1
|
Ellis RJ, Marquine MJ, Kaul M, Fields JA, Schlachetzki JCM. Mechanisms underlying HIV-associated cognitive impairment and emerging therapies for its management. Nat Rev Neurol 2023; 19:668-687. [PMID: 37816937 PMCID: PMC11052664 DOI: 10.1038/s41582-023-00879-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/12/2023]
Abstract
People living with HIV are affected by the chronic consequences of neurocognitive impairment (NCI) despite antiretroviral therapies that suppress viral replication, improve health and extend life. Furthermore, viral suppression does not eliminate the virus, and remaining infected cells may continue to produce viral proteins that trigger neurodegeneration. Comorbidities such as diabetes mellitus are likely to contribute substantially to CNS injury in people living with HIV, and some components of antiretroviral therapy exert undesirable side effects on the nervous system. No treatment for HIV-associated NCI has been approved by the European Medicines Agency or the US Food and Drug Administration. Historically, roadblocks to developing effective treatments have included a limited understanding of the pathophysiology of HIV-associated NCI and heterogeneity in its clinical manifestations. This heterogeneity might reflect multiple underlying causes that differ among individuals, rather than a single unifying neuropathogenesis. Despite these complexities, accelerating discoveries in HIV neuropathogenesis are yielding potentially druggable targets, including excessive immune activation, metabolic alterations culminating in mitochondrial dysfunction, dysregulation of metal ion homeostasis and lysosomal function, and microbiome alterations. In addition to drug treatments, we also highlight the importance of non-pharmacological interventions. By revisiting mechanisms implicated in NCI and potential interventions addressing these mechanisms, we hope to supply reasons for optimism in people living with HIV affected by NCI and their care providers.
Collapse
Affiliation(s)
- Ronald J Ellis
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
| | - María J Marquine
- Department of Medicine, Duke University, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Marcus Kaul
- School of Medicine, Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Jerel Adam Fields
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
2
|
Strogulski NR, Portela LV, Polster BM, Loane DJ. Fundamental Neurochemistry Review: Microglial immunometabolism in traumatic brain injury. J Neurochem 2023; 167:129-153. [PMID: 37759406 PMCID: PMC10655864 DOI: 10.1111/jnc.15959] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
Traumatic brain injury (TBI) is a devastating neurological disorder caused by a physical impact to the brain that promotes diffuse damage and chronic neurodegeneration. Key mechanisms believed to support secondary brain injury include mitochondrial dysfunction and chronic neuroinflammation. Microglia and brain-infiltrating macrophages are responsible for neuroinflammatory cytokine and reactive oxygen species (ROS) production after TBI. Their production is associated with loss of homeostatic microglial functions such as immunosurveillance, phagocytosis, and immune resolution. Beyond providing energy support, mitochondrial metabolic pathways reprogram the pro- and anti-inflammatory machinery in immune cells, providing a critical immunometabolic axis capable of regulating immunologic response to noxious stimuli. In the brain, the capacity to adapt to different environmental stimuli derives, in part, from microglia's ability to recognize and respond to changes in extracellular and intracellular metabolite levels. This capacity is met by an equally plastic metabolism, capable of altering immune function. Microglial pro-inflammatory activation is associated with decreased mitochondrial respiration, whereas anti-inflammatory microglial polarization is supported by increased oxidative metabolism. These metabolic adaptations contribute to neuroimmune responses, placing mitochondria as a central regulator of post-traumatic neuroinflammation. Although it is established that profound neurometabolic changes occur following TBI, key questions related to metabolic shifts in microglia remain unresolved. These include (a) the nature of microglial mitochondrial dysfunction after TBI, (b) the hierarchical positions of different metabolic pathways such as glycolysis, pentose phosphate pathway, glutaminolysis, and lipid oxidation during secondary injury and recovery, and (c) how immunometabolism alters microglial phenotypes, culminating in chronic non-resolving neuroinflammation. In this basic neurochemistry review article, we describe the contributions of immunometabolism to TBI, detail primary evidence of mitochondrial dysfunction and metabolic impairments in microglia and macrophages, discuss how major metabolic pathways contribute to post-traumatic neuroinflammation, and set out future directions toward advancing immunometabolic phenotyping in TBI.
Collapse
Affiliation(s)
- Nathan R. Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Luis V. Portela
- Neurotrauma and Biomarkers Laboratory, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Brian M. Polster
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David J. Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Nassar WM, El-Kholy WM, El-Sawi MR, El-Shafai NM, Alotaibi BS, Ghamry HI, Shukry M. Ameliorative Effect of Thymoquinone and Thymoquinone Nanoparticles against Diazinon-Induced Hepatic Injury in Rats: A Possible Protection Mechanism. TOXICS 2023; 11:783. [PMID: 37755793 PMCID: PMC10536923 DOI: 10.3390/toxics11090783] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023]
Abstract
The health benefits of thymoquinone (TQ) have been a significant focus of numerous studies. However, more research is needed to ascertain whether its nano-form can effectively treat or prevent chronic diseases. In this study, we investigated how thymoquinone and its nanoparticles can mitigate liver damage induced by diazinon in male Wistar rats and explored the intracellular mechanisms involved. Forty-two Wistar male rats (n = 42) were randomly allotted into seven groups. Group 1 served as the control. Group 2 (vehicle) consisted of rats that received corn oil via a gastric tube daily. In Group 3 (TQ), rats were given a daily oral administration of TQ (40 mg/kg bw). Group 4 (thymoquinone nanoparticles, NTQ) included rats that received NTQ (0.5 mg/kg bw) orally for 21 days. Group 5 (DZN) involved rats that were administered diazinon (DZN, 15 mg/kg bw) orally. In Group 6 (TQ + DZN), rats first received TQ orally, followed by DZN. Group 7 (NTQ + DZN) consisted of rats receiving NTQ orally, then DZN. After 21 days of treatment, the rats were euthanized. After oral administration of DZN, liver enzymes were significantly elevated (p < 0.05). Additionally, there were noticeable increases in oxidative injury markers, such as nitric oxide, malondialdehyde, redox oxygen radicals, and overall increases in hydrogen peroxide and liver protein carbonyl concentrations. This was accompanied by the upregulation of apoptotic markers (Bax, caspase9, caspase 3, bax/Bcl2 ratio), inflammatory cytokines (TNF-α, IL-6), and DNA damage. There was also a noteworthy decrease (p < 0.05) in the activities of antioxidant enzymes and anti-apoptotic markers. However, the oral administration of thymoquinone or its nanoparticle form mitigated these diazinon complications; our histopathological findings corroborated our biochemical and molecular observations. In conclusion, the significant antioxidant properties of thymoquinone, or its nanoparticle form, in tandem with the downregulation of apoptotic markers and inflammatory cytokines, provided a protective effect against hepatic dysfunction caused by diazinon.
Collapse
Affiliation(s)
- Walaa M. Nassar
- Physiology Division, Zoology Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Wafaa M. El-Kholy
- Physiology Division, Zoology Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Mamdouh R. El-Sawi
- Physiology Division, Zoology Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Nagi M. El-Shafai
- Nanotechnology Center, Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Badriyah S. Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Heba I. Ghamry
- Nutrition and Food Sciences, Department of Home Economics, Faculty of Home Economics, King Khalid University, P.O. Box 960, Abha 61421, Saudi Arabia
| | - Mustafa Shukry
- Physiology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| |
Collapse
|
4
|
Chen X, Feng Y, Quinn RJ, Pountney DL, Richardson DR, Mellick GD, Ma L. Potassium Channels in Parkinson's Disease: Potential Roles in Its Pathogenesis and Innovative Molecular Targets for Treatment. Pharmacol Rev 2023; 75:758-788. [PMID: 36918260 DOI: 10.1124/pharmrev.122.000743] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/05/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by selective loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region of the midbrain. The loss of neurons results in a subsequent reduction of dopamine in the striatum, which underlies the core motor symptoms of PD. To date, there are no effective treatments to stop, slow, or reverse the pathologic progression of dopaminergic neurodegeneration. This unfortunate predicament is because of the current early stages in understanding the biologic targets and pathways involved in PD pathogenesis. Ion channels have become emerging targets for new therapeutic development for PD due to their essential roles in neuronal function and neuroinflammation. Potassium channels are the most prominent ion channel family and have been shown to be critically important in PD pathology because of their roles in modulating neuronal excitability, neurotransmitter release, synaptic transmission, and neuroinflammation. In this review, members of the subfamilies of voltage-gated K+ channels, inward rectifying K+ channels, and Ca2+-activated K+ channels are described. Evidence of the role of these channels in PD etiology is discussed together with the latest views on related pathologic mechanisms and their potential as biologic targets for developing neuroprotective drugs for PD. SIGNIFICANCE STATEMENT: Parkinson's disease (PD) is the second most common neurodegenerative disorder, featuring progressive degeneration of dopaminergic neurons in the midbrain. It is a multifactorial disease involving multiple risk factors and complex pathobiological mechanisms. Mounting evidence suggests that ion channels play vital roles in the pathogenesis and progression of PD by regulating neuronal excitability and immune cell function. Therefore, they have become "hot" biological targets for PD, as demonstrated by multiple clinical trials of drug candidates targeting ion channels for PD therapy.
