1
|
Batey L, Baumberger B, Khoshbouei H, Hashemi P. Lipopolysaccharide Effects on Neurotransmission: Understanding Implications for Depression. ACS Chem Neurosci 2024; 15:4339-4347. [PMID: 39601433 DOI: 10.1021/acschemneuro.4c00591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Immune activation in the body is well studied; however, much less is known about how peripheral inflammation changes brain chemistry. Because depression and inflammation are close comorbidities, investigating how inflammation affects the brain's chemicals will help us to better understand depression. The levels of the monoamines dopamine, serotonin and norepinephrine are thought to be affected by both inflammation and depression. In this Perspective, we review studies that find chemical changes in the brain after administration of the endotoxin LPS, which is a robust method to induce rapid inflammation. From these studies, we interpreted LPS to reduce dopamine and serotonin and increase norepinephrine levels in various regions in the brain. These changes are not a sign of "dysfunction" but serve an important evolutionary purpose that encourages the body to recover from an immune insult by altering mood.
Collapse
Affiliation(s)
- L Batey
- Department of Bioengineering, Imperial College, South Kensington, London, SW7 2AZ, U.K
| | - B Baumberger
- Department of Bioengineering, Imperial College, South Kensington, London, SW7 2AZ, U.K
| | - H Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida 32610, United States
| | - P Hashemi
- Department of Bioengineering, Imperial College, South Kensington, London, SW7 2AZ, U.K
| |
Collapse
|
2
|
Ghasemi M, Mehranfard N. Neuroprotective actions of norepinephrine in neurological diseases. Pflugers Arch 2024; 476:1703-1725. [PMID: 39136758 DOI: 10.1007/s00424-024-02999-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/24/2024] [Accepted: 07/24/2024] [Indexed: 10/09/2024]
Abstract
Precise control of norepinephrine (NE) levels and NE-receptor interaction is crucial for proper function of the brain. Much evidence for this view comes from experimental studies that indicate an important role for NE in the pathophysiology and treatment of various conditions, including cognitive dysfunction, Alzheimer's disease, Parkinson's disease, multiple sclerosis, and sleep disorders. NE provides neuroprotection against several types of insults in multiple ways. It abrogates oxidative stress, attenuates neuroinflammatory responses in neurons and glial cells, reduces neuronal and glial cell activity, promotes autophagy, and ameliorates apoptotic responses to a variety of insults. It is beneficial for the treatment of neurodegenerative diseases because it improves the generation of neurotrophic factors, promotes neuronal survival, and plays an important role in the regulation of adult neurogenesis. This review aims to present the evidence supporting a principal role for NE in neuroprotection, and molecular mechanisms of neuroprotection.
Collapse
Affiliation(s)
- Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasrin Mehranfard
- Nanokadeh Darooee Samen Private Joint Stock Company, Shafa Street, Urmia, 5715793731, Iran.
| |
Collapse
|
3
|
Merzon E, Magen E, Ashkenazi S, Weizman A, Manor I, Krone B, Green I, Golan-Cohen A, Vinker S, Faraone SV, Israel A. The Association between Glucose 6-Phosphate Dehydrogenase Deficiency and Attention Deficit/Hyperactivity Disorder. Nutrients 2023; 15:4948. [PMID: 38068806 PMCID: PMC10708268 DOI: 10.3390/nu15234948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Glucose-6-phosphate dehydrogenase (G6PD) deficiency, impacting 4.9% of the population and more prevalent in Mediterranean communities, is a common enzymopathy with potential relevance to Attention Deficit/Hyperactivity Disorder (ADHD). This study investigated this association. METHODS The clinical characteristics of 7473 G6PD-deficient patients and 29,892 matched case-controls (selected at a 1:4 ratio) from a cohort of 1,031,354 within the Leumit Health Services database were analyzed using Fisher's exact test for categorical variables and the Mann-Whitney U test for continuous variables. RESULTS In total, 68.7% were male. The mean duration of follow-up was 14.3 ± 6.2 years at a mean age of 29.2 ± 22.3 years. G6PD deficiency was associated with an increased risk of being diagnosed with ADHD (Odds Ratio (OR) = 1.16 [95% CI, 1.08-1.25], p < 0.001), seeking care from adult neurologists (OR = 1.30 [95% CI, 1.22-1.38], p < 0.001), and consulting adult psychiatrists (OR = 1.12 [95% CI, 1.01-1.24], p = 0.048). The use of stimulant medications among G6PD-deficient individuals was 17% higher for the methylphenidate class of drugs (OR = 1.17 [95% CI, 1.08, 1.27], p < 0.001), and there was a 16% elevated risk for amphetamine use (OR = 1.16 [95% CI, 1.03, 1.37], p = 0.047). CONCLUSIONS G6PD deficiency signals an increased risk of ADHD diagnosis, more severe presentations of ADHD and a greater need for psychiatric medications to treat ADHD.
Collapse
Affiliation(s)
- Eugene Merzon
- Adelson School of Medicine, Ariel University, Ariel 40776, Israel;
- Leumit Health Services, Tel Aviv 64738, Israel; (I.G.); (A.G.-C.); (S.V.); (A.I.)
| | - Eli Magen
- Department of Medicine A, Assuta Ashdod University Hospital, Faculty of Health Sciences, Ben Gurion University, Beer Sheba 84990, Israel;
| | - Shai Ashkenazi
- Adelson School of Medicine, Ariel University, Ariel 40776, Israel;
| | - Abraham Weizman
- ADHD Unit, Geha Mental Health Center, Petah Tikva 49100, Israel; (A.W.); (I.M.)
- Department of Psychiatry, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Laboratory of Molecular and Biological Psychiatry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Iris Manor
- ADHD Unit, Geha Mental Health Center, Petah Tikva 49100, Israel; (A.W.); (I.M.)
- Department of Psychiatry, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Beth Krone
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Ilan Green
- Leumit Health Services, Tel Aviv 64738, Israel; (I.G.); (A.G.-C.); (S.V.); (A.I.)
- Department of Family Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Avivit Golan-Cohen
- Leumit Health Services, Tel Aviv 64738, Israel; (I.G.); (A.G.-C.); (S.V.); (A.I.)
- Department of Family Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shlomo Vinker
- Leumit Health Services, Tel Aviv 64738, Israel; (I.G.); (A.G.-C.); (S.V.); (A.I.)
- Department of Family Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Stephen V. Faraone
- Department of Psychiatry, Norton College of Medicine, SUNY Upstate Medical University, Syracuse, NY 13210, USA;
| | - Ariel Israel
- Leumit Health Services, Tel Aviv 64738, Israel; (I.G.); (A.G.-C.); (S.V.); (A.I.)
- Department of Epidemiology and Disease Prevention, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
4
|
Torrillas-de la Cal A, Torres-Sanchez S, Bravo L, Llorca-Torralba M, Garcia-Partida JA, Arroba AI, Berrocoso E. Chemogenetic activation of locus coeruleus neurons ameliorates the severity of multiple sclerosis. J Neuroinflammation 2023; 20:198. [PMID: 37658434 PMCID: PMC10474779 DOI: 10.1186/s12974-023-02865-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/30/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Most current disease-modifying therapies approved for multiple sclerosis (MS) are immunomodulatory drugs that counteract the aberrant activity of the immune system. Hence, new pharmacological interventions that drive anti-inflammatory activity and neuroprotection would represent interesting alternative therapeutic approaches or complementary strategies to treat progressive forms of MS. There is evidence of reduced noradrenaline levels and alterations to locus coeruleus (LC) noradrenergic neurons in MS patients, as well as in animal models of this disease, potentially factors contributing to the pathophysiology. Drugs that enhance noradrenaline appear to have some beneficial effects in MS, suggesting their potential to dampen the underlying pathology and disease progression. METHODS Therefore, we explored the consequences of chronic LC noradrenergic neurons activation by chemogenetics in experimental autoimmune encephalomyelitis (EAE) mice, the most widely used experimental model of MS. LC activation from the onset or the peak of motor symptoms was explored as two different therapeutic approaches, assessing the motor and non-motor behavioral changes as EAE progresses, and studying demyelination, inflammation and glial activation in the spinal cord and cerebral cortex during the chronic phase of EAE. RESULTS LC activation from the onset of motor symptoms markedly alleviated the motor deficits in EAE mice, as well as their anxiety-like behavior and sickness, in conjunction with reduced demyelination and perivascular infiltration in the spinal cord and glial activation in the spinal cord and prefrontal cortex (PFC). When animals exhibited severe paralysis, LC activation produced a modest alleviation of EAE motor symptoms and it enhanced animal well-being, in association with an improvement of the EAE pathology at the spinal cord and PFC level. Interestingly, the reduced dopamine beta-hydroxylase expression associated with EAE in the spinal cord and PFC was reversed through chemogenetic LC activation. CONCLUSION Therefore, clear anti-inflammatory and neuroprotective effects were produced by the selective activation of LC noradrenergic neurons in EAE mice, having greater benefits when LC activation commenced earlier. Overall, these data suggest noradrenergic LC neurons may be targets to potentially alleviate some of the motor and non-motor symptoms in MS.
Collapse
Grants
- #FPU20-03072 "Agencia Estatal de Investigación-Ministerio de Ciencia, Innovación y Universidades"; FPU fellowship
- PID2022-1427850B-I00 "Fondo Europeo de Desarrollo Regional" (FEDER)-UE "A way to build Europe" from the "Ministerio de Economía y Competitividad
- PDC2022-133987-I00 "Fondo Europeo de Desarrollo Regional" (FEDER)-UE "A way to build Europe" from the "Ministerio de Economía y Competitividad
- PY20_00958 "Consejería de Transformación Económica, Industria, Conocimiento y Universidad, Junta de Andalucía"
- CTS-510 "Consejería de Transformación Económica, Industria, Conocimiento y Universidad, Junta de Andalucía"
- CEIJ-003 CEIMAR
- “CIBERSAM”: CIBER-Consorcio Centro de Investigación Biomédica en Red (CB07/09/0033), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación
- “Agencia Estatal de Investigación-Ministerio de Ciencia, Innovación y Universidades”; FPU fellowship
- “Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz-INiBICA” (IN-CO9)
- "Fondo Europeo de Desarrollo Regional” (FEDER)-UE “A way to build Europe” from the “Ministerio de Economía y Competitividad
- Universidad de Cadiz
Collapse
Affiliation(s)
- Alejandro Torrillas-de la Cal
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain
- Ciber de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
| | - Sonia Torres-Sanchez
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain
- Ciber de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
| | - Lidia Bravo
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain
- Ciber de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
| | - Meritxell Llorca-Torralba
- Ciber de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Department of Cell Biology and Histology, University of Cádiz, 11003, Cádiz, Spain
| | - Jose Antonio Garcia-Partida
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain
- Ciber de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
| | - Ana I Arroba
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
- Department of Biomedicine, Biotechnology and Public Health (Immunology Area), University of Cádiz, 11003, Cádiz, Spain
| | - Esther Berrocoso
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003, Cádiz, Spain.
- Ciber de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain.
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain.
| |
Collapse
|
5
|
Hosomoto K, Sasaki T, Yasuhara T, Kameda M, Sasada S, Kin I, Kuwahara K, Kawauchi S, Okazaki Y, Yabuno S, Sugahara C, Kawai K, Nagase T, Tanimoto S, Borlongan CV, Date I. Continuous vagus nerve stimulation exerts beneficial effects on rats with experimentally induced Parkinson's disease: Evidence suggesting involvement of a vagal afferent pathway. Brain Stimul 2023; 16:594-603. [PMID: 36914065 DOI: 10.1016/j.brs.2023.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND Vagus nerve stimulation (VNS) exerts neuroprotective and anti-inflammatory effects in preclinical models of central nervous system disorders, including Parkinson's disease (PD). VNS setting applied for experimental models is limited into single-time or intermittent short-duration stimulation. We developed a VNS device which could deliver continuous stimulation for rats. To date, the effects of vagal afferent- or efferent-selective stimulation on PD using continuous electrical stimulation remains to be determined. OBJECTIVE To investigate the effects of continuous and selective stimulation of vagal afferent or efferent fiber on Parkinsonian rats. METHODS Rats were divided into 5 group: intact VNS, afferent VNS (left VNS in the presence of left caudal vagotomy), efferent VNS (left VNS in the presence of left rostral vagotomy), sham, vagotomy. Rats underwent the implantation of cuff-electrode on left vagus nerve and 6-hydroxydopamine administration into the left striatum simultaneously. Electrical stimulation was delivered just after 6-OHDA administration and continued for 14 days. In afferent VNS and efferent VNS group, the vagus nerve was dissected at distal or proximal portion of cuff-electrode to imitate the selective stimulation of afferent or efferent vagal fiber respectively. RESULTS Intact VNS and afferent VNS reduced the behavioral impairments in cylinder test and methamphetamine-induced rotation test, which were accompanied by reduced inflammatory glial cells in substantia nigra with the increased density of the rate limiting enzyme in locus coeruleus. In contrast, efferent VNS did not exert any therapeutic effects. CONCLUSION Continuous VNS promoted neuroprotective and anti-inflammatory effect in experimental PD, highlighting the crucial role of the afferent vagal pathway in mediating these therapeutic outcomes.
