flavonoids, are capable of exerting improvements in spatial memory via the modulation in hippocampal signaling and protein expression. CONCLUSION Changes in spatial working memory induced by the Champagne supplementation are linked to the effects of absorbed phenolics on cytoskeletal proteins, neurotrophin expression, and the effects of alcohol on the regulation of apoptotic events in the hippocampus and cortex.
Collapse
Affiliation(s)
- Giulia Corona
- 1 Molecular Nutrition Group, Centre for Integrative Neuroscience and Neurodynamics, School of Chemistry, Food and Pharmacy, University of Reading , Reading, United Kingdom
| | | | | | | | | |
Collapse
|
19
|
Dall'Igna OP, Bobermin LD, Souza DO, Quincozes-Santos A. Riluzole increases glutamate uptake by cultured C6 astroglial cells. Int J Dev Neurosci 2013; 31:482-6. [PMID: 23777615 DOI: 10.1016/j.ijdevneu.2013.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 05/24/2013] [Accepted: 06/09/2013] [Indexed: 11/26/2022] Open
Abstract
Riluzole is a drug approved for the treatment of amyotrophic lateral sclerosis (ALS) and may be effective for the treatment of other neurodegenerative and neuropsychiatric disorders. Riluzole exerts diverse actions on the central nervous system, including altering glutamate release and uptake, and therefore act diminishing glutamate extracellular levels, but the underlying mechanism of these actions is still unknown. Here, we demonstrate that riluzole stimulated glutamate uptake and augmented the expression of the glutamate EAAC1 transporter in C6 astroglial cell cultures. The effect of riluzole on glutamate uptake was reduced to below controls when it was co-administered with inhibitors of protein kinase C (PKC; bisindolylmaleimide II), phosphatidylinositol 3-kinase (PI3K; wortmannin) and fibroblast growth factor receptor 1 (FGFR1; PD173074). Riluzole also decreased reactive oxygen species load with no effect on glutathione levels. This study investigates three independent intracellular pathways and the mechanism of action of riluzole on glutamate metabolism.
Collapse
Affiliation(s)
- Oscar P Dall'Igna
- Rua Ramiro Barcelos 2600-Anexo, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, CEP 90035-000, Brazil.
| | | | | | | |
Collapse
|
20
|
Kurauchi Y, Hisatsune A, Isohama Y, Mishima S, Katsuki H. Caffeic acid phenethyl ester protects nigral dopaminergic neurons via dual mechanisms involving haem oxygenase-1 and brain-derived neurotrophic factor. Br J Pharmacol 2012; 166:1151-68. [PMID: 22224485 DOI: 10.1111/j.1476-5381.2012.01833.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Caffeic acid phenethyl ester (CAPE) is a component of honey bee propolis that can induce expression of haem oxygenase-1 (HO-1). Because HO-1 induction has been suggested to protect dopaminergic neurons in the substantia nigra, we examined the effect of CAPE in experimental models of dopaminergic neurodegeneration. EXPERIMENTAL APPROACH Neuroprotective effect of CAPE was investigated in rat organotypic midbrain slice cultures and in vivo, using a mouse model of dopaminergic neurodegeneration induced by intranigral injection of LPS and intrastriatal injection of 6-hydroxydopamine. KEY RESULTS CAPE protected dopaminergic neurons in slice cultures from IFN-γ/LPS-induced injury. The effect of CAPE was inhibited by zinc protoporphyrin IX, an HO-1 inhibitor, and by neutralizing antibody against brain-derived neurotrophic factor (BDNF). A p38 MAPK inhibitor SB203580 prevented activation of NF-E2-related factor 2, attenuated increased expression of HO-1 and BDNF, and blocked the neuroprotective actions of CAPE. In the LPS-injected mouse model, daily intraperitoneal administration of CAPE protected dopaminergic neurons, up-regulated HO-1 and BDNF, and reduced the increase of activated microglia/macrophages. Neuroprotective effects of CAPE against LPS-induced injury was prevented by zinc protoporphyrin IX or anti-BDNF antibody. CAPE protected dopaminergic neurons and alleviated methamphetamine-induced rotational behaviour also in 6-hydroxydopamine hemiparkinsonian mice. CONCLUSION AND IMPLICATIONS CAPE is a novel type of neuroprotective agent whose actions are mediated by both HO-1 and BDNF. These findings may provide novel clues to develop neuroprotective agents for treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Y Kurauchi
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamto, Japan
| | | | | | | | | |
Collapse
|
21
|
