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 PMCID: PMC11660149 DOI: 10.1021/acschemneuro.4c00591] [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: 09/09/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [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
|
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: 3] [Impact Index Per Article: 1.5] [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
|
3
|
Gargano A, Olabiyi BF, Palmisano M, Zimmer A, Bilkei-Gorzo A. Possible role of locus coeruleus neuronal loss in age-related memory and attention deficits. Front Neurosci 2023; 17:1264253. [PMID: 37694113 PMCID: PMC10492095 DOI: 10.3389/fnins.2023.1264253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Aging is associated with a decline in cognitive abilities, including memory and attention. It is generally accepted that age-related histological changes such as increased neuroinflammatory glial activity and a reduction in the number of specific neuronal populations contribute to cognitive aging. Noradrenergic neurons in the locus coeruleus (LC) undergo an approximately 20 % loss during ageing both in humans and mice, but whether this change contributes to cognitive deficits is not known. To address this issue, we asked whether a similar loss of LC neurons in young animals as observed in aged animals impairs memory and attention, cognitive domains that are both influenced by the noradrenergic system and impaired in aging. Methods For that, we treated young healthy mice with DSP-4, a toxin that specifically kills LC noradrenergic neurons. We compared the performance of DSP-4 treated young mice with the performance of aged mice in models of attention and memory. To do this, we first determined the dose of DSP-4, which causes a similar 20 % neuronal loss as is typical in aged animals. Results Young mice treated with DSP-4 showed impaired attention in the presence of distractor and memory deficits in the 5-choice serial reaction time test (5-CSRTT). Old, untreated mice showed severe deficits in both the 5-CSRTT and in fear extinction tests. Discussion Our data now suggest that a reduction in the number of LC neurons contributes to impaired working memory and greater distractibility in attentional tasks but not to deficits in fear extinction. We hypothesize that the moderate loss of LC noradrenergic neurons during aging contributes to attention deficits and working memory impairments.
Collapse
Affiliation(s)
| | | | | | | | - Andras Bilkei-Gorzo
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Venusberg-Campus, Bonn, Germany
| |
Collapse
|
4
|
Meheronnisha SK, Thekkekkara D, Babu A, Tausif YM, Manjula SN. Novel therapeutic targets to halt the progression of Parkinson's disease: an in-depth review on molecular signalling cascades. 3 Biotech 2023; 13:218. [PMID: 37265542 PMCID: PMC10229523 DOI: 10.1007/s13205-023-03637-5] [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: 03/06/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Recent research has focused mostly on understanding and combating the neurodegenerative mechanisms and symptoms of Parkinson's disease (PD). Moreover, developing novel therapeutic targets to halt the progression of PD remains a key focus for researchers. As yet, no agents have been found to have unambiguous evidence of disease-modifying actions in PD. The primary objective of this review is to summarize the promising targets that have recently been uncovered which include histamine 4 receptors, beta2 adrenergic receptor, phosphodiesterase 4, sphingosine-1-phosphate receptor subtype 1, angiotensin receptors, high-mobility group box 1, rabphilin-3A, purinergic 2Y type 12 receptor, colony-stimulating factor-1 receptor, transient receptor potential vanilloid 4, alanine-serine-cysteine transporter 2, G protein-coupled oestrogen receptor, a mitochondrial antiviral signalling protein, glucocerebrosidase, indolamine-2,3-dioxygenase-1, soluble epoxy hydroxylase and dual specificity phosphatase 6. We have also reviewed the molecular signalling cascades of those novel targets which cause the initiation and progression of PD and gathered some emerging disease-modifying agents that could slow the progression of PD. These approaches will assist in the discovery of novel target molecules, for curing disease symptoms and may provide a glimmer of hope for the treatment of PD. As of now, there is no drug available that will completely prevent the progression of PD by inhibiting the pathogenesis involved in PD, and thus, the newer targets and their inhibitors or activators are the major focus for researchers to suppress PD symptomatology. And the major limitations of these targets are the lack of clinical data and less number pre-clinical data, as we have majorly discussed the different targets which all have well reported for other disease pathogenesis. Thus, finding the disease-drug interactions, the molecular mechanisms, and the major side effects will be major challenges for the researchers.
Collapse
Affiliation(s)
- S. K. Meheronnisha
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysore, Karnataka 570015 India
| | - Dithu Thekkekkara
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysore, Karnataka 570015 India
| | - Amrita Babu
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysore, Karnataka 570015 India
| | - Y. Mohammed Tausif
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysore, Karnataka 570015 India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysore, Karnataka 570015 India
| |
Collapse
|
5
|
Wu Y, Chen L, Zhong F, Zhou K, Lu C, Cheng X, Wang S. Cognitive impairment in patients with heart failure: molecular mechanism and therapy. Heart Fail Rev 2023:10.1007/s10741-022-10289-9. [PMID: 36593370 DOI: 10.1007/s10741-022-10289-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 01/04/2023]
Abstract
Heart failure (HF) is associated with multiple organ dysfunction and many comorbidities. Its incidence is high among the elderly and is a major health burden worldwide. Cognitive impairment (CI) is highly prevalent in older patients with HF, which is an abnormality in one or more of the items of cognition, attention, memory, language, psychomotor function, and visual spatial acuity. Studies have shown that the incidence of CI in HF patients is between 13 and 54%, and patients with both conditions have poor self-care ability and prognosis, as well as increased mortality rates. However, the mechanisms of CI development in HF patients are still unclear. In this review, we describe the epidemiology and risk factors as well as measures of improving CI in HF patients. We update the latest pathophysiological mechanisms related to the neurocognitive changes in HF patients, expounding on the mechanisms associated with the development of CI in HF patients.
Collapse
Affiliation(s)
- Yanan Wu
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Liwen Chen
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Feng Zhong
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Kaiyi Zhou
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Chao Lu
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Xiao Cheng
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sheng Wang
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China.
| |
Collapse
|
6
|
Minné D, Marnewick JL, Engel-Hills P. Early Chronic Stress Induced Changes within the Locus Coeruleus in Sporadic Alzheimer's Disease. Curr Alzheimer Res 2023; 20:301-317. [PMID: 37872793 DOI: 10.2174/1567205020666230811092956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 10/25/2023]
Abstract
Chronic exposure to stress throughout the lifespan has been the focus of many studies on Alzheimer's disease (AD) because of the similarities between the biological mechanisms involved in chronic stress and the pathophysiology of AD. In fact, the earliest abnormality associated with the disease is the presence of phosphorylated tau protein in locus coeruleus neurons, a brain structure highly responsive to stress and perceived threat. Here, we introduce allostatic load as a useful concept for understanding many of the complex, interacting neuropathological changes involved in the AD degenerative process. In response to chronic stress, aberrant tau proteins that begin to accumulate within the locus coeruleus decades prior to symptom onset appear to represent a primary pathological event in the AD cascade, triggering a wide range of interacting brain changes involving neuronal excitotoxicity, endocrine alterations, inflammation, oxidative stress, and amyloid plaque exacerbation. While it is acknowledged that stress will not necessarily be the major precipitating factor in all cases, early tau-induced changes within the locus coeruleus-norepinephrine pathway suggests that a therapeutic window might exist for preventative measures aimed at managing stress and restoring balance within the HPA axis.
Collapse
Affiliation(s)
- Donné Minné
- Applied Microbial & Health Biotechnology Institute, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
- Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| | - Jeanine L Marnewick
- Applied Microbial & Health Biotechnology Institute, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| | - Penelope Engel-Hills
- Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| |
Collapse
|
7
|
Gutiérrez IL, Dello Russo C, Novellino F, Caso JR, García-Bueno B, Leza JC, Madrigal JLM. Noradrenaline in Alzheimer's Disease: A New Potential Therapeutic Target. Int J Mol Sci 2022; 23:ijms23116143. [PMID: 35682822 PMCID: PMC9181823 DOI: 10.3390/ijms23116143] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/13/2022] Open
Abstract
A growing body of evidence demonstrates the important role of the noradrenergic system in the pathogenesis of many neurodegenerative processes, especially Alzheimer’s disease, due to its ability to control glial activation and chemokine production resulting in anti-inflammatory and neuroprotective effects. Noradrenaline involvement in this disease was first proposed after finding deficits of noradrenergic neurons in the locus coeruleus from Alzheimer’s disease patients. Based on this, it has been hypothesized that the early loss of noradrenergic projections and the subsequent reduction of noradrenaline brain levels contribute to cognitive dysfunctions and the progression of neurodegeneration. Several studies have focused on analyzing the role of noradrenaline in the development and progression of Alzheimer’s disease. In this review we summarize some of the most relevant data describing the alterations of the noradrenergic system normally occurring in Alzheimer’s disease as well as experimental studies in which noradrenaline concentration was modified in order to further analyze how these alterations affect the behavior and viability of different nervous cells. The combination of the different studies here presented suggests that the maintenance of adequate noradrenaline levels in the central nervous system constitutes a key factor of the endogenous defense systems that help prevent or delay the development of Alzheimer’s disease. For this reason, the use of noradrenaline modulating drugs is proposed as an interesting alternative therapeutic option for Alzheimer’s disease.
Collapse
Affiliation(s)
- Irene L. Gutiérrez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Cinzia Dello Russo
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool L69 3GL, UK
| | - Fabiana Novellino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council, 88100 Catanzaro, Italy
| | - Javier R. Caso
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Borja García-Bueno
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Juan C. Leza
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - José L. M. Madrigal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Correspondence: ; Tel.: +34-91-394-1463
| |
Collapse
|
8
|
Effects of Long-Term Vagus Nerve Electrical Stimulation Therapy on Acute Cerebral Infarction and Neurological Function Recovery in Post MCAO Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8131391. [PMID: 35391930 PMCID: PMC8983242 DOI: 10.1155/2022/8131391] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 02/28/2022] [Accepted: 03/08/2022] [Indexed: 12/28/2022]
Abstract
Background Vagus nerve stimulation therapy is proven to produce neuroprotective effects against central nervous system diseases and reduce neurological injury, having a positive effect on the recovery of neurological functions in mouse model of stroke. Objective This study was aimed at exploring a wider time window for VNS treatment, investigating the long-term behavioral improvement of long-term VNS in mice after pMCAO, and exploring the antiapoptotic properties of VNS and its role in autophagy, all of which may be a permanent deficiency potential mechanism of neuroprotection in hemorrhagic stroke. Methods Permanent focal cerebral ischemia and implantation of vagus nerve stimulator were performed through intracavitary occlusion of the right middle cerebral artery (MCA). The vagus nerve stimulation group received five times vagus nerve stimulation from 6 h after surgery for 5 days. Adhesive removal test and NSS neurological score were used to evaluate the neurological deficit of mice. TTC staining of mouse brain tissue was performed one week after surgery in order to assess the area of cerebral infarction. Additionally, frozen sections were stained with Fluoro-Jade B to observe the apoptotic cells in the ischemic penumbra of brain tissue. Finally, Western blot was used to detect the changes in the levels of apoptosis-related proteins such as cleaved-caspase3 and Bcl-2 and autophagy-related proteins such as mTOR, Beclin-1, and LC3-II in brain tissue. Results VNS can effectively reduce the behavioral score of pMCAO mice; TTC results showed that VNS could effectively reduce the infarct area after pMCAO (P < 0.05). After VNS intervention of the pMCAO group compared with the pMCAO+VNI group, the FJB-positive cells in the VNS group were significantly decreased (P < 0.05); Western Blot analysis showed that the expression of cleaved-caspase3 in the brain tissue of mice increased after pMCAO (P < 0.05), and the expression of Bcl-2 decreased (P < 0.05). This change could be effectively reversed after VNS intervention (P < 0.05). Conclusion VNS could effectively improve the behavioral performance of mice after permanent stroke in addition to significantly reducing the infarct size of the brain tissue. The mechanism may be related to the effective reduction of cell apoptosis and excessive autophagy after pMCAO by VNS.