Collapse
Affiliation(s)
- Xiaoyi Chen
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Yunjiang Feng
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Ronald J Quinn
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Dean L Pountney
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Des R Richardson
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - George D Mellick
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Linlin Ma
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| |
Collapse
|
5
|
Qian C, Fan Y, Zong L, Miao C, Ji LL, Wan L, Jia R, Qin X, Wang Y, Wu Q, Tao XY, Hao L, Hu L, Liu WT. Opening K ATP channels induces inflammatory tolerance and prevents chronic pain. Brain Behav Immun 2023; 107:76-86. [PMID: 36198341 DOI: 10.1016/j.bbi.2022.09.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/30/2022] [Accepted: 09/30/2022] [Indexed: 02/09/2023] Open
Abstract
Current treatments for chronic pain are unsatisfactory, therefore, new therapeutics are urgently needed. Our previous study indicated that KATP channel openers have analgesic effects, but the underlying mechanism has not been elucidated. We speculated that KATP channel openers might increase suppressor of cytokine signaling (SOCS)-3 expression to induce inflammatory tolerance and attenuate chronic pain. Postoperative pain was induced by plantar incision to establish a chronic pain model. Growth arrest-specific 6 (Gas6)-/- and Axl-/- mice were used for signaling studies. The microglia cell line BV-2 was cultured for the in vitro experiments. The KATP channel opener significantly attenuated incision-induced mechanical allodynia in mice associated with the upregulated expression of SOCS3. Opening KATP channels induced the expression of SOCS3 in the Gas6/Axl signaling pathway in microglia, inhibited incision-induced mechanical allodynia by activating the Gas6/Axl-SOCS3 signaling pathway, and induced inflammatory tolerance to relieve neuroinflammation and postoperative pain. We demonstrated that opening of the KATP channel opening activated Gas6/Axl/SOCS3 signaling to induce inflammatory tolerance and relieve chronic pain. We explored a new target for anti-inflammatory and analgesic effects by regulating the innate immune system and provided a theoretical basis for clinical preemptive analgesia.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Pathology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China; Sir Run Run Hospital, Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - Yixin Fan
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Sir Run Run Hospital, Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - Lijuan Zong
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Chen Miao
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Lu-Lu Ji
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Li Wan
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Rumeng Jia
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Xinmiao Qin
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Yu Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Qi Wu
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Xue-You Tao
- Department of Anesthesiology, Yangzhou Maternal and Child Health Hospital Affiliated to Medical College of Yangzhou University, Yangzhou 225001, Jiangsu, China.
| | - Lanxiang Hao
- The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, 224005 Jiangsu, China.
| | - Liang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Wen-Tao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| |
Collapse
|
6
|
Xanthotoxin modulates oxidative stress, inflammation, and MAPK signaling in a rotenone-induced Parkinson's disease model. Life Sci 2022; 310:121129. [DOI: 10.1016/j.lfs.2022.121129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 11/05/2022]
|
7
|
Lv J, Xiao X, Bi M, Tang T, Kong D, Diao M, Jiao Q, Chen X, Yan C, Du X, Jiang H. ATP-sensitive potassium channels: A double-edged sword in neurodegenerative diseases. Ageing Res Rev 2022; 80:101676. [PMID: 35724860 DOI: 10.1016/j.arr.2022.101676] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/15/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022]
Abstract
ATP-sensitive potassium channels (KATP channels), a group of vital channels that link the electrical activity of the cell membrane with cell metabolism, were discovered on the ventricular myocytes of guinea pigs by Noma using the patch-clamp technique in 1983. Subsequently, KATP channels have been found to be expressed in pancreatic β cells, cardiomyocytes, skeletal muscle cells, and nerve cells in the substantia nigra (SN), hippocampus, cortex, and basal ganglia. KATP channel openers (KCOs) diazoxide, nicorandil, minoxidil, and the KATP channel inhibitor glibenclamide have been shown to have anti-hypertensive, anti-myocardial ischemia, and insulin-releasing regulatory effects. Increasing evidence has suggested that KATP channels also play roles in Alzheimer's disease (AD), Parkinson's disease (PD), vascular dementia (VD), Huntington's disease (HD) and other neurodegenerative diseases. KCOs and KATP channel inhibitors protect neurons from injury by regulating neuronal excitability and neurotransmitter release, inhibiting abnormal protein aggregation and Ca2+ overload, reducing reactive oxygen species (ROS) production and microglia activation. However, KATP channels have dual effects in some cases. In this review, we focus on the roles of KATP channels and their related openers and inhibitors in neurodegenerative diseases. This will enable us to precisely take advantage of the KATP channels and provide new ideas for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jirong Lv
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xue Xiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Tingting Tang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Deao Kong
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Meining Diao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Chunling Yan
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China.
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China.
| |
Collapse
|
8
|
Intranasal Exposure to Low-Dose Rotenone Induced Alpha-Synuclein Accumulation and Parkinson's Like Symptoms Without Loss of Dopaminergic Neurons. Neurotox Res 2021; 40:215-229. [PMID: 34817799 DOI: 10.1007/s12640-021-00436-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/21/2022]
Abstract
Epidemiologically Parkinson's disease (PD) is associated with chronic ingestion or inhalation of environmental toxins leading to the development of motor symptoms. Though neurotoxin-based animal models played a major role in understanding diverse pathogenesis, they failed to identify the risk assessment due to uncommon route of toxin exposure. Towards this, the available neurotoxin-based intranasal (i.n.) PD models targeting olfactory bulb (OB) have demonstrated the dopaminergic (DAergic) neurodegeneration in both OB and substantia nigra (SN). Despite that, the studies detecting the alpha-synuclein (α-syn) accumulation in OB and its progression to other brain regions due to inhalation of environmental toxins are still lacking. Herein, we developed oil in water microemulsion of rotenone administered intranasally to the mice at a dose which is not detectable in blood, brain, and olfactory bulb by LCMS method. Our data reveals that 9 weeks of rotenone exposure did not induce olfactory and motor dysfunction. Conversely, after 16 weeks of washout period, rotenone treated mice showed both olfactory and motor impairment, along with α-syn accumulation in the OB and striatum without glial cell activation and loss of dopaminergic neurons. The results depict the progressive nature of the developed model and highlight the role of α-syn in PD like pathology or symptoms. Together, our findings suggest the adverse consequences of early exposure to the environmental toxins on the olfactory system for a shorter period with relevance to the development of synucleinopathy or Parkinson's disease in its later stage.
Collapse
|
9
|
Fu J, Mu G, Liu X, Ou C, Zhao J. Ischemic postconditioning reduces spinal cord ischemia-reperfusion injury through ATP-sensitive potassium channel. Spinal Cord 2021; 60:326-331. [PMID: 34616009 DOI: 10.1038/s41393-021-00714-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 09/17/2021] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN Animal study. OBJECTIVES Explore the neuroprotective effect of remote limb ischemic postconditioning (Post C) in spinal cord ischemic reperfusion injury (SCII) and related mechanisms. SETTING Anesthesiology Laboratory of Southwest Medical University. METHODS We established a rabbit SCII model and processed it with Post C. To evaluate the neural function, spinal cord tissue was taken 48 h later, normal neurons were evaluated by HE staining, and the expression of ATP-sensitive potassium channel (KATP) marker molecule Kir6.2 was detected by Western blot. Immunofluorescence detection of spinal cord Iba-1 expression, ELISA detection of M1 type microglia marker iNOS and M2 type microglia marker Arg, and Western blot detection of NF-κB and IL-1β expression. Through these experiments, we will explore the protective effect of Post C in SCII, observe the changes in the protective effect after using KATP blockers, and verify that Post C can play a neuroprotective effect in SCII by activating KATP. RESULTS We observed that Post C significantly improved exercise ability and the number of spinal motor neurons in the SCII model. Microglia are activated and expression of M1 microglia in the spinal cord was decreased, while M2 was increased. This neuroprotective effect was reversed by the nonspecific KATP inhibitor. CONCLUSION Post C has a neuroprotective effect on SCII, and maybe a protective effect produced by activating KATP to regulate spinal microglia polarization and improve neuroinflammation.
Collapse
Affiliation(s)
- Jia Fu
- Department of Pain Management, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Guo Mu
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, China.
| | - Xiangbo Liu
- Department of Pain Management, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Cehua Ou
- Department of Pain Management, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiaomei Zhao
- Department of Pain Management, Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
10
|
Adu TS, Mabandla MV. Effects of bromelain on striatal neuroinflammation in rat model of Parkinsonism. BRAIN DISORDERS 2021. [DOI: 10.1016/j.dscb.2021.100018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
11
|
Do changes in microglial status underlie neurogenesis impairments and depressive-like behaviours induced by psychological stress? A systematic review in animal models. Neurobiol Stress 2021; 15:100356. [PMID: 34355047 PMCID: PMC8319800 DOI: 10.1016/j.ynstr.2021.100356] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Stress may have a negative effect on mental health and is the primary environmental risk factor in the aetiology of depression. Nevertheless, the neurobiological mechanisms underlying this mood disorder remain poorly characterized. The hippocampus is a target structure of the adverse effects of stress, and hippocampal neurogenesis plays a crucial role. However, we do not know the mechanisms by which stress impacts neurogenesis. Recent studies indicate that changes in neuroinflammation, primarily via microglial cells, may play an essential role in this process. However, the relationship between stress, microglial changes, and alterations in neurogenesis and their involvement in the development of depression is poorly characterized. For this reason, this systematic review aims to synthesise and evaluate current studies that have investigated the relationship between these variables. Taken together, the revised data, although not entirely conclusive, seem to suggest that microglial changes induced by psychological stress regulate neurogenesis and in turn may be responsible for the development of depressive-like behaviours, but other factors that influence these stressful experiences should not be dismissed.