Ju X, Mallet RT, Downey HF, Metzger DB, Jung ME. Intermittent hypoxia conditioning protects mitochondrial cytochrome c oxidase of rat cerebellum from ethanol withdrawal stress. J Appl Physiol (1985) 2012; 112:1706-14. [PMID: 22403345 PMCID: PMC3365408 DOI: 10.1152/japplphysiol.01428.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 03/05/2012] [Indexed: 12/17/2022] Open
Abstract
Intermittent hypoxia (IH) conditioning minimizes neurocognitive impairment and stabilizes brain mitochondrial integrity during ethanol withdrawal (EW) in rats, but the mitoprotective mechanism is unclear. We investigated whether IH conditioning protects a key mitochondrial enzyme, cytochrome c oxidase (COX), from EW stress by inhibiting mitochondrially directed apoptotic pathways involving cytochrome c, Bax, or phosphor-P38 (pP38). Male rats completed two cycles of a 4-wk ethanol diet (6.5%) and 3 wk of EW. An IH program consisting of 5-10 bouts of 5-8 min of mild hypoxia (9.5-10% inspired O(2)) and 4 min of reoxygenation for 20 consecutive days began 3 days before the first EW period. For some animals, vitamin E replaced IH conditioning to test the contributions of antioxidant mechanisms to IH's mitoprotection. During the second EW, cerebellar-related motor function was evaluated by measuring latency of fall from a rotating rod (Rotarod test). After the second EW, COX activity in cerebellar mitochondria was measured by spectrophotometry, and COX, cytochrome c, Bax, and pP38 content were analyzed by immunoblot. Mitochondrial protein oxidation was detected by measuring carbonyl contents and by immunochemistry. Earlier IH conditioning prevented motor impairment, COX inactivation, depletion of COX subunit 4, protein carbonylation, and P38 phosphorylation during EW. IH did not prevent cytochrome c depletion during EW, and Bax content was unaffected by EW ± IH. Vitamin E treatment recapitulated IH protection of COX, and P38 inhibition attenuated protein oxidation during EW. Thus IH protects COX and improves cerebellar function during EW by limiting P38-dependent oxidative damage.
Collapse
Affiliation(s)
- Xiaohua Ju
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107-2699, USA
| | | | | | | | | |
Collapse
|
22
|
In vitro beneficial activation of microglial cells by mechanically-injured astrocytes enhances the synthesis and secretion of BDNF through p38MAPK. Neurochem Int 2012; 61:175-86. [PMID: 22561407 DOI: 10.1016/j.neuint.2012.04.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 03/20/2012] [Accepted: 04/18/2012] [Indexed: 12/30/2022]
Abstract
It has long been promulgated that microglial cells serve beneficial roles in the central nervous system (CNS). The beneficial role of microglial cells is considered to be linked with microglial activation and consequent up-regulation of various trophic factors. However, what triggers microglial activation and consequent elevated level of trophic factors, especially brain-derived neurotrophic factor (BDNF), following traumatic CNS injury has become a crucial but elusive issue. Furthermore, an effort still remains in understanding of the cellular and molecular mechanisms underlying the endogenous neuroprotection of activated microglial cells. In this study, we demonstrated that mechanically-injured astrocyte conditioned medium (ACM) could provoke beneficial activation of microglial cells and thus promote the transcription, synthesis and release of BDNF in cultured microglial cells. The microglia-derived BDNF can exerted a demonstrable biological role in promoting neurite outgrowth and intimate terminal contacts of dorsal root ganglion (DRG) neurons co-cultured with microglial cells. Moreover, ACM induced remarkable p38MAPK phosphorylation in cultured microglial cells that preceded the burst of BDNF. Activating p38-MAPK by anisomycin resulted in salutary effects similar to those seen with ACM, whereas specific inhibition of the p38MAPK by SB203580 abrogated all the positive effects of ACM, including BDNF promotion and subsequent neurite outgrowth of DRG neurite outgrowth of DRG neurons and their intimate terminal contacts with microglial cells. Together, our results indicated that the neuroprotection of the microglial source is mainly caused by micro-environmental soluble molecules released from injured astrocytes, and ACM-induced BDNF production and release from microglial cells may be mediated through p38-MAPK signaling pathway. Therefore, these findings may lay a foundation to further investigations on the microglial beneficial activation role in the repair of traumatic CNS injury and neurodegenerative diseases.