Collapse
|
9
|
James T, Kula B, Choi S, Khan SS, Bekar LK, Smith NA. Locus coeruleus in memory formation and Alzheimer's disease. Eur J Neurosci 2021; 54:6948-6959. [PMID: 33190318 PMCID: PMC8121900 DOI: 10.1111/ejn.15045] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/26/2022]
Abstract
Catecholamine neurons of the locus coeruleus (LC) in the dorsal pontine tegmentum innervate the entire neuroaxis, with signaling actions implicated in the regulation of attention, arousal, sleep-wake cycle, learning, memory, anxiety, pain, mood, and brain metabolism. The co-release of norepinephrine (NE) and dopamine (DA) from LC terminals in the hippocampus plays a role in all stages of hippocampal-memory processing. This catecholaminergic regulation modulates the encoding, consolidation, retrieval, and reversal of hippocampus-based memory. LC neurons in awake animals have two distinct firing modes: tonic firing (explorative) and phasic firing (exploitative). These two firing modes exert different modulatory effects on post-synaptic dendritic spines. In the hippocampus, the firing modes regulate long-term potentiation (LTP) and long-term depression, which differentially regulate the mRNA expression and transcription of plasticity-related proteins (PRPs). These proteins aid in structural alterations of dendritic spines, that is, structural long-term potentiation (sLTP), via expansion and structural long-term depression (sLTD) via contraction of post-synaptic dendritic spines. Given the LC's role in all phases of memory processing, the degeneration of 50% of the LC neuron population occurring in Alzheimer's disease (AD) is a clinically relevant aspect of disease pathology. The loss of catecholaminergic regulation contributes to dysfunction in memory processes along with impaired functions associated with attention and task completion. The multifaceted role of the LC in memory and general task performance and the close correlation of LC degeneration with neurodegenerative disease progression together implicate it as a target for new clinical assessment tools.
Collapse
Affiliation(s)
- Tony James
- George Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Bartosz Kula
- Center for NeuroscienceChildren's National Research InstituteChildren's National HospitalWashingtonDCUSA
| | - Seowon Choi
- Center for NeuroscienceChildren's National Research InstituteChildren's National HospitalWashingtonDCUSA
- Thomas Jefferson High School for Science and TechnologyAlexandriaVAUSA
| | | | - Lane K. Bekar
- Department of Anatomy, Physiology and PharmacologyUniversity of SaskatchewanSaskatoonCanada
| | - Nathan A. Smith
- George Washington University School of Medicine and Health SciencesWashingtonDCUSA
- Center for NeuroscienceChildren's National Research InstituteChildren's National HospitalWashingtonDCUSA
| |
Collapse
|
10
|
Sigurdsson HP, Raw R, Hunter H, Baker MR, Taylor JP, Rochester L, Yarnall AJ. Noninvasive vagus nerve stimulation in Parkinson's disease: current status and future prospects. Expert Rev Med Devices 2021; 18:971-984. [PMID: 34461787 DOI: 10.1080/17434440.2021.1969913] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Parkinson's disease (PD) is a common progressive neurodegenerative disorder with multifactorial etiology. While dopaminergic medication is the standard therapy in PD, it provides limited symptomatic treatment and non-pharmacological interventions are currently being trialed. AREAS COVERED Recent pathophysiological theories of Parkinson's suggest that aggregated α-synuclein form in the gut and spread to nuclei in the brainstem via autonomic connections. In this paper, we review the novel hypothesis that noninvasive vagus nerve stimulation (nVNS), targeting efferent and afferent vagal projections, is a promising therapeutic tool to improve gait and cognitive control and ameliorate non-motor symptoms in people with Parkinson's. We conducted an unstructured search of the literature for any studies employing nVNS in PD as well as for studies examining the efficacy of nVNS on improving cognitive function and where nVNS has been applied to co-occurring conditions in PD. EXPERT OPINION Evidence of nVNS as a novel therapeutic to improve gait in PD is preliminary, but early signs indicate the possibility that nVNS may be useful to target dopa-resistant gait characteristics in early PD. The evidence for nVNS as a therapeutic tool is, however, limited and further studies are needed in both brain health and disease.
Collapse
Affiliation(s)
- Hilmar P Sigurdsson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Rachael Raw
- Department of General Internal Medicine, South Tees Hospitals NHS Foundation Trust, Middlesbrough, UK
| | - Heather Hunter
- Department of Research, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Mark R Baker
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Department of Clinical Neurophysiology, Newcastle upon Tyne NHS Hospitals Foundation Trust, Newcastle upon Tyne, UK
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Lynn Rochester
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Department of Neurosciences, Newcastle upon Tyne NHS Hospitals Foundation Trust, Newcastle upon Tyne, UK
| | - Alison J Yarnall
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Department of Older People's Medicine, Newcastle upon Tyne NHS Hospitals Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
11
|
Matchett BJ, Grinberg LT, Theofilas P, Murray ME. The mechanistic link between selective vulnerability of the locus coeruleus and neurodegeneration in Alzheimer's disease. Acta Neuropathol 2021; 141:631-650. [PMID: 33427939 PMCID: PMC8043919 DOI: 10.1007/s00401-020-02248-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 01/24/2023]
Abstract
Alzheimer's disease (AD) is neuropathologically characterized by the intracellular accumulation of hyperphosphorylated tau and the extracellular deposition of amyloid-β plaques, which affect certain brain regions in a progressive manner. The locus coeruleus (LC), a small nucleus in the pons of the brainstem, is widely recognized as one of the earliest sites of neurofibrillary tangle formation in AD. Patients with AD exhibit significant neuronal loss in the LC, resulting in a marked reduction of its size and function. The LC, which vastly innervates several regions of the brain, is the primary source of the neurotransmitter norepinephrine (NE) in the central nervous system. Considering that NE is a major modulator of behavior, contributing to neuroprotection and suppression of neuroinflammation, degeneration of the LC in AD and the ultimate dysregulation of the LC-NE system has detrimental effects in the brain. In this review, we detail the neuroanatomy and function of the LC, its essential role in neuroprotection, and how this is dysregulated in AD. We discuss AD-related neuropathologic changes in the LC and mechanisms by which LC neurons are selectively vulnerable to insult. Further, we elucidate the neurotoxic effects of LC de-innervation both locally and at projection sites, and how this augments disease pathology, progression and severity. We summarize how preservation of the LC-NE system could be used in the treatment of AD and other neurodegenerative diseases affected by LC degeneration.
Collapse
Affiliation(s)
- Billie J. Matchett
- Neuropathology Laboratory, Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Lea T. Grinberg
- Memory and Aging Center, Department of Neurology, University of California, 675 Nelson Rising Lane, San Francisco, CA 94158 USA
| | - Panos Theofilas
- Memory and Aging Center, Department of Neurology, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA.
| | - Melissa E. Murray
- Neuropathology Laboratory, Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| |
Collapse
|
12
|
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: 2.0] [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]
|
13
|
González-Prieto M, Gutiérrez IL, García-Bueno B, Caso JR, Leza JC, Ortega-Hernández A, Gómez-Garre D, Madrigal JLM. Microglial CX3CR1 production increases in Alzheimer's disease and is regulated by noradrenaline. Glia 2020; 69:73-90. [PMID: 32662924 DOI: 10.1002/glia.23885] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022]
Abstract
The loss of noradrenergic neurons and subsequent reduction of brain noradrenaline (NA) levels are associated with the progression of Alzheimer's disease (AD). This seems to be due mainly to the ability of NA to reduce the activation of microglial cells. We previously observed that NA induces the production of the chemokine Fractalkine/CX3CL1 in neurons. The activation of microglial CX3CR1, sole receptor for CX3CL1, reduces the activation of microglia, which is known to largely contribute to the neuronal damage characteristic of AD. Therefore, alterations of CX3CR1 production in microglia could translate into the enhancement or inhibition of CX3CL1 anti-inflammatory effects. In order to determine if microglial CX3CR1 production is altered in AD and if NA can control it, CX3CR1 expression and synthesis were analyzed in 5xFAD mice and human AD brain samples. In addition, the effects of NA and its reuptake inhibitor reboxetine were analyzed in microglial cultures and mice respectively. Our results indicate that in AD CX3CR1 production is increased in the brain cortex and that reboxetine administration further increases it and enhances microglial reactivity toward amyloid beta plaques. However, direct administration of NA to primary rat microglia or human HMC3 cells inhibits CX3CR1 production, suggesting that microglia responses to NA may be altered in the absence of CX3CL1-producing neurons or other nonmicroglial external factors.
Collapse
Affiliation(s)
- Marta González-Prieto
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| | - Irene L Gutiérrez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| | - Borja García-Bueno
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| | - Javier R Caso
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| | - Juan C Leza
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| | - Adriana Ortega-Hernández
- Vascular Biology Laboratory and Flow Cytometric Unit, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain.,Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Dulcenombre Gómez-Garre
- Vascular Biology Laboratory and Flow Cytometric Unit, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain.,Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - José L M Madrigal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| |
Collapse
|
14
|
Duffy KB, Ray B, Lahiri DK, Tilmont EM, Tinkler GP, Herbert RL, Greig NH, Ingram DK, Ottinger MA, Mattison JA. Effects of Reducing Norepinephrine Levels via DSP4 Treatment on Amyloid-β Pathology in Female Rhesus Macaques (Macaca Mulatta). J Alzheimers Dis 2020; 68:115-126. [PMID: 30689563 DOI: 10.3233/jad-180487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The degeneration in the locus coeruleus associated with Alzheimer's disease suggests an involvement of the noradrenergic system in the disease pathogenesis. The role of depleted norepinephrine was tested in adult and aged rhesus macaques to develop a potential model for testing Alzheimer's disease interventions. Monkeys were injected with the noradrenergic neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4) or vehicle at 0, 3, and 6 months; brains were harvested at 9 months. Reduced norepinephrine in the locus coeruleus was accompanied by decreased dopamine β-hydroxylase staining and increased amyloid-β load in the aged group, and the proportion of potentially toxic amyloid-β42 peptide was increased. Immunohistochemistry revealed no effects on microglia or astrocytes. DSP4 treatment altered amyloid processing, but these changes were not associated with the induction of chronic neuroinflammation. These findings suggest norepinephrine deregulation is an essential component of a nonhuman primate model of Alzheimer's disease, but further refinement is necessary.