Collapse
|
12
|
Jing L, Hou L, Zhang D, Li S, Ruan Z, Zhang X, Hong JS, Wang Q. Microglial Activation Mediates Noradrenergic Locus Coeruleus Neurodegeneration via Complement Receptor 3 in a Rotenone-Induced Parkinson's Disease Mouse Model. J Inflamm Res 2021; 14:1341-1356. [PMID: 33859489 PMCID: PMC8044341 DOI: 10.2147/jir.s299927] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022] Open
Abstract
Background Chronic exposure to the insecticide rotenone can damage dopaminergic neurons and lead to an increased risk of Parkinson’s disease (PD). Whereas it is not clear whether rotenone induces neurodegeneration of noradrenergic locus coeruleus (LC/NE) neurons. Chronic neuroinflammation mediated by microglia has been involved in the pathogenesis of PD. Evidence shows that complement receptor 3 (CR3) is a crucial regulator of microglial activation and related neurodegeneration. However, it is not clear whether CR3 mediates rotenone-elicited degeneration of LC/NE neurons through microglia-mediated neuroinflammation. Materials and Methods Wild type (WT) and CR3 knockout (KO) mice were treated with rotenone. PLX3397 and minocycline were used to deplete or inactivate the microglia. Leukadherin-1 (LA-1) was used to modulate CR3. LC/NE neurodegeneration, microglial phenotype, and expression of CR3 were determined by using immunohistochemistry, Western blot and real-time polymerase chain reaction (PCR) techniques. The glutathione (GSH) and malondialdehyde (MDA) contents were measured by using commercial kits. Results Rotenone exposure led to dose- and time-dependent LC/NE neuronal loss and microglial activation in mice. Depletion of microglia by PLX3397 or inhibition of microglial activation by minocycline significantly reduced rotenone-induced LC/NE neurodegeneration. Mechanistic studies revealed that CR3 played an essential role in the rotenone-induced activation of microglia and neurodegeneration of LC/NE neurons. Rotenone elevated the expression of CR3, and genetic ablation of CR3 markedly reduced rotenone-induced microglial activation and M1 polarization. LA-1 also suppressed rotenone-induced toxic microglial M1 activation. Furthermore, lack of CR3 or treatment with LA-1 reduced oxidative stress in the brainstem of rotenone-intoxicated mice. Finally, we found that mice deficient in CR3 or treated with LA-1 were more resistant to rotenone-induced LC/NE neurodegeneration than WT or vehicle-treated mice, respectively. Conclusion Our results indicate that CR3-mediated microglial activation participates in rotenone-induced LC/NE neurodegeneration, providing novel insight into environmental toxin-induced neurotoxicity and related Parkinsonism.
Collapse
Affiliation(s)
- Lu Jing
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China.,Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Liyan Hou
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Dongdong Zhang
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Sheng Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Zhengzheng Ruan
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Xiaomeng Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Qingshan Wang
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China.,National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| |
Collapse
|
13
|
Zhou XY, Liu J, Xu ZP, Fu Q, Wang PQ, Wang JH, Zhang H. Dexmedetomidine ameliorates postoperative cognitive dysfunction by inhibiting Toll-like receptor 4 signaling in aged mice. Kaohsiung J Med Sci 2020; 36:721-731. [PMID: 32627922 DOI: 10.1002/kjm2.12234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 04/11/2020] [Accepted: 04/26/2020] [Indexed: 11/11/2022] Open
Abstract
Our study aimed to explore the molecular mechanisms involved in the improvement of postoperative cognitive dysfunction (POCD) by dexmedetomidine (DEX). BV2 microglia cells were cultured under normal condition, DEX exposure (0.1 μg/mL), and lipopolysacchride (LPS) treatment (0.1 μg/mL) or with pretreatment of DEX before LPS incubation. For BV2 microglia cells, LPS induced markedly increased release of pro-inflammatory cytokines (interleukin [IL]-1β, IL-6, and tumor necrosis factor-alpha [TNF-α]) and expressions of Toll-like receptor 4 (TLR4) and nuclear factor kappa B (NF-κB), while DEX pretreatment inhibited the LPS-induced production of pro-inflammatory cytokines and expressions of TLR4 and NF-κB. The spatial memory function was impaired in the aged mice following partial hepatectomy since the percentage of time spent in the target quadrant and the number of crossings over the former platform location were reduced. Pretreatment of DEX may attenuate neuroinflammation and improve POCD in aged mice through inhibiting the TLR4-NF-κB signaling pathway in the hippocampus.
Collapse
Affiliation(s)
- Xue-Yue Zhou
- School of Clinical Medicine, Tsinghua University, Beijing, China.,Department of Anesthesiology, Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Jing Liu
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, China
| | - Zhi-Peng Xu
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, China
| | - Qiang Fu
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, China
| | - Pei-Qi Wang
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, China
| | - Jing-Hua Wang
- Department of Anesthesiology, The 305 Hospital of Chinese PLA, Beijing, China
| | - Hong Zhang
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
14
|
Nikbakht F, Khanizadeh AM, Golab F, Baluchnejadmojarad T, Vazifehkhah S, Moeinsadat A. Mitochondrial ATP-sensitive potassium channel, MitoKATP, ameliorates mitochondrial dynamic disturbance induced by temporal lobe epilepsy. J Chem Neuroanat 2020; 113:101808. [PMID: 32497687 DOI: 10.1016/j.jchemneu.2020.101808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/04/2020] [Accepted: 05/26/2020] [Indexed: 01/06/2023]
Abstract
Temporal lobe epilepsy leads to a disturbance in the function and dynamic of the mitochondria. The mitoKATP channel is an important factor in controlling mitochondrial function. In this study, the protective role of mitoKATP was studied in temporal lobe epilepsy through the regulation of mitochondrial dynamic proteins. After induction of epilepsy, 5-HD (the inhibitor of mitoKATP) was administered daily for either 24 or 72 h. The results revealed an imbalance in dynamic proteins after epilepsy, specifically in the first 72 h. The disturbance in the mitochondrial dynamic worsened after blocking mitoKATP. In conclusion, mitoKATP has an important role in balancing mitochondrial dynamic proteins in epilepsy.
Collapse
Affiliation(s)
- Farnaz Nikbakht
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine Iran University of Medical Sciences, Tehran, Iran
| | - Ali Mohammad Khanizadeh
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine Iran University of Medical Sciences, Tehran, Iran.
| | - Fereshteh Golab
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine Iran University of Medical Sciences, Tehran, Iran
| | - Tourandokht Baluchnejadmojarad
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine Iran University of Medical Sciences, Tehran, Iran
| | - Somayeh Vazifehkhah
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Moeinsadat
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Wang DX, Chen AD, Wang QJ, Xin YY, Yin J, Jing YH. Protective effect of metformin against rotenone-induced parkinsonism in mice. Toxicol Mech Methods 2020; 30:350-357. [DOI: 10.1080/15376516.2020.1741053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Dong-Xin Wang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - An-Di Chen
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Qing-Jun Wang
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Science, East China Normal University, Shanghai, PR China
| | - Yue-Yang Xin
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Jie Yin
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
- Key Laboratory of Preclinical Study for New Drugs of Gansu province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| |
Collapse
|
16
|
Mansour RM, Ahmed MAE, El-Sahar AE, El Sayed NS. Montelukast attenuates rotenone-induced microglial activation/p38 MAPK expression in rats: Possible role of its antioxidant, anti-inflammatory and antiapoptotic effects. Toxicol Appl Pharmacol 2018; 358:76-85. [PMID: 30222980 DOI: 10.1016/j.taap.2018.09.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/04/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022]
Abstract
Montelukast (MK),a cysteinyl leukotriene (CysLT1) receptor antagonist, latterly exhibited a remarkable neuroprotective activity in various neurodegenerative disorders. This study aims to elucidate the neuroprotective effect of MK in rotenone-induced Parkinson's disease(PD) model in rats. Ninety six male rats were split into four groups: vehicle control (0.2 ml/kg/48 h, sc), MK (10 mg/kg/day, ip), rotenone (1.5 mg/kg/48 h, sc.) and rotenone pretreated with MK. Rotenone treatment led to significant reduction in motor functioning and elevation in oxidative stress markers. Additionally, upregulation of p38 mitogen-activated protein kinase (p38 MAPK) and CysLT1 receptor expressions were anchored with enhanced striatal microglial activation generating a severe neuro-inflammatory milieu. Furthermore, an augmentation in p53 expression and cleaved caspases-3 activity increased apoptotic neurodegeneration synchronized with reduction of striatal tyrosine hydroxylase (TH) content. Changes in neuronal morphology was also noted. MK administration significantly mitigated motor impairment and rise in oxidative stress mediators. As well, the anti-inflammatory activity of MK was manifested by hindering the principal controller of inflammatory pathway, nuclear factor-kappa B, followed by its downstream pro-inflammatory cytokines (tumor necrosis factor-alpha and interleukin-1 beta), by attenuating striatal microglial activation and hampering the expression of both p38 MAPK and CysLT1. Moreover, MK revealed a decline in p53 expression with its downstream cleaved caspases-3 which resulted in preservation of striatal TH terminals as verified by increased striatal TH content and improvement in the histopathological changes incited by rotenone. In conclusion, MK endowed neuroprotective effects in rotenone-induced PD animal model via attenuation of microglial cell activation and p38 MAPK expression.