Collapse
Affiliation(s)
- Kakeru Hosomoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan.
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Masahiro Kameda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan; Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Susumu Sasada
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Ittetsu Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Ken Kuwahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Satoshi Kawauchi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Yosuke Okazaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Satoru Yabuno
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Chiaki Sugahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Koji Kawai
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Takayuki Nagase
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Shun Tanimoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Cesario V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL, 33611, USA
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| |
Collapse
|
6
|
Song S, Tu D, Meng C, Liu J, Wilson B, Wang Q, Shih YYI, Gao HM, Hong JS. Dysfunction of the noradrenergic system drives inflammation, α-synucleinopathy, and neuronal loss in mouse colon. Front Immunol 2023; 14:1083513. [PMID: 36845109 PMCID: PMC9950510 DOI: 10.3389/fimmu.2023.1083513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Clinical and pathological evidence revealed that α-synuclein (α-syn) pathology seen in PD patients starts in the gut and spreads via anatomically connected structures from the gut to the brain. Our previous study demonstrated that depletion of central norepinephrine (NE) disrupted brain immune homeostasis, producing a spatiotemporal order of neurodegeneration in the mouse brain. The purpose of this study was 1) to determine the role of peripheral noradrenergic system in the maintenance of gut immune homeostasis and in the pathogenesis of PD and 2) to investigate whether NE-depletion induced PD-like α-syn pathological changes starts from the gut. For these purposes, we investigated time-dependent changes of α-synucleinopathy and neuronal loss in the gut following a single injection of DSP-4 (a selective noradrenergic neurotoxin) to A53T-SNCA (human mutant α-syn) over-expression mice. We found DPS-4 significantly reduced the tissue level of NE and increased immune activities in gut, characterized by increased number of phagocytes and proinflammatory gene expression. Furthermore, a rapid-onset of α-syn pathology was observed in enteric neurons after 2 weeks and delayed dopaminergic neurodegeneration in the substantia nigra was detected after 3-5 months, associated with the appearance of constipation and impaired motor function, respectively. The increased α-syn pathology was only observed in large, but not in the small, intestine, which is similar to what was observed in PD patients. Mechanistic studies reveal that DSP-4-elicited upregulation of NADPH oxidase (NOX2) initially occurred only in immune cells during the acute intestinal inflammation stage, and then spread to enteric neurons and mucosal epithelial cells during the chronic inflammation stage. The upregulation of neuronal NOX2 correlated well with the extent of α-syn aggregation and subsequent enteric neuronal loss, suggesting that NOX2-generated reactive oxygen species play a key role in α-synucleinopathy. Moreover, inhibiting NOX2 by diphenyleneiodonium or restoring NE function by salmeterol (a β2-receptor agonist) significantly attenuated colon inflammation, α-syn aggregation/propagation, and enteric neurodegeneration in the colon and ameliorated subsequent behavioral deficits. Taken together, our model of PD shows a progressive pattern of pathological changes from the gut to the brain and suggests a potential role of the noradrenergic dysfunction in the pathogenesis of PD.
Collapse
Affiliation(s)
- Sheng Song
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, United States
| | - Dezhen Tu
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Institute for Brain Sciences, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of medicine, Nanjing University, Nanjing, China
| | - Chengbo Meng
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Jie Liu
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Belinda Wilson
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Yen-Yu Ian Shih
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, United States
| | - Hui-Ming Gao
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Institute for Brain Sciences, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of medicine, Nanjing University, Nanjing, China
| | - Jau-Shyong Hong
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| |
Collapse
|
7
|
Oshaghi M, Kourosh-Arami M, Roozbehkia M. Role of neurotransmitters in immune-mediated inflammatory disorders: a crosstalk between the nervous and immune systems. Neurol Sci 2023; 44:99-113. [PMID: 36169755 DOI: 10.1007/s10072-022-06413-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/14/2022] [Indexed: 02/07/2023]
Abstract
Immune-mediated inflammatory diseases (IMIDs) are a group of common heterogeneous disorders, characterized by an alteration of cellular homeostasis. Primarily, it has been shown that the release and diffusion of neurotransmitters from nervous tissue could result in signaling through lymphocyte cell-surface receptors and the modulation of immune function. This finding led to the idea that the neurotransmitters could serve as immunomodulators. It is now manifested that neurotransmitters can also be released from leukocytes and act as autocrine or paracrine modulators. Increasing data indicate that there is a crosstalk between inflammation and alterations in neurotransmission. The primary goal of this review is to demonstrate how these two pathways may converge at the level of the neuron and glia to involve in IMID. We review the role of neurotransmitters in IMID. The different effects that these compounds exert on a variety of immune cells are also reviewed. Current and future developments in understanding the cross-talk between the immune and nervous systems will undoubtedly identify new ways for treating immune-mediated diseases utilizing agonists or antagonists of neurotransmitter receptors.
Collapse
Affiliation(s)
- Mojgan Oshaghi
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Maryam Roozbehkia
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Zhang X, Tu D, Li S, Li N, Li D, Gao Y, Tian L, Liu J, Zhang X, Hong JS, Hou L, Zhao J, Wang Q. A novel synthetic peptide SVHRSP attenuates dopaminergic neurodegeneration by inhibiting NADPH oxidase-mediated neuroinflammation in experimental models of Parkinson's disease. Free Radic Biol Med 2022; 188:363-374. [PMID: 35760232 DOI: 10.1016/j.freeradbiomed.2022.06.241] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 01/21/2023]
Abstract
Current treatment of Parkinson's disease (PD) ameliorates symptoms but fails to block disease progression. This study was conducted to explore the protective effects of SVHRSP, a synthetic heat-resistant peptide derived from scorpion venom, against dopaminergic neurodegeneration in experimental models of PD. Results showed that SVHRSP dose-dependently reduced the loss of dopaminergic neuron in the nigrostriatal pathway and motor impairments in both rotenone and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTP/p)-induced mouse PD models. Microglial activation and imbalance of M1/M2 polarization were also abrogated by SVHRSP in both models. In rotenone-treated primary midbrain neuron-glial cultures, loss of dopaminergic neuron and microglial activation were mitigated by SVHRSP. Furthermore, lipopolysaccharide (LPS)-elicited microglial activation, M1 polarization and related dopaminergic neurodegeneration in primary cultures were also abrogated by SVHRSP, suggesting that inhibition of microglial activation contributed to SVHRSP-afforded neuroprotection. Mechanistic studies revealed that SVHRSP blocked both LPS- and rotenone-induced microglial NADPH oxidase (NOX2) activation by preventing membrane translocation of cytosolic subunit p47phox. NOX2 knockdown by siRNA markedly attenuated the inhibitory effects of SVHRSP against LPS- and rotenone-induced gene expressions of proinflammatory factors and related neurotoxicity. Altogether, SVHRSP protects dopaminergic neurons by blocking NOX2-mediated microglial activation in experimental PD models, providing experimental basis for the screening of clinical therapeutic drugs for PD.
Collapse
Affiliation(s)
- Xiaomeng Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Dezhen Tu
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Sheng Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Na Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Donglai Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Yun Gao
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Lu Tian
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Jianing Liu
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Xuan Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Jau-Shyong Hong
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Liyan Hou
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China.
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
9
|
Jensen RK, Pedersen H, Lorentzen J, Laursen NS, Vorup-Jensen T, Andersen GR. Structural insights into the function-modulating effects of nanobody binding to the integrin receptor α Mβ 2. J Biol Chem 2022; 298:102168. [PMID: 35738398 PMCID: PMC9287160 DOI: 10.1016/j.jbc.2022.102168] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 01/12/2023] Open
Abstract
The integrin receptor αMβ2 mediates phagocytosis of complement-opsonized objects, adhesion to the extracellular matrix, and transendothelial migration of leukocytes. However, the mechanistic aspects of αMβ2 signaling upon ligand binding are unclear. Here, we present the first atomic structure of the human αMβ2 headpiece fragment in complex with the nanobody (Nb) hCD11bNb1 at a resolution of 3.2 Å. We show that the receptor headpiece adopts the closed conformation expected to exhibit low ligand affinity. The crystal structure indicates that in the R77H αM variant, associated with systemic lupus erythematosus, the modified allosteric relationship between ligand binding and integrin outside–inside signaling is due to subtle conformational effects transmitted over a distance of 40 Å. Furthermore, we found the Nb binds to the αI domain of the αM subunit in an Mg2+-independent manner with low nanomolar affinity. Biochemical and biophysical experiments with purified proteins demonstrated that the Nb acts as a competitive inhibitor through steric hindrance exerted on the thioester domain of complement component iC3b attempting to bind the αM subunit. Surprisingly, we show that the Nb stimulates the interaction of cell-bound αMβ2 with iC3b, suggesting that it may represent a novel high-affinity proteinaceous αMβ2-specific agonist. Taken together, our data suggest that the iC3b–αMβ2 complex may be more dynamic than predicted from the crystal structure of the core complex. We propose a model based on the conformational spectrum of the receptor to reconcile these observations regarding the functional consequences of hCD11bNb1 binding to αMβ2.
Collapse
Affiliation(s)
- Rasmus K Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | - Henrik Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | - Josefine Lorentzen
- Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | | | | | | |
Collapse
|
10
|
Borges JMP, de Jesus LB, Dos Santos Souza C, da Silva VDA, Costa SL, de Fátima Dias Costa M, El-Bachá RS. Astrocyte Reaction to Catechol-Induced Cytotoxicity Relies on the Contact with Microglia Before Isolation. Neurotox Res 2022; 40:973-994. [PMID: 35708826 DOI: 10.1007/s12640-022-00528-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 10/18/2022]
Abstract
Astrocytes preserve the brain microenvironment homeostasis in order to protect other brain cells, mainly neurons, against damages. Glial cells have specific functions that are important in the context of neuronal survival in different models of central nervous system (CNS) diseases. Microglia are among these cells, secreting several molecules that can modulate astrocyte functions. Although 1,2-dihydroxybenzene (catechol) is a neurotoxic monoaromatic compound of exogenous origin, several endogenous molecules also present the catechol group. This study compared two methods to obtain astrocyte-enriched cultures from newborn Wistar rats of both sexes. In the first technique (P1), microglial cells began to be removed early 48 h after primary mixed glial cultures were plated. In the second one (P2), microglial cells were late removed 7 to 10 days after plating. Both cultures were exposed to catechol for 72 h. Catechol was more cytotoxic to P1 cultures than to P2, decreasing cellularity and changing the cell morphology. Microglial-conditioned medium (MCM) protected P1 cultures and inhibited the catechol autoxidation. P2 cultures, as well as P1 in the presence of 20% MCM, presented long, dense, and fibrillary processes positive for glial fibrillary acidic protein, which retracted the cytoplasm when exposed to catechol. The Ngf and Il1beta transcription increased in P1, meanwhile astrocytes expressed more Il10 in P2. Catechol decreased Bdnf and Il10 in P2 cultures, and it decreased the expression of Il1beta in both conditions. A prolonged contact with microglia before isolation of astrocyte-enriched cultures modifies astrocyte functions and morphology, protecting these cells against catechol-induced cytotoxicity.
Collapse
Affiliation(s)
- Julita Maria Pereira Borges
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), 40.110-902, Salvador, Bahia (BA), Brazil. .,Department of Science and Technology, Southwest Bahia State University (UESB), 45.208-409, Jequie, BA, Brazil.
| | - Lívia Bacelar de Jesus
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), 40.110-902, Salvador, Bahia (BA), Brazil
| | - Cleide Dos Santos Souza
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), 40.110-902, Salvador, Bahia (BA), Brazil
| | - Victor Diogenes Amaral da Silva
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), 40.110-902, Salvador, Bahia (BA), Brazil
| | - Silvia Lima Costa
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), 40.110-902, Salvador, Bahia (BA), Brazil
| | - Maria de Fátima Dias Costa
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), 40.110-902, Salvador, Bahia (BA), Brazil
| | - Ramon Santos El-Bachá
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia (UFBA), 40.110-902, Salvador, Bahia (BA), Brazil.
| |
Collapse
|
11
|
Revuelta M, Urrutia J, Villarroel A, Casis O. Microglia-Mediated Inflammation and Neural Stem Cell Differentiation in Alzheimer's Disease: Possible Therapeutic Role of K V1.3 Channel Blockade. Front Cell Neurosci 2022; 16:868842. [PMID: 35530176 PMCID: PMC9070300 DOI: 10.3389/fncel.2022.868842] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/31/2022] [Indexed: 12/14/2022] Open
Abstract
Increase of deposits of amyloid β peptides in the extracellular matrix is landmark during Alzheimer’s Disease (AD) due to the imbalance in the production vs. clearance. This accumulation of amyloid β deposits triggers microglial activation. Microglia plays a dual role in AD, a protective role by clearing the deposits of amyloid β peptides increasing the phagocytic response (CD163, IGF-1 or BDNF) and a cytotoxic role, releasing free radicals (ROS or NO) and proinflammatory cytokines (TNF-α, IL-1β) in response to reactive gliosis activated by the amyloid β aggregates. Microglia activation correlated with an increase KV1.3 channels expression, protein levels and current density. Several studies highlight the importance of KV1.3 in the activation of inflammatory response and inhibition of neural progenitor cell proliferation and neuronal differentiation. However, little is known about the pathways of this activation in neural stem cells differentiation and proliferation and the role in amyloid β accumulation. In recent studies using in vitro cells derived from mice models, it has been demonstrated that KV1.3 blockers inhibit microglia-mediated neurotoxicity in culture reducing the expression and production of the pro-inflammatory cytokines IL-1β and TNF-α through the NF-kB and p38MAPK pathway. Overall, we conclude that KV1.3 blockers change the course of AD development, reducing microglial cytotoxic activation and increasing neural stem cell differentiation. However, further investigations are needed to establish the specific pathway and to validate the use of this blocker as therapeutic treatment in Alzheimer patients.