Collapse
Affiliation(s)
- Kara B Duffy
- Animal and Avian Sciences Department, University of Maryland, College Park, MD, USA
| | - Balmiki Ray
- Myriad Neuroscience (Assurex Health), Mason, OH, USA (present address).,Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Debomoy K Lahiri
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Edward M Tilmont
- Translational Gerontology Branch, National Institute on Aging, NIH Animal Center, Dickerson, MD, USA
| | - Gregory P Tinkler
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Richard L Herbert
- Clinical Medicine Branch, National Institute of Allergy and Infectious Disease, NIH, Dickerson, MD, USA
| | - Nigel H Greig
- Translational Gerontology Branch, NIA/NIH, Baltimore, MD, USA
| | - Donald K Ingram
- Nutritional Neuroscience and Aging Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Mary Ann Ottinger
- Animal and Avian Sciences Department, University of Maryland, College Park, MD, USA.,Department of Biology and Biochemistry, University of Houston, Houston, TX, USA (present address)
| | - Julie A Mattison
- Translational Gerontology Branch, National Institute on Aging, NIH Animal Center, Dickerson, MD, USA
| |
Collapse
|
15
|
Chen X, Gumina G, Virga KG. Recent Advances in Drug Repurposing for Parkinson's Disease. Curr Med Chem 2019; 26:5340-5362. [PMID: 30027839 DOI: 10.2174/0929867325666180719144850] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 12/25/2022]
Abstract
As a long-term degenerative disorder of the central nervous system that mostly affects older people, Parkinson's disease is a growing health threat to our ever-aging population. Despite remarkable advances in our understanding of this disease, all therapeutics currently available only act to improve symptoms but cannot stop the disease progression. Therefore, it is essential that more effective drug discovery methods and approaches are developed, validated, and used for the discovery of disease-modifying treatments for Parkinson's disease. Drug repurposing, also known as drug repositioning, or the process of finding new uses for existing or abandoned pharmaceuticals, has been recognized as a cost-effective and timeefficient way to develop new drugs, being equally promising as de novo drug discovery in the field of neurodegeneration and, more specifically for Parkinson's disease. The availability of several established libraries of clinical drugs and fast evolvement in disease biology, genomics and bioinformatics has stimulated the momentums of both in silico and activity-based drug repurposing. With the successful clinical introduction of several repurposed drugs for Parkinson's disease, drug repurposing has now become a robust alternative approach to the discovery and development of novel drugs for this disease. In this review, recent advances in drug repurposing for Parkinson's disease will be discussed.
Collapse
Affiliation(s)
- Xin Chen
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC 29325, United States
| | - Giuseppe Gumina
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC 29325, United States
| | - Kristopher G Virga
- Department of Pharmaceutical Sciences, William Carey University School of Pharmacy, Biloxi, MS 39532, United States
| |
Collapse
|
16
|
Jang HS, Kim J, Padanilam BJ. Renal sympathetic nerve activation via α 2-adrenergic receptors in chronic kidney disease progression. Kidney Res Clin Pract 2019; 38:6-14. [PMID: 30831675 PMCID: PMC6481969 DOI: 10.23876/j.krcp.18.0143] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/11/2018] [Accepted: 12/15/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic kidney disease (CKD) is increasing worldwide without an effective therapeutic strategy. Sympathetic nerve activation is implicated in CKD progression, as well as cardiovascular dysfunction. Renal denervation is beneficial for controlling blood pressure (BP) and improving renal function through reduction of sympathetic nerve activity in patients with resistant hypertension and CKD. Sympathetic neurotransmitter norepinephrine (NE) via adrenergic receptor (AR) signaling has been implicated in tissue homeostasis and various disease progressions, including CKD. Increased plasma NE level is a predictor of survival and the incidence of cardiovascular events in patients with end-stage renal disease, as well as future renal injury in subjects with normal BP and renal function. Our recent data demonstrate that NE derived from renal nerves causes renal inflammation and fibrosis progression through alpha-2 adrenergic receptors (α2-AR) in renal fibrosis models independent of BP. Sympathetic nerve activation-associated molecular mechanisms and signals seem to be critical for the development and progression of CKD, but the exact role of sympathetic nerve activation in CKD progression remains undefined. This review explores the current knowledge of NE-α2-AR signaling in renal diseases and offers prospective views on developing therapeutic strategies targeting NE-AR signaling in CKD progression.
Collapse
Affiliation(s)
- Hee-Seong Jang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jinu Kim
- Department of Anatomy, Jeju National University School of Medicine, Jeju, Korea.,Department of Biomedicine and Drug Development, Jeju National University, Jeju, Korea
| | - Babu J Padanilam
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, Section of Nephrology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
17
|
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: 1.1] [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
|
18
|
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: 53] [Impact Index Per Article: 7.6] [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
|
19
|
Phillips C, Fahimi A. Immune and Neuroprotective Effects of Physical Activity on the Brain in Depression. Front Neurosci 2018; 12:498. [PMID: 30093853 PMCID: PMC6070639 DOI: 10.3389/fnins.2018.00498] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/03/2018] [Indexed: 12/13/2022] Open
Abstract
Physical activity-a lifestyle factor that is associated with immune function, neuroprotection, and energy metabolism-modulates the cellular and molecular processes in the brain that are vital for emotional and cognitive health, collective mechanisms that can go awry in depression. Physical activity optimizes the stress response, neurotransmitter level and function (e.g., serotonergic, noradrenergic, dopaminergic, and glutamatergic), myokine production (e.g., interleukin-6), transcription factor levels and correlates [e.g., peroxisome proliferator-activated receptor C coactivator-1α [PGC-1α], mitochondrial density, nitric oxide pathway activity, Ca2+ signaling, reactive oxygen specie production, and AMP-activated protein kinase [AMPK] activity], kynurenine metabolites, glucose regulation, astrocytic health, and growth factors (e.g., brain-derived neurotrophic factor). Dysregulation of these interrelated processes can effectuate depression, a chronic mental illness that affects millions of individuals worldwide. Although the biogenic amine model has provided some clinical utility in understanding chronic depression, a need remains to better understand the interrelated mechanisms that contribute to immune dysfunction and the means by which various therapeutics mitigate them. Fortunately, convergent evidence suggests that physical activity improves emotional and cognitive function in persons with depression, particularly in those with comorbid inflammation. Accordingly, the aims of this review are to (1) underscore the link between inflammatory correlates and depression, (2) explicate immuno-neuroendocrine foundations, (3) elucidate evidence of neurotransmitter and cytokine crosstalk in depressive pathobiology, (4) determine the immunomodulatory effects of physical activity in depression, (5) examine protocols used to effectuate the positive effects of physical activity in depression, and (6) highlight implications for clinicians and scientists. It is our contention that a deeper understanding of the mechanisms by which inflammation contributes to the pathobiology of depression will translate to novel and more effective treatments, particularly by identifying relevant patient populations that can benefit from immune-based therapies within the context of personalized medicine.
Collapse
Affiliation(s)
- Cristy Phillips
- Physical Therapy, Arkansas State University, Jonesboro, AR, United States
- Physical Therapy, University of Tennessee Health Science Center, Memphis, TN, United States
| | | |
Collapse
|
20
|
Kalinin S, González-Prieto M, Scheiblich H, Lisi L, Kusumo H, Heneka MT, Madrigal JLM, Pandey SC, Feinstein DL. Transcriptome analysis of alcohol-treated microglia reveals downregulation of beta amyloid phagocytosis. J Neuroinflammation 2018; 15:141. [PMID: 29759078 PMCID: PMC5952855 DOI: 10.1186/s12974-018-1184-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 04/29/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Microglial activation contributes to the neuropathology associated with chronic alcohol exposure and withdrawal, including the expression of inflammatory and anti-inflammatory genes. In the current study, we examined the transcriptome of primary rat microglial cells following incubation with alcohol alone, or alcohol together with a robust inflammatory stimulus. METHODS Primary microglia were prepared from mixed rat glial cultures. Cells were incubated with 75 mM ethanol alone or with proinflammatory cytokines ("TII": IL1β, IFNγ, and TNFα). Isolated mRNA was used for RNAseq analysis and qPCR. Effects of alcohol on phagocytosis were determined by uptake of oligomeric amyloid beta. RESULTS Alcohol induced nitrite production in control cells and increased nitrite production in cells co-treated with TII. RNAseq analysis of microglia exposed for 24 h to alcohol identified 312 differentially expressed mRNAs ("Alc-DEs"), with changes confirmed by qPCR analysis. Gene ontology analysis identified phagosome as one of the highest-ranking KEGG pathways including transcripts regulating phagocytosis. Alcohol also increased several complement-related mRNAs that have roles in phagocytosis, including C1qa, b, and c; C3; and C3aR1. RNAseq analysis identified over 3000 differentially expressed mRNAs in microglia following overnight incubation with TII; and comparison to the group of Alc-DEs revealed 87 mRNAs modulated by alcohol but not by TII, including C1qa, b, and c. Consistent with observed changes in phagocytosis-related mRNAs, the uptake of amyloid beta1-42, by primary microglia, was reduced by alcohol. CONCLUSIONS Our results define alterations that occur to microglial gene expression following alcohol exposure and suggest that alcohol effects on phagocytosis could contribute to the development of Alzheimer's disease.
