201
|
Mogali S, Askalsky P, Madera G, Jones JD, Comer SD. Minocycline attenuates oxycodone-induced positive subjective responses in non-dependent, recreational opioid users. Pharmacol Biochem Behav 2021; 209:173241. [PMID: 34298029 DOI: 10.1016/j.pbb.2021.173241] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 01/28/2023]
Abstract
BACKGROUND Recent data suggest that glial cells may be involved in the analgesic effects and abuse liability of opioids. Preclinical studies have demonstrated that mu-opioid-receptor-selective agonists, such as oxycodone, activate glia and increase the release of cytokines, causing a suppression of opioid-induced analgesic effects. Preclinical studies also show that certain medications, such as the broad-spectrum tetracycline antibiotic minocycline, inhibit opioid-induced glial activation and thereby enhance the analgesic effects of opioids. Importantly, minocycline reduces the rewarding effects of opioids at the same doses that it enhances opioid-induced analgesia. AIMS The purpose of the present study was to assess the effects of acute administration of minocycline on the subjective, physiological, and analgesic effects of oxycodone in human research volunteers. DESIGN This study was a within-subject, randomized, double-blind outpatient study. Participants completed five separate sessions in which they received 0, 100, or 200 mg minocycline (MINO) simultaneously with either 0 or 40 mg oxycodone (OXY). The subjective, physiological, and analgesic effects of OXY were measured before and repeatedly after drug administration. SETTINGS AND PARTICIPANTS Participants were between 21 and 45 years of age, non-treatment seeking, non-dependent recreational opioid users (N = 12). This study was conducted between 2013 and 2014 at the New York State Psychiatric Institute in New York, NY. FINDINGS MINO 100 and 200 mg were safe and well-tolerated in combination with OXY 40 mg. MINO 200 mg administered with OXY 40 mg attenuated OXY-induced positive subjective effects such as "Good Effect" and "Liking" compared to OXY alone. MINO did not alter the physiological or analgesic effects of OXY. CONCLUSIONS MINO may attenuate the abuse liability of mu-opioid-receptor-selective agonists.
Collapse
Affiliation(s)
- S Mogali
- Division on Substance Use Disorders, New York State Psychiatric Institute, Department of Psychiatry, Vagelos College of Physicians and Surgeons of Columbia University, 1051 Riverside Dr., Unit 66, New York, NY 10032, United States of America.
| | - P Askalsky
- NYU Langone School of Medicine, Department of Psychiatry, New York, NY 10016, United States of America
| | - G Madera
- Weill Cornell Medical College, 515 East 71st Street, New York, NY 10021, United States of America
| | - J D Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute, Department of Psychiatry, Vagelos College of Physicians and Surgeons of Columbia University, 1051 Riverside Dr., Unit 66, New York, NY 10032, United States of America
| | - S D Comer
- Division on Substance Use Disorders, New York State Psychiatric Institute, Department of Psychiatry, Vagelos College of Physicians and Surgeons of Columbia University, 1051 Riverside Dr., Unit 66, New York, NY 10032, United States of America
| |
Collapse
|
202
|
Mendes MS, Le L, Atlas J, Brehm Z, Ladron-de-Guevara A, Matei E, Lamantia C, McCall MN, Majewska AK. The role of P2Y12 in the kinetics of microglial self-renewal and maturation in the adult visual cortex in vivo. eLife 2021; 10:e61173. [PMID: 34250902 PMCID: PMC8341987 DOI: 10.7554/elife.61173] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 07/10/2021] [Indexed: 12/31/2022] Open
Abstract
Microglia are the brain's resident immune cells with a tremendous capacity to autonomously self-renew. Because microglial self-renewal has largely been studied using static tools, its mechanisms and kinetics are not well understood. Using chronic in vivo two-photon imaging in awake mice, we confirm that cortical microglia show limited turnover and migration under basal conditions. Following depletion, however, microglial repopulation is remarkably rapid and is sustained by the dynamic division of remaining microglia, in a manner that is largely independent of signaling through the P2Y12 receptor. Mathematical modeling of microglial division demonstrates that the observed division rates can account for the rapid repopulation observed in vivo. Additionally, newly born microglia resemble mature microglia within days of repopulation, although morphological maturation is different in newly born microglia in P2Y12 knock out mice. Our work suggests that microglia rapidly locally and that newly born microglia do not recapitulate the slow maturation seen in development but instead take on mature roles in the CNS.
Collapse
Affiliation(s)
- Monique S Mendes
- Department of Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Linh Le
- Department of Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Jason Atlas
- Department of Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Zachary Brehm
- Department of Biostatistics, University of Rochester Medical CenterRochesterUnited States
| | - Antonio Ladron-de-Guevara
- Department of Neuroscience, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Evelyn Matei
- Department of Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Cassandra Lamantia
- Department of Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Matthew N McCall
- Department of Biostatistics, University of Rochester Medical CenterRochesterUnited States
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester Medical CenterRochesterUnited States
- Center for Visual Science, University of RochesterRochesterUnited States
| |
Collapse
|
203
|
El Gaamouch F, Liu K, Lin HY, Wu C, Wang J. Development of grape polyphenols as multi-targeting strategies for Alzheimer's disease. Neurochem Int 2021; 147:105046. [PMID: 33872681 PMCID: PMC8178246 DOI: 10.1016/j.neuint.2021.105046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is by far the most prevalent neurodegenerative disease of aging and is a major burden for patients, caregivers, and the overall health care system. The complexity of AD pathophysiology and the lack of deep understanding of disease mechanisms impeded the development of AD therapy. Currently approved treatments for AD only modestly improve cognitive function but do not modify disease course. The lack of pharmacological approaches has led to the consideration of alternative strategies to prevent or to slow down the progression of AD. There has been a growing interest in the scientific community regarding the impact of diet and nutrition on AD. Grape derived nutraceuticals and phytochemical compounds have demonstrated anti-amyloidogenic, antioxidative, anti-inflammatory and neurotrophic properties and present as potential novel strategies for AD treatment. In this review, we summarize promising grape derived polyphenols that have been shown to modulate AD pathophysiology including amyloid plaques and neurofibrillary tangles formation, AD-induced oxidative stress, neuroinflammation and synaptic dysfunction.
Collapse
Affiliation(s)
- Farida El Gaamouch
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA; Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, 10468, USA
| | - Kalena Liu
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA; Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, 10468, USA
| | - Hsiao-Yun Lin
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA; Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, 10468, USA
| | - Clark Wu
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jun Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA; Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, 10468, USA.
| |
Collapse
|
204
|
Huang Y, Huang W, Yang G, Wang R, Ma L. Design and synthesis of novel diosgenin-triazole hybrids targeting inflammation as potential neuroprotective agents. Bioorg Med Chem Lett 2021; 43:128092. [PMID: 33964436 DOI: 10.1016/j.bmcl.2021.128092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/19/2021] [Accepted: 05/01/2021] [Indexed: 12/09/2022]
Abstract
Alzheimer's disease is a progressive neurodegenerative disease, and its incidence is expected to increase as the global population ages. Recent studies provide increasing evidence that inflammation plays a key role in the pathogenesis and progression of AD. Diosgenin, an active ingredient in Dioscorea nipponica Makino, is a promising bioactive lead compound in the treatment of Alzheimer's disease, which exhibited anti-inflammatory activity. To search for more efficient anti-Alzheimer agents, a series of novel diosgenin-triazolyl hybrids were designed, synthesized, and their neuroprotective effects against oxygen-glucose deprivation-induced neurotoxicity and LPS-induced NO production were evaluated. Most of these new hybrids displayed better activities than DIO. In particular, the promising compound L6 not only demonstrated an excellent neuroprotective effect but also showed the best anti-inflammatory activity. The structure-activity relationship study illustrated that the introduction of benzyl or phenyl triazole did improve the activity, and the introduction of benzyl triazole was better than that of phenyl triazole. The results we obtained showed that the diosgenin skeleton could be a promising structural template for the development of new anti-Alzheimer drug candidates, and compound L6 has the potential to be an important lead compound for further research.
Collapse
Affiliation(s)
- Yi Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weiwei Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Guixiang Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Rui Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
205
|
B Cells in Neuroinflammation: New Perspectives and Mechanistic Insights. Cells 2021; 10:cells10071605. [PMID: 34206848 PMCID: PMC8305155 DOI: 10.3390/cells10071605] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, the role of B cells in neurological disorders has substantially expanded our perspectives on mechanisms of neuroinflammation. The success of B cell-depleting therapies in patients with CNS diseases such as neuromyelitis optica and multiple sclerosis has highlighted the importance of neuroimmune crosstalk in inflammatory processes. While B cells are essential for the adaptive immune system and antibody production, they are also major contributors of pro- and anti-inflammatory cytokine responses in a number of inflammatory diseases. B cells can contribute to neurological diseases through peripheral immune mechanisms, including production of cytokines and antibodies, or through CNS mechanisms following compartmentalization. Emerging evidence suggests that aberrant pro- or anti-inflammatory B cell populations contribute to neurological processes, including glial activation, which has been implicated in the pathogenesis of several neurodegenerative diseases. In this review, we summarize recent findings on B cell involvement in neuroinflammatory diseases and discuss evidence to support pathogenic immunomodulatory functions of B cells in neurological disorders, highlighting the importance of B cell-directed therapies.
Collapse
|
206
|
Qian X, Ji F, Ng KK, Koh AJ, Loo BRY, Townsend MC, Pasternak O, Tay SH, Zhou JH, Mak A. Brain white matter extracellular free-water increases are related to reduced neurocognitive function in systemic lupus erythematosus. Rheumatology (Oxford) 2021; 61:1166-1174. [PMID: 34156469 DOI: 10.1093/rheumatology/keab511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/12/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Brain white matter (WM) microstructural changes evaluated by diffusion MRI were well documented in patients with systemic lupus erythematosus (SLE). Yet, conventional diffusion tensor imaging technique fails to differentiate WM changes that originate from tissue alterations from those due to increased extracellular free water (FW) related to neuroinflammation, microvascular disruption, atrophy, or other extracellular processes. Here, we sought to delineate changes in WM tissue microstructure and extracellular FW volume and examine their relationships with neurocognitive function in SLE patients. METHODS Twenty SLE patients (16 females, aged 36.0±10.6) without clinically-overt neuropsychiatric manifestation and 61 healthy controls (HC) (29 females, aged 29.2±9.4) underwent diffusion MRI and computerized neuropsychological assessments cross-sectionally. The FW imaging method was applied to compare microstructural tissue changes and extracellular FW volume of the brain WM between SLE patients and HC. Association between extracellular FW changes and neurocognitive performance was studied. RESULTS SLE patients had higher WM extracellular FW compared to HC (family-wise-error-corrected p < 0.05) while no group difference was found in FW-corrected tissue compartment and structural connectivity metrics. Extracellular FW increases in SLE patients were associated with poorer neurocognitive performance that probed sustained attention (p = 0.022) and higher cumulative glucocorticoid dose (p = 0.0041). Such findings remained robust after controlling for age, gender, IQ, and total WM volume. CONCLUSIONS The association between WM extracellular FW increases and reduced neurocognitive performance suggest possible microvascular degradation and/or neuroinflammation in SLE patients with clinically-inactive disease. The mechanistic impact of cumulative glucocorticoids on WM FW deserves further evaluation.