Collapse
Affiliation(s)
- Miren Revuelta
- Department of Physiology, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Janire Urrutia
- Department of Physiology, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Alvaro Villarroel
- Instituto Biofisika, Consejo Superior de Investigaciones Científicas (CSIC)-University of the Basque Country/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Spain
| | - Oscar Casis
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| |
Collapse
|
12
|
Barut J, Rafa-Zabłocka K, Jurga AM, Bagińska M, Nalepa I, Parlato R, Kreiner G. Genetic lesions of the noradrenergic system trigger induction of oxidative stress and inflammation in the ventral midbrain. Neurochem Int 2022; 155:105302. [PMID: 35150790 DOI: 10.1016/j.neuint.2022.105302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/07/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor deficits caused by the loss of dopaminergic neurons in the substantia nigra (SN) and ventral tegmental area (VTA). However, clinical data revealed that not only the dopaminergic system is affected in PD. Postmortem studies showed degeneration of noradrenergic cells in the locus coeruleus (LC) to an even greater extent than that observed in the SN/VTA. Pharmacological models support the concept that modification of noradrenergic transmission can influence the PD-like phenotype induced by neurotoxins. Nevertheless, there are no existing data on animal models regarding the distant impact of noradrenergic degeneration on intact SN/VTA neurons. The aim of this study was to create a transgenic mouse model with endogenously evoked progressive degeneration restricted to noradrenergic neurons and investigate its long-term impact on the dopaminergic system. To this end, we selectively ablated the transcription initiation factor-IA (TIF-IA) in neurons expressing dopamine β-hydroxylase (DBH) by the Cre-loxP system. This mutation mimics a condition of nucleolar stress affecting neuronal survival. TIF-IADbhCre mice were characterized by selective, progressive degeneration of noradrenergic neurons, followed by phenotypic alterations associated with sympathetic system impairment. Our studies did not show any loss of tyrosine hydroxylase (TH)-positive cells in the SN/VTA of mutant mice; however, we observed increased indices of oxidative stress, enhanced markers of glial cell activation, inflammatory processes and isolated degenerating cells positive for FluoroJade C. These results were supported by gene expression profiling of VTA and SN from TIF-IADbhCre mice, revealing that 34 out of 246 significantly regulated genes in the SN/VTA were related to PD. Overall, our results shed new light on the possible negative influence of noradrenergic degeneration on dopaminergic neurons, reinforcing the neuroprotective role of noradrenaline.
Collapse
Affiliation(s)
- Justyna Barut
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland
| | - Katarzyna Rafa-Zabłocka
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland
| | - Agnieszka M Jurga
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland
| | - Monika Bagińska
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland
| | - Irena Nalepa
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland
| | - Rosanna Parlato
- Division of Neurodegenerative Disorders, Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim Heidelberg University, Mannheim, Germany; Institute of Anatomy and Cell Biology, University of Heidelberg, 69120, Heidelberg, Germany.
| | - Grzegorz Kreiner
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland.
| |
Collapse
|
13
|
Ko DWK. Transcutaneous vagus nerve stimulation (tVNS) as a potential therapeutic application for neurodegenerative disorders - A focus on dysautonomia in Parkinson's disease. Auton Neurosci 2021; 235:102858. [PMID: 34365230 DOI: 10.1016/j.autneu.2021.102858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 11/29/2022]
Abstract
The understandings of pathogenic processes in major neurodegenerative diseases has significantly advanced in recent years, with evidence showing pathological spread of intraneuronal proteinaceous inclusions as a fundamental factor. In Parkinson's disease (PD), the culprit protein has been identified as α-synuclein as the main component for mediating progressive neurodegeneration. With severe pathology evident in the autonomic nervous system prior to clinical manifestations of PD, pathogenic spread can occur from the peripheral nervous system through key nuclei, such as the anterior olfactory nucleus and dorsal motor nucleus of the glossopharyngeal and vagal nerves, gradually reaching the brainstem, midbrain and cerebral cortex. With this understanding and the proposed involvement of the vagus nerve in disease progression in PD, notably occurring prior to characterized clinical motor features, it raises intriguing questions as to whether vagal nerve pathology can be accurately detected, and importantly used as a reliable marker for determining early neurodegeneration. Along with this is the potential use of vagus nerve neuromodulation for treatment of early disease symptoms like dysautonomia, for modulating sympatho-vagal imbalances and easing severe comorbidities of the disease. In this article, we take a closer look at the pathogenic transmission processes in neurodegenerative disorders that impact the vagus nerve, and how vagus nerve neuromodulation can be potentially applied as a therapeutic approach for major neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniel W K Ko
- Neuropix Company Ltd, Core F, Cyberport 3, 100 Cyberport Road, Hong Kong Special Administrative Region.
| |
Collapse
|
14
|
Vagus Nerve Stimulation with Mild Stimulation Intensity Exerts Anti-Inflammatory and Neuroprotective Effects in Parkinson's Disease Model Rats. Biomedicines 2021; 9:biomedicines9070789. [PMID: 34356853 PMCID: PMC8301489 DOI: 10.3390/biomedicines9070789] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/17/2022] Open
Abstract
Background: The major surgical treatment for Parkinson’s disease (PD) is deep brain stimulation (DBS), but a less invasive treatment is desired. Vagus nerve stimulation (VNS) is a relatively safe treatment without cerebral invasiveness. In this study, we developed a wireless controllable electrical stimulator to examine the efficacy of VNS on PD model rats. Methods: Adult female Sprague-Dawley rats underwent placement of a cuff-type electrode and stimulator on the vagus nerve. Following which, 6-hydroxydopamine (6-OHDA) was administered into the left striatum to prepare a PD model. VNS was started immediately after 6-OHDA administration and continued for 14 days. We evaluated the therapeutic effects of VNS with behavioral and immunohistochemical outcome assays under different stimulation intensity (0.1, 0.25, 0.5 and 1 mA). Results: VNS with 0.25–0.5 mA intensity remarkably improved behavioral impairment, preserved dopamine neurons, reduced inflammatory glial cells, and increased noradrenergic neurons. On the other hand, VNS with 0.1 mA and 1 mA intensity did not display significant therapeutic efficacy. Conclusions: VNS with 0.25–0.5 mA intensity has anti-inflammatory and neuroprotective effects on PD model rats induced by 6-OHDA administration. In addition, we were able to confirm the practicality and effectiveness of the new experimental device.
Collapse
|
15
|
Jensen RK, Bajic G, Sen M, Springer TA, Vorup-Jensen T, Andersen GR. Complement Receptor 3 Forms a Compact High-Affinity Complex with iC3b. THE JOURNAL OF IMMUNOLOGY 2021; 206:3032-3042. [PMID: 34117107 DOI: 10.4049/jimmunol.2001208] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 04/13/2021] [Indexed: 11/19/2022]
Abstract
Complement receptor 3 (CR3, also known as Mac-1, integrin αMβ2, or CD11b/CD18) is expressed on a subset of myeloid and certain activated lymphoid cells. CR3 is essential for the phagocytosis of complement-opsonized particles such as pathogens and apoptotic or necrotic cells opsonized with the complement fragment iC3b and, to a lesser extent, C3dg. Although the interaction between the iC3b thioester domain and the ligand binding CR3 αM I-domain is structurally and functionally well characterized, the nature of additional CR3-iC3b interactions required for phagocytosis of complement-opsonized objects remains obscure. In this study, we analyzed the interaction between iC3b and the 150-kDa headpiece fragment of the CR3 ectodomain. Surface plasmon resonance experiments demonstrated a 30 nM affinity of the CR3 headpiece for iC3b compared with 515 nM for the iC3b thioester domain, whereas experiments monitoring binding of iC3b to CR3-expressing cells suggested an affinity of 50 nM for the CR3-iC3b interaction. Small angle x-ray scattering analysis revealed that iC3b adopts an extended but preferred conformation in solution. Upon interaction with CR3, iC3b rearranges to form a compact receptor-ligand complex. Overall, the data suggest that the iC3b-CR3 interaction is of high affinity and relies on minor contacts formed between CR3 and regions outside the iC3b thioester domain. Our results rationalize the more efficient phagocytosis elicited by iC3b than by C3dg and pave the way for the development of specific therapeutics for the treatment of inflammatory and neurodegenerative diseases that do not interfere with the recognition of noncomplement CR3 ligands.
Collapse
Affiliation(s)
- Rasmus K Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Goran Bajic
- Laboratory of Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA.,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mehmet Sen
- Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Timothy A Springer
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA; and
| | | | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark;
| |
Collapse
|
16
|
O'Neill E, Griffin ÉW, O'Sullivan R, Murray C, Ryan L, Yssel J, Harkin A, Cunningham C. Acute neuroinflammation, sickness behavior and working memory responses to acute systemic LPS challenge following noradrenergic lesion in mice. Brain Behav Immun 2021; 94:357-368. [PMID: 33307172 DOI: 10.1016/j.bbi.2020.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/16/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
Locus coeruleus (LC)-derived noradrenaline is important in cognition and decreases with age, but the impact of prior noradrenaline deficiency on vulnerability to inflammation-induced acute cognitive dysfunction is unclear. Here we assessed whether noradrenergic depletion, in female mice, impacted upon inflammation, locomotor activity and working memory directly after acute systemic immune challenge with bacterial lipopolysaccharide (LPS), a paradigm we have previously used to capture delirium-like acute cognitive deficits. Mice received 2 doses of the LC-selective noradrenergic toxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4; 50 mg/kg i.p.) and were challenged, 2 weeks later, with LPS (100 μg/kg i.p.). DSP-4 dramatically reduced noradrenaline concentrations and tyrosine hydroxylase-positive afferents in the frontal cortex and hippocampus. This did not significantly alter numbers of Pu.1-positive microglia, Iba1-positive microglial morphology or mRNA expression of microglia-associated gene transcripts (Tyrobp, Sall1, Cd68, Sra2, Clec7a) in the hippocampus or frontal cortex and produced modest reductions in Cx3cr1 and P2ry12. LPS induced blood and brain cytokine levels, cFOS activation and locomotor responses that were highly similar in DSP-4- and vehicle-treated mice, although LPS-induced plasma TNF-α was significantly reduced in those treated with DSP-4. Importantly, prior noradrenergic depletion did not predispose to LPS-induced T-maze working memory deficits. The data demonstrate that significant depletion of noradrenaline in the hippocampus and frontal cortex does not prompt acutely exaggerated neuroinflammation or leave the brain vulnerable to acute, transient working memory deficits upon low dose LPS challenge. These findings have implications for our understanding of the impact of systemic inflammation on the aging and vulnerable brain during septic encephalopathy and delirium.
Collapse
Affiliation(s)
- Eoin O'Neill
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Éadaoin W Griffin
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Ruairi O'Sullivan
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Carol Murray
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Lucy Ryan
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Justin Yssel
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew Harkin
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Colm Cunningham
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
17
|
Urrutia PJ, Bórquez DA, Núñez MT. Inflaming the Brain with Iron. Antioxidants (Basel) 2021; 10:antiox10010061. [PMID: 33419006 PMCID: PMC7825317 DOI: 10.3390/antiox10010061] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023] Open
Abstract
Iron accumulation and neuroinflammation are pathological conditions found in several neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Iron and inflammation are intertwined in a bidirectional relationship, where iron modifies the inflammatory phenotype of microglia and infiltrating macrophages, and in turn, these cells secrete diffusible mediators that reshape neuronal iron homeostasis and regulate iron entry into the brain. Secreted inflammatory mediators include cytokines and reactive oxygen/nitrogen species (ROS/RNS), notably hepcidin and nitric oxide (·NO). Hepcidin is a small cationic peptide with a central role in regulating systemic iron homeostasis. Also present in the cerebrospinal fluid (CSF), hepcidin can reduce iron export from neurons and decreases iron entry through the blood-brain barrier (BBB) by binding to the iron exporter ferroportin 1 (Fpn1). Likewise, ·NO selectively converts cytosolic aconitase (c-aconitase) into the iron regulatory protein 1 (IRP1), which regulates cellular iron homeostasis through its binding to iron response elements (IRE) located in the mRNAs of iron-related proteins. Nitric oxide-activated IRP1 can impair cellular iron homeostasis during neuroinflammation, triggering iron accumulation, especially in the mitochondria, leading to neuronal death. In this review, we will summarize findings that connect neuroinflammation and iron accumulation, which support their causal association in the neurodegenerative processes observed in AD and PD.