Collapse
Affiliation(s)
- Sergey Kalinin
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Marta González-Prieto
- Department of Pharmacology, University Complutense, Centro de Investigacion Biomedica en Red de Salud Mental (CIBERSAM), Madrid, 28040 Spain
| | - Hannah Scheiblich
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn, 53127 Bonn, Germany
| | - Lucia Lisi
- Institute of Pharmacology, Catholic University Medical School, 00168 Rome, Italy
| | - Handojo Kusumo
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Michael T. Heneka
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn, 53127 Bonn, Germany
| | - Jose L. M. Madrigal
- Department of Pharmacology, University Complutense, Centro de Investigacion Biomedica en Red de Salud Mental (CIBERSAM), Madrid, 28040 Spain
| | - Subhash C. Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612 USA
- Department of Veterans Affairs, Jesse Brown VA Medical Center, Chicago, IL 60612 USA
| | - Douglas L. Feinstein
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612 USA
- Department of Veterans Affairs, Jesse Brown VA Medical Center, Chicago, IL 60612 USA
| |
Collapse
|
21
|
Treatment with the noradrenaline re-uptake inhibitor atomoxetine alone and in combination with the α2-adrenoceptor antagonist idazoxan attenuates loss of dopamine and associated motor deficits in the LPS inflammatory rat model of Parkinson's disease. Brain Behav Immun 2018; 69:456-469. [PMID: 29339319 DOI: 10.1016/j.bbi.2018.01.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/04/2018] [Accepted: 01/11/2018] [Indexed: 01/14/2023] Open
Abstract
The impact of treatment with the noradrenaline (NA) re-uptake inhibitor atomoxetine and the α2-adrenoceptor (AR) antagonist idazoxan in an animal model of Parkinson's disease (PD) was assessed. Concurrent systemic treatment with atomoxetine and idazoxan, a combination which serves to enhance the extra-synaptic availability of NA, exerts anti-inflammatory and neuroprotective effects following delivery of an inflammatory stimulus, the bacterial endotoxin, lipopolysaccharide (LPS) into the substantia nigra. Lesion-induced deficits in motor function (akinesia, forelimb-use asymmetry) and striatal dopamine (DA) loss were rescued to varying degrees depending on the treatment. Treatment with atomoxetine following LPS-induced lesion to the substantia nigra, yielded a robust anti-inflammatory effect, suppressing microglial activation and expression of the pro-inflammatory cytokine TNF-α whilst increasing the expression of neurotrophic factors. Furthermore atomoxetine treatment prevented loss of tyrosine hydroxylase (TH) positive nigral dopaminergic neurons and resulted in functional improvements in motor behaviours. Atomoxetine alone was sufficient to achieve most of the observed effects. In combination with idazoxan, an additional improvement in the impairment of contralateral limb use 7 days post lesion and a reduction in amphetamine-mediated rotational asymmetry 14 days post-lesion was observed, compared to atomoxetine or idazoxan treatments alone. The results indicate that increases in central NA tone has the propensity to regulate the neuroinflammatory phenotype in vivo and may act as an endogenous neuroprotective mechanism where inflammation contributes to the progression of DA loss. In accordance with this, the clinical use of agents such as NA re-uptake inhibitors and α2-AR antagonists may prove useful in enhancing the endogenous neuroimmunomodulatory potential of NA in conditions associated with brain inflammation.
Collapse
|
22
|
Assessing disease-modifying effects of norepinephrine in Down syndrome and Alzheimer's disease. Brain Res 2017; 1702:3-11. [PMID: 29102776 DOI: 10.1016/j.brainres.2017.09.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/18/2017] [Indexed: 11/23/2022]
Abstract
Building upon the knowledge that a number of important brain circuits undergo significant degeneration in Alzheimer's disease, numerous recent studies suggest that the norepinephrine-ergic system in the brainstem undergoes significant alterations early in the course of both Alzheimer's disease and Down syndrome. Massive projections from locus coeruleus neurons to almost the entire brain, extensive innervation of brain capillaries, and widespread distribution of noradrenergic receptors enable the norepinephrine-ergic system to play a crucial role in neural processes, including cognitive function. These anatomical and functional characteristics support the role of the norepinephrine-ergic system as an important target for developing new therapies for cognitive dysfunction. Careful neuropathological examinations using postmortem samples from individuals with Alzheimer's disease have implicated the role of the norepinephrine-ergic system in the etiopathogenesis of Alzheimer's disease. Furthermore, numerous studies have supported the existence of a strong interaction between norepinephrine-ergic and neuroimmune systems. We explore the interaction between the two systems that could play a role in the disease-modifying effects of norepinephrine in Alzheimer's disease and Down syndrome.
Collapse
|
23
|
Farrand AQ, Helke KL, Gregory RA, Gooz M, Hinson VK, Boger HA. Vagus nerve stimulation improves locomotion and neuronal populations in a model of Parkinson's disease. Brain Stimul 2017; 10:1045-1054. [PMID: 28918943 PMCID: PMC5675746 DOI: 10.1016/j.brs.2017.08.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/15/2017] [Accepted: 08/22/2017] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive, neurodegenerative disorder with no disease-modifying therapies, and symptomatic treatments are often limited by debilitating side effects. In PD, locus coeruleus noradrenergic (LC-NE) neurons degenerate prior to substantia nigra dopaminergic (SN-DA) neurons. Vagus nerve stimulation (VNS) activates LC neurons, and decreases pro-inflammatory markers, allowing improvement of LC targets, making it a potential PD therapeutic. OBJECTIVE To assess therapeutic potential of VNS in a PD model. METHODS To mimic the progression of PD degeneration, rats received a systemic injection of noradrenergic neurotoxin DSP-4, followed one week later by bilateral intrastriatal injection of dopaminergic neurotoxin 6-hydroxydopamine. At this time, a subset of rats also had vagus cuffs implanted. After eleven days, rats received a precise VNS regimen twice a day for ten days, and locomotion was measured during each afternoon session. Immediately following final stimulation, rats were euthanized, and left dorsal striatum, bilateral SN and LC were sectioned for immunohistochemical detection of monoaminergic neurons (tyrosine hydroxylase, TH), α-synuclein, astrocytes (GFAP) and microglia (Iba-1). RESULTS VNS significantly increased locomotion of lesioned rats. VNS also resulted in increased expression of TH in striatum, SN, and LC; decreased SN α-synuclein expression; and decreased expression of glial markers in the SN and LC of lesioned rats. Additionally, saline-treated rats after VNS, had higher LC TH and lower SN Iba-1. CONCLUSIONS Our findings of increased locomotion, beneficial effects on LC-NE and SN-DA neurons, decreased α-synuclein density in SN TH-positive neurons, and neuroinflammation suggest VNS has potential as a novel PD therapeutic.
Collapse
Affiliation(s)
- Ariana Q Farrand
- Dept of Neuroscience and Center on Aging, Medical University of South Carolina, 173 Ashley Ave, BSB403, MSC510, Charleston, SC 29425, USA
| | - Kristi L Helke
- Dept of Comparative Medicine, Medical University of South Carolina, 114 Doughty St, STB 648, MSC 777, Charleston, SC 29425, USA; Dept of Pathology, Medical University of South Carolina, 165 Ashley Ave, Children's Hospital 309, MSC 908, Charleston, SC 29425, USA
| | - Rebecca A Gregory
- Dept of Neuroscience and Center on Aging, Medical University of South Carolina, 173 Ashley Ave, BSB403, MSC510, Charleston, SC 29425, USA; Dept of Comparative Medicine, Medical University of South Carolina, 114 Doughty St, STB 648, MSC 777, Charleston, SC 29425, USA
| | - Monika Gooz
- Dept of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President St, DDB 507, MSC 139, Charleston, SC 29425, USA
| | - Vanessa K Hinson
- Dept of Neurology, Medical University of South Carolina, 96 Jonathan Lucas St, CSB 309, MSC 606, Charleston, SC 29425, USA
| | - Heather A Boger
- Dept of Neuroscience and Center on Aging, Medical University of South Carolina, 173 Ashley Ave, BSB403, MSC510, Charleston, SC 29425, USA.
| |
Collapse
|
24
|
Yi B, Jahangir A, Evans AK, Briggs D, Ravina K, Ernest J, Farimani AB, Sun W, Rajadas J, Green M, Feinberg EN, Pande VS, Shamloo M. Discovery of novel brain permeable and G protein-biased beta-1 adrenergic receptor partial agonists for the treatment of neurocognitive disorders. PLoS One 2017; 12:e0180319. [PMID: 28746336 PMCID: PMC5529018 DOI: 10.1371/journal.pone.0180319] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/14/2017] [Indexed: 01/09/2023] Open
Abstract
The beta-1 adrenergic receptor (ADRB1) is a promising therapeutic target intrinsically involved in the cognitive deficits and pathological features associated with Alzheimer's disease (AD). Evidence indicates that ADRB1 plays an important role in regulating neuroinflammatory processes, and activation of ADRB1 may produce neuroprotective effects in neuroinflammatory diseases. Novel small molecule modulators of ADRB1, engineered to be highly brain permeable and functionally selective for the G protein with partial agonistic activity, could have tremendous value both as pharmacological tools and potential lead molecules for further preclinical development. The present study describes our ongoing efforts toward the discovery of functionally selective partial agonists of ADRB1 that have potential therapeutic value for AD and neuroinflammatory disorders, which has led to the identification of the molecule STD-101-D1. As a functionally selective agonist of ADRB1, STD-101-D1 produces partial agonistic activity on G protein signaling with an EC50 value in the low nanomolar range, but engages very little beta-arrestin recruitment compared to the unbiased agonist isoproterenol. STD-101-D1 also inhibits the tumor necrosis factor α (TNFα) response induced by lipopolysaccharide (LPS) both in vitro and in vivo, and shows high brain penetration. Other than the therapeutic role, this newly identified, functionally selective, partial agonist of ADRB1 is an invaluable research tool to study mechanisms of G protein-coupled receptor signal transduction.