Collapse
Affiliation(s)
- Riham M Mansour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Manufacturing, Misr University for Science and Technology (MUST), 6th October City, Giza, Egypt
| | - Maha A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Manufacturing, Misr University for Science and Technology (MUST), 6th October City, Giza, Egypt
| | - Ayman E El-Sahar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt.
| |
Collapse
|
17
|
De Miranda BR, Rocha EM, Bai Q, El Ayadi A, Hinkle D, Burton EA, Timothy Greenamyre J. Astrocyte-specific DJ-1 overexpression protects against rotenone-induced neurotoxicity in a rat model of Parkinson's disease. Neurobiol Dis 2018; 115:101-114. [PMID: 29649621 PMCID: PMC5943150 DOI: 10.1016/j.nbd.2018.04.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 02/07/2023] Open
Abstract
DJ-1 is a redox-sensitive protein with several putative functions important in mitochondrial physiology, protein transcription, proteasome regulation, and chaperone activity. High levels of DJ-1 immunoreactivity are reported in astrocytes surrounding pathology associated with idiopathic Parkinson's disease, possibly reflecting the glial response to oxidative damage. Previous studies showed that astrocytic over-expression of DJ-1 in vitro prevented oxidative stress and mitochondrial dysfunction in primary neurons. Based on these observations, we developed a pseudotyped lentiviral gene transfer vector with specific tropism for CNS astrocytes in vivo to overexpress human DJ-1 protein in astroglial cells. Following vector delivery to the substantia nigra and striatum of adult Lewis rats, the DJ-1 transgene was expressed robustly and specifically within astrocytes. There was no observable transgene expression in neurons or other glial cell types. Three weeks after vector infusion, animals were exposed to rotenone to induce Parkinson's disease-like pathology, including loss of dopaminergic neurons, accumulation of endogenous α-synuclein, and neuroinflammation. Animals over-expressing hDJ-1 in astrocytes were protected from rotenone-induced neurodegeneration, and displayed a marked reduction in neuronal oxidative stress and microglial activation. In addition, α-synuclein accumulation and phosphorylation were decreased within substantia nigra dopaminergic neurons in DJ-1-transduced animals, and expression of LAMP-2A, a marker of chaperone mediated autophagy, was increased. Together, these data indicate that astrocyte-specific overexpression of hDJ-1 protects neighboring neurons against multiple pathologic features of Parkinson's disease and provides the first direct evidence in vivo of a cell non-autonomous neuroprotective function of astroglial DJ-1.
Collapse
Affiliation(s)
- Briana R De Miranda
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Qing Bai
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Amina El Ayadi
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - David Hinkle
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States; Geriatric Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States; Geriatric Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States.
| |
Collapse
|
18
|
Kabba JA, Xu Y, Christian H, Ruan W, Chenai K, Xiang Y, Zhang L, Saavedra JM, Pang T. Microglia: Housekeeper of the Central Nervous System. Cell Mol Neurobiol 2018; 38:53-71. [PMID: 28534246 PMCID: PMC11481884 DOI: 10.1007/s10571-017-0504-2] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/16/2017] [Indexed: 12/17/2022]
Abstract
Microglia, of myeloid origin, play fundamental roles in the control of immune responses and the maintenance of central nervous system homeostasis. These cells, just like peripheral macrophages, may be activated into M1 pro-inflammatory or M2 anti-inflammatory phenotypes by appropriate stimuli. Microglia do not respond in isolation, but form part of complex networks of cells influencing each other. This review addresses the complex interaction of microglia with each cell type in the brain: neurons, astrocytes, cerebrovascular endothelial cells, and oligodendrocytes. We also highlight the participation of microglia in the maintenance of homeostasis in the brain, and their roles in the development and progression of age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- John Alimamy Kabba
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Yazhou Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Handson Christian
- Department of Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Wenchen Ruan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Kitchen Chenai
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yun Xiang
- Department of Laboratory Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, People's Republic of China
| | - Luyong Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Juan M Saavedra
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC, 20057, USA
| | - Tao Pang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China.
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC, 20057, USA.
| |
Collapse
|
19
|
|
20
|
Afshin-Majd S, Bashiri K, Kiasalari Z, Baluchnejadmojarad T, Sedaghat R, Roghani M. Acetyl- l -carnitine protects dopaminergic nigrostriatal pathway in 6-hydroxydopamine-induced model of Parkinson’s disease in the rat. Biomed Pharmacother 2017; 89:1-9. [DOI: 10.1016/j.biopha.2017.02.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 01/27/2017] [Accepted: 02/07/2017] [Indexed: 01/16/2023] Open
|
21
|
Sesamin imparts neuroprotection against intrastriatal 6-hydroxydopamine toxicity by inhibition of astroglial activation, apoptosis, and oxidative stress. Biomed Pharmacother 2017; 88:754-761. [DOI: 10.1016/j.biopha.2017.01.123] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 12/12/2022] Open
|
22
|
Zhao XJ, Zhao Z, Yang DD, Cao LL, Zhang L, Ji J, Gu J, Huang JY, Sun XL. Activation of ATP-sensitive potassium channel by iptakalim normalizes stress-induced HPA axis disorder and depressive behaviour by alleviating inflammation and oxidative stress in mouse hypothalamus. Brain Res Bull 2017; 130:146-155. [DOI: 10.1016/j.brainresbull.2017.01.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/24/2017] [Accepted: 01/30/2017] [Indexed: 12/19/2022]
|
23
|
Opening of the Adenosine Triphosphate-sensitive Potassium Channel Attenuates Morphine Tolerance by Inhibiting JNK and Astrocyte Activation in the Spinal Cord. Clin J Pain 2017; 32:617-23. [PMID: 26626290 DOI: 10.1097/ajp.0000000000000299] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVES In the present study, we investigated the role of adenosine triphosphate (ATP)-sensitive potassium (KATP) channels in chronic morphine tolerance. MATERIALS AND METHODS Male mice were injected intrathecally with morphine or saline, respectively (each in 10 μL). Different doses of the KATP opener cromakalim (0.3, 1, or 3 μg/10 μL/mouse) were administered 15 minutes before the morphine (10 μg/10 μL/mouse) challenge daily for 7 consecutive days. Half an hour after morphine injection, the tail-flick latency was measured to evaluate the antinociceptive effect of morphine. On the seventh day, mice were euthanized with sodium pentobarbital (100 mg/kg) at 1 hour after morphine injection, and their spinal cords were removed for the assays of Western blot, immunofluorescence, and quantitative real-time polymerase chain reaction. RESULTS Opening of the KATP channel attenuates chronic morphine tolerance, suppresses astrocyte activation inhibits the increase in interleukin-1β at the transcriptional and the translational levels, and reduces the upregulation of phosphorylated c-Jun N-terminal kinase mitogen-activated protein kinase in the spinal cord after chronic morphine treatment. Moreover, transcriptional levels of spinal cord astrocyte KATP channel subunits, named the inwardly rectifying potassium (Kir) 6.1 and sulfonylurea receptor 1, are decreased in morphine-tolerant mice. DISCUSSION Cromakalim suppresses morphine-induced astrocyte activation significantly by suppressing the c-Jun N-terminal kinase pathway, resulting in a reduced release of interleukin-1β and the attenuation of morphine chronic antinociceptive tolerance.
Collapse
|
24
|
Development and characterization of a human embryonic stem cell-derived 3D neural tissue model for neurotoxicity testing. Toxicol In Vitro 2016; 38:124-135. [PMID: 27729293 DOI: 10.1016/j.tiv.2016.10.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/27/2016] [Accepted: 10/05/2016] [Indexed: 02/08/2023]
Abstract
Alternative models for more rapid compound safety testing are of increasing demand. With emerging techniques using human pluripotent stem cells, the possibility of generating human in vitro models has gained interest, as factors related to species differences could be potentially eliminated. When studying potential neurotoxic effects of a compound it is of crucial importance to have both neurons and glial cells. We have successfully developed a protocol for generating in vitro 3D human neural tissues, using neural progenitor cells derived from human embryonic stem cells. These 3D neural tissues can be maintained for two months and undergo progressive differentiation. We showed a gradual decreased expression of early neural lineage markers, paralleled by an increase in markers specific for mature neurons, astrocytes and oligodendrocytes. At the end of the two-month culture period the neural tissues not only displayed synapses and immature myelin sheaths around axons, but electrophysiological measurements also showed spontaneous activity. Neurotoxicity testing - comparing non-neurotoxic to known neurotoxic model compounds - showed an expected increase in the marker of astroglial reactivity after exposure to known neurotoxicants methylmercury and trimethyltin. Although further characterization and refinement of the model is required, these results indicate its potential usefulness for in vitro neurotoxicity testing.
Collapse
|
25
|
Kandil EA, Abdelkader NF, El-Sayeh BM, Saleh S. Imipramine and amitriptyline ameliorate the rotenone model of Parkinson's disease in rats. Neuroscience 2016; 332:26-37. [PMID: 27365173 DOI: 10.1016/j.neuroscience.2016.06.040] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 12/12/2022]
Abstract
Amitriptyline (AMI), a commonly prescribed tricyclic antidepressant (TCA) to parkinsonian patients, specifically showed a significant delay in dopaminergic therapy initiation and improvement in motor disability in parkinsonian patients. Moreover, it was recently shown that AMI has neuroprotective properties; however, the mechanisms underlying this effect in Parkinson's disease (PD) are not fully understood. The current study aimed to investigate the possible neuroprotective mechanisms afforded by AMI in the rotenone model of PD and to assess whether another TCA member, imipramine (IMI), shows a corresponding effect. Rats were allocated into seven groups. Four groups were given either saline, dimethyl sulfoxide, AMI or IMI. Three rotenone groups were either untreated or treated with AMI or IMI. Rats receiving rotenone exhibited motor impairment in open field and rotarod tests. Additionally, immunohistochemical examination revealed dopaminergic neuronal damage in substantia nigra. Besides, striatal monoamines and brain derived neurotrophic factor levels were declined. Furthermore, oxidative stress, microglial activation and inflammation were evident in the striata. Pretreatment of rotenone groups with AMI or IMI prevented rotenone-induced neuronal degeneration and increased striatal dopamine level with motor recovery. These effects were accompanied by restoring striatal monoamines and brain-derived neurotrophic factor levels, as well as reducing oxidative damage, microglial activation and expression of proinflammatory cytokines and inducible nitric oxide synthase. The present results suggest that modulation of noradrenaline and serotonin levels, up-regulation of neurotrophin, inhibition of glial activation, anti-oxidant and anti-inflammatory activities could serve as important mechanisms underlying the neuroprotective effects of the used drugs in the rotenone model of PD.