Collapse
Affiliation(s)
- Pamela J. Urrutia
- Department of Biology, Faculty of Sciences, Universidad de Chile, 7800024 Santiago, Chile;
| | - Daniel A. Bórquez
- Center for Biomedical Research, Faculty of Medicine, Universidad Diego Portales, 8370007 Santiago, Chile;
| | - Marco Tulio Núñez
- Department of Biology, Faculty of Sciences, Universidad de Chile, 7800024 Santiago, Chile;
- Correspondence: ; Tel.: +56-2-29787360
| |
Collapse
|
18
|
Song S, Liu J, Zhang F, Hong JS. Norepinephrine depleting toxin DSP-4 and LPS alter gut microbiota and induce neurotoxicity in α-synuclein mutant mice. Sci Rep 2020; 10:15054. [PMID: 32929122 PMCID: PMC7490385 DOI: 10.1038/s41598-020-72202-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
This study examined the genetic mutation and toxicant exposure in producing gut microbiota alteration and neurotoxicity. Homozygous α-synuclein mutant (SNCA) mice that overexpress human A53T protein and littermate wild-type mice received a single injection of LPS (2 mg/kg) or a selective norepinephrine depleting toxin DSP-4 (50 mg/kg), then the motor activity, dopaminergic neuron loss, colon gene expression and gut microbiome were examined 13 months later. LPS and DSP-4 decreased rotarod and wirehang activity, reduced dopaminergic neurons in substantia nigra pars compacta (SNpc), and SNCA mice were more vulnerable. SNCA mice had 1,000-fold higher human SNCA mRNA expression in the gut, and twofold higher gut expression of NADPH oxidase (NOX2) and translocator protein (TSPO). LPS further increased expression of TSPO and IL-6 in SNCA mice. Both LPS and DSP-4 caused microbiome alterations, and SNCA mice were more susceptible. The altered colon microbiome approximated clinical findings in PD patients, characterized by increased abundance of Verrucomicrobiaceae, and decreased abundance of Prevotellaceae, as evidenced by qPCR with 16S rRNA primers. The Firmicutes/Bacteroidetes ratio was increased by LPS in SNCA mice. This study demonstrated a critical role of α-synuclein and toxins interactions in producing gut microbiota disruption, aberrant gut pro-inflammatory gene expression, and dopaminergic neuron loss.
Collapse
Affiliation(s)
- Sheng Song
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Jie Liu
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA. .,Key Lab for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| | - Feng Zhang
- Key Lab for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Jau-Shyong Hong
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
19
|
Albertini G, Etienne F, Roumier A. Regulation of microglia by neuromodulators: Modulations in major and minor modes. Neurosci Lett 2020; 733:135000. [DOI: 10.1016/j.neulet.2020.135000] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
|
20
|
Farrand AQ, Verner RS, McGuire RM, Helke KL, Hinson VK, Boger HA. Differential effects of vagus nerve stimulation paradigms guide clinical development for Parkinson's disease. Brain Stimul 2020; 13:1323-1332. [PMID: 32629028 DOI: 10.1016/j.brs.2020.06.078] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/15/2020] [Accepted: 06/30/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Vagus nerve stimulation (VNS) modifies brain rhythms in the locus coeruleus (LC) via the solitary nucleus. Degeneration of the LC in Parkinson's disease (PD) is an early catalyst of the spreading neurodegenerative process, suggesting that stimulating LC output with VNS has the potential to modify disease progression. We previously showed in a lesion PD model that VNS delivered twice daily reduced neuroinflammation and motor deficits, and attenuated tyrosine hydroxylase (TH)-positive cell loss. OBJECTIVE The goal of this study was to characterize the differential effects of three clinically-relevant VNS paradigms in a PD lesion model. METHODS Eleven days after DSP-4 (N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine, noradrenergic lesion, administered systemically)/6-OHDA (6-hydroxydopamine, dopaminergic lesion, administered intrastriatally) rats were implanted with VNS devices, and received either low-frequency VNS, standard-frequency VNS, or high-frequency microburst VNS. After 10 days of treatment and behavioral assessment, rats were euthanized, right prefrontal cortex (PFC) was dissected for norepinephrine assessment, and the left striatum, bilateral substantia nigra (SN), and LC were sectioned for immunohistochemical detection of catecholamine neurons, α-synuclein, astrocytes, and microglia. RESULTS At higher VNS frequencies, specifically microburst VNS, greater improvements occurred in motor function, attenuation of TH-positive cell loss in SN and LC, and norepinephrine concentration in the PFC. Additionally, higher VNS frequencies resulted in lower intrasomal α-synuclein accumulation and glial density in the SN. CONCLUSIONS These data indicate that higher stimulation frequencies provided the greatest attenuation of behavioral and pathological markers in this PD model, indicating therapeutic potential for these VNS paradigms.
Collapse
Affiliation(s)
- Ariana Q Farrand
- Department of Neuroscience and Center on Aging, Medical University of South Carolina, 173 Ashley Ave, BSB Suite 403, MSC 510, Charleston, SC, 29425, USA
| | - Ryan S Verner
- Neuromodulation Division of LivaNova, PLC, 100 Cyberonics Blvd, Houston, TX, 77058, USA
| | - Ryan M McGuire
- Neuromodulation Division of LivaNova, PLC, 100 Cyberonics Blvd, Houston, TX, 77058, USA
| | - Kristi L Helke
- Department of Comparative Medicine, 114 Doughty St, STB 648, MSC 777; Department of Pathology and Laboratory Medicine, 165 Ashley Ave, Children's Hospital 309, MSC 908, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Vanessa K Hinson
- Department of Neurology, Medical University of South Carolina, 96 Jonathan Lucas St, CSB 309, MSC 606, Charleston, SC, 29425, USA
| | - Heather A Boger
- Department of Neuroscience and Center on Aging, Medical University of South Carolina, 173 Ashley Ave, BSB Suite 403, MSC 510, Charleston, SC, 29425, USA.
| |
Collapse
|
21
|
Hou L, Qu X, Qiu X, Huang R, Zhao X, Wang Q. Integrin CD11b mediates locus coeruleus noradrenergic neurodegeneration in a mouse Parkinson's disease model. J Neuroinflammation 2020; 17:148. [PMID: 32375810 PMCID: PMC7201626 DOI: 10.1186/s12974-020-01823-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 04/22/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The loss of locus coeruleus noradrenergic (LC/NE) neurons in the brainstem is reported in multiple neurodegenerative disorders, including Parkinson's disease (PD). However, the mechanisms remain unclear. Strong evidence suggested that microglia-mediated neuroinflammation contributes to neurodegeneration in PD. We recently recognized integrin CD11b, the α-chain of macrophage antigen complex-1 (Mac-1, also called CR3), as a key regulator for microglial activation. However, whether CD11b is involved in LC/NE neurodegeneration in PD remains to be investigated. METHODS LC/NE neurodegeneration and microglial activation were compared between wild type (WT) and CD11b KO mice after treated with paraquat and maneb, two pesticides that widely used to create PD model. The role of NLRP3 inflammasome in CD11b-mediated microglial dysfunction and LC/NE neurodegeneration was further explored. LC/NE neurodegeneration, microglial phenotype, and NLRP3 inflammasome activation were determined by using Western blot, immunohistochemistry, and RT-PCR technologies. RESULTS Paraquat and maneb co-exposure elevated the expressions of CD11b in the brainstem of mice, and CD11b knockout significantly reduced LC/NE neurodegeneration induced by paraquat and maneb. Mitigated microglial activation and gene expressions of proinflammatory cytokines were also observed in paraquat and maneb-treated CD11b-/- mice. Mechanistically, CD11b-mediated NLRP3 inflammasome activation contributes to paraquat and maneb-induced LC/NE neurodegeneration. Compared with WT controls, CD11b deficiency reduced paraquat and maneb-induced NLRP3 expression, caspase-1 activation, and interleukin-1β production in mice. Furthermore, inhibition of NLRP3 inflammasome by glybenclamide, a sulfonylurea inhibitor of NLRP3 inflammasome, was found to be able to suppress microglial proinflammatory activation and nuclear factor-κB activation induced by paraquat and maneb. Moreover, reduced reactive oxygen species production, NADPH oxidase, and inducible nitric oxide synthase expressions as well as 4-hydroxynonenal and malondialdehyde levels were detected in combined glybenclamide and paraquat and maneb-treated mice compared with paraquat and maneb alone group. Finally, we found that glybenclamide treatment ameliorated LC/NE neurodegeneration and α-synuclein aggregation in paraquat and maneb-treated mice. CONCLUSION Our findings suggested that CD11b mediates LC/NE neurodegeneration through NLRP3 inflammation-dependent microglial proinflammatory activation in a two pesticide-induced mouse PD model, providing a novel insight into the immune pathogenesis of LC/NE neuronal damage in related disorders.
Collapse
Affiliation(s)
- Liyan Hou
- School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Xingyue Qu
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Xiaofei Qiu
- Qingdao Municipal Center for Disease Control & Prevention/Qingdao Institute of Preventive Medicine, Qingdao, 266033, China.,School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Ruixue Huang
- School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Xiulan Zhao
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, Dalian, 116044, China. .,National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
22
|
Paredes-Rodriguez E, Vegas-Suarez S, Morera-Herreras T, De Deurwaerdere P, Miguelez C. The Noradrenergic System in Parkinson's Disease. Front Pharmacol 2020; 11:435. [PMID: 32322208 PMCID: PMC7157437 DOI: 10.3389/fphar.2020.00435] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
Nowadays it is well accepted that in Parkinson’s disease (PD), the neurodegenerative process occurs in stages and that damage to other areas precedes the neuronal loss in the substantia nigra pars compacta, which is considered a pathophysiological hallmark of PD. This heterogeneous and progressive neurodegeneration may explain the diverse symptomatology of the disease, including motor and non-motor alterations. In PD, one of the first areas undergoing degeneration is the locus coeruleus (LC). This noradrenergic nucleus provides extensive innervation throughout the brain and plays a fundamental neuromodulator role, participating in stress responses, emotional memory, and control of motor, sensory, and autonomic functions. Early in the disease, LC neurons suffer modifications that can condition the effectiveness of pharmacological treatments, and importantly, can lead to the appearance of common non-motor symptomatology. The noradrenergic system also exerts anti-inflammatory and neuroprotective effect on the dopaminergic degeneration and noradrenergic damage can consequently condition the progress of the disease. From the pharmacological point of view, it is also important to understand how the noradrenergic system performs in PD, since noradrenergic medication is often used in these patients, and drug interactions can take place when combining them with the gold standard drug therapy in PD, L-3,4-dihydroxyphenylalanine (L-DOPA). This review provides an overview about the functional status of the noradrenergic system in PD and its contribution to the efficacy of pharmacological-based treatments. Based on preclinical and clinical publications, a special attention will be dedicated to the most prevalent non-motor symptoms of the disease.
Collapse
Affiliation(s)
- Elena Paredes-Rodriguez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Sergio Vegas-Suarez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Philippe De Deurwaerdere
- Centre National de la Recherche scientifique, Institut des Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA UMR 5287), Bordeaux, France
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| |
Collapse
|
23
|
Chen C, Zhang BB, Hu AL, Li H, Liu J, Zhang F. Protective role of cinnabar and realgar in Hua-Feng-Dan against LPS plus rotenone-induced neurotoxicity and disturbance of gut microbiota in rats. JOURNAL OF ETHNOPHARMACOLOGY 2020; 247:112299. [PMID: 31606537 DOI: 10.1016/j.jep.2019.112299] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/11/2019] [Accepted: 10/09/2019] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hua-Feng-Dan (HFD) is a traditional Chinese medicine used for neurological disorders. HFD contains cinnabar (HgS) and realgar (As4S4). The ethnopharmacological basis of cinnabar and realgar in HFD is not known. AIM OF THE STUDY To address the role of cinnabar and realgar in HFD-produced neuroprotection against neurodegenerative diseases and disturbance of gut microbiota. MATERIALS AND METHODS Lipopolysaccharide (LPS) plus rotenone (ROT)-elicited rat dopaminergic (DA) neuronal damage loss was performed as a Parkinson's disease animal model. Rats were given a single injection of LPS. Four months later, rats were challenged with the threshold dose of ROT. The clinical dose of HFD was administered via feed, starting from ROT administration for 46 days. Behavioral dysfunction was detected by rotarod and Y-maze tests. DA neuron loss and microglial activation were assessed via immunohistochemical staining and western bolt analysis. The colon content was collected to extract bacterial DNA followed by real-time PCR analysis with 16S rRNA primers. RESULTS LPS plus ROT induced neurotoxicity, as evidenced by DA neuron loss in substantia nigra, impaired behavioral functions and increased microglial activation. HFD-original (containing 10% cinnabar and 10% realgar) rescued loss of DA neurons, improved behavioral dysfunction and attenuated microglial activation. Compared with HFD-original, HFD-reduced (3% cinnabar and 3% realgar) was also effective, but to be a less extent, while HFD-removed (without cinnabar and realgar) was ineffective. In analysis of gut microbiome, the increased Verrucomicrobiaceae and Lactobacteriaceae, and the decreased Enterobacteeriaceae by LPS plus ROT were ameliorated by HFD-original, and to be the less extent by HFD-reduced. CONCLUSION Cinnabar and realgar are active ingredients in HFD to exert beneficial effects in a neurodegenerative model and gut microbiota.