Collapse
MESH Headings
- Adrenergic beta-1 Receptor Agonists/chemistry
- Adrenergic beta-1 Receptor Agonists/pharmacokinetics
- Adrenergic beta-1 Receptor Agonists/therapeutic use
- Alzheimer Disease/drug therapy
- Alzheimer Disease/metabolism
- Animals
- Brain/metabolism
- CHO Cells
- Cell Line, Tumor
- Cells, Cultured
- Cricetinae
- Cricetulus
- Crystallography, X-Ray
- Drug Discovery
- GTP-Binding Proteins/metabolism
- Humans
- Magnetic Resonance Spectroscopy
- Male
- Mice, Inbred C57BL
- Models, Chemical
- Models, Molecular
- Molecular Structure
- Neurocognitive Disorders/drug therapy
- Neurocognitive Disorders/metabolism
- Permeability
- Phenyl Ethers/chemistry
- Phenyl Ethers/pharmacokinetics
- Phenyl Ethers/therapeutic use
- Propanolamines/chemistry
- Propanolamines/pharmacokinetics
- Propanolamines/therapeutic use
- Protein Binding
- Rats, Sprague-Dawley
- Receptors, Adrenergic, beta-1/chemistry
- Receptors, Adrenergic, beta-1/metabolism
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Bitna Yi
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Alam Jahangir
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Andrew K. Evans
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Denise Briggs
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Kristine Ravina
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Jacqueline Ernest
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Amir B. Farimani
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Wenchao Sun
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Jayakumar Rajadas
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Michael Green
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Evan N. Feinberg
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Vijay S. Pande
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, United States of America
- * E-mail:
| |
Collapse
|
25
|
Noradrenaline induces CX3CL1 production and release by neurons. Neuropharmacology 2017; 114:146-155. [DOI: 10.1016/j.neuropharm.2016.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/29/2016] [Accepted: 12/02/2016] [Indexed: 02/07/2023]
|
26
|
Ardestani PM, Evans AK, Yi B, Nguyen T, Coutellier L, Shamloo M. Modulation of neuroinflammation and pathology in the 5XFAD mouse model of Alzheimer's disease using a biased and selective beta-1 adrenergic receptor partial agonist. Neuropharmacology 2017; 116:371-386. [PMID: 28089846 DOI: 10.1016/j.neuropharm.2017.01.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 12/25/2022]
Abstract
Degeneration of noradrenergic neurons occurs at an early stage of Alzheimer's Disease (AD). The noradrenergic system regulates arousal and learning and memory, and has been implicated in regulating neuroinflammation. Loss of noradrenergic tone may underlie AD progression at many levels. We have previously shown that acute administration of a partial agonist of the beta-1 adrenergic receptor (ADRB1), xamoterol, restores behavioral deficits in a mouse model of AD. The current studies examined the effects of chronic low dose xamoterol on neuroinflammation, pathology, and behavior in the pathologically aggressive 5XFAD transgenic mouse model of AD. In vitro experiments in cells expressing human beta adrenergic receptors demonstrate that xamoterol is highly selective for ADRB1 and functionally biased for the cAMP over the β-arrestin pathway. Data demonstrate ADRB1-mediated attenuation of TNF-α production with xamoterol in primary rat microglia culture following LPS challenge. Finally, two independent cohorts of 5XFAD and control mice were administered xamoterol from approximately 4.0-6.5 or 7.0-9.5 months, were tested in an array of behavioral tasks, and brains were examined for evidence of neuroinflammation, and amyloid beta and tau pathology. Xamoterol reduced mRNA expression of neuroinflammatory markers (Iba1, CD74, CD14 and TGFβ) and immunohistochemical evidence for microgliosis and astrogliosis. Xamoterol reduced amyloid beta and tau pathology as measured by regional immunohistochemistry. Behavioral deficits were not observed for 5XFAD mice. In conclusion, chronic administration of a selective, functionally biased, partial agonist of ADRB1 is effective in reducing neuroinflammation and amyloid beta and tau pathology in the 5XFAD model of AD.
Collapse
Affiliation(s)
- Pooneh Memar Ardestani
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304, USA
| | - Andrew K Evans
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304, USA
| | - Bitna Yi
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304, USA
| | - Tiffany Nguyen
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304, USA
| | - Laurence Coutellier
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304, USA
| | - Mehrdad Shamloo
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304, USA.
| |
Collapse
|
27
|
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: 1.2] [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
|
28
|
Feinstein DL, Kalinin S, Braun D. Causes, consequences, and cures for neuroinflammation mediated via the locus coeruleus: noradrenergic signaling system. J Neurochem 2016; 139 Suppl 2:154-178. [PMID: 26968403 DOI: 10.1111/jnc.13447] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 12/31/2022]
Abstract
Aside from its roles in as a classical neurotransmitter involved in regulation of behavior, noradrenaline (NA) has other functions in the CNS. This includes restricting the development of neuroinflammatory activation, providing neurotrophic support to neurons, and providing neuroprotection against oxidative stress. In recent years, it has become evident that disruption of physiological NA levels or signaling is a contributing factor to a variety of neurological diseases and conditions including Alzheimer's disease (AD) and Multiple Sclerosis. The basis for dysregulation in these diseases is, in many cases, due to damage occurring to noradrenergic neurons present in the locus coeruleus (LC), the major source of NA in the CNS. LC damage is present in AD, multiple sclerosis, and a large number of other diseases and conditions. Studies using animal models have shown that experimentally induced lesion of LC neurons exacerbates neuropathology while treatments to compensate for NA depletion, or to reduce LC neuronal damage, provide benefit. In this review, we will summarize the anti-inflammatory and neuroprotective actions of NA, summarize examples of how LC damage worsens disease, and discuss several approaches taken to treat or prevent reductions in NA levels and LC neuronal damage. Further understanding of these events will be of value for the development of treatments for AD, multiple sclerosis, and other diseases and conditions having a neuroinflammatory component. The classical neurotransmitter noradrenaline (NA) has critical roles in modulating behaviors including those involved in sleep, anxiety, and depression. However, NA can also elicit anti-inflammatory responses in glial cells, can increase neuronal viability by inducing neurotrophic factor expression, and can reduce neuronal damage due to oxidative stress by scavenging free radicals. NA is primarily produced by tyrosine hydroxylase (TH) expressing neurons in the locus coeruleus (LC), a relatively small brainstem nucleus near the IVth ventricle which sends projections throughout the brain and spinal cord. It has been known for close to 50 years that LC neurons are lost during normal aging, and that loss is exacerbated in neurological diseases including Parkinson's disease and Alzheimer's disease. LC neuronal damage and glial activation has now been documented in a variety of other neurological conditions and diseases, however, the causes of LC damage and cell loss remain largely unknown. A number of approaches have been developed to address the loss of NA and increased inflammation associated with LC damage, and several methods are being explored to directly minimize the extent of LC neuronal cell loss or function. In this review, we will summarize some of the consequences of LC loss, consider several factors that likely contribute to that loss, and discuss various ways that have been used to increase NA or to reduce LC damage. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Douglas L Feinstein
- Department of Anesthesiology, University of Illinois, Chicago, IL, USA. .,Jesse Brown VA Medical Center, Chicago, IL, USA.
| | - Sergey Kalinin
- Department of Anesthesiology, University of Illinois, Chicago, IL, USA.,Jesse Brown VA Medical Center, Chicago, IL, USA
| | - David Braun
- Department of Anesthesiology, University of Illinois, Chicago, IL, USA.,Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
29
|
Neuroprotection and reduced gliosis by atomoxetine pretreatment in a gerbil model of transient cerebral ischemia. J Neurol Sci 2015; 359:373-80. [DOI: 10.1016/j.jns.2015.11.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/06/2015] [Accepted: 11/15/2015] [Indexed: 11/23/2022]
|
30
|
Bartus RT, Bétourné A, Basile A, Peterson BL, Glass J, Boulis NM. β2-Adrenoceptor agonists as novel, safe and potentially effective therapies for Amyotrophic lateral sclerosis (ALS). Neurobiol Dis 2015; 85:11-24. [PMID: 26459114 DOI: 10.1016/j.nbd.2015.10.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/25/2015] [Accepted: 10/08/2015] [Indexed: 02/04/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a chronic and progressive neuromuscular disease for which no cure exists and better treatment options are desperately needed. We hypothesize that currently approved β2-adrenoceptor agonists may effectively treat the symptoms and possibly slow the progression of ALS. Although β2-agonists are primarily used to treat asthma, pharmacologic data from animal models of neuromuscular diseases suggest that these agents may have pharmacologic effects of benefit in treating ALS. These include inhibiting protein degradation, stimulating protein synthesis, inducing neurotrophic factor synthesis and release, positively modulating microglial and systemic immune function, maintaining the structural and functional integrity of motor endplates, and improving energy metabolism. Moreover, stimulation of β2-adrenoceptors can activate a range of downstream signaling events in many different cell types that could account for the diverse array of effects of these agents. The evidence supporting the possible therapeutic benefits of β2-agonists is briefly reviewed, followed by a more detailed review of clinical trials testing the efficacy of β-agonists in a variety of human neuromuscular maladies. The weight of evidence of the potential benefits from treating these diseases supports the hypothesis that β2-agonists may be efficacious in ALS. Finally, ways to monitor and manage the side effects that may arise with chronic administration of β2-agonists are evaluated. In sum, effective, safe and orally-active β2-agonists may provide a novel and convenient means to reduce the symptoms of ALS and possibly delay disease progression, affording a unique opportunity to repurpose these approved drugs for treating ALS, and rapidly transforming the management of this serious, unmet medical need.
Collapse
Affiliation(s)
| | | | | | | | - Jonathan Glass
- Dept Neurology and Emory ALS Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Nicholas M Boulis
- Dept Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
31
|
Genestine M, Lin L, Durens M, Yan Y, Jiang Y, Prem S, Bailoor K, Kelly B, Sonsalla PK, Matteson PG, Silverman J, Crawley JN, Millonig JH, DiCicco-Bloom E. Engrailed-2 (En2) deletion produces multiple neurodevelopmental defects in monoamine systems, forebrain structures and neurogenesis and behavior. Hum Mol Genet 2015. [PMID: 26220976 DOI: 10.1093/hmg/ddv301] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Many genes involved in brain development have been associated with human neurodevelopmental disorders, but underlying pathophysiological mechanisms remain undefined. Human genetic and mouse behavioral analyses suggest that ENGRAILED-2 (EN2) contributes to neurodevelopmental disorders, especially autism spectrum disorder. In mouse, En2 exhibits dynamic spatiotemporal expression in embryonic mid-hindbrain regions where monoamine neurons emerge. Considering their importance in neuropsychiatric disorders, we characterized monoamine systems in relation to forebrain neurogenesis in En2-knockout (En2-KO) mice. Transmitter levels of serotonin, dopamine and norepinephrine (NE) were dysregulated from Postnatal day 7 (P7) to P21 in En2-KO, though NE exhibited the greatest abnormalities. While NE levels were reduced ∼35% in forebrain, they were increased 40 -: 75% in hindbrain and cerebellum, and these patterns paralleled changes in locus coeruleus (LC) fiber innervation, respectively. Although En2 promoter was active in Embryonic day 14.5 -: 15.5 LC neurons, expression diminished thereafter and gene deletion did not alter brainstem NE neuron numbers. Significantly, in parallel with reduced NE levels, En2-KO forebrain regions exhibited reduced growth, particularly hippocampus, where P21 dentate gyrus granule neurons were decreased 16%, suggesting abnormal neurogenesis. Indeed, hippocampal neurogenic regions showed increased cell death (+77%) and unexpectedly, increased proliferation. Excess proliferation was restricted to early Sox2/Tbr2 progenitors whereas increased apoptosis occurred in differentiating (Dcx) neuroblasts, accompanied by reduced newborn neuron survival. Abnormal neurogenesis may reflect NE deficits because intra-hippocampal injections of β-adrenergic agonists reversed cell death. These studies suggest that disruption of hindbrain patterning genes can alter monoamine system development and thereby produce forebrain defects that are relevant to human neurodevelopmental disorders.