Collapse
Affiliation(s)
- Xing Qian
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Fang Ji
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kwun Kei Ng
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Amelia Jialin Koh
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Beatrice Rui Yi Loo
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mary Charlotte Townsend
- Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ofer Pasternak
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sen Hee Tay
- Division of Rheumatology, Department of Medicine, National University Hospital, National University Health System, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore.,Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore, Singapore
| | - Anselm Mak
- Division of Rheumatology, Department of Medicine, National University Hospital, National University Health System, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
207
|
Passaro AP, Lebos AL, Yao Y, Stice SL. Immune Response in Neurological Pathology: Emerging Role of Central and Peripheral Immune Crosstalk. Front Immunol 2021; 12:676621. [PMID: 34177918 PMCID: PMC8222736 DOI: 10.3389/fimmu.2021.676621] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation is a key component of neurological disorders and is an important therapeutic target; however, immunotherapies have been largely unsuccessful. In cases where these therapies have succeeded, particularly multiple sclerosis, they have primarily focused on one aspect of the disease and leave room for improvement. More recently, the impact of the peripheral immune system is being recognized, since it has become evident that the central nervous system is not immune-privileged, as once thought. In this review, we highlight key interactions between central and peripheral immune cells in neurological disorders. While traditional approaches have examined these systems separately, the immune responses and processes in neurological disorders consist of substantial crosstalk between cells of the central and peripheral immune systems. Here, we provide an overview of major immune effector cells and the role of the blood-brain barrier in regard to neurological disorders and provide examples of this crosstalk in various disorders, including stroke and traumatic brain injury, multiple sclerosis, neurodegenerative diseases, and brain cancer. Finally, we propose targeting central-peripheral immune interactions as a potential improved therapeutic strategy to overcome failures in clinical translation.
Collapse
Affiliation(s)
- Austin P. Passaro
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Division of Neuroscience, Biomedical Health and Sciences Institute, University of Georgia, Athens, GA, United States
| | - Abraham L. Lebos
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Department of Biochemistry and Microbiology, University of Georgia, Athens, GA, United States
| | - Yao Yao
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, United States
| | - Steven L. Stice
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Division of Neuroscience, Biomedical Health and Sciences Institute, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, United States
| |
Collapse
|
208
|
Peng Y, Tao H, Wang S, Xiao J, Wang Y, Su H. Dietary intervention with edible medicinal plants and derived products for prevention of Alzheimer's disease: A compendium of time-tested strategy. J Funct Foods 2021; 81:104463. [DOI: 10.1016/j.jff.2021.104463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
209
|
Mendes MS, Majewska AK. An overview of microglia ontogeny and maturation in the homeostatic and pathological brain. Eur J Neurosci 2021; 53:3525-3547. [PMID: 33835613 PMCID: PMC8225243 DOI: 10.1111/ejn.15225] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/17/2021] [Accepted: 04/01/2021] [Indexed: 12/21/2022]
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and are increasingly recognized as critical players in development, brain homeostasis, and disease pathogenesis. The lifespan, maintenance, proliferation, and turnover of microglia are important factors that regulate microglial behavior and affect their roles in the CNS. However, emerging evidence suggests that microglia are morphologically and phenotypically distinct in different brain areas, at different ages, and during disease. Ongoing research focuses on understanding how microglia acquire specific phenotypes in response to extrinsic cues in the environment and how phenotypes are specified by intrinsic properties of different populations of microglia. With the development of pharmacological and genetic tools that allow the investigation of microglia in vivo, there have been considerable advances in understanding molecular signatures of both homeostatic microglia and those reacting to injury and disease. Here, we review the master gene regulators that define microglia as well as discuss the evidence that microglia are heterogeneous and fall into distinct clusters that display specific intrinsic properties and perform unique tasks in different settings. Taken together, the information presented supports the idea that microglia morphology and transcriptional heterogeneity should be considered when studying the complex nature of microglia and their roles in brain health and disease.
Collapse
Affiliation(s)
- Monique S Mendes
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Center for Visual Science, University of Rochester, Rochester, NY, USA
| |
Collapse
|
210
|
Chen X, Drew J, Berney W, Lei W. Neuroprotective Natural Products for Alzheimer's Disease. Cells 2021; 10:1309. [PMID: 34070275 PMCID: PMC8225186 DOI: 10.3390/cells10061309] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/17/2021] [Accepted: 05/22/2021] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is the number one neurovegetative disease, but its treatment options are relatively few and ineffective. In efforts to discover new strategies for AD therapy, natural products have aroused interest in the research community and in the pharmaceutical industry for their neuroprotective activity, targeting different pathological mechanisms associated with AD. A wide variety of natural products from different origins have been evaluated preclinically and clinically for their neuroprotective mechanisms in preventing and attenuating the multifactorial pathologies of AD. This review mainly focuses on the possible neuroprotective mechanisms from natural products that may be beneficial in AD treatment and the natural product mixtures or extracts from different sources that have demonstrated neuroprotective activity in preclinical and/or clinical studies. It is believed that natural product mixtures or extracts containing multiple bioactive compounds that can work additively or synergistically to exhibit multiple neuroprotective mechanisms might be an effective approach in AD drug discovery.
Collapse
Affiliation(s)
- Xin Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA
| | - Joshua Drew
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA
| | - Wren Berney
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA
| | - Wei Lei
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Presbyterian College, Clinton, SC 29325, USA
| |
Collapse
|
211
|
Xu JJ, Guo S, Xue R, Xiao L, Kou JN, Liu YQ, Han JY, Fu JJ, Wei N. Adalimumab ameliorates memory impairments and neuroinflammation in chronic cerebral hypoperfusion rats. Aging (Albany NY) 2021; 13:14001-14014. [PMID: 34030135 PMCID: PMC8202885 DOI: 10.18632/aging.203009] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/05/2021] [Indexed: 01/31/2023]
Abstract
Vascular dementia (VaD) is the second most common type of dementia worldwide. Although there are five FDA-approved drugs for the treatment of Alzheimer's disease (AD), none of them have been applied to treat VaD. Adalimumab is a TNF-α inhibitor that is used for the treatment of autoimmune diseases such as rheumatoid arthritis. In a recent retrospective case-control study, the application of adalimumab for rheumatoid or psoriasis was shown to decrease the risk of AD. However, whether adalimumab can be used for the treatment of VaD is not clear. In this study, we used 2VO surgery to generate a VaD rat model and treated the rats with adalimumab or vehicle. We demonstrated that VaD rats treated with adalimumab exhibited significant improvements in memory. In addition, adalimumab treatment significantly alleviated neuronal loss in the hippocampi of VaD rats. Moreover, adalimumab significantly reduced microglial activation and reversed M1/M2 polarization in VaD rats. Furthermore, adalimumab treatment suppressed the activity of NF-κB, an important neuroinflammatory transcription factor. Finally, adalimumab displayed a protective role against oxidative stress in VaD rats. Our results indicate that adalimumab may be applied for the treatment of human patients with VaD.
Collapse
Affiliation(s)
- Jing-Jing Xu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People’s Republic of China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People’s Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People’s Republic of China
| | - Si Guo
- Department of Medical Laboratory, Henan Provincial People’s Hospital, Zhengzhou, Henan 450003, People’s Republic of China
- Department of Medical Laboratory of Central China Fuwai Hospital, Zhengzhou, Henan 450003, People’s Republic of China
- Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan 450003, People’s Republic of China
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People’s Republic of China
| | - Lin Xiao
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People’s Republic of China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People’s Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People’s Republic of China
| | - Jun-Na Kou
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People’s Republic of China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People’s Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People’s Republic of China
| | - Yu-Qiong Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People’s Republic of China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People’s Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People’s Republic of China
| | - Jun-Ya Han
- Department of Pathology, People’s Hospital of Zhengzhou, Zhengzhou 450000, People’s Republic of China
| | - Jing-Jie Fu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People’s Republic of China
| | - Na Wei
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, People’s Republic of China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou 450002, People’s Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450002, People’s Republic of China
| |
Collapse
|
212
|
Churchill NW, Hutchison MG, Graham SJ, Schweizer TA. Insular Connectivity Is Associated With Self-Appraisal of Cognitive Function After a Concussion. Front Neurol 2021; 12:653442. [PMID: 34093401 PMCID: PMC8175663 DOI: 10.3389/fneur.2021.653442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
Concussion is associated with acute cognitive impairments, with declines in processing speed and reaction time being common. In the clinical setting, these issues are identified via symptom assessments and neurocognitive test (NCT) batteries. Practice guidelines recommend integrating both symptoms and NCTs into clinical decision-making, but correlations between these measures are often poor. This suggests that many patients experience difficulties in the self-appraisal of cognitive issues. It is presently unclear what neural mechanisms give rise to appraisal mismatch after a concussion. One promising target is the insula, which regulates aspects of cognition, particularly interoception and self-monitoring. The present study tested the hypothesis that appraisal mismatch is due to altered functional connectivity of the insula to frontal and midline structures, with hypo-connectivity leading to under-reporting of cognitive issues and hyper-connectivity leading to over-reporting. Data were collected from 59 acutely concussed individuals and 136 normative controls, including symptom assessments, NCTs and magnetic resonance imaging (MRI) data. Analysis of resting-state functional MRI supported the hypothesis, identifying insular networks that were associated with appraisal mismatch in concussed athletes that included frontal, sensorimotor, and cingulate connections. Subsequent analysis of diffusion tensor imaging also determined that symptom over-reporting was associated with reduced fractional anisotropy and increased mean diffusivity of posterior white matter. These findings provide new insights into the mechanisms of cognitive appraisal mismatch after a concussion. They are of particular interest given the central role of symptom assessments in the diagnosis and clinical management of concussion.
Collapse
Affiliation(s)
- Nathan W Churchill
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Neuroscience Research Program, St. Michael's Hospital, Toronto, ON, Canada
| | - Michael G Hutchison
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Simon J Graham
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences Platform, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Tom A Schweizer
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Neuroscience Research Program, St. Michael's Hospital, Toronto, ON, Canada.,Faculty of Medicine (Neurosurgery), University of Toronto, Toronto, ON, Canada.,The Institute of Biomaterials and Biomedical Engineering (IBBME) at the University of Toronto, Toronto, ON, Canada
| |
Collapse
|
213
|
Lu J, Zhou W, Dou F, Wang C, Yu Z. TRPV1 sustains microglial metabolic reprogramming in Alzheimer's disease. EMBO Rep 2021; 22:e52013. [PMID: 33998138 DOI: 10.15252/embr.202052013] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/19/2021] [Accepted: 04/01/2021] [Indexed: 12/31/2022] Open
Abstract
As the brain-resident innate immune cells, reactive microglia are a major pathological feature of Alzheimer's disease (AD). However, the exact role of microglia is still unclear in AD pathogenesis. Here, using metabolic profiling, we show that microglia energy metabolism is significantly suppressed during chronic Aβ-tolerant processes including oxidative phosphorylation and aerobic glycolysis via the mTOR-AKT-HIF-1α pathway. Pharmacological activation of TRPV1 rescues Aβ-tolerant microglial dysfunction, the AKT/mTOR pathway activity, and metabolic impairments and restores the immune responses including phagocytic activity and autophagy function. Amyloid pathology and memory impairment are accelerated in microglia-specific TRPV1-knockout APP/PS1 mice. Finally, we showed that metabolic boosting with TRPV1 agonist decreases amyloid pathology and reverses memory deficits in AD mice model. These results indicate that TRPV1 is an important target regulating metabolic reprogramming for microglial functions in AD treatment.