Collapse
Affiliation(s)
- Ce Chen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Bin-Bin Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - An-Ling Hu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Huan Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jie Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
24
|
O'Neill E, Yssel JD, McNamara C, Harkin A. Pharmacological targeting of β 2 -adrenoceptors is neuroprotective in the LPS inflammatory rat model of Parkinson's disease. Br J Pharmacol 2020; 177:282-297. [PMID: 31506926 PMCID: PMC6989960 DOI: 10.1111/bph.14862] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/20/2019] [Accepted: 08/27/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Chronic inflammation may play a role in the pathogenesis of Parkinson's disease (PD). Noradrenaline is an endogenous neurotransmitter with anti-inflammatory properties. In the present investigation, we assessed the immunomodulatory and neuroprotective efficacy of pharmacologically targeting the CNS noradrenergic system in a rat model of PD. EXPERIMENTAL APPROACH The impact of treatment with the β2 -adrenoceptor agonists clenbuterol and formoterol was assessed in the intranigral LPS rat model of PD. The immunomodulatory potential of formoterol to influence the CNS response to systemic inflammation was also assessed. KEY RESULTS LPS-induced deficits in motor function (akinesia and forelimb-use asymmetry) and nigrostriatal dopamine loss were rescued by both agents. Treatment with the noradrenaline reuptake inhibitor atomoxetine reduced striatal dopamine loss and motor deficits following intranigral LPS injection. Co-treatment with the β2 -adrenoceptor antagonist ICI 118,551 attenuated the protective effects of atomoxetine. Systemic LPS challenge exacerbated reactive microgliosis, IL-1β production, dopamine cell loss in the substantia nigra, nerve terminal degeneration in the striatum, and associated motor impairments in animals that previously received intranigral LPS. This exacerbation was attenuated by formoterol treatment. CONCLUSION AND IMPLICATIONS The results indicate that pharmacologically targeting β2 -adrenoceptors has the propensity to regulate the neuroinflammatory phenotype in vivo and may be a potential neuroprotective strategy where inflammation contributes to the progression of dopaminergic neurodegeneration. In accordance with this, clinical agents such as β2 -adrenoceptor agonists may prove useful as immunomodulatory agents in the treatment of neurodegenerative conditions associated with brain inflammation.
Collapse
Affiliation(s)
- Eoin O'Neill
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of NeuroscienceTrinity College DublinDublin 2Ireland
| | - Justin D. Yssel
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of NeuroscienceTrinity College DublinDublin 2Ireland
| | - Caoimhe McNamara
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of NeuroscienceTrinity College DublinDublin 2Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of NeuroscienceTrinity College DublinDublin 2Ireland
| |
Collapse
|
25
|
Lesion of the Locus Coeruleus Damages Learning and Memory Performance in Paraquat and Maneb-induced Mouse Parkinson’s Disease Model. Neuroscience 2019; 419:129-140. [DOI: 10.1016/j.neuroscience.2019.09.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 09/07/2019] [Accepted: 09/09/2019] [Indexed: 11/18/2022]
|
26
|
Song S, Wang Q, Jiang L, Oyarzabal E, Riddick NV, Wilson B, Moy SS, Shih YYI, Hong JS. Noradrenergic dysfunction accelerates LPS-elicited inflammation-related ascending sequential neurodegeneration and deficits in non-motor/motor functions. Brain Behav Immun 2019; 81:374-387. [PMID: 31247288 PMCID: PMC6754798 DOI: 10.1016/j.bbi.2019.06.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 06/20/2019] [Accepted: 06/22/2019] [Indexed: 02/06/2023] Open
Abstract
The loss of central norepinephrine (NE) released by neurons of the locus coeruleus (LC) occurs with aging, and is thought to be an important factor in producing the many of the nonmotor symptoms and exacerbating the degenerative process in animal models of Parkinson's disease (PD). We hypothesize that selectively depleting noradrenergic LC neurons prior to the induction of chronic neuroinflammation may not only accelerate the rate of progressive neurodegeneration throughout the brain, but may exacerbate nonmotor and motor behavioral phenotypes that recapitulate symptoms of PD. For this reason, we used a "two-hit" mouse model whereby brain NE were initially depleted by DSP-4 one week prior to exposing mice to LPS. We found that pretreatment with DSP-4 potentiated LPS-induced sequential neurodegeneration in SNpc, hippocampus, and motor cortex, but not in VTA and caudate/putamen. Mechanistic study revealed that DSP-4 enhanced LPS-induced microglial activation and subsequently elevated neuronal oxidative stress in affected brain regions in a time-dependent pattern. To further characterize the effects of DSP-4 on non-motor and motor symptoms in the LPS model, physiological and behavioral tests were performed at different time points following injection. Consistent with the enhanced neurodegeneration, DSP-4 accelerated the progressive deficits of non-motor symptoms including hyposmia, constipation, anxiety, sociability, exaggerated startle response and impaired learning. Furthermore, notable decreases of motor functions, including decreased rotarod activity, grip strength, and gait disturbance, were observed in treated mice. In summary, our studies provided not only an accelerated "two-hit" PD model that recapitulates the features of sequential neuron loss and the progression of motor/non-motor symptoms of PD, but also revealed the critical role of early LC noradrenergic neuron damage in the pathogenesis of PD-like symptoms.
Collapse
Affiliation(s)
- Sheng Song
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA,Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
| | - Qingshan Wang
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA,Department of Toxicology, School of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Lulu Jiang
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA,Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, China
| | - Esteban Oyarzabal
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA,Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
| | - Natallia V. Riddick
- Department of Psychiatry and Carolina Institute for Developmental Disabilities, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Belinda Wilson
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Sheryl S. Moy
- Department of Psychiatry and Carolina Institute for Developmental Disabilities, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Yen-Yu Ian Shih
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
| | - Jau-Shyong Hong
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA.
| |
Collapse
|
27
|
Central Noradrenergic Agonists in the Treatment of Ischemic Stroke-an Overview. Transl Stroke Res 2019; 11:165-184. [PMID: 31327133 DOI: 10.1007/s12975-019-00718-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 02/03/2023]
Abstract
Ischemic stroke is the leading cause of morbidity and mortality with a significant health burden worldwide and few treatment options. Among the short- and long-term effects of ischemic stroke is the cardiovascular sympathetic autonomic dysfunction, presented in part as the by-product of the ischemic damage to the noradrenergic centers of the brain. Unlike high levels in the plasma, the brain may face suboptimal levels of norepinephrine (NE), with adverse effects on the clinical and functional outcomes of ischemic stroke. The intravenous administration of NE and other sympathomimetic agents, in an attempt to increase cerebral perfusion pressure, often aggravates the ischemia-induced rise in blood pressure (BP) with life-threatening consequences for stroke patients, the majority of whom present with hypertension at the time of admission. Unlike the systemic administration, the central administration of NE reduces BP while exerting anti-inflammatory and neuroprotective effects. These characteristics of centrally administered NE, combined with the short latency of response, make it an ideal candidate for use in the acute phase of stroke, followed by the use of centrally acting noradrenergic agonists, such as NE reuptake inhibitors and B2-adrenergic receptor agonists for stroke rehabilitation. In addition, a number of nonpharmacological strategies, such as transcutaneous vagus nerve stimulation (tVNS) and trigeminal nerve stimulation (TNS), have the potential to enhance the central noradrenergic functional activities and improve stroke clinical outcomes. Many factors could influence the efficacy of the noradrenergic treatment in stroke patients. These factors include the type of the noradrenergic agent; the dose, frequency, and duration of administration; the timing of administration in relation to the acute event; and the site and characteristics of the ischemic lesions. Having this knowledge, combined with the better understanding of the regulation of noradrenergic receptors in different parts of the brain, would pave the path for the successful use of the centrally acting noradrenergic agents in the management of ischemic stroke.
Collapse
|
28
|
Lechtenberg KJ, Meyer ST, Doyle JB, Peterson TC, Buckwalter MS. Augmented β2-adrenergic signaling dampens the neuroinflammatory response following ischemic stroke and increases stroke size. J Neuroinflammation 2019; 16:112. [PMID: 31138227 PMCID: PMC6540436 DOI: 10.1186/s12974-019-1506-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/09/2019] [Indexed: 12/20/2022] Open
Abstract
Background Ischemic stroke provokes a neuroinflammatory response and simultaneously promotes release of epinephrine and norepinephrine by the sympathetic nervous system. This increased sympathetic outflow can act on β2-adrenergic receptors expressed by immune cells such as brain-resident microglia and monocyte-derived macrophages (MDMs), but the effect on post-stroke neuroinflammation is unknown. Thus, we investigated how changes in β2-adrenergic signaling after stroke onset influence the microglia/MDM stroke response, and the specific importance of microglia/MDM β2-adrenergic receptors to post-stroke neuroinflammation. Methods To investigate the effects of β2-adrenergic receptor manipulation on post-stroke neuroinflammation, we administered the β2-adrenergic receptor agonist clenbuterol to mice 3 h after the onset of photothrombotic stroke. We immunostained to quantify microglia/MDM numbers and proliferation and to assess morphology and activation 3 days later. We assessed stroke outcomes by measuring infarct volume and functional motor recovery and analyzed gene expression levels of neuroinflammatory molecules. Finally, we evaluated changes in cytokine expression and microglia/MDM response in brains of mice with selective knockout of the β2-adrenergic receptor from microglia and monocyte-lineage cells. Results We report that clenbuterol treatment after stroke onset causes enlarged microglia/MDMs and impairs their proliferation, resulting in reduced numbers of these cells in the peri-infarct cortex by 1.7-fold at 3 days after stroke. These changes in microglia/MDMs were associated with increased infarct volume in clenbuterol-treated animals. In mice that had the β2-adrenergic receptor specifically knocked out of microglia/MDMs, there was no change in morphology or numbers of these cells after stroke. However, knockdown of β2-adrenergic receptors in microglia and MDMs resulted in increased expression of TNFα and IL-10 in peri-infarct tissue, while stimulation of β2-adrenergic receptors with clenbuterol had the opposite effect, suppressing TNFα and IL-10 expression. Conclusions We identified β2-adrenergic receptor signaling as an important regulator of the neuroimmune response after ischemic stroke. Increased β2-adrenergic signaling after stroke onset generally suppressed the microglia/MDM response, reducing upregulation of both pro- and anti-inflammatory cytokines, and increasing stroke size. In contrast, diminished β2-adrenergic signaling in microglia/MDMs augmented both pro- and anti-inflammatory cytokine expression after stroke. The β2-adrenergic receptor may therefore present a therapeutic target for improving the post-stroke neuroinflammatory and repair process.
Collapse
Affiliation(s)
- Kendra J Lechtenberg
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA
| | - Scott T Meyer
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA
| | - Janelle B Doyle
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA
| | - Todd C Peterson
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA. .,Department of Neurosurgery, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA.
| |
Collapse
|
29
|
TIA1 regulates the generation and response to toxic tau oligomers. Acta Neuropathol 2019; 137:259-277. [PMID: 30465259 PMCID: PMC6377165 DOI: 10.1007/s00401-018-1937-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/31/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022]
Abstract
RNA binding proteins (RBPs) are strongly linked to the pathophysiology of motor neuron diseases. Recent studies show that RBPs, such as TIA1, also contribute to the pathophysiology of tauopathy. RBPs co-localize with tau pathology, and reduction of TIA1 protects against tau-mediated neurodegeneration. The mechanism through which TIA1 reduction protects against tauopathy, and whether TIA1 modulates the propagation of tau, are unknown. Previous studies indicate that the protective effect of TIA1 depletion correlates with both the reduction of oligomeric tau and the reduction of pathological TIA1 positive tau inclusions. In the current report, we used a novel tau propagation approach to test whether TIA1 is required for producing toxic tau oligomers and whether TIA1 reduction would provide protection against the spread of these oligomers. The approach used young PS19 P301S tau mice at an age at which neurodegeneration would normally not yet occur and seeding oligomeric or fibrillar tau by injection into hippocampal CA1 region. We find that propagation of exogenous tau oligomers (but not fibrils) across the brain drives neurodegeneration in this model. We demonstrate that TIA1 reduction essentially brackets the pathophysiology of tau, being required for the production of tau oligomers, as well as regulating the response of neurons to propagated toxic tau oligomers. These results indicate that RNA binding proteins modulate the pathophysiology of tau at multiple levels and provide insights into possible therapeutic approaches to reduce the spread of neurodegeneration in tauopathy.