Collapse
Affiliation(s)
- Esraa A Kandil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Noha F Abdelkader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Bahia M El-Sayeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Samira Saleh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
26
|
Vitamin K2 suppresses rotenone-induced microglial activation in vitro. Acta Pharmacol Sin 2016; 37:1178-89. [PMID: 27498777 DOI: 10.1038/aps.2016.68] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/30/2016] [Indexed: 02/06/2023]
Abstract
AIM Increasing evidence has shown that environmental factors such as rotenone and paraquat induce neuroinflammation, which contributes to the pathogenesis of Parkinson's disease (PD). In this study, we investigated the molecular mechanisms underlying the repression by menaquinone-4 (MK-4), a subtype of vitamin K2, of rotenone-induced microglial activation in vitro. METHODS A microglial cell line (BV2) was exposed to rotenone (1 μmol/L) with or without MK-4 treatment. The levels of TNF-α or IL-1β in 100 μL of cultured media of BV2 cells were measured using ELISA kits. BV2 cells treated with rotenone with or without MK4 were subjected to mitochondrial membrane potential, ROS production, immunofluorescence or immunoblot assays. The neuroblastoma SH-SY5Y cells were treated with conditioned media (CM) of BV2 cells that were exposed to rotenone with or without MK-4 treatment, and the cell viability was assessed using MTT assay. RESULTS In rotenone-treated BV2 cells, MK-4 (0.5-20 μmol/L) dose-dependently suppressed the upregulation in the expression of iNOS and COX-2 in the cells, as well as the production of TNF-α and IL-1β in the cultured media. MK-4 (5-20 μmol/L) significantly inhibited rotenone-induced nuclear translocation of NF-κB in BV2 cells. MK-4 (5-20 μmol/L) significantly inhibited rotenone-induced p38 activation, ROS production, and caspase-1 activation in BV2 cells. MK-4 (5-20 μmol/L) also restored the mitochondrial membrane potential that had been damaged by rotenone. Exposure to CM from rotenone-treated BV2 cells markedly decreased the viability of SH-SY5Y cells. However, this rotenone-activated microglia-mediated death of SH-SY5Y cells was significantly attenuated when the BV2 cells were co-treated with MK-4 (5-20 μmol/L). CONCLUSION Vitamin K2 can directly suppress rotenone-induced activation of microglial BV2 cells in vitro by repressing ROS production and p38 activation.
Collapse
|
27
|
Mirzaie M, Khalili M, Kiasalari Z, Roghani M. Neuroprotective and Antiapoptotic Potential of Trigonelline in a Striatal 6-Hydroxydopamine Rat Model of Parkinson’s Disease. NEUROPHYSIOLOGY+ 2016. [DOI: 10.1007/s11062-016-9586-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
28
|
Gore PR, Prajapati CP, Mahajan UB, Goyal SN, Belemkar S, Ojha S, Patil CR. Protective Effect of Thymoquinone against Cyclophosphamide-Induced Hemorrhagic Cystitis through Inhibiting DNA Damage and Upregulation of Nrf2 Expression. Int J Biol Sci 2016; 12:944-53. [PMID: 27489498 PMCID: PMC4971733 DOI: 10.7150/ijbs.15781] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/06/2016] [Indexed: 12/17/2022] Open
Abstract
Cyclophosphamide (CYP) induced hemorrhagic cystitis is a dose-limiting side effect involving increased oxidative stress, inflammatory cytokines and suppressed activity of nuclear factor related erythroid 2-related factor (Nrf2). Thymoquinone (TQ), an active constituent of Nigella sativa seeds, is reported to increase the expression of Nrf2, exert antioxidant action, and anti-inflammatory effects in the experimental animals. The present study was designed to explore the effects of TQ on CYP-induced hemorrhagic cystitis in Balb/c mice. Cystitis was induced by a single intraperitoneal injection of CYP (200 mg/kg). TQ was administered intraperitoneally at 5, 10 and 20 mg/kg doses twice a day, for three days before and three days after the CYP administration. The efficacy of TQ was determined in terms of the protection against the CYP-induced histological perturbations in the bladder tissue, reduction in the oxidative stress, and inhibition of the DNA fragmentation. Immunohistochemistry was performed to examine the expression of Nrf2. TQ protected against CYP-induced oxidative stress was evident from significant reduction in the lipid peroxidation, restoration of the levels of reduced glutathione, catalase and superoxide dismutase activities. TQ treatment significantly reduced the DNA damage evident as reduced DNA fragmentation. A significant decrease in the cellular infiltration, edema, epithelial denudation and hemorrhage were observed in the histological observations. There was restoration and rise in the Nrf2 expression in the bladder tissues of mice treated with TQ. These results confirm that, TQ ameliorates the CYP-induced hemorrhagic cystitis in mice through reduction in the oxidative stress, inhibition of the DNA damage and through increased expression of Nrf2 in the bladder tissues.
Collapse
Affiliation(s)
- Prashant R Gore
- 1. Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, District-Dhule, Maharashtra, 425405, India
| | - Chaitali P Prajapati
- 1. Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, District-Dhule, Maharashtra, 425405, India
| | - Umesh B Mahajan
- 1. Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, District-Dhule, Maharashtra, 425405, India
| | - Sameer N Goyal
- 1. Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, District-Dhule, Maharashtra, 425405, India
| | - Sateesh Belemkar
- 2. School of Pharmacy and Technology Management, SVKM's NMIMS, MPTP, Shirpur, District- Dhule, Maharashtra, 425405, India
| | - Shreesh Ojha
- 3. Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates, University, Al Ain, Abu Dhabi 17666, United Arab Emirates
| | - Chandragouda R Patil
- 1. Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, District-Dhule, Maharashtra, 425405, India
| |
Collapse
|
29
|
Wang S, Zhang H, Xu Y. Crosstalk between microglia and T cells contributes to brain damage and recovery after ischemic stroke. Neurol Res 2016; 38:495-503. [DOI: 10.1080/01616412.2016.1188473] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
30
|
Kiasalari Z, Baluchnejadmojarad T, Roghani M. Hypericum Perforatum Hydroalcoholic Extract Mitigates Motor Dysfunction and is Neuroprotective in Intrastriatal 6-Hydroxydopamine Rat Model of Parkinson's Disease. Cell Mol Neurobiol 2016; 36:521-30. [PMID: 26119304 PMCID: PMC11482409 DOI: 10.1007/s10571-015-0230-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/20/2015] [Indexed: 12/31/2022]
Abstract
Parkinson's disease is the second most common neurodegenerative disorder with selective and progressive decline of nigral dopaminergic neurons. Hypericum perforatum L. (H. perforatum, St. John's wort) has been traditionally used for management of different disorders, especially mild-to-moderate depression. This study was conducted to evaluate the effect of H. perforatum extract against unilateral striatal 6-hydroxydopamine (6-OHDA) toxicity and to unmask some involved mechanisms. Intrastriatal 6-OHDA-lesioned rats were treated with H. perforatum hydroalcoholic extract at a dose of 200 mg/kg/day started 1 week pre-surgery for 1 week post-surgery. The extract attenuated apomorphine-induced rotational behavior, decreased the latency to initiate and the total time on the narrow beam task, lowered striatal level of malondialdehyde and enhanced striatal catalase activity and reduced glutathione content, normalized striatal expression of glial fibrillary acidic protein, tumor necrosis factor α with no significant effect on mitogen-activated protein kinase, lowered nigral DNA fragmentation, and prevented damage of nigral dopaminergic neurons with a higher striatal tyrosine hydroxylase immunoreactivity. These findings reveal the beneficial effect of H. perforatum via attenuation of DNA fragmentation, astrogliosis, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Zahra Kiasalari
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| | | | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| |
Collapse
|
31
|
Aquaporin-4 mediates communication between astrocyte and microglia: Implications of neuroinflammation in experimental Parkinson’s disease. Neuroscience 2016; 317:65-75. [DOI: 10.1016/j.neuroscience.2016.01.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/05/2016] [Accepted: 01/05/2016] [Indexed: 01/08/2023]
|
32
|
Protective Effect of Oral Hesperetin Against Unilateral Striatal 6-Hydroxydopamine Damage in the Rat. Neurochem Res 2015; 41:1065-72. [DOI: 10.1007/s11064-015-1796-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 12/01/2015] [Accepted: 12/08/2015] [Indexed: 12/12/2022]
|
33
|
Nelson PT, Jicha GA, Wang WX, Ighodaro E, Artiushin S, Nichols CG, Fardo DW. ABCC9/SUR2 in the brain: Implications for hippocampal sclerosis of aging and a potential therapeutic target. Ageing Res Rev 2015; 24:111-25. [PMID: 26226329 PMCID: PMC4661124 DOI: 10.1016/j.arr.2015.07.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/24/2015] [Indexed: 01/06/2023]
Abstract
The ABCC9 gene and its polypeptide product, SUR2, are increasingly implicated in human neurologic disease, including prevalent diseases of the aged brain. SUR2 proteins are a component of the ATP-sensitive potassium ("KATP") channel, a metabolic sensor for stress and/or hypoxia that has been shown to change in aging. The KATP channel also helps regulate the neurovascular unit. Most brain cell types express SUR2, including neurons, astrocytes, oligodendrocytes, microglia, vascular smooth muscle, pericytes, and endothelial cells. Thus it is not surprising that ABCC9 gene variants are associated with risk for human brain diseases. For example, Cantu syndrome is a result of ABCC9 mutations; we discuss neurologic manifestations of this genetic syndrome. More common brain disorders linked to ABCC9 gene variants include hippocampal sclerosis of aging (HS-Aging), sleep disorders, and depression. HS-Aging is a prevalent neurological disease with pathologic features of both neurodegenerative (aberrant TDP-43) and cerebrovascular (arteriolosclerosis) disease. As to potential therapeutic intervention, the human pharmacopeia features both SUR2 agonists and antagonists, so ABCC9/SUR2 may provide a "druggable target", relevant perhaps to both HS-Aging and Alzheimer's disease. We conclude that more work is required to better understand the roles of ABCC9/SUR2 in the human brain during health and disease conditions.