Collapse
Affiliation(s)
- Jia Lu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Fangfang Dou
- Basic Research Department, Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenfei Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihua Yu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
214
|
Li H, McLaurin KA, Illenberger JM, Mactutus CF, Booze RM. Microglial HIV-1 Expression: Role in HIV-1 Associated Neurocognitive Disorders. Viruses 2021; 13:924. [PMID: 34067600 PMCID: PMC8155894 DOI: 10.3390/v13050924] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 11/16/2022] Open
Abstract
The persistence of HIV-1 viral reservoirs in the brain, despite treatment with combination antiretroviral therapy (cART), remains a critical roadblock for the development of a novel cure strategy for HIV-1. To enhance our understanding of viral reservoirs, two complementary studies were conducted to (1) evaluate the HIV-1 mRNA distribution pattern and major cell type expressing HIV-1 mRNA in the HIV-1 transgenic (Tg) rat, and (2) validate our findings by developing and critically testing a novel biological system to model active HIV-1 infection in the rat. First, a restricted, region-specific HIV-1 mRNA distribution pattern was observed in the HIV-1 Tg rat. Microglia were the predominant cell type expressing HIV-1 mRNA in the HIV-1 Tg rat. Second, we developed and critically tested a novel biological system to model key aspects of HIV-1 by infusing F344/N control rats with chimeric HIV (EcoHIV). In vitro, primary cultured microglia were treated with EcoHIV revealing prominent expression within 24 h of infection. In vivo, EcoHIV expression was observed seven days after stereotaxic injections. Following EcoHIV infection, microglia were the major cell type expressing HIV-1 mRNA, results that are consistent with observations in the HIV-1 Tg rat. Within eight weeks of infection, EcoHIV rats exhibited neurocognitive impairments and synaptic dysfunction, which may result from activation of the NogoA-NgR3/PirB-RhoA signaling pathway and/or neuroinflammation. Collectively, these studies enhance our understanding of HIV-1 viral reservoirs in the brain and offer a novel biological system to model HIV-associated neurocognitive disorders and associated comorbidities (i.e., drug abuse) in rats.
Collapse
Affiliation(s)
| | | | | | | | - Rosemarie M. Booze
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (H.L.); (K.A.M.); (J.M.I.); (C.F.M.)
| |
Collapse
|
215
|
Postolache TT, Wadhawan A, Can A, Lowry CA, Woodbury M, Makkar H, Hoisington AJ, Scott AJ, Potocki E, Benros ME, Stiller JW. Inflammation in Traumatic Brain Injury. J Alzheimers Dis 2021; 74:1-28. [PMID: 32176646 DOI: 10.3233/jad-191150] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is an increasing evidence that inflammation contributes to clinical and functional outcomes in traumatic brain injury (TBI). Many successful target-engaging, lesion-reducing, symptom-alleviating, and function-improving interventions in animal models of TBI have failed to show efficacy in clinical trials. Timing and immunological context are paramount for the direction, quality, and intensity of immune responses to TBI and the resulting neuroanatomical, clinical, and functional course. We present components of the immune system implicated in TBI, potential immune targets, and target-engaging interventions. The main objective of our article is to point toward modifiable molecular and cellular mechanisms that may modify the outcomes in TBI, and contribute to increasing the translational value of interventions that have been identified in animal models of TBI.
Collapse
Affiliation(s)
- Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD, USA
| | - Abhishek Wadhawan
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Saint Elizabeths Hospital, Department of Psychiatry, Washington, DC, USA
| | - Adem Can
- School of Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Christopher A Lowry
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Margaret Woodbury
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Hina Makkar
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrew J Hoisington
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Systems Engineering and Management, Air Force Institute of Technology, Wright-Patterson AFB, OH, USA
| | - Alison J Scott
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Eileen Potocki
- VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Michael E Benros
- Copenhagen Research Center for Mental Health-CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - John W Stiller
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Maryland State Athletic Commission, Baltimore, MD, USA.,Saint Elizabeths Hospital, Neurology Consultation Services, Washington, DC, USA
| |
Collapse
|
216
|
Peters van Ton AM, Duindam HB, van Tuijl J, Li WW, Dieker HJ, Riksen NP, Meijer FA, Kessels RP, Kohn N, van der Hoeven JG, Pickkers P, Rijpkema M, Abdo WF. Neuroinflammation in cognitive decline post-cardiac surgery (the FOCUS study): an observational study protocol. BMJ Open 2021; 11:e044062. [PMID: 33980522 PMCID: PMC8118022 DOI: 10.1136/bmjopen-2020-044062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Postoperative cognitive dysfunction occurs frequently after coronary artery bypass grafting (CABG). The underlying mechanisms remain poorly understood, but neuroinflammation might play a pivotal role. We hypothesise that systemic inflammation induced by the surgical trauma could activate the innate immune (glial) cells of the brain. This could lead to an exaggerated neuroinflammatory cascade, resulting in neuronal dysfunction and loss of neuronal cells. Therefore, the aims of this study are to assess neuroinflammation in vivo presurgery and postsurgery in patients undergoing major cardiac surgery and investigate whether there is a relationship of neuroinflammation to cognitive outcomes, changes to brain structure and function, and systemic inflammation. METHODS AND ANALYSIS The FOCUS study is a prospective, single-centre observational study, including 30 patients undergoing elective on-pump CABG. Translocator protein (TSPO) positron emission tomography neuroimaging will be performed preoperatively and postoperatively using the second generation tracer 18F-DPA-714 to assess the neuroinflammatory response. In addition, a comprehensive cerebral MRI will be performed presurgery and postsurgery, in order to discover newly developed brain and vascular wall lesions. Up to 6 months postoperatively, serial extensive neurocognitive assessments will be performed and blood will be obtained to quantify systemic inflammatory responses and peripheral immune cell activation. ETHICS AND DISSEMINATION Patients do not benefit directly from engaging in the study, but imaging neuroinflammation is considered safe and no side effects are expected. The study protocol obtained ethical approval by the Medical Research Ethics Committee region Arnhem-Nijmegen. This work will be published in peer-reviewed international medical journals and presented at medical conferences. TRIAL REGISTRATION NUMBER NCT04520802.
Collapse
Affiliation(s)
- Annemieke M Peters van Ton
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Harmke B Duindam
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Julia van Tuijl
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Wilson Wl Li
- Department of Cardiothoracic Surgery, Radboud university medical center, Nijmegen, The Netherlands
| | - Hendrik-Jan Dieker
- Department of Cardiology, Radboud university medical center, Nijmegen, The Netherlands
| | - Niels P Riksen
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Fj Anton Meijer
- Department of Medical Imaging, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Roy Pc Kessels
- Department of Medical Psychology, Radboud university medical center, Nijmegen, The Netherlands
- Donders Center for Cognition, Radboud University, Nijmegen, The Netherlands
| | - Nils Kohn
- Donders Institute for Brain, Cognition and Behaviour, Cognitive Neuroscience, Radboud university medical center, Nijmegen, The Netherlands
| | - Johannes G van der Hoeven
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Mark Rijpkema
- Department of Medical Imaging, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Wilson F Abdo
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
217
|
Edler MK, Mhatre-Winters I, Richardson JR. Microglia in Aging and Alzheimer's Disease: A Comparative Species Review. Cells 2021; 10:1138. [PMID: 34066847 PMCID: PMC8150617 DOI: 10.3390/cells10051138] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are the primary immune cells of the central nervous system that help nourish and support neurons, clear debris, and respond to foreign stimuli. Greatly impacted by their environment, microglia go through rapid changes in cell shape, gene expression, and functional behavior during states of infection, trauma, and neurodegeneration. Aging also has a profound effect on microglia, leading to chronic inflammation and an increase in the brain's susceptibility to neurodegenerative processes that occur in Alzheimer's disease. Despite the scientific community's growing knowledge in the field of neuroinflammation, the overall success rate of drug treatment for age-related and neurodegenerative diseases remains incredibly low. Potential reasons for the lack of translation from animal models to the clinic include the use of a single species model, an assumption of similarity in humans, and ignoring contradictory data or information from other species. To aid in the selection of validated and predictive animal models and to bridge the translational gap, this review evaluates similarities and differences among species in microglial activation and density, morphology and phenotype, cytokine expression, phagocytosis, and production of oxidative species in aging and Alzheimer's disease.
Collapse
Affiliation(s)
- Melissa K. Edler
- Department of Anthropology, School of Biomedical Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44240, USA;
| | - Isha Mhatre-Winters
- School of Biomedical Sciences, College of Arts and Sciences, Kent State University, Kent, OH 44240, USA;
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Jason R. Richardson
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
218
|
Bao W, Xie F, Zuo C, Guan Y, Huang YH. PET Neuroimaging of Alzheimer's Disease: Radiotracers and Their Utility in Clinical Research. Front Aging Neurosci 2021; 13:624330. [PMID: 34025386 PMCID: PMC8134674 DOI: 10.3389/fnagi.2021.624330] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's Disease (AD), the leading cause of senile dementia, is a progressive neurodegenerative disorder affecting millions of people worldwide and exerting tremendous socioeconomic burden on all societies. Although definitive diagnosis of AD is often made in the presence of clinical manifestations in late stages, it is now universally believed that AD is a continuum of disease commencing from the preclinical stage with typical neuropathological alterations appearing decades prior to its first symptom, to the prodromal stage with slight symptoms of amnesia (amnestic mild cognitive impairment, aMCI), and then to the terminal stage with extensive loss of basic cognitive functions, i.e., AD-dementia. Positron emission tomography (PET) radiotracers have been developed in a search to meet the increasing clinical need of early detection and treatment monitoring for AD, with reference to the pathophysiological targets in Alzheimer's brain. These include the pathological aggregations of misfolded proteins such as β-amyloid (Aβ) plagues and neurofibrillary tangles (NFTs), impaired neurotransmitter system, neuroinflammation, as well as deficient synaptic vesicles and glucose utilization. In this article we survey the various PET radiotracers available for AD imaging and discuss their clinical applications especially in terms of early detection and cognitive relevance.
Collapse
Affiliation(s)
- Weiqi Bao
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Fang Xie
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Yiyun Henry Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
219
|
Arrona Cardoza P, Spillane MB, Morales Marroquin E. Alzheimer's disease and gut microbiota: does trimethylamine N-oxide (TMAO) play a role? Nutr Rev 2021; 80:271-281. [PMID: 33942080 DOI: 10.1093/nutrit/nuab022] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that affects memory and cognitive function. Clinical evidence has put into question our current understanding of AD development, propelling researchers to look into further avenues. Gut microbiota has emerged as a potential player in AD pathophysiology. Lifestyle factors, such as diet, can have negative effects on the gut microbiota and thus host health. A Western-type diet has been highlighted as a risk factor for both gut microbiota alteration as well as AD development. The gut-derived trimethylamine N-oxide (TMAO) has been previously implied in the development of cardiovascular diseases with recent evidence suggesting a plausible role of TMAO in AD development. Therefore, the main goal of the present review is to provide the reader with potential mechanisms of action through which consumption of a Western-type diet could increase AD risk, by acting through microbiota-produced TMAO. Although a link between TMAO and AD is far from definitive, this review will serve as a call for research into this new area of research.