Collapse
|
30
|
Low-Grade Inflammation Aggravates Rotenone Neurotoxicity and Disrupts Circadian Clock Gene Expression in Rats. Neurotox Res 2018; 35:421-431. [PMID: 30328585 DOI: 10.1007/s12640-018-9968-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/08/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023]
Abstract
A single injection of LPS produced low-grade neuroinflammation leading to Parkinson's disease (PD) in mice several months later. Whether such a phenomenon occurs in rats and whether such low-grade neuroinflammation would aggravate rotenone (ROT) neurotoxicity and disrupts circadian clock gene/protein expressions were examined in this study. Male rats were given two injections of LPS (2.5-7.5 mg/kg), and neuroinflammation and dopamine neuron loss were evident 3 months later. Seven months after a single LPS (5 mg/kg) injection, rats received low doses of ROT (0.5 mg/kg, sc, 5 times/week for 4 weeks) to examine low-grade neuroinflammation on ROT toxicity. LPS plus ROT produced more pronounced non-motor and motor dysfunctions than LPS or ROT alone in behavioral tests, and decreased mitochondrial complex 1 activity, together with aggravated neuroinflammation and neuron loss. The expressions of clock core genes brain and muscle Arnt-like protein-1 (Bmal1), locomotor output cycles kaput (Clock), and neuronal PAS domain protein-2 (Npas2) were decreased in LPS, ROT, and LPS plus ROT groups. The expressions of circadian feedback genes Periods (Per1 and Per2) were also decreased, but Cryptochromes (Cry1 and Cry2) were unaltered. The circadian clock target genes nuclear receptor Rev-Erbα (Nr1d1), and D-box-binding protein (Dbp) expressions were also decreased. Consistent with the transcript levels, circadian clock protein BMAL1, CLOCK, NR1D1, and DBP were also decreased. Thus, LPS-induced chronic low-grade neuroinflammation potentiated ROT neurotoxicity and disrupted circadian clock gene/protein expression, suggesting a role of disrupted circadian in PD development and progression. Graphical Abstract ᅟ.
Collapse
|
31
|
Sharma M, Flood PM. β-arrestin2 regulates the anti-inflammatory effects of Salmeterol in lipopolysaccharide-stimulated BV2 cells. J Neuroimmunol 2018; 325:10-19. [PMID: 30352316 DOI: 10.1016/j.jneuroim.2018.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/11/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022]
Abstract
Microglial activation contributes to chronic inflammation and neuronal loss in progressive neurodegenerative disorders such as Parkinson's disease (PD). Thus, treatments suppressing microglial activation may have therapeutic benefits to prevent neuronal loss in neurodegenerative diseases. Our previous findings show that Salmeterol, a long-acting β2-adrenergic receptor (β2-AR) agonist, is neuroprotective in two distinct animal models of PD, including where lipopolysaccharide (LPS) from E. coli was used to initiate chronic neurodegeneration. Salmeterol was found to be a potent inhibitor of dopaminergic neurodegeneration by regulating the production of pro-inflammatory mediators from activated microglial cells. In the present study, we investigated the molecular basis of the anti-inflammatory effects of Salmeterol on LPS-activated murine microglial BV2 cells. BV2 cells were pretreated with Salmeterol and followed by stimulation with LPS. Salmeterol inhibited LPS-induced release of the pro-inflammatory mediators such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and nitric oxide from BV2 cells. Additionally, Salmeterol suppressed nuclear translocation of nuclear factor kappa-B (NF-κB) p65 by inhibiting the IκB-α degradation and TAK1 (transforming growth factor-beta-activated kinase1) phosphorylation. We have also found that Salmeterol increases the expression of β-arrestin2 and enhances the interaction between β-arrestin2 and TAB1 (TAK1-binding protein), reduced TAK1/TAB1 mediated activation of NFκB and expression of pro-inflammatory genes. Furthermore, silencing of β-arrestin2 abrogates the anti-inflammatory effects of Salmeterol in LPS-stimulated BV2 cells. Our findings suggest that the anti-inflammatory properties of Salmeterol is β-arrestin2 dependent and also offers novel therapeutics targeting inflammatory pathways to prevent microglial cell activation and neuronal loss in neuroinflammatory diseases like PD.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| | - Patrick M Flood
- Departments of Dentistry and Medical Microbiology and Immunology, and Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
32
|
Song S, Jiang L, Oyarzabal EA, Wilson B, Li Z, Shih YYI, Wang Q, Hong JS. Loss of Brain Norepinephrine Elicits Neuroinflammation-Mediated Oxidative Injury and Selective Caudo-Rostral Neurodegeneration. Mol Neurobiol 2018; 56:2653-2669. [PMID: 30051353 DOI: 10.1007/s12035-018-1235-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
Environmental toxicant exposure has been strongly implicated in the pathogenesis of Parkinson's disease (PD). Clinical manifestations of non-motor and motor symptoms in PD stem from decades of progressive neurodegeneration selectively afflicting discrete neuronal populations along a caudo-rostral axis. However, recapitulating this spatiotemporal neurodegenerative pattern in rodents has been unsuccessful. The purpose of this study was to generate such animal PD models and delineate mechanism underlying the ascending neurodegeneration. Neuroinflammation, oxidative stress, and neuronal death in mice brains were measured at different times following a single systemic injection of lipopolysaccharide (LPS). We demonstrate that LPS produced an ascending neurodegeneration that temporally afflicted neurons initially in the locus coeruleus (LC), followed by substantia nigra, and lastly the primary motor cortex and hippocampus. To test the hypothesis that LPS-elicited early loss of noradrenergic LC neurons may underlie this ascending pattern, we used a neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) to deplete brain norepinephrine. DSP-4 injection resulted in a time-dependent ascending degenerative pattern similar to that generated by the LPS model. Mechanistic studies revealed that increase in nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-2 (NOX2)-dependent superoxide/reactive oxygen species (ROS) production plays a key role in both LPS- and DSP-4-elicited neurotoxicity. We found that toxin-elicited chronic neuroinflammation, oxidative neuronal injuries, and neurodegeneration were greatly suppressed in mice deficient in NOX2 gene or treated with NOX2-specific inhibitor. Our studies document the first rodent PD model recapturing the ascending neurodegenerative pattern of PD patients and provide convincing evidence that the loss of brain norepinephrine is critical in initiating and maintaining chronic neuroinflammation and the discrete neurodegeneration in PD.
Collapse
Affiliation(s)
- Sheng Song
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Mail Drop F1-01Research Triangle Park, North Carolina, 27709, USA
| | - Lulu Jiang
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Mail Drop F1-01Research Triangle Park, North Carolina, 27709, USA.,Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, China
| | - Esteban A Oyarzabal
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Mail Drop F1-01Research Triangle Park, North Carolina, 27709, USA.,Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Belinda Wilson
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Mail Drop F1-01Research Triangle Park, North Carolina, 27709, USA
| | - Zibo Li
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yen-Yu Ian Shih
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Qingshan Wang
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Mail Drop F1-01Research Triangle Park, North Carolina, 27709, USA. .,Department of Toxicology, School of Public Health, Dalian Medical University, Dalian, Liaoning, China.
| | - Jau-Shyong Hong
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Mail Drop F1-01Research Triangle Park, North Carolina, 27709, USA.
| |
Collapse
|
33
|
Abstract
SIGNIFICANCE Oxidative stress increases in the brain with aging and neurodegenerative diseases. Previous work emphasized irreversible oxidative damage in relation to cognitive impairment. This research has evolved to consider a continuum of alterations, from redox signaling to oxidative damage, which provides a basis for understanding the onset and progression of cognitive impairment. This review provides an update on research linking redox signaling to altered function of neural circuits involved in information processing and memory. Recent Advances: Starting in middle age, redox signaling triggers changes in nervous system physiology described as senescent physiology. Hippocampal senescent physiology involves decreased cell excitability, altered synaptic plasticity, and decreased synaptic transmission. Recent studies indicate N-methyl-d-aspartate and ryanodine receptors and Ca2+ signaling molecules as molecular substrates of redox-mediated senescent physiology. CRITICAL ISSUES We review redox homeostasis mechanisms and consider the chemical character of reactive oxygen and nitrogen species and their role in regulating different transmitter systems. In this regard, senescent physiology may represent the co-opting of pathways normally responsible for feedback regulation of synaptic transmission. Furthermore, differences across transmitter systems may underlie differential vulnerability of brain regions and neuronal circuits to aging and disease. FUTURE DIRECTIONS It will be important to identify the intrinsic mechanisms for the shift in oxidative/reductive processes. Intrinsic mechanism will depend on the transmitter system, oxidative stressors, and expression/activity of antioxidant enzymes. In addition, it will be important to identify how intrinsic processes interact with other aging factors, including changes in inflammatory or hormonal signals. Antioxid. Redox Signal. 28, 1724-1745.
Collapse
Affiliation(s)
- Ashok Kumar
- 1 Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Brittney Yegla
- 1 Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Thomas C Foster
- 1 Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida.,2 Genetics and Genomics Program, Genetics Institute, University of Florida , Gainesville, Florida
| |
Collapse
|
34
|
Tong H, Zhang X, Meng X, Lu L, Mai D, Qu S. Simvastatin Inhibits Activation of NADPH Oxidase/p38 MAPK Pathway and Enhances Expression of Antioxidant Protein in Parkinson Disease Models. Front Mol Neurosci 2018; 11:165. [PMID: 29872377 PMCID: PMC5972184 DOI: 10.3389/fnmol.2018.00165] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/01/2018] [Indexed: 11/13/2022] Open
Abstract
Evidence suggests that oxidative stress is involved in the pathogenesis of Parkinson disease (PD). Simvastatin has been suggested to protect against oxidative stress in several diseases. However, the molecular mechanisms by which simvastatin protects against neuropathology and oxidative damage in PD are poorly elucidated. In this study, we aimed to investigate the potential neuroprotective effects of simvastatin owing to its anti-oxidative properties in 6-hydroxydopamine (6-OHDA)-treated SH-SY5Y cells and mice. The results of 2′,7′-dichlorodihydrofluorescein diacetate (DCFH-DA) fluorescence and CCK-8 assay demonstrated that simvastatin reduced intracellular reactive oxygen species (ROS) levels and reversed apoptosis in 6-OHDA-treated SH-SY5Y cells. Mechanistic studies revealed that 6-OHDA-induced activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase/p38 mitogen-activated protein kinase (MAPK) pathway was inhibited and nuclear factor-κB (NF-κB) nuclear transcription decreased in SH-SY5Y cells after simvastatin treatment. Enhanced expression levels of superoxide dismutase (SOD), heme oxygenase-1 (HO-1), peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) and glutamate-cysteine ligase modifier subunit (GCLM) were observed after simvastatin treatment in 6-OHDA-treated SH-SY5Y cells. In vivo studies revealed that administration of simvastatin by gavage decreased limb-use asymmetry and apomorphine-induced rotations in 6-OHDA-lesioned mice. Simvastatin increased dopaminergic neurons and reduced protein tyrosine nitration and gliosis in the midbrain of PD mice. An inhibitory effect on activation of the NADPH oxidase/p38 MAPK was observed, and increased antioxidant protein expression in the midbrain were seen in the simvastatin plus 6-OHDA group compared with the 6-OHDA-lesioned group. Taken together, these results demonstrate that simvastatin might inhibit the activation of NADPH oxidase/p38 MAPK pathway, enhance antioxidant protein expression and protect against oxidative stress, thereby providing a novel antioxidant mechanism that has therapeutic validity.
Collapse
Affiliation(s)
- Huichun Tong
- Clinical Medicine Research Center, Shunde Hospital, Southern Medical University, Foshan, China
| | - Xiuping Zhang
- Teaching Center of Experimental Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xingjun Meng
- Clinical Medicine Research Center, Shunde Hospital, Southern Medical University, Foshan, China
| | - Lingli Lu
- Clinical Medicine Research Center, Shunde Hospital, Southern Medical University, Foshan, China
| | - Dongmei Mai
- Clinical Medicine Research Center, Shunde Hospital, Southern Medical University, Foshan, China
| | - Shaogang Qu
- Clinical Medicine Research Center, Shunde Hospital, Southern Medical University, Foshan, China
| |
Collapse
|
35
|
Cao L, Blekemolen MC, Tintor N, Cornelissen BJC, Takken FLW. The Fusarium oxysporum Avr2-Six5 Effector Pair Alters Plasmodesmatal Exclusion Selectivity to Facilitate Cell-to-Cell Movement of Avr2. MOLECULAR PLANT 2018; 11:691-705. [PMID: 29481865 DOI: 10.1016/j.molp.2018.02.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 05/03/2023]
Abstract
Pathogens use effector proteins to manipulate their hosts. During infection of tomato, the fungus Fusarium oxysporum secretes the effectors Avr2 and Six5. Whereas Avr2 suffices to trigger I-2-mediated cell death in heterologous systems, both effectors are required for I-2-mediated disease resistance in tomato. How Six5 participates in triggering resistance is unknown. Using bimolecular fluorescence complementation assays we found that Avr2 and Six5 interact at plasmodesmata. Single-cell transformation revealed that a 2xRFP marker protein and Avr2-GFP only move to neighboring cells in the presence of Six5. Six5 alone does not alter plasmodesmatal transduction as 2xRFP was only translocated in the presence of both effectors. In SIX5-expressing transgenic plants, the distribution of virally expressed Avr2-GFP, and subsequent onset of I-2-mediated cell death, differed from that in wild-type tomato. Taken together, our data show that in the presence of Six5, Avr2 moves from cell to cell, which in susceptible plants contributes to virulence, but in I-2 containing plants induces resistance.