Collapse
Affiliation(s)
- Peter T Nelson
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA; University of Kentucky, Department of Pathology, Lexington, KY 40536, USA.
| | - Gregory A Jicha
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA; University of Kentucky, Department of Neurology, Lexington, KY, 40536, USA
| | - Wang-Xia Wang
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA
| | - Eseosa Ighodaro
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA
| | - Sergey Artiushin
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - David W Fardo
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA; Department of Biostatistics, Lexington, KY, 40536, USA
| |
Collapse
|
34
|
Ji J, Yan H, Chen ZZ, Zhao Z, Yang DD, Sun XL, Shi YP. Iptakalim protects against ischemic injury by improving neurovascular unit function in the mouse brain. Clin Exp Pharmacol Physiol 2015; 42:766-71. [DOI: 10.1111/1440-1681.12426] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Juan Ji
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | - Hui Yan
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | - Zheng-Zhen Chen
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | - Zhan Zhao
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | - Dan-Dan Yang
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | - Xiu-Lan Sun
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | | |
Collapse
|
35
|
Zhu R, Bi LQ, Wu SL, Li L, Kong H, Xie WP, Wang H, Meng ZL. Iptakalim attenuates hypoxia-induced pulmonary arterial hypertension in rats by endothelial function protection. Mol Med Rep 2015; 12:2945-52. [PMID: 25936382 DOI: 10.3892/mmr.2015.3695] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 01/09/2015] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to investigate the protective effects of iptakalim, an adenosine triphosphate (ATP)-sensitive potassium channel opener, on the inflammation of the pulmonary artery and endothelial cell injury in a hypoxia-induced pulmonary arterial hypertension (PAH) rat model. Ninety-six Sprague-Dawley rats were placed into normobaric hypoxia chambers for four weeks and were treated with iptakalim (1.5 mg/kg/day) or saline for 28 days. The right ventricle systolic pressures (RVSP) were measured and small pulmonary arterial morphological alterations were analyzed with hematoxylin and eosin staining. Enzyme-linked immunosorbent assay (ELISA) was performed to analyze the content of interleukin (IL)-1β and IL-10. Immunohistochemical analysis for ED1(+) monocytes was performed to detect the inflammatory cells surrounding the pulmonary arterioles. Western blot analysis was performed to analyze the expression levels of platelet endothelial cell adhesion molecule-1 (PECAM-1) and endothelial nitric oxide synthase (eNOS) in the lung tissue. Alterations in small pulmonary arteriole morphology and the ultrastructure of pulmonary arterial endothelial cells were observed via light and transmission electron microscopy, respectively. Iptakalim significantly attenuated the increase in mean pulmonary artery pressure, RVSP, right ventricle to left ventricle plus septum ratio and small pulmonary artery wall remodeling in hypoxia-induced PAH rats. Iptakalim also prevented an increase in IL-1β and a decrease in IL-10 in the peripheral blood and lung tissue, and alleviated inflammatory cell infiltration in hypoxia-induced PAH rats. Furthermore, iptakalim enhanced PECAM-1 and eNOS expression and prevented the endothelial cell injury induced by hypoxic stimuli. Iptakalim suppressed the pulmonary arteriole and systemic inflammatory responses and protected against the endothelial damage associated with the upregulation of PECAM-1 and eNOS, suggesting that iptakalim may represent a potential therapeutic agent for PAH.
Collapse
Affiliation(s)
- Rong Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Li-Qing Bi
- Geriatric Intensive Care Unit, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Su-Ling Wu
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lan Li
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hui Kong
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei-Ping Xie
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hong Wang
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zi-Li Meng
- Department of Respiratory Medicine, The Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
36
|
Cebrián C, Loike JD, Sulzer D. Neuroinflammation in Parkinson's disease animal models: a cell stress response or a step in neurodegeneration? Curr Top Behav Neurosci 2015; 22:237-270. [PMID: 25293443 DOI: 10.1007/7854_2014_356] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The motor symptoms of Parkinson's disease are due to the progressive degeneration of dopaminergic neurons in the substantia nigra. Multiple neuroinflammatory processes are exacerbated in Parkinson's disease, including glial-mediated reactions, increased expression of proinflammatory substances, and lymphocytic infiltration, particularly in the substantia nigra. Neuroinflammation is also implicated in the neurodegeneration and consequent behavioral symptoms of many Parkinson's disease animal models, although it is not clear whether these features emulate pathogenic steps in the genuine disorder or if some inflammatory features provide protective stress responses. Here, we compare and summarize findings on neuroinflammatory responses and effects on behavior in a wide range of toxin-based, inflammatory and genetic Parkinson's disease animal models.
Collapse
Affiliation(s)
- Carolina Cebrián
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | | | | |
Collapse
|
37
|
von Wrangel C, Schwabe K, John N, Krauss JK, Alam M. The rotenone-induced rat model of Parkinson's disease: behavioral and electrophysiological findings. Behav Brain Res 2014; 279:52-61. [PMID: 25446762 DOI: 10.1016/j.bbr.2014.11.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/29/2014] [Accepted: 11/04/2014] [Indexed: 12/21/2022]
Abstract
Exposure to rotenone leads to parkinsonian features, such as loss of dopaminergic neurons in the substantia nigra and motor impairment, however, the validity of this model has recently been questioned. In rodent and monkey models of Parkinson's disease (PD) abnormal neuronal activity in the basal ganglia motor loop has been described, with hyperactivity of the subthalamic nucleus (STN) similar to that found in PD. The present study aims at providing new and more specific evidence for the validity of the rotenone rat model of PD by examining whether neuronal activity in the STN is altered. Male Sprague Dawley rats were treated with rotenone injections (2.5mg/kg bodyweight intraperitoneally) for 60 days. Behavioral analysis showed an impairment in the rotarod and hanging wire test in the rotenone group (p<0.05), accompanied by a decline in tyrosine hydroxylase immunoreactive neurons in the nigro-striatal region (p<0.001). Thereafter, single unit (SU) activities and local field potentials were recorded in the STN in urethane anesthetized rats. The SU analysis revealed a higher neuronal discharge rate (p<0.001), more bursts per minute (p=0.006) and a higher oscillatory activity (p=0.008) in the STN of rotenone treated rats. Spectral analysis showed an increase of relative beta power in the STN as well as in the motor cortex. We found electrophysiological key features of PD pathology and pathophysiology in the STN of rotenone treated rats. Therefore, the rotenone-induced rat model of PD deserves further attention since it covers more aspects than dopamine depletion and implies the reproducibility of PD specific features.
Collapse
Affiliation(s)
| | - Kerstin Schwabe
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Nadine John
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Joachim K Krauss
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Mesbah Alam
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
38
|
Bordt EA, Polster BM. NADPH oxidase- and mitochondria-derived reactive oxygen species in proinflammatory microglial activation: a bipartisan affair? Free Radic Biol Med 2014; 76:34-46. [PMID: 25091898 PMCID: PMC4252610 DOI: 10.1016/j.freeradbiomed.2014.07.033] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/19/2014] [Accepted: 07/24/2014] [Indexed: 12/18/2022]
Abstract
Microglia are the resident immune cells of the brain and play major roles in central nervous system development, maintenance, and disease. Brain insults cause microglia to proliferate, migrate, and transform into one or more activated states. Classical M1 activation triggers the production of proinflammatory factors such as tumor necrosis factor-α, interleukin-1β (IL-1β), nitric oxide, and reactive oxygen species (ROS), which, in excess, can exacerbate brain injury. The mechanisms underlying microglial activation are not fully understood, yet reactive oxygen species are increasingly implicated as mediators of microglial activation. In this review, we highlight studies linking reactive oxygen species, in particular hydrogen peroxide derived from NADPH oxidase-generated superoxide, to the classical activation of microglia. In addition, we critically evaluate controversial evidence suggesting a specific role for mitochondrial reactive oxygen species in the activation of the NLRP3 inflammasome, a multiprotein complex that mediates the production of IL-1β and IL-18. Finally, the limitations of common techniques used to implicate mitochondrial ROS in microglial and inflammasome activation, such as the use of the mitochondrially targeted ROS indicator MitoSOX and the mitochondrially targeted antioxidant MitoTEMPO, are also discussed.