Collapse
Affiliation(s)
- Pablo Arrona Cardoza
- P. Arrona Cardoza is with the Tecnológico de Monterrey, School of Medicine and Health Science, Monterrey, Nuevo Leon, Mexico. M.B Spillane is with the H.C. Drew School of Health and Human Performance, McNeese State University, Lake Charles, Louisiana, USA. E. Morales Marroquin is with the School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA. E. Morales Marroquin is with the Center for Pediatric Population Health, UTHealth School of Public Health and Children's Health System of Texas, Dallas, Texas, USA
| | - Micheil B Spillane
- P. Arrona Cardoza is with the Tecnológico de Monterrey, School of Medicine and Health Science, Monterrey, Nuevo Leon, Mexico. M.B Spillane is with the H.C. Drew School of Health and Human Performance, McNeese State University, Lake Charles, Louisiana, USA. E. Morales Marroquin is with the School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA. E. Morales Marroquin is with the Center for Pediatric Population Health, UTHealth School of Public Health and Children's Health System of Texas, Dallas, Texas, USA
| | - Elisa Morales Marroquin
- P. Arrona Cardoza is with the Tecnológico de Monterrey, School of Medicine and Health Science, Monterrey, Nuevo Leon, Mexico. M.B Spillane is with the H.C. Drew School of Health and Human Performance, McNeese State University, Lake Charles, Louisiana, USA. E. Morales Marroquin is with the School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA. E. Morales Marroquin is with the Center for Pediatric Population Health, UTHealth School of Public Health and Children's Health System of Texas, Dallas, Texas, USA
| |
Collapse
|
220
|
Kveštak D, Juranić Lisnić V, Lisnić B, Tomac J, Golemac M, Brizić I, Indenbirken D, Cokarić Brdovčak M, Bernardini G, Krstanović F, Rožmanić C, Grundhoff A, Krmpotić A, Britt WJ, Jonjić S. NK/ILC1 cells mediate neuroinflammation and brain pathology following congenital CMV infection. J Exp Med 2021; 218:e20201503. [PMID: 33630019 PMCID: PMC7918636 DOI: 10.1084/jem.20201503] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/11/2020] [Accepted: 01/29/2021] [Indexed: 12/31/2022] Open
Abstract
Congenital human cytomegalovirus (cHCMV) infection of the brain is associated with a wide range of neurocognitive sequelae. Using infection of newborn mice with mouse cytomegalovirus (MCMV) as a reliable model that recapitulates many aspects of cHCMV infection, including disseminated infection, CNS infection, altered neurodevelopment, and sensorineural hearing loss, we have previously shown that mitigation of inflammation prevented alterations in cerebellar development, suggesting that host inflammatory factors are key drivers of neurodevelopmental defects. Here, we show that MCMV infection causes a dramatic increase in the expression of the microglia-derived chemokines CXCL9/CXCL10, which recruit NK and ILC1 cells into the brain in a CXCR3-dependent manner. Surprisingly, brain-infiltrating innate immune cells not only were unable to control virus infection in the brain but also orchestrated pathological inflammatory responses, which lead to delays in cerebellar morphogenesis. Our results identify NK and ILC1 cells as the major mediators of immunopathology in response to virus infection in the developing CNS, which can be prevented by anti-IFN-γ antibodies.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Brain/immunology
- Brain/pathology
- Brain/virology
- Chemokine CXCL10/genetics
- Chemokine CXCL10/immunology
- Chemokine CXCL10/metabolism
- Chemokine CXCL9/genetics
- Chemokine CXCL9/immunology
- Chemokine CXCL9/metabolism
- Cytomegalovirus/immunology
- Cytomegalovirus/physiology
- Cytomegalovirus Infections/immunology
- Cytomegalovirus Infections/virology
- Gene Expression Regulation/immunology
- Humans
- Immunity, Innate/immunology
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/virology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocytes/immunology
- Lymphocytes/metabolism
- Mice, 129 Strain
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Microglia/immunology
- Microglia/metabolism
- Microglia/virology
- Receptors, CXCR3/genetics
- Receptors, CXCR3/immunology
- Receptors, CXCR3/metabolism
- Mice
Collapse
Affiliation(s)
- Daria Kveštak
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Vanda Juranić Lisnić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Berislav Lisnić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jelena Tomac
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Mijo Golemac
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Daniela Indenbirken
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | | | - Giovanni Bernardini
- Department of Molecular Medicine, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, Rome, Italy
| | - Fran Krstanović
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Carmen Rožmanić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Adam Grundhoff
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Astrid Krmpotić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - William J. Britt
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| | - Stipan Jonjić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
221
|
Somaa F. A Review of the Application of Hyperbaric Oxygen Therapy in Alzheimer's Disease. J Alzheimers Dis 2021; 81:1361-1367. [PMID: 33935095 DOI: 10.3233/jad-210157] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is considered as the most common cause of dementia in elderly population. While the exact mechanism of AD has not been discovered, hyperbolic oxygen therapy (HBOT) has been proven to be effective in the treatment of this degenerative disease. The objectives of this article are to review the literature available on molecular and physiological mechanisms underlying HBOT and its efficacy in treating AD and to review the effectiveness of HBOT as an alternate treatment intervention in both human and animal models. 391 full text articles were included in the review after literature search between 1980-2021 from two online data base (ScienceDirect and PubMed). The following key words were used: 'hyperbaric oxygen therapy' and 'Alzheimer disease.' Based on the outcomes of clinical and experimental studies, this review advocates the use of HBOT for the treatment of AD. This review explores future directions and recommends further research into a treatment protocol that will maintain long-term cognitive health of AD patients.
Collapse
Affiliation(s)
- Fahad Somaa
- Occupational Therapy Department, Faculty of Medical Rehabilitation Sciences, King AbdulAziz University, Jeddah, Saudi Arabia
| |
Collapse
|
222
|
Hall S, Deurveilher S, Robertson GS, Semba K. Homeostatic state of microglia in a rat model of chronic sleep restriction. Sleep 2021; 43:5849344. [PMID: 32474610 DOI: 10.1093/sleep/zsaa108] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/13/2020] [Indexed: 12/29/2022] Open
Abstract
Chronic sleep restriction (CSR) negatively impacts brain functions. Whether microglia, the brain's resident immune cells, play any role is unknown. We studied microglia responses to CSR using a rat model featuring slowly rotating wheels (3 h on/1 h off), which was previously shown to induce both homeostatic and adaptive responses in sleep and attention. Adult male rats were sleep restricted for 27 or 99 h. Control rats were housed in locked wheels. After 27 and/or 99 h of CSR, the number of cells immunoreactive for the microglia marker ionized calcium-binding adaptor molecule-1 (Iba1) and the density of Iba1 immunoreactivity were increased in 4/10 brain regions involved in sleep/wake regulation and cognition, including the prelimbic cortex, central amygdala, perifornical lateral hypothalamic area, and dorsal raphe nucleus. CSR neither induced mitosis in microglia (assessed with bromodeoxyuridine) nor impaired blood-brain barrier permeability (assessed with Evans Blue). Microglia appeared ramified in all treatment groups and, when examined quantitatively in the prelimbic cortex, their morphology was not affected by CSR. After 27 h, but not 99 h, of CSR, mRNA levels of the anti-inflammatory cytokine interleukin-10 were increased in the frontal cortex. Pro-inflammatory cytokine mRNA levels (tumor necrosis factor-α, interleukin-1β, and interleukin-6) were unchanged. Furthermore, cortical microglia were not immunoreactive for several pro- and anti-inflammatory markers tested, but were immunoreactive for the purinergic P2Y12 receptor. These results suggest that microglia respond to CSR while remaining in a physiological state and may contribute to the previously reported homeostatic and adaptive responses to CSR.
Collapse
Affiliation(s)
- Shannon Hall
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Samüel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - George S Robertson
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada.,Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada.,Department of Psychiatry, Dalhousie University, Halifax, NS, Canada.,Department of Psychology & Neuroscience, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
223
|
Noshy PA, Azouz RA. Neuroprotective effect of hesperidin against emamectin benzoate-induced neurobehavioral toxicity in rats. Neurotoxicol Teratol 2021; 86:106981. [PMID: 33838246 DOI: 10.1016/j.ntt.2021.106981] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/22/2021] [Accepted: 04/02/2021] [Indexed: 12/13/2022]
Abstract
Emamectin Benzoate (EMB) is an avermectin insecticide widely used in agriculture and veterinary medicine. Hesperidin (HSP) is a flavanone glycoside predominantly found in citrus fruits and has various beneficial health effects. The current research was conducted to study the neurobehavioral toxic effects of EMB in rats and also to evaluate the protective effect of HSP against these toxic effects. Sixty Sprague-Dawley rats were randomly divided into 4 equal groups: control group, EMB group, HSP group, and EMB + HSP group. EMB (8.8. mg/kg) and/or HSP (100 mg/kg) were administered daily by gavage for 8 weeks. The behavioral assessment demonstrated the adverse effects of EMB on the behavioral, motor, and cognitive brain functions. Exposure to EMB also decreased the activity of antioxidants (catalase and reduced glutathione) and increased the malondialdehyde level in nervous tissue. Moreover, EMB increased the level of inflammatory cytokines (tumor necrosis factor-α and interleukin-1β) and decreased brain-derived neurotrophic factor (BDNF) levels in rats' brains. On the other hand, concurrent administration of HSP ameliorated the toxic effects of EMB as indicated by improvements in neural functions and reduction of oxidative stress and inflammation. The study concluded that exposure to EMB induces toxic effects in the brain of rats and that HSP has a protective effect against these toxic effects.
Collapse
Affiliation(s)
- Peter A Noshy
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| | - Rehab A Azouz
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
224
|
Nitsch L, Schneider L, Zimmermann J, Müller M. Microglia-Derived Interleukin 23: A Crucial Cytokine in Alzheimer's Disease? Front Neurol 2021; 12:639353. [PMID: 33897596 PMCID: PMC8058463 DOI: 10.3389/fneur.2021.639353] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/15/2021] [Indexed: 01/26/2023] Open
Abstract
Neuronal cell death, amyloid β plaque formation and development of neurofibrillary tangles are among the characteristics of Alzheimer's disease (AD). In addition to neurodegeneration, inflammatory processes such as activation of microglia and astrocytes are crucial in the pathogenesis and progression of AD. Cytokines are essential immune mediators of the immune response in AD. Recent data suggest a role of interleukin 23 (IL-23) and its p40 subunit in the pathogenesis of AD and corresponding animal models, in particular concerning microglia activation and amyloid β plaque formation. Moreover, in animal models, the injection of anti-p40 antibodies resulted in reduced amyloid β plaque formation and improved cognitive performance. Here, we discuss the pathomechanism of IL-23 mediated inflammation and its role in AD.