Collapse
Affiliation(s)
- Lingxue Cao
- Molecular Plant Pathology, Swammerdam Institute for Life Science, University of Amsterdam, 1098 XH Amsterdam, the Netherlands
| | - Mila C Blekemolen
- Molecular Plant Pathology, Swammerdam Institute for Life Science, University of Amsterdam, 1098 XH Amsterdam, the Netherlands
| | - Nico Tintor
- Molecular Plant Pathology, Swammerdam Institute for Life Science, University of Amsterdam, 1098 XH Amsterdam, the Netherlands
| | - Ben J C Cornelissen
- Molecular Plant Pathology, Swammerdam Institute for Life Science, University of Amsterdam, 1098 XH Amsterdam, the Netherlands
| | - Frank L W Takken
- Molecular Plant Pathology, Swammerdam Institute for Life Science, University of Amsterdam, 1098 XH Amsterdam, the Netherlands.
| |
Collapse
|
36
|
Hou L, Che Y, Sun F, Wang Q. Taurine protects noradrenergic locus coeruleus neurons in a mouse Parkinson's disease model by inhibiting microglial M1 polarization. Amino Acids 2018; 50:547-556. [PMID: 29508060 DOI: 10.1007/s00726-018-2547-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 02/23/2018] [Indexed: 10/17/2022]
Abstract
Beyond nigrostriatal dopaminergic system, the noradrenergic locus coeruleus (LC/NE) neurons are also degenerated in patients with Parkinson's disease (PD), the second most common neurodegenerative disorder. We previously reported that microglia-mediated neuroinflammation contributes to LC/NE neurodegeneration. The purpose of this study is aimed to test whether taurine, an endogenous amino acid, could be able to protect LC/NE neurons through inhibition of microglial activation using paraquat and maneb-induced mouse PD model. Taurine (150 mg/kg) was administrated (i.p) to mice 30 min prior to paraquat (10 mg/kg) and maneb (30 mg/kg) intoxication for consecutive 6 weeks (twice per week). The results clearly demonstrated that paraquat and maneb co-exposure resulted in loss of tyrosine hydroxylase-positive neurons in the LC in mice, which was significantly ameliorated by taurine. Mechanistically, inhibition of microglia-mediated neuroinflammation contributed to taurine-afforded neuroprotection. Taurine attenuated paraquat and maneb-induced microglial activation and M1 polarization as well as release of proinflammatory cytokines in brainstem of mice. Taurine also abrogated microglial NADPH oxidase activation and oxidative damage in paraquat and maneb-treated mice. Furthermore, inhibition of nuclear factor-κB (NF-κB) but not signal transducers and activators of transcription 1/3 (STAT1/3) signaling pathway participated in taurine-inhibited microglial activation. Collectively, taurine exerted LC/NE neuroprotection against microglia-mediated neurotoxicity. The robust neuroprotective effects of taurine suggest that taurine may be a promising candidate for potential therapy for patients suffering from PD.
Collapse
Affiliation(s)
- Liyan Hou
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Yuning Che
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Fuqiang Sun
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China.
| |
Collapse
|
37
|
Hou L, Wang K, Zhang C, Sun F, Che Y, Zhao X, Zhang D, Li H, Wang Q. Complement receptor 3 mediates NADPH oxidase activation and dopaminergic neurodegeneration through a Src-Erk-dependent pathway. Redox Biol 2018; 14:250-260. [PMID: 28978491 PMCID: PMC5975223 DOI: 10.1016/j.redox.2017.09.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/17/2017] [Accepted: 09/24/2017] [Indexed: 01/08/2023] Open
Abstract
Microglial NADPH oxidase (Nox2) plays a key role in chronic neuroinflammation and related dopaminergic neurodegeneration in Parkinson's disease (PD). However, the mechanisms behind Nox2 activation remain unclear. Here, we revealed the critical role of complement receptor 3 (CR3), a microglia-specific pattern recognition receptor, in Nox2 activation and subsequent dopaminergic neurodegeneration by using paraquat and maneb-induced PD model. Suppression or genetic deletion of CR3 impeded paraquat and maneb-induced activation of microglial Nox2, which was associated with attenuation of dopaminergic neurodegeneration. Mechanistic inquiry revealed that blocking CR3 reduced paraquat and maneb-induced membrane translocation of Nox2 cytosolic subunit p47phox, an essential step for Nox2 activation. Src and Erk (extracellular regulated protein kinases) were subsequently recognized as the downstream signals of CR3. Moreover, inhibition of Src or Erk impaired Nox2 activation in response to paraquat and maneb co-exposure. Finally, we found that CR3-deficient mice were more resistant to paraquat and maneb-induced Nox2 activation and nigral dopaminergic neurodegeneration as well as motor dysfunction than the wild type controls. Taken together, our results showed that CR3 regulated Nox2 activation and dopaminergic neurodegeneration through a Src-Erk-dependent pathway in a two pesticide-induced PD model, providing novel insights into the immune pathogenesis of PD.
Collapse
Affiliation(s)
- Liyan Hou
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Ke Wang
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Cong Zhang
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Fuqiang Sun
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Yuning Che
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Xiulan Zhao
- Institute of Toxicology, School of Public Health, Shandong University, 44 West Wenhua Road, Jinan, Shandong, China
| | - Dan Zhang
- State Key Laboratory of Natural Products and Functions, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Huihua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China.
| |
Collapse
|
38
|
Zhang C, Hou L, Yang J, Che Y, Sun F, Li H, Wang Q. 2,5-Hexanedione induces dopaminergic neurodegeneration through integrin α Mβ2/NADPH oxidase axis-mediated microglial activation. Cell Death Dis 2018; 9:60. [PMID: 29352205 PMCID: PMC5833449 DOI: 10.1038/s41419-017-0091-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/08/2017] [Accepted: 10/23/2017] [Indexed: 02/06/2023]
Abstract
Recent study demonstrated that chronic exposure to solvents increases the risk of Parkinson's disease (PD), the second most common neurodegenerative disorder characterized by progressive dopaminergic neurodegeneration in the substantia nigra (SN). n-Hexane, a widely used organic solvent, displays central-peripheral neurotoxicity, which is mainly mediated by its active metabolite, 2,5-hexanedione (HD). However, whether HD exposure contributes to PD remains unclear. In this study, we found that rats exposed to HD displayed progressive dopaminergic neurodegeneration in the nigrostriatal system. Microglial activation was also detected in HD-treated rats, which occurred prior to degeneration of dopaminergic neurons. Moreover, depletion of microglia markedly reduced HD-induced dopaminergic neurotoxicity. Mechanistic study revealed an essential role of microglial integrin αMβ2-NADPH oxidase (NOX2) axis in HD-elicited neurotoxicity. HD activated NOX2 by inducing membrane translocation of NOX2 cytosolic subunit, p47phox. Integrin αMβ2 was critical for HD-induced NOX2 activation since inhibition or genetic deletion of αMβ2 attenuated NOX2-generated superoxide and p47phox membrane translocation in response to HD. Src and Erk, two downstream signals of αMβ2, were recognized to bridge HD/αMβ2-mediated NOX2 activation. Finally, pharmacological inhibition of αMβ2-NOX2 axis attenuated HD-induced microglial activation and dopaminergic neurodegeneration. Our findings revealed that HD exposure damaged nigrostriatal dopaminergic system through αMβ2-NOX2 axis-mediated microglial activation, providing, for the first time, experimental evidence for n-hexane exposure contributing to the etiology of PD.
Collapse
Affiliation(s)
- Cong Zhang
- 0000 0000 9558 1426grid.411971.bSchool of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian, 116044 China
| | - Liyan Hou
- 0000 0000 9558 1426grid.411971.bSchool of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian, 116044 China
| | - Jie Yang
- 0000 0000 9558 1426grid.411971.bSchool of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian, 116044 China
| | - Yuning Che
- 0000 0000 9558 1426grid.411971.bSchool of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian, 116044 China
| | - Fuqiang Sun
- 0000 0000 9558 1426grid.411971.bSchool of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian, 116044 China
| | - Huihua Li
- 0000 0000 9558 1426grid.411971.bSchool of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian, 116044 China ,grid.452435.1Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingshan Wang
- 0000 0000 9558 1426grid.411971.bSchool of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian, 116044 China
| |
Collapse
|
39
|
Griffin ÉW, Yssel JD, O’Neill E, Ryan KJ, Boyle N, Harper P, Harkin A, Connor T. The β2-adrenoceptor agonist clenbuterol reduces the neuroinflammatory response, neutrophil infiltration and apoptosis following intra-striatal IL-1β administration to rats. Immunopharmacol Immunotoxicol 2018; 40:99-106. [DOI: 10.1080/08923973.2017.1418882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Éadaoin W. Griffin
- Neuroimmunology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland
| | - Justin D. Yssel
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Eoin O’Neill
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| | - Katie J. Ryan
- Neuroimmunology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland
| | - Noreen Boyle
- Neuroimmunology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland
| | - Peter Harper
- Neuroimmunology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| | - Thomas Connor
- Neuroimmunology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland
| |
Collapse
|
40
|
O'Neill E, Harkin A. Targeting the noradrenergic system for anti-inflammatory and neuroprotective effects: implications for Parkinson's disease. Neural Regen Res 2018; 13:1332-1337. [PMID: 30106035 PMCID: PMC6108217 DOI: 10.4103/1673-5374.235219] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Degeneration of the locus coeruleus noradrenergic system is thought to play a key role in the pathogenesis of Parkinson's disease (PD), whereas pharmacological approaches to increase noradrenaline bioavailability may provide neuroprotection. Noradrenaline inhibits microglial activation and suppresses pro-inflammatory mediator production (e.g., tumor necrosis factor-α, interleukin-1β & inducible nitric oxide synthase activity), thus limiting the cytotoxicity of midbrain dopaminergic neurons in response to an inflammatory stimulus. Neighbouring astrocyte populations promote a neurotrophic environment in response to β2-adrenoceptor (β2-AR) stimulation via the production of growth factors (e.g., brain derived neurotrophic factor, cerebral dopamine neurotrophic factor & glial cell derived neurotrophic factor which have shown promising neuroprotective and neuro-restorative effects in the nigrostriatal dopaminergic system. More recent findings have demonstrated a role for the β2-AR in down-regulating expression levels of the human α-synuclein gene SNCA and relative α-synuclein protein abundance. Given that α-synuclein is a major protein constituent of Lewy body pathology, a hallmark neuropathological feature in Parkinson's disease, these findings could open up new avenues for pharmacological intervention strategies aimed at alleviating the burden of α-synucleinopathies in the Parkinsonian brain. In essence, the literature reviewed herein supports our hypothesis of a tripartite neuroprotective role for noradrenaline in combating PD-related neuropathology and motor dysfunction via (1) inhibiting nigral microglial activation & pro-inflammatory mediator production, (2) promoting the synthesis of neurotrophic factors from midbrain astrocytes and (3) downregulating α-synuclein gene expression and protein abundance in a β2-AR-dependent manner. Thus, taken together, either pharmacologically enhancing extra-synaptic noradrenaline bioavailability or targeting glial β2-ARs directly makes itself as a promising treatment option aimed at slowing/halting PD progression.
Collapse
Affiliation(s)
- Eoin O'Neill
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| |
Collapse
|
41
|
|
42
|
Wang S, Irving G, Jiang L, Wang H, Li M, Wang X, Han W, Xu Y, Yang Y, Zeng T, Song F, Zhao X, Xie K. Oxidative Stress Mediated Hippocampal Neuron Apoptosis Participated in Carbon Disulfide-Induced Rats Cognitive Dysfunction. Neurochem Res 2016; 42:583-594. [PMID: 27900598 DOI: 10.1007/s11064-016-2113-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/06/2016] [Accepted: 11/15/2016] [Indexed: 10/20/2022]
Abstract
Occupational exposure to carbon disulfide (CS2) exhibits central nervous systems toxicity. But the mechanism is unclear. The present study was designed to investigate the relationship between the CNS damage and cognitive dysfunction caused by CS2, and eventually reveal the possible oxidative-related mechanism of hippocampus pathological changes in CS2 exposed rats. Male Wistar rats were administrated with CS2 at dosage of 200, 400 and 600 mg/kg for consecutive 20 days, respectively. Cognitive performances were evaluated by Morris water maze tests. Thionin and immunohistochemical analysis were used to investigate the hippocampal neuron damage, and the expression of apoptosis related proteins (cleaved-caspase 3, Bax and Bcl-2) were detected to explore the possible mechanisms of neuronal loss. Oxidative stress parameters were checked by commercial assay kits. Rats exposed to CS2 displayed cognitive dysfunction manifested as decreased spatial learning ability and memory lesion. Pathological changes and significant neuron loss were observed in hippocampus, especially in CA1 and CA3 sub-regions. Mitochondria-dependent apoptosis pathway was implicated in the CS2-induced neuronal loss which was demonstrated by the up-regulation of cleaved-caspase 3 and Bax accompanied with down-regulation of Bcl-2. Furthermore, extensive oxidative stress induced by CS2 was also revealed by the measurement of ROS, RNS, MDA, GSH&GSSG and antioxidant enzymes (CAT, T-SOD, and GSH-Px). Our study suggested that oxidative stress mediated hippocampal neuron apoptosis might play an important role in CS2 induced CNS damage and cognitive dysfunction.