Collapse
|
23
|
Anti-allodynic effect of the flavonoid myricetin in a rat model of neuropathic pain: Involvement of p38 and protein kinase C mediated modulation of Ca2+
channels. Eur J Pain 2012; 14:992-8. [DOI: 10.1016/j.ejpain.2010.04.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 03/18/2010] [Accepted: 04/14/2010] [Indexed: 02/04/2023]
|
24
|
Bellingham MC. A review of the neural mechanisms of action and clinical efficiency of riluzole in treating amyotrophic lateral sclerosis: what have we learned in the last decade? CNS Neurosci Ther 2011; 17:4-31. [PMID: 20236142 DOI: 10.1111/j.1755-5949.2009.00116.x] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating and fatal neurodegenerative disease of adults which preferentially attacks the neuromotor system. Riluzole has been used as the only approved treatment for amyotrophic lateral sclerosis since 1995, but its mechanism(s) of action in slowing the progression of this disease remain obscure. Searching PubMed for "riluzole" found 705 articles published between January 1996 and June 2009. A systematic review of this literature found that riluzole had a wide range of effects on factors influencing neural activity in general, and the neuromotor system in particular. These effects occurred over a large dose range (<1 μM to >1 mM). Reported neural effects of riluzole included (in approximate ascending order of dose range): inhibition of persistent Na(+) current = inhibition of repetitive firing < potentiation of calcium-dependent K(+) current < inhibition of neurotransmitter release < inhibition of fast Na(+) current < inhibition of voltage-gated Ca(2+) current = promotion of neuronal survival or growth factors < inhibition of voltage-gated K(+) current = modulation of two-pore K(+) current = modulation of ligand-gated neurotransmitter receptors = potentiation of glutamate transporters. Only the first four of these effects commonly occurred at clinically relevant concentrations of riluzole (plasma levels of 1-2 μM with three- to four-fold higher concentrations in brain tissue). Treatment of human ALS patients or transgenic rodent models of ALS with riluzole most commonly produced a modest but significant extension of lifespan. Riluzole treatment was well tolerated in humans and animals. In animals, despite in vitro evidence that riluzole may inhibit rhythmic motor behaviors, in vivo administration of riluzole produced relatively minor effects on normal respiration parameters, but inhibited hypoxia-induced gasping. This effect may have implications for the management of hypoventilation and sleep-disordered breathing during end-stage ALS in humans.
Collapse
Affiliation(s)
- Mark C Bellingham
- School of Biomedical Sciences, University of Queensland, Brisbane, Qld. 4072, Australia.
| |
Collapse
|
25
|
Hamatake M, Miyazaki N, Sudo K, Matsuda M, Sadakata T, Furuya A, Ichisaka S, Hata Y, Nakagawa C, Nagata KI, Furuichi T, Katoh-Semba R. Phase advance of the light-dark cycle perturbs diurnal rhythms of brain-derived neurotrophic factor and neurotrophin-3 protein levels, which reduces synaptophysin-positive presynaptic terminals in the cortex of juvenile rats. J Biol Chem 2011; 286:21478-87. [PMID: 21527636 DOI: 10.1074/jbc.m110.195859] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
In adult rat brains, brain-derived neurotrophic factor (BDNF) rhythmically oscillates according to the light-dark cycle and exhibits unique functions in particular brain regions. However, little is known of this subject in juvenile rats. Here, we examined diurnal variation in BDNF and neurotrophin-3 (NT-3) levels in 14-day-old rats. BDNF levels were high in the dark phase and low in the light phase in a majority of brain regions. In contrast, NT-3 levels demonstrated an inverse phase relationship that was limited to the cerebral neocortex, including the visual cortex, and was most prominent on postnatal day 14. An 8-h phase advance of the light-dark cycle and sleep deprivation induced an increase in BDNF levels and a decrease in NT-3 levels in the neocortex, and the former treatment reduced synaptophysin expression and the numbers of synaptophysin-positive presynaptic terminals in cortical layer IV and caused abnormal BDNF and NT-3 rhythms 1 week after treatment. A similar reduction of synaptophysin expression was observed in the cortices of Bdnf gene-deficient mice and Ca(2+)-dependent activator protein for secretion 2 gene-deficient mice with abnormal free-running rhythm and autistic-like phenotypes. In the latter mice, no diurnal variation in BDNF levels was observed. These results indicate that regular rhythms of BDNF and NT-3 are essential for correct cortical network formation in juvenile rodents.
Collapse
Affiliation(s)
- Michiko Hamatake
- Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|