Collapse
Affiliation(s)
- Louisa Nitsch
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Linda Schneider
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Marcus Müller
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
225
|
Wang WY, Xie L, Zou XS, Li N, Yang YG, Wu ZJ, Tian XY, Zhao GY, Chen MH. Inhibition of extracellular signal-regulated kinase/calpain-2 pathway reduces neuroinflammation and necroptosis after cerebral ischemia-reperfusion injury in a rat model of cardiac arrest. Int Immunopharmacol 2021; 93:107377. [PMID: 33517223 DOI: 10.1016/j.intimp.2021.107377] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (CIRI) is the leading cause of poor neurological prognosis after cardiopulmonary resuscitation (CPR). We previously reported that the extracellular signal-regulated kinase (ERK) activation mediates CIRI. Here, we explored the potential ERK/calpain-2 pathway role in CIRI using a rat model of cardiac arrest (CA). METHODS Adult male Sprague-Dawley rats suffered from CA/CPR-induced CIRI, received saline, DMSO, PD98059 (ERK1/2 inhibitor, 0.3 mg/kg), or MDL28170 (calpain inhibitor, 3.0 mg/kg) after spontaneous circulation recovery. The survival rate and the neurological deficit score (NDS) were utilized to assess the brain function. Hematoxylin stain, Nissl staining, and transmission electron microscopy were used to evaluate the neuron injury. The expression levels of p-ERK, ERK, calpain-2, neuroinflammation-related markers (GFAP, Iba1, IL-1β, TNF-α), and necroptosis proteins (TNFR1, RIPK1, RIPK3, p-MLKL, and MLKL) in the brain tissues were determined by western blotting and immunohistochemistry. Fluorescent multiplex immunohistochemistry was used to analyze the p-ERK, calpain-2, and RIPK3 co-expression in neurons, and RIPK3 expression levels in microglia or astrocytes. RESULTS At 24 h after CA/CPR, the rats in the saline-treated and DMSO groups presented with injury tissue morphology, low NDS, ERK/calpain-2 pathway activation, and inflammatory cytokine and necroptosis protein over-expression in the brain tissue. After PD98059 and MDL28170 treatment, the brain function was improved, while inflammatory response and necroptosis were suppressed by ERK/calpain-2 pathway inhibition. CONCLUSION Inflammation activation and necroptosis involved in CA/CPR-induced CIRI were regulated by the ERK/calpain-2 signaling pathway. Inhibition of that pathway can reduce neuroinflammation and necroptosis after CIRI in the CA model rats.
Collapse
Affiliation(s)
- Wen-Yan Wang
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, People's Republic of China
| | - Lu Xie
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530000, People's Republic of China
| | - Xin-Sen Zou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, People's Republic of China
| | - Nuo Li
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530000, People's Republic of China
| | - Ye-Gui Yang
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, People's Republic of China
| | - Zhi-Jiang Wu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, People's Republic of China
| | - Xin-Yue Tian
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, People's Republic of China
| | - Gao-Yang Zhao
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, People's Republic of China
| | - Meng-Hua Chen
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, People's Republic of China.
| |
Collapse
|
226
|
Churchill NW, Hutchison MG, Graham SJ, Schweizer TA. Disturbances in Brain Physiology Due to Season Play: A Multi-Sport Study of Male and Female University Athletes. Front Physiol 2021; 12:653603. [PMID: 33868020 PMCID: PMC8044759 DOI: 10.3389/fphys.2021.653603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/01/2021] [Indexed: 11/16/2022] Open
Abstract
High-performance university athletes experience frequent exertion, resulting in disrupted biological homeostasis, but it is unclear to what extent brain physiology is affected. We examined whether athletes without overtraining symptoms show signs of increased neurophysiological stress over the course of a single athletic season, and whether the effects are modified by demographic factors of age, sex and concussion history, and sport-related factors of contact exposure and season length. Fifty-three university-level athletes were recruited from multiple sports at a single institution and followed longitudinally from beginning of season (BOS) to end of season (EOS) and 1 month afterwards, with a subset followed up at the subsequent beginning of season. MRI was used to comprehensively assess white matter (WM) diffusivity, cerebral blood flow (CBF), and brain activity, while overtraining symptoms were assessed with Hooper’s Index (HI). Although athletes did not report increased HI scores, they showed significantly increased white matter diffusivity and decreased CBF at EOS and 1 month afterwards, with recovery at follow-up. Global brain activity was not significantly altered though, highlighting the ability of the brain to adapt to exercise-related stressors. Male athletes had greater white matter diffusivity at EOS, but female athletes had greater declines in CBF at 1 month afterwards. Post-season changes in MRI measures were not related to change in HI score, age, concussion history, contact exposure, or length of athletic season. Hence, the brain shows substantial but reversible neurophysiological changes due to season play in the absence of overtraining symptoms, with effects that are sex-dependent but otherwise insensitive to demographic variations. These findings provide new insights into the effects of training and competitive play on brain health.
Collapse
Affiliation(s)
- Nathan W Churchill
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Neuroscience Research Program, St. Michael's Hospital, Toronto, ON, Canada
| | - Michael G Hutchison
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Simon J Graham
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Center, Toronto, ON, Canada
| | - Tom A Schweizer
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Neuroscience Research Program, St. Michael's Hospital, Toronto, ON, Canada.,Faculty of Medicine (Neurosurgery), University of Toronto, Toronto, ON, Canada.,The Institute of Biomaterials & Biomedical Engineering (IBBME) at the University of Toronto, Toronto, ON, Canada
| |
Collapse
|
227
|
Welcome MO, Mastorakis NE. The taste of neuroinflammation: Molecular mechanisms linking taste sensing to neuroinflammatory responses. Pharmacol Res 2021; 167:105557. [PMID: 33737243 DOI: 10.1016/j.phrs.2021.105557] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
Evidence indicates a critical role of neuroinflammatory response as an underlying pathophysiological process in several central nervous system disorders, including neurodegenerative diseases. However, the molecular mechanisms that trigger neuroinflammatory processes are not fully known. The discovery of bitter taste receptors in regions other than the oral cavity substantially increased research interests on their functional roles in extra-oral tissues. It is now widely accepted that bitter taste receptors, for instance, in the respiratory, intestinal, reproductive and urinary tracts, are crucial not only for sensing poisonous substances, but also, act as immune sentinels, mobilizing defense mechanisms against pathogenic aggression. The relatively recent discovery of bitter taste receptors in the brain has intensified research investigation on the functional implication of cerebral bitter taste receptor expression. Very recent data suggest that responses of bitter taste receptors to neurotoxins and microbial molecules, under normal condition, are necessary to prevent neuroinflammatory reactions. Furthermore, emerging data have revealed that downregulation of key components of the taste receptor signaling cascade leads to increased oxidative stress and inflammasome signaling in neurons that ultimately culminate in neuroinflammation. Nevertheless, the mechanisms that link taste receptor mediated surveillance of the extracellular milieu to neuroinflammatory responses are not completely understood. This review integrates new data on the molecular mechanisms that link bitter taste receptor sensing to neuroinflammatory responses. The role of bitter taste receptor-mediated sensing of toxigenic substances in brain disorders is also discussed. The therapeutic significance of targeting these receptors for potential treatment of neurodegenerative diseases is also highlighted.
Collapse
Affiliation(s)
- Menizibeya O Welcome
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, Abuja, Nigeria.
| | | |
Collapse
|
228
|
Delgado A, Cholevas C, Theoharides TC. Neuroinflammation in Alzheimer's disease and beneficial action of luteolin. Biofactors 2021; 47:207-217. [PMID: 33615581 DOI: 10.1002/biof.1714] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), already the world's most common form of dementia, is projected to continue increasing in prevalence over the next several decades. The current lack of understanding of the pathogenesis of AD has hampered the development of effective treatments. Historically, AD research has been predicated on the amyloid cascade hypothesis (ACH), which attributes disease progression to the build-up of amyloid protein. However, multiple clinical studies of drugs interfering with ACH have failed to show any benefit demonstrating that AD etiology is more complex than previously thought. Here we review the current literature on the emerging key role of neuroinflammation, especially activation of microglia, in AD pathogenesis. Moreover, we provide compelling evidence that certain flavonoids, especially luteolin formulated in olive pomace oil together with hydroxytyrosol, offers a reasonable prophylactic treatment approach due to its many beneficial actions.
Collapse
Affiliation(s)
- Alejandro Delgado
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Biomedical Sciences Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Christos Cholevas
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
- BrainGate, Thessaloniki, Greece
| | - Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Biomedical Sciences Program, Tufts University School of Medicine, Boston, Massachusetts, USA
- BrainGate, Thessaloniki, Greece
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts, USA
- Department of Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
229
|
Marucci G, Ben DD, Lambertucci C, Navia AM, Spinaci A, Volpini R, Buccioni M. Combined Therapy of A 1AR Agonists and A 2AAR Antagonists in Neuroinflammation. Molecules 2021; 26:1188. [PMID: 33672225 PMCID: PMC7926490 DOI: 10.3390/molecules26041188] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's, Parkinson's, and multiple sclerosis are neurodegenerative diseases related by neuronal degeneration and death in specific areas of the central nervous system. These pathologies are associated with neuroinflammation, which is involved in disease progression, and halting this process represents a potential therapeutic strategy. Evidence suggests that microglia function is regulated by A1 and A2A adenosine receptors (AR), which are considered as neuroprotective and neurodegenerative receptors, respectively. The manuscript's aim is to elucidate the role of these receptors in neuroinflammation modulation through potent and selective A1AR agonists (N6-cyclopentyl-2'- or 3'-deoxyadenosine substituted or unsubstituted in 2 position) and A2AAR antagonists (9-ethyl-adenine substituted in 8 and/or in 2 position), synthesized in house, using N13 microglial cells. In addition, the combined therapy of A1AR agonists and A2AAR antagonists to modulate neuroinflammation was evaluated. Results showed that A1AR agonists were able, to varying degrees, to prevent the inflammatory effect induced by cytokine cocktail (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and interferon (IFN)-γ), while A2AAR antagonists showed a good ability to counteract neuroinflammation. Moreover, the effect achieved by combining the two most effective compounds (1 and 6) in doses previously found to be non-effective was greater than the treatment effect of each of the two compounds used separately at maximal dose.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Michela Buccioni
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, 62032 Camerino, MC, Italy; (G.M.); (D.D.B.); (C.L.); (A.M.N.); (A.S.); (R.V.)
| |
Collapse
|
230
|
Sharma S, Tiarks G, Haight J, Bassuk AG. Neuropathophysiological Mechanisms and Treatment Strategies for Post-traumatic Epilepsy. Front Mol Neurosci 2021; 14:612073. [PMID: 33708071 PMCID: PMC7940684 DOI: 10.3389/fnmol.2021.612073] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death in young adults and a risk factor for acquired epilepsy. Severe TBI, after a period of time, causes numerous neuropsychiatric and neurodegenerative problems with varying comorbidities; and brain homeostasis may never be restored. As a consequence of disrupted equilibrium, neuropathological changes such as circuit remodeling, reorganization of neural networks, changes in structural and functional plasticity, predisposition to synchronized activity, and post-translational modification of synaptic proteins may begin to dominate the brain. These pathological changes, over the course of time, contribute to conditions like Alzheimer disease, dementia, anxiety disorders, and post-traumatic epilepsy (PTE). PTE is one of the most common, devastating complications of TBI; and of those affected by a severe TBI, more than 50% develop PTE. The etiopathology and mechanisms of PTE are either unknown or poorly understood, which makes treatment challenging. Although anti-epileptic drugs (AEDs) are used as preventive strategies to manage TBI, control acute seizures and prevent development of PTE, their efficacy in PTE remains controversial. In this review, we discuss novel mechanisms and risk factors underlying PTE. We also discuss dysfunctions of neurovascular unit, cell-specific neuroinflammatory mediators and immune response factors that are vital for epileptogenesis after TBI. Finally, we describe current and novel treatments and management strategies for preventing PTE.