Collapse
Affiliation(s)
- Shuo Wang
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Gleniece Irving
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Lulu Jiang
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Hui Wang
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Ming Li
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Xujing Wang
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Wenting Han
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Yongpeng Xu
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Yilin Yang
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Fuyong Song
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Xiulan Zhao
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Keqin Xie
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
43
|
Schneble N, Schmidt C, Bauer R, Müller JP, Monajembashi S, Wetzker R. Phosphoinositide 3-kinase γ ties chemoattractant- and adrenergic control of microglial motility. Mol Cell Neurosci 2016; 78:1-8. [PMID: 27825984 DOI: 10.1016/j.mcn.2016.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 09/15/2016] [Accepted: 11/03/2016] [Indexed: 12/18/2022] Open
Abstract
Microglial motility is tightly controlled by multitude of agonistic and antagonistic factors. Chemoattractants, released after infection or damage of the brain, provoke directed migration of microglia to the pathogenic incident. In contrast, noradrenaline and other stress hormones have been shown to suppress microglial movement. Here we asked for the signaling reactions involved in the positive and negative control of microglial motility. Using pharmacological and genetic approaches we identified the lipid kinase activity of phosphoinositide 3-kinase species γ (PI3Kγ) as an essential mediator of microglial migration provoked by the complement component C5a and other chemoattractants. Inhibition of PI3Kγ lipid kinase activity by protein kinase A was disclosed as mechanism causing suppression of microglial migration by noradrenaline. Together these data characterize PI3Kγ as a nodal point in the control of microglial motility.
Collapse
Affiliation(s)
- Nadine Schneble
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital of Jena, Hans -Knöll -Straße 2, 07745 Jena, Germany.
| | - Caroline Schmidt
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital of Jena, Hans -Knöll -Straße 2, 07745 Jena, Germany.
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital of Jena, Hans -Knöll -Straße 2, 07745 Jena, Germany.
| | - Jörg P Müller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital of Jena, Hans -Knöll -Straße 2, 07745 Jena, Germany.
| | - Shamci Monajembashi
- Leibnitz Institute for Age Research, Fritz Lipmann Institute (FLI), Beutenberg-Straße 11, 07745 Jena, Germany.
| | - Reinhard Wetzker
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital of Jena, Hans -Knöll -Straße 2, 07745 Jena, Germany.
| |
Collapse
|
44
|
Atzori M, Cuevas-Olguin R, Esquivel-Rendon E, Garcia-Oscos F, Salgado-Delgado RC, Saderi N, Miranda-Morales M, Treviño M, Pineda JC, Salgado H. Locus Ceruleus Norepinephrine Release: A Central Regulator of CNS Spatio-Temporal Activation? Front Synaptic Neurosci 2016; 8:25. [PMID: 27616990 PMCID: PMC4999448 DOI: 10.3389/fnsyn.2016.00025] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 08/05/2016] [Indexed: 12/22/2022] Open
Abstract
Norepinephrine (NE) is synthesized in the Locus Coeruleus (LC) of the brainstem, from where it is released by axonal varicosities throughout the brain via volume transmission. A wealth of data from clinics and from animal models indicates that this catecholamine coordinates the activity of the central nervous system (CNS) and of the whole organism by modulating cell function in a vast number of brain areas in a coordinated manner. The ubiquity of NE receptors, the daunting number of cerebral areas regulated by the catecholamine, as well as the variety of cellular effects and of their timescales have contributed so far to defeat the attempts to integrate central adrenergic function into a unitary and coherent framework. Since three main families of NE receptors are represented-in order of decreasing affinity for the catecholamine-by: α2 adrenoceptors (α2Rs, high affinity), α1 adrenoceptors (α1Rs, intermediate affinity), and β adrenoceptors (βRs, low affinity), on a pharmacological basis, and on the ground of recent studies on cellular and systemic central noradrenergic effects, we propose that an increase in LC tonic activity promotes the emergence of four global states covering the whole spectrum of brain activation: (1) sleep: virtual absence of NE, (2) quiet wake: activation of α2Rs, (3) active wake/physiological stress: activation of α2- and α1-Rs, (4) distress: activation of α2-, α1-, and β-Rs. We postulate that excess intensity and/or duration of states (3) and (4) may lead to maladaptive plasticity, causing-in turn-a variety of neuropsychiatric illnesses including depression, schizophrenic psychoses, anxiety disorders, and attention deficit. The interplay between tonic and phasic LC activity identified in the LC in relationship with behavioral response is of critical importance in defining the short- and long-term biological mechanisms associated with the basic states postulated for the CNS. While the model has the potential to explain a large number of experimental and clinical findings, a major challenge will be to adapt this hypothesis to integrate the role of other neurotransmitters released during stress in a centralized fashion, like serotonin, acetylcholine, and histamine, as well as those released in a non-centralized fashion, like purines and cytokines.
Collapse
Affiliation(s)
- Marco Atzori
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis PotosíSan Luis Potosí, Mexico; School for Behavior and Brain Sciences, University of Texas at DallasRichardson, TX, USA
| | - Roberto Cuevas-Olguin
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí San Luis Potosí, Mexico
| | - Eric Esquivel-Rendon
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí San Luis Potosí, Mexico
| | | | - Roberto C Salgado-Delgado
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí San Luis Potosí, Mexico
| | - Nadia Saderi
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí San Luis Potosí, Mexico
| | - Marcela Miranda-Morales
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí San Luis Potosí, Mexico
| | - Mario Treviño
- Laboratory of Cortical Plasticity and Learning, Universidad de Guadalajara Guadalajara, Mexico
| | - Juan C Pineda
- Electrophysiology Laboratory, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán Mérida, Mexico
| | - Humberto Salgado
- Electrophysiology Laboratory, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán Mérida, Mexico
| |
Collapse
|
45
|
Neurons and astroglia govern microglial endotoxin tolerance through macrophage colony-stimulating factor receptor-mediated ERK1/2 signals. Brain Behav Immun 2016; 55:260-272. [PMID: 27132056 PMCID: PMC4899166 DOI: 10.1016/j.bbi.2016.04.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 04/20/2016] [Accepted: 04/26/2016] [Indexed: 11/20/2022] Open
Abstract
Endotoxin tolerance (ET) is a reduced responsiveness of innate immune cells like macrophages/monocytes to an endotoxin challenge following a previous encounter with the endotoxin. Although ET in peripheral systems has been well studied, little is known about ET in the brain. The present study showed that brain immune cells, microglia, being different from peripheral macrophages, displayed non-cell autonomous mechanisms in ET formation. Specifically, neurons and astroglia were indispensable for microglial ET. Macrophage colony-stimulating factor (M-CSF) secreted from these non-immune cells was essential for governing microglial ET. Neutralization of M-CSF deprived the neuron-glia conditioned medium of its ability to enable microglia to form ET when microglia encountered two lipopolysaccharide (LPS) treatments. Recombinant M-CSF protein rendered enriched microglia refractory to the second LPS challenge leading to microglial ET. Activation of microglial M-CSF receptor (M-CSFR; also known as CSF1R) and the downstream ERK1/2 signals was responsible for M-CSF-mediated microglial ET. Endotoxin-tolerant microglia in neuron-glia cultures displayed M2-like polarized phenotypes, as shown by upregulation of M2 marker Arg-1, elevated production of anti-inflammatory cytokine interleukin 10, and decreased secretion of pro-inflammatory mediators (tumor necrosis factor α, nitric oxide, prostaglandin E2 and interleukin 1β). Endotoxin-tolerant microglia protected neurons against LPS-elicited inflammatory insults, as shown by reduced neuronal damages in LPS pre-treatment group compared with the group without LPS pre-treatment. Moreover, while neurons and astroglia became injured during chronic neuroinflammation, microglia failed to form ET. Thus, this study identified a distinct non-cell autonomous mechanism of microglial ET. Interactions of M-CSF secreted by neurons and astroglia with microglial M-CSFR programed microglial ET. Loss of microglial ET could be an important pathogenetic mechanism of inflammation-associated neuronal damages.
Collapse
|
46
|
von Bernhardi R, Eugenín-von Bernhardi L, Eugenín J. Microglial cell dysregulation in brain aging and neurodegeneration. Front Aging Neurosci 2015; 7:124. [PMID: 26257642 PMCID: PMC4507468 DOI: 10.3389/fnagi.2015.00124] [Citation(s) in RCA: 377] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 06/22/2015] [Indexed: 12/29/2022] Open
Abstract
Aging is the main risk factor for neurodegenerative diseases. In aging, microglia undergoes phenotypic changes compatible with their activation. Glial activation can lead to neuroinflammation, which is increasingly accepted as part of the pathogenesis of neurodegenerative diseases, including Alzheimer’s disease (AD). We hypothesize that in aging, aberrant microglia activation leads to a deleterious environment and neurodegeneration. In aged mice, microglia exhibit an increased expression of cytokines and an exacerbated inflammatory response to pathological changes. Whereas LPS increases nitric oxide (NO) secretion in microglia from young mice, induction of reactive oxygen species (ROS) predominates in older mice. Furthermore, there is accumulation of DNA oxidative damage in mitochondria of microglia during aging, and also an increased intracellular ROS production. Increased ROS activates the redox-sensitive nuclear factor kappa B, which promotes more neuroinflammation, and can be translated in functional deficits, such as cognitive impairment. Mitochondria-derived ROS and cathepsin B, are also necessary for the microglial cell production of interleukin-1β, a key inflammatory cytokine. Interestingly, whereas the regulatory cytokine TGFβ1 is also increased in the aged brain, neuroinflammation persists. Assessing this apparent contradiction, we have reported that TGFβ1 induction and activation of Smad3 signaling after inflammatory stimulation are reduced in adult mice. Other protective functions, such as phagocytosis, although observed in aged animals, become not inducible by inflammatory stimuli and TGFβ1. Here, we discuss data suggesting that mitochondrial and endolysosomal dysfunction could at least partially mediate age-associated microglial cell changes, and, together with the impairment of the TGFβ1-Smad3 pathway, could result in the reduction of protective activation and the facilitation of cytotoxic activation of microglia, resulting in the promotion of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | | | - Jaime Eugenín
- Laboratory of Neural Systems, Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile (USACH) Santiago, Chile
| |
Collapse
|
47
|
Ishii Y, Yamaizumi A, Kawakami A, Islam A, Choudhury ME, Takahashi H, Yano H, Tanaka J. Anti-inflammatory effects of noradrenaline on LPS-treated microglial cells: Suppression of NFκB nuclear translocation and subsequent STAT1 phosphorylation. Neurochem Int 2015; 90:56-66. [PMID: 26190182 DOI: 10.1016/j.neuint.2015.07.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/13/2015] [Accepted: 07/14/2015] [Indexed: 12/22/2022]
Abstract
Noradrenaline (NA) has marked anti-inflammatory effects on activated microglial cells. The present study was conducted to elucidate the mechanisms underlying the NA effects using rat primary cultured microglial cells. NA, an α1 agonist, phenylephrine (Phe) and a β2 agonist, terbutaline (Ter) suppressed lipopolysaccharide (LPS)-induced nitric oxide (NO) release by microglia and prevented neuronal degeneration in LPS-treated neuron-microglia coculture. The agents suppressed expression of mRNA encoding proinflammatory mediators. Both an α1-selective blocker terazocine and a β2-selective blocker butoxamine overcame the suppressive effects of NA. cAMP-dependent kinase (PKA) inhibitors did not abolish the suppressive NA effects. LPS decreased IκB leading to NFκB translocation into nuclei, then induced phosphorylation of signal transducer and activator of transcription 1 (STAT1) and expression of interferon regulatory factor 1 (IRF1). NA inhibited LPS-induced these changes. When NFκB expression was knocked down with siRNA, LPS-induced STAT1 phosphorylation and IRF1 expression was abolished. NA did not suppress IL-6 induced STAT1 phosphorylation and IRF1 expression. These results suggest that one of the critical mechanisms underlying the anti-inflammatory effects of NA is the inhibition of NFκB translocation. Although inhibitory effects of NA on STAT1 phosphorylation and IRF1 expression may contribute to the overall suppressive effects of NA, these may be the downstream events of inhibitory effects on NFκB. Since NA, Phe and Ter exerted almost the same effects and PKA inhibitors did not show significant antagonistic effects, the suppression by NA might not be dependent on specific adrenergic receptors and cAMP-dependent signaling pathway.
Collapse
Affiliation(s)
- Yurika Ishii
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Japan
| | - Ayaka Yamaizumi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Japan
| | - Ayu Kawakami
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Japan
| | - Afsana Islam
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Japan
| | - Mohammed E Choudhury
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Japan
| | - Hisaaki Takahashi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Japan; Center for Advanced Research and Education, Asahikawa Medical University, Japan
| | - Hajime Yano
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Japan.
| |
Collapse
|