Collapse
Affiliation(s)
- Matthieu Genestine
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers
| | - Lulu Lin
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, Graduate School of Biological Sciences, Rutgers
| | - Madel Durens
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, Graduate School of Biological Sciences, Rutgers
| | - Yan Yan
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, Graduate School of Biological Sciences, Rutgers
| | - Yiqin Jiang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers
| | - Smrithi Prem
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers
| | - Kunal Bailoor
- Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Brian Kelly
- Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Patricia K Sonsalla
- Department of Neurology, Rutgers Robert Wood Johnson Medical School, Rutgers
| | - Paul G Matteson
- Center for Advanced Biotechnology and Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Jill Silverman
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Jacqueline N Crawley
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - James H Millonig
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, Center for Advanced Biotechnology and Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, Graduate School of Biological Sciences, Rutgers, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA and
| |
Collapse
|
32
|
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: 40] [Impact Index Per Article: 4.0] [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
|
33
|
Braun D, Madrigal JLM, Feinstein DL. Noradrenergic regulation of glial activation: molecular mechanisms and therapeutic implications. Curr Neuropharmacol 2014; 12:342-52. [PMID: 25342942 PMCID: PMC4207074 DOI: 10.2174/1570159x12666140828220938] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 07/12/2014] [Accepted: 07/16/2014] [Indexed: 01/07/2023] Open
Abstract
It has been known for many years that the endogenous neurotransmitter noradrenaline (NA) exerts anti-inflammatory and neuroprotective effects both in vitro and in vivo. In many cases the site of action of NA are beta-adrenergic receptors (βARs), causing an increase in intracellular levels of cAMP which initiates a broad cascade of events including suppression of inflammatory transcription factor activities, alterations in nuclear localization of proteins, and induction of patterns of gene expression mediated through activity of the CREB transcription factor. These changes lead not only to reduced inflammatory events, but also contribute to neuroprotective actions of NA by increasing expression of neurotrophic substances including BDNF, GDNF, and NGF. These properties have prompted studies to determine if treatments with drugs to raise CNS NA levels could provide benefit in various neurological conditions and diseases having an inflammatory component. Moreover, increasing evidence shows that disruptions in endogenous NA levels occurs in several diseases and conditions including Alzheimer's disease (AD), Parkinson's disease (PD), Down's syndrome, posttraumatic stress disorder (PTSD), and multiple sclerosis (MS), suggesting that damage to NA producing neurons is a common factor that contributes to the initiation or progression of neuropathology. Methods to increase NA levels, or to reduce damage to noradrenergic neurons, therefore represent potential preventative as well as therapeutic approaches to disease.
Collapse
Affiliation(s)
- David Braun
- Department of Anesthesiology, University of Illinois at Chicago, Chicago IL, USA, 60612
| | - Jose L M Madrigal
- Departamento de Farmacología, Universidad Complutense de Madrid, Spain
| | - Douglas L Feinstein
- Department of Anesthesiology, University of Illinois at Chicago, Chicago IL, USA, 60612 ; Jesse Brown VA Medical Center, Chicago IL, USA, 60612
| |
Collapse
|
34
|
Potential benefits of therapeutic use of β2-adrenergic receptor agonists in neuroprotection and Parkinsonμs disease. J Immunol Res 2014; 2014:103780. [PMID: 24741572 PMCID: PMC3987873 DOI: 10.1155/2014/103780] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 12/03/2013] [Accepted: 12/04/2013] [Indexed: 11/30/2022] Open
Abstract
The β2-adrenergic receptor (β2AR) is a seven-transmembrane (7TM) G-protein coupled receptor that is expressed on cells of the pulmonary, cardiac, skeletal muscle, and immune systems. Previous work has shown that stimulation of this receptor on immune cells has profound effects on the regulatory activity of both adaptive and innate immune cells. This review examines the functional dichotomy associated with stimulation of β2AR and microglial cells. As well, recent studies targeting these receptors with long-acting agonists are considered with respect to their therapeutic potential in management of Parkinsonμs disease.
Collapse
|
35
|
Hinojosa AE, Caso JR, García-Bueno B, Leza JC, Madrigal JLM. Dual effects of noradrenaline on astroglial production of chemokines and pro-inflammatory mediators. J Neuroinflammation 2013; 10:81. [PMID: 23837880 PMCID: PMC3708781 DOI: 10.1186/1742-2094-10-81] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/29/2013] [Indexed: 11/23/2022] Open
Abstract
Background Noradrenaline (NA) is known to limit neuroinflammation. However, the previously described induction by NA of a chemokine involved in the progression of immune/inflammatory processes, such as chemokine (C-C motif) ligand 2 (CCL2)/monocyte chemotactic protein-1 (MCP-1), apparently contradicts NA anti-inflammatory actions. In the current study we analyzed NA regulation of astroglial chemokine (C-X3-C motif) ligand 1 (CX3CL1), also known as fractalkine, another chemokine to which both neuroprotective and neurodegenerative actions have been attributed. In addition, NA effects on other chemokines and pro-inflammatory mediators were also analyzed. Methods Primary astrocyte-enriched cultures were obtained from neonatal Wistar rats. These cells were incubated for different time durations with combinations of NA and lipopolysaccharide (LPS). The expression and synthesis of different proteins was measured by RT-PCR and enzyme-linked immunosorbent assay (ELISA) or enzyme immunoassays. Data were analyzed by one-way analysis of variance (ANOVA), followed by Newman-Keuls multiple comparison tests. Results The data presented here show that in control conditions, NA induces the production of CX3CL1 in rat cultured astrocytes, but in the presence of an inflammatory stimulus, such as LPS, NA has the opposite effect inhibiting CX3CL1 production. This inversion of NA effect was also observed for MCP-1. Based on the observation of this dual action, NA regulation of different chemokines and pro-inflammatory cytokines was also analyzed, observing that in most cases NA exerts an inhibitory effect in the presence of LPS. One characteristic exception was the induction of cyclooxygenase-2 (COX-2), where a summative effect was detected for both LPS and NA. Conclusion These data suggest that NA effects on astrocytes can adapt to the presence of an inflammatory agent reducing the production of certain cytokines, while in basal conditions NA may have the opposite effect and help to maintain moderate levels of these cytokines.
Collapse
Affiliation(s)
- Ara E Hinojosa
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Investigación Hospital 12 de Octubre, Avenida Complutense s/n, Madrid, Spain
| | | | | | | | | |
Collapse
|
36
|
Gerecke KM, Kolobova A, Allen S, Fawer JL. Exercise protects against chronic restraint stress-induced oxidative stress in the cortex and hippocampus. Brain Res 2013; 1509:66-78. [DOI: 10.1016/j.brainres.2013.02.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 01/12/2013] [Accepted: 02/15/2013] [Indexed: 02/08/2023]
|
37
|
Khanam R, Najfi H, Akhtar M, Vohora D. Evaluation of venlafaxine on glucose homeostasis and oxidative stress in diabetic mice. Hum Exp Toxicol 2012; 31:1244-1250. [PMID: 22751285 DOI: 10.1177/0960327112446840] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Depression occurs frequently with diabetes affecting the quality of life. All major classes of antidepressants have been shown to have a direct pharmacologic effect on metabolic function, which further worsens glycemic control. There were no reports on the effects of venlafaxine on glucose levels and oxidative stress in diabetic animals. The present study evaluated the effects of venlafaxine (8 and 16 mg/kg per d) on glucose homeostasis along with oxidative stress in brain in diabetic mice (streptozotocin (STZ), 40 mg/kg per d for 5 days). We observed that 21 days of administration of venlafaxine (8 and 16 mg/kg per d) in diabetic mice significantly enhanced swimming in normal and STZ-treated mice with a corresponding reduction in immobility. No significant difference in blood glucose levels was observed in diabetic and normal mice following venlafaxine treatment. Venlafaxine (16 mg/kg) reversed STZ-induced elevated thiobarbituric acid reactive substance (TBARS) levels and also restored the glutathione (GSH) levels in diabetic mice. Venlafaxine (8 and 16 mg/kg) per se does not produce any significant effect in normal animals. The results indicate a dose-dependent antidepressant action of venlafaxine in diabetes-induced depressive mice. Furthermore, the blood glucose levels were not significantly altered in normal and diabetic mice. In addition, venlafaxine exhibited a decrease in TBARS and elevation in GSH levels in mice brain. Venlafaxine drug treatment appears to be safer for depression associated with diabetes.
Collapse
Affiliation(s)
- R Khanam
- Department of Pharmacology, Jamia Hamdard University, New Delhi, India.
| | | | | | | |
Collapse
|
38
|
De Paola M, Mariani A, Bigini P, Peviani M, Ferrara G, Molteni M, Gemma S, Veglianese P, Castellaneta V, Boldrin V, Rossetti C, Chiabrando C, Forloni G, Mennini T, Fanelli R. Neuroprotective effects of toll-like receptor 4 antagonism in spinal cord cultures and in a mouse model of motor neuron degeneration. Mol Med 2012; 18:971-81. [PMID: 22562723 DOI: 10.2119/molmed.2012.00020] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 04/30/2012] [Indexed: 12/13/2022] Open
Abstract
Sustained inflammatory reactions are common pathological events associated with neuron loss in neurodegenerative diseases. Reported evidence suggests that Toll-like receptor 4 (TLR4) is a key player of neuroinflammation in several neurodegenerative diseases. However, the mechanisms by which TLR4 mediates neurotoxic signals remain poorly understood. We investigated the role of TLR4 in in vitro and in vivo settings of motor neuron degeneration. Using primary cultures from mouse spinal cords, we characterized both the proinflammatory and neurotoxic effects of TLR4 activation with lipopolysaccharide (activation of microglial cells, release of proinflammatory cytokines and motor neuron death) and the protective effects of a cyanobacteria-derived TLR4 antagonist (VB3323). With the use of TLR4-deficient cells, a critical role of the microglial component with functionally active TLR4 emerged in this setting. The in vivo experiments were carried out in a mouse model of spontaneous motor neuron degeneration, the wobbler mouse, where we preliminarily confirmed a protective effect of TLR4 antagonism. Compared with vehicle- and riluzole-treated mice, those chronically treated with VB3323 showed a decrease in microglial activation and morphological alterations of spinal cord neurons and a better performance in the paw abnormality and grip-strength tests. Taken together, our data add new understanding of the role of TLR4 in mediating neurotoxicity in the spinal cord and suggest that TLR4 antagonists could be considered in future studies as candidate protective agents for motor neurons in degenerative diseases.