Collapse
Affiliation(s)
- Shaunik Sharma
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Grant Tiarks
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Joseph Haight
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Alexander G Bassuk
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
231
|
Retinal Inflammation, Cell Death and Inherited Retinal Dystrophies. Int J Mol Sci 2021; 22:ijms22042096. [PMID: 33672611 PMCID: PMC7924201 DOI: 10.3390/ijms22042096] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a group of retinal disorders that cause progressive and severe loss of vision because of retinal cell death, mainly photoreceptor cells. IRDs include retinitis pigmentosa (RP), the most common IRD. IRDs present a genetic and clinical heterogeneity that makes it difficult to achieve proper treatment. The progression of IRDs is influenced, among other factors, by the activation of the immune cells (microglia, macrophages, etc.) and the release of inflammatory molecules such as chemokines and cytokines. Upregulation of tumor necrosis factor alpha (TNFα), a pro-inflammatory cytokine, is found in IRDs. This cytokine may influence photoreceptor cell death. Different cell death mechanisms are proposed, including apoptosis, necroptosis, pyroptosis, autophagy, excessive activation of calpains, or parthanatos for photoreceptor cell death. Some of these cell death mechanisms are linked to TNFα upregulation and inflammation. Therapeutic approaches that reduce retinal inflammation have emerged as useful therapies for slowing down the progression of IRDs. We focused this review on the relationship between retinal inflammation and the different cell death mechanisms involved in RP. We also reviewed the main anti-inflammatory therapies for the treatment of IRDs.
Collapse
|
232
|
Sangaran PG, Ibrahim ZA, Chik Z, Mohamed Z, Ahmadiani A. LPS Preconditioning Attenuates Apoptosis Mechanism by Inhibiting NF-κB and Caspase-3 Activity: TLR4 Pre-activation in the Signaling Pathway of LPS-Induced Neuroprotection. Mol Neurobiol 2021; 58:2407-2422. [PMID: 33421016 DOI: 10.1007/s12035-020-02227-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
Neuroinflammation, an inflammatory response within the nervous system, has been shown to be implicated in the progression of various neurodegenerative diseases. Recent in vivo studies showed that lipopolysaccharide (LPS) preconditioning provides neuroprotection by activating Toll-like receptor 4 (TLR4), one of the members for pattern recognition receptor (PRR) family that play critical role in host response to tissue injury, infection, and inflammation. Pre-exposure to low dose of LPS could confer a protective state against cellular apoptosis following subsequent stimulation with LPS at higher concentration, suggesting a role for TLR4 pre-activation in the signaling pathway of LPS-induced neuroprotection. However, the precise molecular mechanism associated with this protective effect is not well understood. In this article, we provide an overall review of the current state of our knowledge about LPS preconditioning in attenuating apoptosis mechanism and conferring neuroprotection via TLR4 signaling pathway.
Collapse
Affiliation(s)
- Pushpa Gandi Sangaran
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Zaridatul Aini Ibrahim
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Zamri Chik
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Abolhassan Ahmadiani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Evin, PO Box 19839-63113, Tehran, Iran.
| |
Collapse
|
233
|
Sobue A, Komine O, Hara Y, Endo F, Mizoguchi H, Watanabe S, Murayama S, Saito T, Saido TC, Sahara N, Higuchi M, Ogi T, Yamanaka K. Microglial gene signature reveals loss of homeostatic microglia associated with neurodegeneration of Alzheimer's disease. Acta Neuropathol Commun 2021; 9:1. [PMID: 33402227 PMCID: PMC7786928 DOI: 10.1186/s40478-020-01099-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/06/2020] [Indexed: 02/08/2023] Open
Abstract
Microglia-mediated neuroinflammation has been implicated in the pathogenesis of Alzheimer’s disease (AD). Although microglia in aging and neurodegenerative disease model mice show a loss of homeostatic phenotype and activation of disease-associated microglia (DAM), a correlation between those phenotypes and the degree of neuronal cell loss has not been clarified. In this study, we performed RNA sequencing of microglia isolated from three representative neurodegenerative mouse models, AppNL-G-F/NL-G-F with amyloid pathology, rTg4510 with tauopathy, and SOD1G93A with motor neuron disease by magnetic activated cell sorting. In parallel, gene expression patterns of the human precuneus with early Alzheimer’s change (n = 11) and control brain (n = 14) were also analyzed by RNA sequencing. We found that a substantial reduction of homeostatic microglial genes in rTg4510 and SOD1G93A microglia, whereas DAM genes were uniformly upregulated in all mouse models. The reduction of homeostatic microglial genes was correlated with the degree of neuronal cell loss. In human precuneus with early AD pathology, reduced expression of genes related to microglia- and oligodendrocyte-specific markers was observed, although the expression of DAM genes was not upregulated. Our results implicate a loss of homeostatic microglial function in the progression of AD and other neurodegenerative diseases. Moreover, analyses of human precuneus also suggest loss of microglia and oligodendrocyte functions induced by early amyloid pathology in human.
Collapse
|
234
|
Graykowski D, Cudaback E. Don't know what you got till it's gone: microglial depletion and neurodegeneration. Neural Regen Res 2021; 16:1921-1927. [PMID: 33642360 PMCID: PMC8343303 DOI: 10.4103/1673-5374.308078] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In the central nervous system, immunologic surveillance and response are carried out, in large part, by microglia. These resident macrophages derive from myeloid precursors in the embryonic yolk sac, migrating to the brain and eventually populating local tissue prior to blood-brain barrier formation. Preserved for the duration of lifespan, microglia serve the host as more than just a central arm of innate immunity, also contributing significantly to the development and maintenance of neurons and neural networks, as well as neuroregeneration. The critical nature of these varied functions makes the characterization of key roles played by microglia in neurodegenerative disorders, especially Alzheimer's disease, of paramount importance. While genetic models and rudimentary pharmacologic approaches for microglial manipulation have greatly improved our understanding of central nervous system health and disease, significant advances in the selective and near complete in vitro and in vivo depletion of microglia for neuroscience application continue to push the boundaries of research. Here we discuss the research efficacy and utility of various microglial depletion strategies, including the highly effective CSF1R inhibitor models, noteworthy insights into the relationship between microglia and neurodegeneration, and the potential for therapeutic repurposing of microglial depletion and repopulation.
Collapse
Affiliation(s)
- David Graykowski
- Department of Health Sciences, DePaul University, Chicago, IL, USA
| | - Eiron Cudaback
- Department of Health Sciences, DePaul University, Chicago, IL, USA
| |
Collapse
|
235
|
Zhao Y, Yi Y, Gu B, Wang H, Ma J, Guo Z. Echinacoside protects adenine-induced uremic rats from sciatic nerve damage by up-regulating α-Klotho. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2021; 21:413-421. [PMID: 34465681 PMCID: PMC8426649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
OBJECTIVES To investigate the therapeutic effect of Echinacoside on uremia-induced sciatic nerve injury and explore the specific molecular mechanism and role of α-Klotho. METHODS SD rats were given continuous gavage of adenine to prepare a uremia-induced sciatic nerve injury model. The model was given either Echinacoside or α-Klotho by gavage. Histopathological changes of kidney and sciatic nerve were detected by H&E staining. The changes of creatinine, urea nitrogen, and urine protein were detected by biochemical detection. The changes of IL-1β and IL-18 were detected by ELISA. Nerve activity-related indicators were detected by biochemical detection. Changes in related mRNA and protein expression were detected by qPCR and western blot. RESULTS Creatinine, urea nitrogen, urine protein, and malondialdehyde (MDA) in the model group were significantly increased and inhibited by Echinacoside and α-Klotho treatment with Echinacoside dose-dependence. Meanwhile, the activities of ATP concentration, potassium adenosine triphosphate (Na+, K+ ATPase), succinate dehydrogenase (SDH), glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) showed opposite trends. CONCLUSIONS Echinacoside can significantly relieve uremia-induced sciatic nerve injury in rats. Its specific molecular mechanism is related to the inhibition of the classical cellular pyroptosis pathway, which is likely achieved by promoting α-Klotho expression.
Collapse
Affiliation(s)
- Yingdan Zhao
- Department of Nephrology, Changhai Hospital of Second Military Medical University, Shanghai, China
| | - Yang Yi
- Department of Nephrology, Jing’ an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Bo Gu
- Department of Nephrology, Jing’ an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Hanqing Wang
- Department of Nephrology, Jing’ an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jun Ma
- Department of Nephrology, Jing’ an District Centre Hospital of Shanghai, Fudan University, Shanghai, China,Jun Ma, Department of Nephrology, Jing’ an District Centre Hospital of Shanghai, Fudan University, Shanghai, 200040, China E-mail:
| | - Zhiyong Guo
- Department of Nephrology, Changhai Hospital of Second Military Medical University, Shanghai, China
| |
Collapse
|
236
|
Zhang W, Tian T, Gong SX, Huang WQ, Zhou QY, Wang AP, Tian Y. Microglia-associated neuroinflammation is a potential therapeutic target for ischemic stroke. Neural Regen Res 2021; 16:6-11. [PMID: 32788440 PMCID: PMC7818879 DOI: 10.4103/1673-5374.286954] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Microglia-associated neuroinflammation plays an important role in the pathophysiology of ischemic stroke. Microglial activation and polarization, and the inflammatory response mediated by these cells play important roles in the development, progression and outcome of brain injury after ischemic stroke. Currently, there is no effective strategy for treating ischemic stroke in clinical practice. Therefore, it is clinically important to study the role and regulation of microglia in stroke. In this review, we discuss the involvement of microglia in the neuroinflammatory process in ischemic stroke, with the aim of providing a better understanding of the relationship between ischemic stroke and microglia.
Collapse
Affiliation(s)
- Wan Zhang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Tian Tian
- Department of Clinical Laboratory, the First Hospital of Changsha, Changsha, Hunan Province, China
| | - Shao-Xin Gong
- Department of Pathology, the First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Wen-Qian Huang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Qin-Yi Zhou
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| |
Collapse
|
237
|
Pilli D, Zou A, Dawes R, Lopez JA, Tea F, Liyanage G, Lee FX, Merheb V, Houston SD, Pillay A, Jones HF, Ramanathan S, Mohammad S, Kelleher AD, Alexander SI, Dale RC, Brilot F. Pro-inflammatory dopamine-2 receptor-specific T cells in paediatric movement and psychiatric disorders. Clin Transl Immunology 2020; 9:e1229. [PMID: 33425355 PMCID: PMC7780098 DOI: 10.1002/cti2.1229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/09/2020] [Accepted: 11/29/2020] [Indexed: 12/13/2022] Open
Abstract
Objectives A dysregulated inflammatory response against the dopamine‐2 receptor (D2R) has been implicated in movement and psychiatric disorders. D2R antibodies were previously reported in a subset of these patients; however, the role of T cells in these disorders remains unknown. Our objective was to identify and characterise pro‐inflammatory D2R‐specific T cells in movement and psychiatric disorders. Methods Blood from paediatric patients with movement and psychiatric disorders of suspected autoimmune and neurodevelopmental aetiology (n = 24) and controls (n = 16) was cultured in vitro with a human D2R peptide library, and D2R‐specific T cells were identified by flow cytometric quantification of CD4+CD25+CD134+ T cells. Cytokine secretion was analysed using a cytometric bead array and ELISA. HLA genotypes were examined in D2R‐specific T‐cell‐positive patients. D2R antibody seropositivity was determined using a flow cytometry live cell‐based assay. Results Three immunodominant regions of D2R, amino acid (aa)121–131, aa171–181 and aa396–416, specifically activated CD4+ T cells in 8/24 patients. Peptides corresponding to these regions were predicted to bind with high affinity to the HLA of the eight positive patients and had also elicited the secretion of pro‐inflammatory cytokines IL‐2, IFN‐ γ, TNF, IL‐6, IL‐17A and IL‐17F. All eight patients were seronegative for D2R antibodies. Conclusion Autoreactive D2R‐specific T cells and a pro‐inflammatory Th1 and Th17 cytokine profile characterise a subset of paediatric patients with movement and psychiatric disorders, further underpinning the theory of immune dysregulation in these disorders. These findings offer new perspectives into the neuroinflammatory mechanisms of movement and psychiatric disorders and can influence patient diagnosis and treatment.