Collapse
Affiliation(s)
- Evan A Bordt
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research, and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Brian M Polster
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research, and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
39
|
Jiang BP, Le L, Xu LJ, Xiao PG. Minocycline inhibits ICAD degradation and the NF-κB activation induced by 6-OHDA in PC12 cells. Brain Res 2014; 1586:1-11. [PMID: 25195972 DOI: 10.1016/j.brainres.2014.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/30/2014] [Accepted: 08/01/2014] [Indexed: 01/08/2023]
Abstract
6-Hydroxydopamine (6-OHDA) is a neurotoxin that is commonly employed to induce lesions of the dopaminergic pathways to generating experimental models of Parkinson's disease (PD) in rodents. Antioxidant and anti-inflammatory therapy approaches have been the focus of attention in the treatment of neurodegenerative. PD and Alzheimer's diseases, and oxidative stress have been implicated in these diseases. In this study, we investigated the neuroprotective effects of minocycline and the signalling pathway that is possibly involved in a PC12 cell model of PD. The results indicated that 6-OHDA cytotoxicity was accompanied by an increment in lactate dehydrogenase (LDH) release, an increase in caspase-3 protein activity, an increase in ROS generation, MDA content and decrease in the SOD, CAT activities and cell viability. Moreover, treatment with 6-OHDA alone for 24h resulted in ICAD degradation, increased nuclear translocation of NF-κB, and increased p53 expression. However, pretreatment with minocycline (5, 10, 20 µM) for 24h significantly reduced LDH release, reduced caspase-3 protein production, reduced ROS production, MDA content and attenuated the decrease in SOD, CAT activities and cell viability. Additionally, minocycline (20 µM) markedly decreased the levels of cleaved ICAD protein, down-regulated p53 activity and inhibited the nuclear translocation of NF-κB. The neuroprotective effects of minocycline were attributable to its potent antioxidant activities, which prevented the nuclear translocation of NF-κB and the subsequent promotion of cell death. Therefore, the present study supports the notion that minocycline may be a promising neuroprotective agent for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Bao-Ping Jiang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Key Laboratory of Bioactive Substances and Resources Unilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Liang Le
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Li-Jia Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Key Laboratory of Bioactive Substances and Resources Unilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
| | - Pei-Gen Xiao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Key Laboratory of Bioactive Substances and Resources Unilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| |
Collapse
|
40
|
Iptakalim confers an antidepressant effect in a chronic mild stress model of depression through regulating neuro-inflammation and neurogenesis. Int J Neuropsychopharmacol 2014; 17:1501-10. [PMID: 24621884 DOI: 10.1017/s1461145714000285] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Depression is a serious mental disorder in the world, but the underlying mechanisms remain unclear and the effective cures are scarce. Iptakalim (Ipt), an ATP-sensitive potassium (K-ATP) channel opener that can cross the blood-brain barrier freely, has been demonstrated to inhibit neuro-inflammation and enhance adult hippocampal neurogenesis. But it is unknown whether Ipt is beneficial to therapy of depression by modulating neurogenesis and neuro-inflammation. This study aimed to determine the potential antidepressant efficacy of Ipt in a chronic mild stress (CMS) mouse model of depression. We showed that treatment with Ipt (10 mg/kg/day, i.p) for 4 wk restored the decrease of sucrose preference and shortened the immobile time in forced swimming tests (FST) and tail suspension tests (TST) in CMS model mice. We further found that Ipt reversed the CMS-induced reduction of the adult hippocampal neurogenesis and improved cerebral insulin signalling in the CMS mice. Furthermore, Ipt negatively regulated nod-like receptor protein 3 (NLRP3) expression and, in turn, inhibited microglia-mediated neuro-inflammation by suppressing the activation of NLRP3-inflammasome/caspase-1/interleukin 1β axis in the hippocampus of CMS mice. Taken together, our findings demonstrate that Ipt plays a potential antidepressant role in CMS model mice through regulating neuro-inflammation and neurogenesis, which will provide potential for Ipt in terms of opening up novel therapeutic avenues for depression.
Collapse
|
41
|
Zhang XY, Chen L, Yang Y, Xu DM, Zhang SR, Li CT, Zheng W, Yu SY, Wei EQ, Zhang LH. Regulation of rotenone-induced microglial activation by 5-lipoxygenase and cysteinyl leukotriene receptor 1. Brain Res 2014; 1572:59-71. [DOI: 10.1016/j.brainres.2014.05.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 04/15/2014] [Accepted: 05/13/2014] [Indexed: 12/11/2022]
|
42
|
Carnosine exerts neuroprotective effect against 6-hydroxydopamine toxicity in hemiparkinsonian rat. Mol Neurobiol 2014; 51:1064-70. [PMID: 24939694 DOI: 10.1007/s12035-014-8771-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/01/2014] [Indexed: 10/25/2022]
Abstract
Parkinson's disease (PD) is the second most common disorder of the central nervous system due to the degeneration of mesencephalic dopaminergic neurons. Current treatments for PD have a symptomatic relief strategy with no prevention of disease progression. Due to the neuroprotective and antiapoptotic potential of the natural dipeptide carnosine, this study was conducted to assess its beneficial effect in 6-hydroxydopamine (6-OHDA)-induced model of PD in rat. Unilateral intrastriatal 6-OHDA-lesioned rats received i.p. carnosine at a dose of 250 mg/kg twice at an interval of 24 h, which started presurgery. Apomorphine caused contralateral rotations, a significant reduction in the number of Nissl-stained neurons on the left side of the substantia nigra, and increased apoptosis was observed with enhanced oxidative stress burden in 6-OHDA-lesioned rats. Carnosine pretreatment significantly reduced rotations, attenuated apoptosis, and restored malondialdehyde and nitrite content and catalase activity with no significant effect on reduced glutathione (GSH). These results indicate that prelesion administration of carnosine could exert neuroprotection against 6-OHDA toxicity, and this may be of benefit in patients with early PD.
Collapse
|
43
|
Lee JE, Park JH, Jang SJ, Koh HC. Rosiglitazone inhibits chlorpyrifos-induced apoptosis via modulation of the oxidative stress and inflammatory response in SH-SY5Y cells. Toxicol Appl Pharmacol 2014; 278:159-71. [PMID: 24793810 DOI: 10.1016/j.taap.2014.04.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 04/04/2014] [Accepted: 04/22/2014] [Indexed: 01/21/2023]
Abstract
Oxidative stress can lead to expression of inflammatory transcription factors, which are important regulatory elements in the induction of inflammatory responses. One of the transcription factors, nuclear transcription factor kappa-B (NF-κB) plays a significant role in the inflammation regulatory process. Inflammatory cell death has been implicated in neuronal cell death in some neurodegenerative disorders such as Parkinson's disease (PD). In this study, we investigated the molecular mechanisms underlying apoptosis initiated by chlorpyrifos (CPF)-mediated oxidative stress. Based on the cytotoxic mechanism of CPF, we examined the neuroprotective effects of rosiglitazone (RGZ), a peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist, against CPF-induced neuronal cell death. The treatment of SH-SY5Y cells with CPF induced oxidative stress. In addition, CPF activated the p38, JNK and ERK mitogen-activated protein kinases (MAPKs), and induced increases in the inflammatory genes such as COX-2 and TNF-α. CPF also induced nuclear translocation of NF-κB and inhibitors of NF-κB abolished the CPF-induced COX-2 expression. Pretreatment with RGZ significantly reduced ROS generation and enhanced HO-1 expression in CPF-exposed cells. RGZ blocked the activation of both p38 and JNK signaling, while ERK activation was strengthened. RGZ also attenuated CPF-induced cell death through the reduction of NF-κB-mediated proinflammatory factors. Results from this study suggest that RGZ may exert an anti-apoptotic effect against CPF-induced cytotoxicity by attenuation of oxidative stress as well as inhibition of the inflammatory cascade via inactivation of signaling by p38 and JNK, and NF-κB.
Collapse
Affiliation(s)
- Jeong Eun Lee
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul, Republic of Korea; Hanyang Biomedical Research Institute, Seoul, Republic of Korea
| | - Jae Hyeon Park
- Hanyang Biomedical Research Institute, Seoul, Republic of Korea; Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Sea Jeong Jang
- Hanyang Biomedical Research Institute, Seoul, Republic of Korea; Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hyun Chul Koh
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul, Republic of Korea; Hanyang Biomedical Research Institute, Seoul, Republic of Korea; Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Yang C, Yang L, Liu Y. Soluble complement complex C5b-9 promotes microglia activation. J Neuroimmunol 2013; 267:16-9. [PMID: 24434076 DOI: 10.1016/j.jneuroim.2013.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 11/22/2013] [Accepted: 11/28/2013] [Indexed: 01/23/2023]
Abstract
Soluble C5b-9 has been described as a pro-inflammatory mediator that triggers cell activation rather than inducing cell death. Microglia is the most important immune cell involved in inflammatory response in the CNS. Although microglia activation induced by various stimuli has been well characterized, the role of C5b-9 in microglia has not been well studied. In the current experiment, we utilized assembled functional C5b-9 to treat microglia and analyzed the function. We found that soluble C5b-9 could promote microglia activation by up-regulation of costimulatory molecules and increase cytokine secretion. Our results suggested that soluble C5b-9 possessed immunoregulatory potential on microglia.