Collapse
Affiliation(s)
- Massimiliano De Paola
- Department of Environmental Health Sciences, Mario Negri Institute for Pharmacological Research, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Qin Z, Luo J, VandeVrede L, Tavassoli E, Fa' M, Teich AF, Arancio O, Thatcher GRJ. Design and synthesis of neuroprotective methylthiazoles and modification as NO-chimeras for neurodegenerative therapy. J Med Chem 2012; 55:6784-801. [PMID: 22779770 DOI: 10.1021/jm300353r] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Learning and memory deficits in Alzheimer's disease (AD) result from synaptic failure and neuronal loss, the latter caused in part by excitotoxicity and oxidative stress. A therapeutic approach is described that uses NO-chimeras directed at restoration of both synaptic function and neuroprotection. 4-Methylthiazole (MZ) derivatives were synthesized, based upon a lead neuroprotective pharmacophore acting in part by GABA(A) receptor potentiation. MZ derivatives were assayed for protection of primary neurons against oxygen-glucose deprivation and excitotoxicity. Selected neuroprotective derivatives were incorporated into NO-chimera prodrugs, coined nomethiazoles. To provide proof of concept for the nomethiazole drug class, selected examples were assayed for restoration of synaptic function in hippocampal slices from AD-transgenic mice, reversal of cognitive deficits, and brain bioavailability of the prodrug and its neuroprotective MZ metabolite. Taken together, the assay data suggest that these chimeric nomethiazoles may be of use in treatment of multiple components of neurodegenerative disorders, such as AD.
Collapse
Affiliation(s)
- Zhihui Qin
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612-7231, United States
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Hiraki T, Baker W, Greenberg JH. Effect of vagus nerve stimulation during transient focal cerebral ischemia on chronic outcome in rats. J Neurosci Res 2012; 90:887-94. [PMID: 22420043 DOI: 10.1002/jnr.22812] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The aim of this study was to investigate the effect of vagus nerve stimulation (VNS) on infarct volume and neurological recovery up to 3 weeks following transient focal cerebral ischemia. Transient ischemia was produced by filament occlusion of the proximal middle cerebral artery (MCA) in rats. The right vagus nerve was stimulated starting 30 min after MCA occlusion and consisted of 30-sec pulse trains (20 Hz) delivered to the animal's right vagus nerve every 5 min for a total period of 60 min (n = 10). All the procedures were duplicated, but no stimulus was delivered, in a control group (n = 10). Neurological evaluations were performed in all animals at 24 hr, 48 hr, 1 week, 2 weeks, and 3 weeks after MCA occlusion; animals were euthanized; and neuronal damage was evaluated in hematoxylin-eosin-stained sections. The ischemic lesion volume was smaller in the VNS-treated animals in comparison with the nonstimulated group (P < 0.02). Although the functional score in both treated and untreated groups improved over the 3-week observation period (P < 0.001), there was still a statistically significant improvement reszulting from VNS treatment compared with control animals (P < 0.05). Cerebral blood flow changes in the MCA territory during ischemia did not differ between the VNS-treated animals (31.9% ± 10.4% of baseline) and control animals (29.9% ± 9.1%; P = 0.6). Stimulation of the vagus nerve for only a brief period early in ischemia provides neuroprotection in transient ischemia, with neuroprotection persisting for at least 3 weeks.
Collapse
Affiliation(s)
- Teruyuki Hiraki
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104-6063, USA
| | | | | |
Collapse
|
41
|
Polak PE, Kalinin S, Braun D, Sharp A, Lin SX, Feinstein DL. The vincamine derivative vindeburnol provides benefit in a mouse model of multiple sclerosis: effects on the Locus coeruleus. J Neurochem 2012; 121:206-16. [PMID: 22288774 DOI: 10.1111/j.1471-4159.2012.07673.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The endogenous neurotransmitter noradrenaline (NA) plays several roles in maintaining brain homeostasis, including exerting anti-inflammatory and neuroprotective effects. The primary source of NA in the CNS are tyrosine hydroxylase (TH)-positive neurons located in the Locus coeruleus (LC) which send projections throughout the brain and spinal cord. We recently demonstrated that dysregulation of the LC:Noradrenergic system occurs in experimental autoimmune encephalomyelitis as well as in MS patients, associated with damage occurring to LC neurons. Vindeburnol, a structural analog of the cerebral vasodilator vincamine, was previously reported to increase TH expression and activity in LC neurons. Female C57BL/6 mice were immunized with myelin oligodendrocyte glycoprotein (MOG)(35-55) peptide, and treated with vindeburnol at the first appearance of clinical signs. Clinical signs continued to increase for about 1 week, at which point mice in the vehicle group continued to worsen while vindeburnol-treated mice showed improvement. Pro-inflammatory cytokine production from splenic T cells was not reduced by vindeburnol suggesting primarily central actions of treatment. In the cerebellum, vindeburnol decreased astrocyte activation and reduced the number of demyelinated regions. Vindeburnol reduced astrocyte activation in the LC, reduced TH+ neuronal hypertrophy, increased expression of several genes involved in LC survival and maturation, and increased NA levels in the spinal cord. These results suggest that treatments with drugs such as vindeburnol which target LC survival or function could be of benefit in MS patients.
Collapse
Affiliation(s)
- Paul E Polak
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
42
|
Hinojosa AE, García-Bueno B, Leza JC, Madrigal JLM. Regulation of CCL2/MCP-1 production in astrocytes by desipramine and atomoxetine: involvement of α2 adrenergic receptors. Brain Res Bull 2011; 86:326-33. [PMID: 21963947 DOI: 10.1016/j.brainresbull.2011.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 09/12/2011] [Accepted: 09/19/2011] [Indexed: 01/22/2023]
Abstract
Having previously observed that noradrenaline activation of β adrenergic receptors induces the synthesis of the chemokine monocyte chemoattractant protein (CCL2/MCP-1) in astrocytes, it is our interest to analyze the mechanisms involved in this process, particularly the possible effect of noradrenaline-modulating drugs. The treatment of primary rat astrocyte cultures with the noradrenaline transporter inhibitors desipramine or atomoxetine induced the expression and synthesis of CCL2/MCP-1 in these cells. This effect of both drugs in vitro suggests that CCL2/MCP-1 expression could also be modulated by some mechanism independent of the elevation of brain noradrenaline levels. This was confirmed by measuring a reduction in CCL2/MCP-1 production by the treatment with the α2 adrenergic receptor agonist clonidine. Accordingly, the blockade of α2 adrenergic receptors with yohimbine potentiated the production of MCP-1 stimulated by the activation of β receptors. While the activation of β adrenergic receptors and the subsequent elevation of cAMP levels seem to be the main pathway for noradrenaline to induce CCL2/MCP-1 in astrocytes, our data indicate that the α2 adrenergic receptors also regulate CCL2/MCP-1 expression working as inhibitory mediators.
Collapse
Affiliation(s)
- Ara E Hinojosa
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid and Centro de Investigación Biomédica en Red de Salud Mental, 28040 Madrid, Spain
| | | | | | | |
Collapse
|
43
|
Hinojosa AE, Garcia-Bueno B, Leza JC, Madrigal JLM. CCL2/MCP-1 modulation of microglial activation and proliferation. J Neuroinflammation 2011; 8:77. [PMID: 21729288 PMCID: PMC3146846 DOI: 10.1186/1742-2094-8-77] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 07/05/2011] [Indexed: 01/08/2023] Open
Abstract
Background Monocyte chemoattractant protein (CCL2/MCP-1) is a chemokine that attracts cells involved in the immune/inflammatory response. As microglia are one of the main cell types sustaining inflammation in brain, we proposed here to analyze the direct effects of MCP-1 on cultured primary microglia. Methods Primary microglia and neuronal cultures were obtained from neonatal and embryonic Wistar rats, respectively. Microglia were incubated with different concentrations of recombinant MCP-1 and LPS. Cell proliferation was quantified by measuring incorporation of bromodeoxyuridine (BrdU). Nitrite accumulation was measured using the Griess assay. The expression and synthesis of different proteins was measured by RT-PCR and ELISA. Cell death was quantified by measuring release of LDH into the culture medium. Results MCP-1 treatment (50 ng/ml, 24 h) did not induce morphological changes in microglial cultures. Protein and mRNA levels of different cytokines were measured, showing that MCP-1 was not able to induce proinflammatory cytokines (IL-1β, IL6, MIP-1α), either by itself or in combination with LPS. A similar lack of effect was observed when measuring inducible nitric oxide synthase (NOS2) expression or accumulation of nitrites in the culture media as a different indicator of microglial activation. MCP-1 was also unable to alter the expression of different trophic factors that were reduced by LPS treatment. In order to explore the possible release of other products by microglia and their potential neurotoxicity, neurons were co-cultured with microglia: no death of neurons could be detected when treated with MCP-1. However, the presence of MCP-1 induced proliferation of microglia, an effect opposite to that observed with LPS. Conclusion These data indicate that, while causing migration and proliferation of microglia, MCP-1 does not appear to directly activate an inflammatory response in this cell type, and therefore, other factors may be necessary to cause the changes that result in the neuronal damage commonly observed in situations where MCP-1 levels are elevated.
Collapse
Affiliation(s)
- Ara E Hinojosa
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Avenida Complutense s/n, Madrid, Spain
| | | | | | | |
Collapse
|
44
|
Polak PE, Kalinin S, Feinstein DL. Locus coeruleus damage and noradrenaline reductions in multiple sclerosis and experimental autoimmune encephalomyelitis. Brain 2011; 134:665-77. [PMID: 21297130 DOI: 10.1093/brain/awq362] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The endogenous neurotransmitter noradrenaline exerts anti-inflammatory and neuroprotective effects in vitro and in vivo. Several studies report that noradrenaline levels are altered in the central nervous system of patients with multiple sclerosis and rodents with experimental autoimmune encephalomyelitis, which could contribute to pathology. Since the major source of noradrenaline are neurons in the locus coeruleus, we hypothesized that alterations in noradrenaline levels are a consequence of stress or damage to locus coeruleus neurons. In C57BL/6 mice immunized with myelin oligodendrocyte glycoprotein peptide 35-55 to develop chronic disease, cortical and spinal cord levels of noradrenaline were significantly reduced versus control mice. Immunohistochemical staining revealed increased astrocyte activation in the ventral portion of the locus coeruleus in immunized mice. The immunized mice showed neuronal damage in the locus coeruleus detected by a reduction of average cell size of tyrosine hydroxylase stained neurons. Analysis of the locus coeruleus of multiple sclerosis and control brains showed a significant increase in astrocyte activation, a reduction in noradrenaline levels, and neuronal stress indicated by hypertrophy of tyrosine hydroxylase stained cell bodies. However, the magnitude of these changes was not correlated with extent of demyelination or of cellular infiltrates. Together these findings demonstrate the presence of inflammation and neuronal stress in multiple sclerosis as well as in experimental autoimmune encephalomyelitis. Since reduced noradrenaline levels could be permissive for increased inflammation and neuronal damage, these results suggest that methods to raise noradrenaline levels or increase locus coeruleus function may be of benefit in treating multiple sclerosis.