Collapse
Affiliation(s)
- Deepti Pilli
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Alicia Zou
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Ruebena Dawes
- Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Genomic Medicine Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia
| | - Joseph A Lopez
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Fiona Tea
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Ganesha Liyanage
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,School of Medical Sciences Discipline of Applied Medical Science Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Fiona Xz Lee
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia
| | - Vera Merheb
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia
| | - Samuel D Houston
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,School of Biomedical Engineering The University of Sydney Sydney NSW Australia
| | - Aleha Pillay
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia
| | - Hannah F Jones
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Sudarshini Ramanathan
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Shekeeb Mohammad
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | | | - Stephen I Alexander
- Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Centre for Kidney Research Children's Hospital at Westmead Sydney NSW Australia
| | - Russell C Dale
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Brain and Mind Centre The University of Sydney Sydney NSW Australia
| | - Fabienne Brilot
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Discipline of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,School of Medical Sciences Discipline of Applied Medical Science Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Brain and Mind Centre The University of Sydney Sydney NSW Australia
| |
Collapse
|
238
|
Montaser AB, Järvinen J, Löffler S, Huttunen J, Auriola S, Lehtonen M, Jalkanen A, Huttunen KM. L-Type Amino Acid Transporter 1 Enables the Efficient Brain Delivery of Small-Sized Prodrug across the Blood-Brain Barrier and into Human and Mouse Brain Parenchymal Cells. ACS Chem Neurosci 2020; 11:4301-4315. [PMID: 33228353 DOI: 10.1021/acschemneuro.0c00564] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Membrane transporters have long been utilized to improve the oral, hepatic, and renal (re)absorption. In the brain, however, the transporter-mediated drug delivery has not yet been fully achieved due to the complexity of the blood-brain barrier (BBB). Because L-type amino acid transporter 1 (LAT1) is a good candidate to improve the brain delivery, we developed here four novel LAT1-utilizing prodrugs of four nonsteroidal anti-inflammatory drugs. As a result, all the prodrugs were able to cross the BBB and localize into the brain cells. The brain uptake of salicylic acid (SA) was improved five times, not only across the mouse BBB but also into the cultured mouse and human brain cells. The naproxen prodrug was also transported efficiently into the mouse brain achieving less peripheral exposure, but the brain release of naproxen from the prodrug was not improved. Contrarily, the high plasma protein binding of the flurbiprofen prodrug and the premature bioconversion of the ibuprofen prodrug in the mouse blood hindered the efficient brain delivery. Thus, the structure of the parent drug affects the successful brain delivery of the LAT1-utilizing prodrugs, and the small-sized LAT1-utilizing prodrug of SA constituted a successful model to specifically deliver its parent drug across the mouse BBB and into the cultured mouse and human brain cells.
Collapse
Affiliation(s)
- Ahmed B. Montaser
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Juulia Järvinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Susanne Löffler
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Aaro Jalkanen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
239
|
Wang X, Yu H, Wang C, Liu Y, You J, Wang P, Xu G, Shen H, Yao H, Lan X, Zhao R, Wu X, Zhang G. Chronic ethanol exposure induces neuroinflammation in H4 cells through TLR3 / NF-κB pathway and anxiety-like behavior in male C57BL/6 mice. Toxicology 2020; 446:152625. [PMID: 33161052 DOI: 10.1016/j.tox.2020.152625] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/22/2020] [Accepted: 11/01/2020] [Indexed: 02/08/2023]
Abstract
Chronic alcoholism has become a major public health problem. Long-term and excessive drinking can lead to a variety of diseases. Chronic ethanol exposure can induce neuroinflammation and anxiety-like behavior, and this may be induced through the Toll-like receptor 3/nuclear factor-κB (TLR3/NF-κB) pathway. Animal experiments were performed using healthy adult male C57BL/6 N mice given 10 % (m/V) or 20 % ethanol solution as the only choice of drinkable fluid for 60, 90 or 180 d. In cell culture experiments, H4 human glioma cells were treated with 100 mM ethanol for 2 d, with the TLR3 gene silenced by RNAi and NF-κB inhibited by ammonium pyrrolidine dithiocarbamate (PDTC, 10 μM). After treatment with ethanol solution for a specific time, the anxiety-like behavior of the mice was tested using the open field test and the elevated plus maze test. Western blotting was used to detect the expression of TLR3, TLR4, NF-κB, IL-1β, IL-6, and TNF-α in the mouse hippocampus and H4 cells. The expression of IL-1β, IL-6 and TNF-α in the supernatant of cell culture medium was detected by ELISA. The open field test showed a decrease in time spent in the central area, and the elevated plus maze test showed a decrease in activity time in the open arm region. These behavioral tests indicated that ethanol caused anxiety-like behavior in mice. The expression levels of TLR3, TLR4, NF-κB, IL-1β, IL-6, and TNF-α increased after ethanol exposure in both the hippocampus of mice and H4 cells. Silencing of the TLR3 gene by RNAi or inhibition of NF-κB by PDTC attenuated the ethanol-induced increase in the expression of inflammatory factors in H4 cells. These findings indicated that chronic ethanol exposure increases the expression of TLR3 and NF-κB and produces neuroinflammation and anxiety-like behavior in male C57BL/6 mice and that ethanol-induced neuroinflammation can be caused through the TLR3/NF-κB pathway.
Collapse
Affiliation(s)
- Xiaolong Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Hao Yu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Changliang Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China; The People's Procuratorate of Liaoning Province Judicial Authentication Center, Shenyang, Liaoning, 110032, PR China; Collaborative Laboratory of Intelligentized Forensic Science (CLIFS), Shenyang, Liaoning, 110032, PR China
| | - Yang Liu
- The People's Procuratorate of Liaoning Province Judicial Authentication Center, Shenyang, Liaoning, 110032, PR China; Collaborative Laboratory of Intelligentized Forensic Science (CLIFS), Shenyang, Liaoning, 110032, PR China
| | - Jiabin You
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Pengfei Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Guohui Xu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Hui Shen
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Hui Yao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Xinze Lan
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Rui Zhao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Xu Wu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China.
| | - Guohua Zhang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China.
| |
Collapse
|
240
|
Ruffini N, Klingenberg S, Schweiger S, Gerber S. Common Factors in Neurodegeneration: A Meta-Study Revealing Shared Patterns on a Multi-Omics Scale. Cells 2020; 9:E2642. [PMID: 33302607 PMCID: PMC7764447 DOI: 10.3390/cells9122642] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are heterogeneous, progressive diseases with frequently overlapping symptoms characterized by a loss of neurons. Studies have suggested relations between neurodegenerative diseases for many years (e.g., regarding the aggregation of toxic proteins or triggering endogenous cell death pathways). We gathered publicly available genomic, transcriptomic, and proteomic data from 177 studies and more than one million patients to detect shared genetic patterns between the neurodegenerative diseases on three analyzed omics-layers. The results show a remarkably high number of shared differentially expressed genes between the transcriptomic and proteomic levels for all conditions, while showing a significant relation between genomic and proteomic data between AD and PD and AD and ALS. We identified a set of 139 genes being differentially expressed in several transcriptomic experiments of all four diseases. These 139 genes showed overrepresented gene ontology (GO) Terms involved in the development of neurodegeneration, such as response to heat and hypoxia, positive regulation of cytokines and angiogenesis, and RNA catabolic process. Furthermore, the four analyzed neurodegenerative diseases (NDDs) were clustered by their mean direction of regulation throughout all transcriptomic studies for this set of 139 genes, with the closest relation regarding this common gene set seen between AD and HD. GO-Term and pathway analysis of the proteomic overlap led to biological processes (BPs), related to protein folding and humoral immune response. Taken together, we could confirm the existence of many relations between Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis on transcriptomic and proteomic levels by analyzing the pathways and GO-Terms arising in these intersections. The significance of the connection and the striking relation of the results to processes leading to neurodegeneration between the transcriptomic and proteomic data for all four analyzed neurodegenerative diseases showed that exploring many studies simultaneously, including multiple omics-layers of different neurodegenerative diseases simultaneously, holds new relevant insights that do not emerge from analyzing these data separately. Furthermore, the results shed light on processes like the humoral immune response that have previously been described only for certain diseases. Our data therefore suggest human patients with neurodegenerative diseases should be addressed as complex biological systems by integrating multiple underlying data sources.
Collapse
Affiliation(s)
- Nicolas Ruffini
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
- Leibniz Institute for Resilience Research, Leibniz Association, Wallstraße 7, 55122 Mainz, Germany
| | - Susanne Klingenberg
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susann Schweiger
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susanne Gerber
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| |
Collapse
|
241
|
Gao S, Cheng QC, Hu YG, Tan ZZ, Chen L, Liu SW, Kang QY, Wei T. LncRNA AK148321 alleviates neuroinflammation in LPS-stimulated BV2 microglial cell through regulating microRNA-1199-5p/HSPA5 axis. Life Sci 2020; 266:118863. [PMID: 33301806 DOI: 10.1016/j.lfs.2020.118863] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
AIMS Dysregulated long non-coding RNA (lncRNA) expression is closely related to neuroinflammation, leading to multiple neurodegenerative diseases. In this study, we investigated the function and regulation of lncRNA AK148321 in neuroinflammation using an in vitro lipopolysaccharide (LPS)-stimulated BV2 microglial cell system. METHODS Expression of AK148321 was analyzed by qPCR. Inflammatory cytokine expression levels were determined by ELISA assay. The interaction between AK148321, microRNA (miRNA), and its target gene was validated by luciferase reporter assay and RNA immunoprecipitation (RIP). Cell apoptosis was analyzed by Annexin V/PI staining. RESULTS LPS treatment suppressed AK148321 expression in BV2 cells. Overexpression of AK148321 inhibited LPS-induced BV2 microglial cell activation and decreased the expression of inflammatory cytokine TNF-α and IL-1β. AK148321 function as a competing endogenous RNA (ceRNA) by sponging microRNA-1199-5p (MiR-1199-5p). In LPS-stimulated BV2 cells, AK148321 exerted its inhibitory function via negatively modulating miR-1199-5p expression. Moreover, we identified that Heat Shock Protein Family A Member 5 (HSPA5) was a direct target of miR-1199-5p. RIP assay using the anti-Ago2 antibody further validated the relationship among AK148321, miR-1199-5p and HSPA5. The AK148321/miR-1199-5p/HSPA5 axis regulated the neuroinflammation in LPS-induced BV2 microglial cells. Microglial cell culture supernatant from LPS-stimulated, AK148321-overexpressing BV2 cells suppressed the cell apoptosis of mouse hippocampal neuronal cell HT22, while HSPA5 knockdown abrogated the suppression effect. CONCLUSION Our findings suggest that AK148321 alleviates neuroinflammation in LPS-stimulated BV2 microglial cells through miR-1199-5p/HSPA5 axis.