Collapse
Affiliation(s)
- Chao Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Li Yang
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Yong Liu
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
45
|
Gao F, Chen D, Hu Q, Wang G. Rotenone directly induces BV2 cell activation via the p38 MAPK pathway. PLoS One 2013; 8:e72046. [PMID: 23977201 PMCID: PMC3748029 DOI: 10.1371/journal.pone.0072046] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Accepted: 07/06/2013] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. Although its pathogenesis is still unclear, increasing evidence suggests that mitochondrial dysfunction induced by environmental toxins, such as mitochondrial complex I inhibitors, plays a significant role in the disease process. The microglia in PD brains are highly activated, and inflammation is also an essential element in PD pathogenesis. However, the means by which these toxins activate microglia is still unclear. In the present study, we found that rotenone, a mitochondrial complex I inhibitor, could directly activate microglia via the nuclear factor kappa B (NF-κB) signaling pathway, thereby inducing significantly increased expression of inflammatory cytokines. We further observed that rotenone induced caspase-1 activation and mature IL-1β release, both of which are strictly dependent on p38 mitogen-activated protein kinase (MAPK). The activation of p38 is associated with the presence of reactive oxygen species (ROS) produced by rotenone. Removal of these ROS abrogated the activation of the microglia. Therefore, our data suggest that the environmental toxin rotenone can directly activate microglia through the p38 MAPK pathway.
Collapse
Affiliation(s)
- Feng Gao
- Laboratory of Molecular Neuropathology, Key Laboratory of Brain Function and Diseases and School of Life Sciences, University of Science & Technology of China, Chinese Academy of Sciences, Hefei, China
- * E-mail: (FG); (GH)
| | - Dong Chen
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Soochow University College of Pharmaceutical Sciences, Suzhou, China
| | - Qingsong Hu
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Soochow University College of Pharmaceutical Sciences, Suzhou, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Key Laboratory of Brain Function and Diseases and School of Life Sciences, University of Science & Technology of China, Chinese Academy of Sciences, Hefei, China
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Soochow University College of Pharmaceutical Sciences, Suzhou, China
- * E-mail: (FG); (GH)
| |
Collapse
|
46
|
Inhibition of lipopolysaccharide-induced proinflammatory responses by Buddleja officinalis extract in BV-2 microglial cells via negative regulation of NF-kB and ERK1/2 signaling. Molecules 2013; 18:9195-206. [PMID: 23912273 PMCID: PMC6270610 DOI: 10.3390/molecules18089195] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 07/24/2013] [Accepted: 07/25/2013] [Indexed: 11/16/2022] Open
Abstract
Buddleja officinalis has been traditionally used in the supportive treatment of inflammatory and neuronal diseases in Korea and China. Although several reports have shown the anti-inflammatory effects of Buddleja officinalis, the anti-neuroinflammatory effect has remained unclear. In this study, we aimed to investigate the inhibitory effects of flower buds of B. officinalis Maximowicz water extract (BOWE) on LPS-induced inflammatory processes in BV-2 microglial cells. BOWE dose-dependently inhibited the production of nitric oxide as well as iNOS mRNA expression. Moreover, BOWE prevented IL-1β and IL-6 mRNA expression. However, BOWE had no effect on LPS-induced COX-2 or TNF-α mRNA expression. The extract also had no effect on LPS-stimulated p38 MAPK, JNK, and c-Jun phosphorylation, whereas ERK1/2 phosphorylation was strongly inhibited by BOWE. BOWE also inhibited the LPS-induced degradation of IκB-α, and LPS-induced phosphorylation of p65 NF-κB protein. These data indicate that BOWE inhibited the nitric oxide production and pro-inflammatory gene expression in BV-2 microglial cells, possibly through a negative regulation of the NF-κB and ERK1/2 pathways. Further identification of the direct target molecule(s) of BOWE is required to support its use as an anti-neuroinflammatory agent against the neurodegenerative disorders.
Collapse
|
47
|
Emmrich JV, Hornik TC, Neher JJ, Brown GC. Rotenone induces neuronal death by microglial phagocytosis of neurons. FEBS J 2013; 280:5030-8. [PMID: 23789887 DOI: 10.1111/febs.12401] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 06/08/2013] [Accepted: 06/11/2013] [Indexed: 12/21/2022]
Abstract
Rotenone, a common pesticide and inhibitor of mitochondrial complex I, induces microglial activation and loss of dopaminergic neurons in models of Parkinson's disease. However, the mechanisms of rotenone neurotoxicity are still poorly defined. Here, we used primary neuronal/glial cultures prepared from rat cerebella to investigate the contribution of microglia to neuronal cell death induced by low concentrations of rotenone. Rotenone at 2.5 nm induced neuronal loss over several days without increasing the numbers of necrotic or apoptotic neurons, and neuronal loss/death could be prevented by selective removal of microglia. Rotenone increased microglial proliferation and phagocytic activity, without increasing tumour necrosis factor-α release. Rotenone-induced neuronal loss/death could be prevented by inhibition of phagocytic signalling between neurons and microglia with: cyclo(Arg-Gly-Asp-d-Phe-Val) (to block the microglial vitronectin receptor); MRS2578 (to block the microglial P2Y6 receptor); or either annexin V or an antibody against phosphatidylserine (to block exposed phosphatidylserine, a well-characterized neuronal 'eat-me' signal). As inhibition of phagocytosis by five different means prevented neuronal loss without increasing neuronal death, these data indicate that rotenone neurotoxicity is at least partially mediated by microglial phagocytosis of otherwise viable neurons (phagoptosis). Thus, neuronal loss in Parkinson's disease and other neurological diseases might be prevented by blocking phagocytic signalling.
Collapse
|
48
|
Ferris CF, Marella M, Smerkers B, Barchet TM, Gershman B, Matsuno-Yagi A, Yagi T. A phenotypic model recapitulating the neuropathology of Parkinson's disease. Brain Behav 2013; 3:351-66. [PMID: 24381808 PMCID: PMC3869678 DOI: 10.1002/brb3.138] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 03/08/2013] [Accepted: 03/18/2013] [Indexed: 12/21/2022] Open
Abstract
This study was undertaken to develop a phenotypic model recapitulating the neuropathology of Parkinson's disease (PD). Such a model would show loss of dopamine in the basal ganglia, appearance of Lewy bodies, and the early stages of motor dysfunction. The model was developed by subcutaneously injecting biodegradable microspheres of rotenone, a complex I inhibitor in 8-9 month old, ovariectomized Long-Evans rats. Animals were observed for changes in body weight and motor activity. At the end of 11-12 weeks animals were euthanized and the brains examined for histopathological changes. Rotenone treated animals gain weight and appear normal and healthy as compared to controls but showed modest hypokinesia around 5-6 weeks posttreatment. Animals showed loss of dopaminergic (DA) neurons and the appearance of putative Lewy bodies in the substantia nigra. Neuroinflammation and oxidative stress were evidenced by the appearance of activated microglia, iron precipitates, and 8-oxo-2'-deoxyguanosine a major product of DNA oxidation. The dorsal striatum, the projection site of midbrain DA neurons, showed a significant reduction in tyrosine hydroxylase immunostaining, together with an increase in reactive astrocytes, an early sign of DA nerve terminal damage. Levels of vesicular monoamine transporter 2 (VMAT2) were significantly reduced in the dorsal striatum; however, there was an unexpected increase in dopamine transporter (DAT) levels. Old, ovariectomized females treated with rotenone microspheres present with normal weight gain and good health but a modest hypokinesia. Accompanying this behavioral phenotype are a constellation of neuropathologies characteristic of PD that include loss of DA neurons, microglia activation, oxidative damage to nuclear DNA, iron deposition, and appearance of putative Lewy bodies. This phenotypic model recapitulating the neuropathology of Parkinson's disease could provide insight into early mechanisms of pathogenesis and could aid in the identification of biomarkers to identify patients in early stage, PD.
Collapse
Affiliation(s)
- Craig F Ferris
- Center for Translational NeuroImaging, Northeastern University Boston, Massachusetts
| | - Mathieu Marella
- Division of Biochemistry, Department of Molecular and Experimental Medicine, The Scripps Research Institute La Jolla, California
| | - Brian Smerkers
- State University of New York Upstate Medical University Syracuse, New York
| | - Thomas M Barchet
- Center for Translational NeuroImaging, Northeastern University Boston, Massachusetts
| | | | - Akemi Matsuno-Yagi
- Division of Biochemistry, Department of Molecular and Experimental Medicine, The Scripps Research Institute La Jolla, California
| | - Takao Yagi
- Division of Biochemistry, Department of Molecular and Experimental Medicine, The Scripps Research Institute La Jolla, California
| |
Collapse
|
49
|
Taylor JM, Main BS, Crack PJ. Neuroinflammation and oxidative stress: Co-conspirators in the pathology of Parkinson’s disease. Neurochem Int 2013; 62:803-19. [DOI: 10.1016/j.neuint.2012.12.016] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 12/20/2012] [Accepted: 12/26/2012] [Indexed: 12/21/2022]
|
50
|
Jalali-Nadoushan M, Roghani M. Alpha-lipoic acid protects against 6-hydroxydopamine-induced neurotoxicity in a rat model of hemi-parkinsonism. Brain Res 2013; 1505:68-74. [DOI: 10.1016/j.brainres.2013.01.054] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Revised: 01/30/2013] [Accepted: 01/31/2013] [Indexed: 10/27/2022]
|