Collapse
Affiliation(s)
- Paul E Polak
- Department of Anaesthesiology, University of Illinois at Chicago, Jesse Brown Medical Centre, Chicago, IL 60612, USA
| | | | | |
Collapse
|
45
|
Markus T, Hansson SR, Cronberg T, Cilio C, Wieloch T, Ley D. β-Adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices. J Neuroinflammation 2010; 7:94. [PMID: 21172031 PMCID: PMC3017519 DOI: 10.1186/1742-2094-7-94] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 12/20/2010] [Indexed: 01/16/2023] Open
Abstract
Background Inflammation acting in synergy with brain ischemia aggravates perinatal ischemic brain damage. The sensitizing effect of pro-inflammatory exposure prior to hypoxia is dependent on signaling by TNF-α through TNF receptor (TNFR) 1. Adrenoceptor (AR) activation is known to modulate the immune response and synaptic transmission. The possible protective effect of α˜ and β˜AR activation against neuronal damage caused by tissue ischemia and inflammation, acting in concert, was evaluated in murine hippocampal organotypic slices treated with lipopolysaccharide (LPS) and subsequently subjected to oxygen-glucose deprivation (OGD). Method Hippocampal slices from mice were obtained at P6, and were grown in vitro for 9 days on nitrocellulose membranes. Slices were treated with β1(dobutamine)-, β2(terbutaline)-, α1(phenylephrine)- and α2(clonidine)-AR agonists (5 and 50 μM, respectively) during LPS (1 μg/mL, 24 h) -exposure followed by exposure to OGD (15 min) in a hypoxic chamber. Cell death in the slice CA1 region was assessed by propidium iodide staining of dead cells. Results Exposure to LPS + OGD caused extensive cell death from 4 up to 48 h after reoxygenation. Co-incubation with β1-agonist (50 μM) during LPS exposure before OGD conferred complete protection from cell death (P < 0.001) whereas the β2-agonist (50 μM) was partially protective (p < 0.01). Phenylephrine was weakly protective while no protection was attained by clonidine. Exposure to both β1- and β2-agonist during LPS exposure decreased the levels of secreted TNF-α, IL-6 and monocyte chemoattractant protein-1 and prevented microglia activation in the slices. Dobutamine remained neuroprotective in slices exposed to pure OGD as well as in TNFR1-/- and TNFR2-/- slices exposed to LPS followed by OGD. Conclusions Our data demonstrate that activation of both β1- and β2-receptors is neuroprotective and may offer mechanistic insights valuable for development of neuro-protective strategies in neonates.
Collapse
|
46
|
Norepinephrine promotes microglia to uptake and degrade amyloid beta peptide through upregulation of mouse formyl peptide receptor 2 and induction of insulin-degrading enzyme. J Neurosci 2010; 30:11848-57. [PMID: 20810904 DOI: 10.1523/jneurosci.2985-10.2010] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Locus ceruleus (LC) is the main subcortical site of norepinephrine synthesis. In Alzheimer's disease (AD) patients and rodent models, degeneration of LC neurons and reduced levels of norepinephrine in LC projection areas are significantly correlated with the increase in amyloid plaques, neurofibrillary tangles, and severity of dementia. Activated microglia play a pivotal role in the progression of AD by either clearing amyloid beta peptide (Abeta) deposits through uptake of Abeta or releasing cytotoxic substances and proinflammatory cytokines. Here, we investigated the effect of norepinephrine on Abeta uptake and clearance by murine microglia and explored the underlying mechanisms. We found that murine microglia cell line N9 and primary microglia expressed beta(2) adrenergic receptor (AR) but not beta(1) and beta(3)AR. Norepinephrine and isoproterenol upregulated the expression of Abeta receptor mFPR2, a mouse homolog of human formyl peptide receptor FPR2, through activation of beta(2)AR in microglia. Norepinephrine also induced mFPR2 expression in mouse brain. Activation of beta(2)AR in microglia promoted Abeta(42) uptake through upregulation of mFPR2 and enhanced spontaneous cell migration but had no effect on cell migration in response to mFPR2 agonists. Furthermore, activation of beta(2)AR on microglia induced the expression of insulin-degrading enzyme and increased the degradation of Abeta(42). Mechanistic studies showed that isoproterenol induced mFPR2 expression through ERK1/2-NF-kappaB and p38-NF-kappaB signaling pathways. These findings suggest that noradrenergic innervation from LC is needed to maintain adequate Abeta uptake and clearance by microglia, and norepinephrine is a link between neuron and microglia to orchestrate the host response to Abeta in AD.
Collapse
|
47
|
Noradrenaline reuptake inhibitors inhibit expression of chemokines IP-10 and RANTES and cell adhesion molecules VCAM-1 and ICAM-1 in the CNS following a systemic inflammatory challenge. J Neuroimmunol 2010; 220:34-42. [PMID: 20061033 DOI: 10.1016/j.jneuroim.2009.12.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 12/12/2009] [Accepted: 12/14/2009] [Indexed: 11/20/2022]
Abstract
Evidence suggests that noradrenaline has a tonic anti-inflammatory action in the central nervous system (CNS) via its ability to inhibit expression of inflammatory mediators from glial cells. Consequently it is suggested that noradrenaline may play an endogenous neuroprotective role in CNS disorders where inflammatory events contribute to pathology. Infiltration of peripheral immune cells into the brain is driven by increased chemokine and cell adhesion molecule (CAM) expression, and is known to exacerbate neuroinflammation and thereby contribute to the disease process in a number of neurodegenerative disease states. Here we demonstrate that treatment of rats with the noradrenaline reuptake inhibitors (NRIs) desipramine and atomoxetine, agents that increase extracellular noradrenaline in the CNS, suppressed chemokine and cell adhesion molecule (CAM) expression in rat brain following a systemic challenge with bacterial lipopolysaccharide (LPS). Specifically, these agents reduced expression of the chemokines, interferon-inducible protein-10 (IP-10, CXCL-10) and regulated upon activation normal T-cell expressed and secreted (RANTES, CCL-5), and the CAMs, vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule (ICAM-1) in cortex and hippocampus. The inhibitory action of NRIs on chemokines and CAM expression was mimicked by in vitro exposure of cultured glial cells to noradrenaline, but not to the NRIs themselves. These data indicate that the suppressive action of NRIs on chemokine and CAM expression that occurs in vivo is due to increased noradrenaline availability at glial cells, as opposed to a direct action of the drugs on glial cells per se. These results support the theory that noradrenaline has anti-inflammatory properties, and agents that increase noradrenaline availability in vivo can play a role in combating brain inflammation by reducing expression of chemokines and CAMs; molecules that facilitate leucocyte influx into the CNS.
Collapse
|
48
|
Schlachetzki JCM, Fiebich BL, Haake E, de Oliveira ACP, Candelario-Jalil E, Heneka MT, Hüll M. Norepinephrine enhances the LPS-induced expression of COX-2 and secretion of PGE2 in primary rat microglia. J Neuroinflammation 2010; 7:2. [PMID: 20064241 PMCID: PMC2819253 DOI: 10.1186/1742-2094-7-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2009] [Accepted: 01/11/2010] [Indexed: 12/26/2022] Open
Abstract
Background Recent studies suggest an important role for neurotransmitters as modulators of inflammation. Neuroinflammatory mediators such as cytokines and molecules of the arachidonic acid pathway are generated and released by microglia. The monoamine norepinephrine reduces the production of cytokines by activated microglia in vitro. However, little is known about the effects of norepinephrine on prostanoid synthesis. In the present study, we investigate the role of norepinephrine on cyclooxygenase- (COX-)2 expression/synthesis and prostaglandin (PG)E2 production in rat primary microglia. Results Interestingly, norepinephrine increased COX-2 mRNA, but not protein expression. Norepinephrine strongly enhanced COX-2 expression and PGE2 production induced by lipopolysaccharide (LPS). This effect is likely to be mediated by β-adrenoreceptors, since β-, but not α-adrenoreceptor agonists produced similar results. Furthermore, β-adrenoreceptor antagonists blocked the enhancement of COX-2 levels induced by norepinephrine and β-adrenoreceptor agonists. Conclusions Considering that PGE2 displays different roles in neuroinflammatory and neurodegenerative disorders, norepinephrine may play an important function in the modulation of these processes in pathophysiological conditions.
Collapse
|
49
|
McNamee EN, Ryan KM, Kilroy D, Connor TJ. Noradrenaline induces IL-1ra and IL-1 type II receptor expression in primary glial cells and protects against IL-1beta-induced neurotoxicity. Eur J Pharmacol 2009; 626:219-28. [PMID: 19818755 DOI: 10.1016/j.ejphar.2009.09.054] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 09/18/2009] [Accepted: 09/28/2009] [Indexed: 10/20/2022]
Abstract
The pro-inflammatory cytokine interleukin-1beta (IL-1beta) plays a key role in initiating an immune response within the central nervous system (CNS), and is thought to be a significant contributor to the neurodegenerative process. The actions of IL-1beta can be regulated by interleukin-1 receptor antagonist (IL-1ra), which prevents IL-1beta from acting on the IL-1 type I receptor (IL-1RI). Another negative regulator of the IL-1 system is the IL-1 type II receptor (IL-1RII); a decoy receptor that serves to sequester IL-1. Consequently, pharmacological strategies that tip the balance in favour of IL-1ra and IL-1RII may be of therapeutic benefit. Evidence suggests that the neurotransmitter noradrenaline elicits anti-inflammatory actions in the CNS, and consequently may play an endogenous neuroprotective role. Here we report that noradrenaline induces production of IL-1ra and IL-1RII from primary rat mixed glial cells. In contrast, noradrenaline did not alter IL-1beta expression, or expression of IL-1RI or the IL-1 type I receptor accessory protein (IL-1RAcp); both of which are required for IL-1 signalling. Our results demonstrate that the ability of noradrenaline to induce IL-1ra and IL-1RII is mediated via beta-adrenoceptor activation and downstream activation of protein kinase A and extracellular signal-regulated kinase (ERK). In parallel with its ability to increase IL-1ra and IL-1RII, noradrenaline prevented neurotoxicity in cortical primary neurons induced by conditioned medium from IL-1beta treated mixed glial cells. These data indicate that noradrenaline negatively regulates IL-1 system in glial cells and has neuroprotective properties in situations where IL-1 contributes to pathology.
Collapse
Affiliation(s)
- Eoin N McNamee
- Neuroimmunology Research Group, Department of Physiology, School of Medicine & Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | | | | | | |
Collapse
|
50
|
Wang QW, Rowan MJ, Anwyl R. Inhibition of LTP by beta-amyloid is prevented by activation of β2 adrenoceptors and stimulation of the cAMP/PKA signalling pathway. Neurobiol Aging 2009; 30:1608-13. [DOI: 10.1016/j.neurobiolaging.2007.12.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Revised: 12/10/2007] [Accepted: 12/11/2007] [Indexed: 10/22/2022]
|