Collapse
Affiliation(s)
- Shan Gao
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Qiao-Chu Cheng
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Ya-Guang Hu
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Zi-Zhu Tan
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Li Chen
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Si-Wei Liu
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Qian-Yan Kang
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Ting Wei
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
242
|
Cserép C, Pósfai B, Dénes Á. Shaping Neuronal Fate: Functional Heterogeneity of Direct Microglia-Neuron Interactions. Neuron 2020; 109:222-240. [PMID: 33271068 DOI: 10.1016/j.neuron.2020.11.007] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/13/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
The functional contribution of microglia to normal brain development, healthy brain function, and neurological disorders is increasingly recognized. However, until recently, the nature of intercellular interactions mediating these effects remained largely unclear. Recent findings show microglia establishing direct contact with different compartments of neurons. Although communication between microglia and neurons involves intermediate cells and soluble factors, direct membrane contacts enable a more precisely regulated, dynamic, and highly effective form of interaction for fine-tuning neuronal responses and fate. Here, we summarize the known ultrastructural, molecular, and functional features of direct microglia-neuron interactions and their roles in brain disease.
Collapse
Affiliation(s)
- Csaba Cserép
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary
| | - Balázs Pósfai
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary; Szentágothai János Doctoral School of Neurosciences, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary.
| |
Collapse
|
243
|
Bjørklund G, Tinkov AA, Hosnedlová B, Kizek R, Ajsuvakova OP, Chirumbolo S, Skalnaya MG, Peana M, Dadar M, El-Ansary A, Qasem H, Adams JB, Aaseth J, Skalny AV. The role of glutathione redox imbalance in autism spectrum disorder: A review. Free Radic Biol Med 2020; 160:149-162. [PMID: 32745763 DOI: 10.1016/j.freeradbiomed.2020.07.017] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/22/2022]
Abstract
The role of glutathione in autism spectrum disorder (ASD) is emerging as a major topic, due to its role in the maintenance of the intracellular redox balance. Several studies have implicated glutathione redox imbalance as a leading factor in ASD, and both ASD and many other neurodevelopmental disorders involve low levels of reduced glutathione (GSH), high levels of oxidized glutathione (GSSG), and abnormalities in the expressions of glutathione-related enzymes in the blood or brain. Glutathione metabolism, through its impact on redox environment or redox-independent mechanisms, interferes with multiple mechanisms involved in ASD pathogenesis. Glutathione-mediated regulation of glutamate receptors [e.g., N-methyl-d-aspartate (NMDA) receptor], as well as the role of glutamate as a substrate for glutathione synthesis, may be involved in the regulation of glutamate excitotoxicity. However, the interaction between glutathione and glutamate in the pathogenesis of brain diseases may vary from synergism to antagonism. Modulation of glutathione is also associated with regulation of redox-sensitive transcription factors nuclear factor kappa B (NF-κB) and activator protein 1 (AP-1) and downstream signaling (proinflammatory cytokines and inducible enzymes), thus providing a significant impact on neuroinflammation. Mitochondrial dysfunction, as well as neuronal apoptosis, may also provide a significant link between glutathione metabolism and ASD. Furthermore, it has been recently highlighted that glutathione can affect and modulate DNA methylation and epigenetics. Review analysis including research studies meeting the required criteria for analysis showed statistically significant differences between the plasma GSH and GSSG levels as well as GSH:GSSG ratio in autistic patients compared with healthy individuals (P = 0.0145, P = 0.0150 and P = 0.0202, respectively). Therefore, the existing data provide a strong background on the role of the glutathione system in ASD pathogenesis. Future research is necessary to investigate the role of glutathione redox signaling in ASD, which could potentially also lead to promising therapeutics.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo I Rana, Norway.
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Božena Hosnedlová
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic; CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno, Czech Republic
| | - Rene Kizek
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic; CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno, Czech Republic; Faculty of Pharmacy with Division of Laboratory Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Olga P Ajsuvakova
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy; CONEM Scientific Secretary, Verona, Italy
| | - Margarita G Skalnaya
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | | | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Afaf El-Ansary
- Medicinal Chemistry Department, King Saud University, Riyadh, Saudi Arabia; Autism Research and Treatment Center, Riyadh, Saudi Arabia; CONEM Saudi Autism Research Group, King Saud University, Riyadh, Saudi Arabia
| | - Hanan Qasem
- Autism Research and Treatment Center, Riyadh, Saudi Arabia; CONEM Saudi Autism Research Group, King Saud University, Riyadh, Saudi Arabia
| | - James B Adams
- School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, USA
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway
| | - Anatoly V Skalny
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| |
Collapse
|
244
|
Kaur N, Chugh H, Sakharkar MK, Dhawan U, Chidambaram SB, Chandra R. Neuroinflammation Mechanisms and Phytotherapeutic Intervention: A Systematic Review. ACS Chem Neurosci 2020; 11:3707-3731. [PMID: 33146995 DOI: 10.1021/acschemneuro.0c00427] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Neuroinflammation is indicated in the pathogenesis of several acute and chronic neurological disorders. Acute lesions in the brain parenchyma induce intense and highly complex neuroinflammatory reactions with similar mechanisms among various disease prototypes. Microglial cells in the CNS sense tissue damage and initiate inflammatory responses. The cellular and humoral constituents of the neuroinflammatory reaction to brain injury contribute significantly to secondary brain damage and neurodegeneration. Inflammatory cascades such as proinflammatory cytokines from invading leukocytes and direct cell-mediated cytotoxicity between lymphocytes and neurons are known to cause "collateral damage" in models of acute brain injury. In addition to degeneration and neuronal cell loss, there are secondary inflammatory mechanisms that modulate neuronal activity and affect neuroinflammation which can even be detected at the behavioral level. Hence, several of health conditions result from these pathogenetic conditions which are underlined by progressive neuronal function loss due to chronic inflammation and oxidative stress. In the first part of this Review, we discuss critical neuroinflammatory mediators and their pathways in detail. In the second part, we review the phytochemicals which are considered as potential therapeutic molecules for treating neurodegenerative diseases with an inflammatory component.
Collapse
Affiliation(s)
- Navrinder Kaur
- Drug Discovery and Development Laboratory, Department of Chemistry, University of Delhi, New Delhi-110007, India
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi-110007, India
| | - Heerak Chugh
- Drug Discovery and Development Laboratory, Department of Chemistry, University of Delhi, New Delhi-110007, India
| | - Meena K. Sakharkar
- College of Pharmacy and Nutrition, University of Sasketchwan, Saskatoon S7N 5E5, Canada
| | - Uma Dhawan
- Department of Biomedical Science, Bhaskaracharya College of Applied Sciences, University of Delhi, New Delhi-110075, India
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), S.S. Nagar, Mysuru-570015, India
- Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research JSS AHER, Mysuru-570015, India
| | - Ramesh Chandra
- Drug Discovery and Development Laboratory, Department of Chemistry, University of Delhi, New Delhi-110007, India
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi-110007, India
| |
Collapse
|
245
|
Zhang M, He Q, Chen G, Li PA. Suppression of NLRP3 Inflammasome, Pyroptosis, and Cell Death by NIM811 in Rotenone-Exposed Cells as an in vitro Model of Parkinson's Disease. NEURODEGENER DIS 2020; 20:73-83. [PMID: 33176317 DOI: 10.1159/000511207] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/28/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by the selective death of dopaminergic neurons in the substantia nigra. Recently, NLRP3 inflammasome and pyroptosis were found to be associated with PD. Cyclosporine A (CsA), an immunosuppressant, reduces neuronal death in PD. However, CsA could hardly pass through the blood-brain barrier (BBB) and high dose is associated with severe side effects and toxicity. N-methyl-4-isoleucine-cyclosporine (NIM811) is a CsA derivate that can pass through the BBB. However, little is known about its effect on PD. OBJECTIVE The objectives of this study were to explore the mechanism of rotenone-induced cell damage and to examine the protective effects of NIM811 on the neurotoxicity of a Parkinson-like in vitro model induced by rotenone. METHODS Murine hippocampal HT22 cells were cultured with the mitochondrial complex I inhibitor rotenone, a widely used pesticide that has been used for many years as a tool to induce a PD model in vitro and in vivo and proven to be reproducible. NIM811 was added to the culture media 3 h prior to the rotenone incubation. Cell viability was determined by resazurin assay, reactive oxygen species (ROS) production by dihydroethidine (DHE), and mitochondrial membrane potential by tetramethyl rhodamine methyl ester (TMRM). TUNEL and caspase-1 immunofluorescent double staining was used to detect pyroptosis. NLRP3, caspase-1, pro-caspase-1, GSDMD, and interleukin-18 (IL-18) were measured using Western blotting after 24 h of rotenone incubation. The reactivity of interleukin-1β (IL-1β) was determined by ELISA. RESULTS Our results demonstrated that rotenone caused more than 40% of cell death, increased ROS production, and reduced mitochondrial membrane potential, while NIM811 reversed these alterations. Immunofluorescent double staining showed that rotenone increased the percentage of caspase-1 and TUNEL double-labelled cells, an indication of pyroptosis, after 24 h of incubation. The protein expression of NLRP3, caspase-1, pro-caspase-1, GSDMD, IL-18, and IL-1β was significantly increased after 24 h of rotenone incubation. NIM811 suppressed rotenone-induced pyroptosis and downregulated the protein expression of NLRP3, caspase-1, pro-caspase-1, GSDMD, IL-1β, and IL-18. CONCLUSION These results provide evidence that rotenone activates the NLRP3 inflammomere and induces pyroptosis. NIM811 protects the cell from rotenone-induced damage and inhibits NLRP3 inflammasome and pyroptosis. NIM811 might serve as a potential therapeutic drug in the treatment of PD.
Collapse
Affiliation(s)
- Minghao Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.,Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, North Carolina, USA.,Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Qingping He
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Guisheng Chen
- Department of Neurology, Neuroscience Center, General Hospital of Ningxia Medical University, Key Laboratory of Craniocerebral Diseases, Yinchuan, China
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, North Carolina, USA,
| |
Collapse
|
246
|
Borsom EM, Lee K, Cope EK. Do the Bugs in Your Gut Eat Your Memories? Relationship between Gut Microbiota and Alzheimer's Disease. Brain Sci 2020; 10:E814. [PMID: 33153085 PMCID: PMC7693835 DOI: 10.3390/brainsci10110814] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023] Open
Abstract
The human microbiota is composed of trillions of microbial cells inhabiting the oral cavity, skin, gastrointestinal (GI) tract, airways, and reproductive organs. The gut microbiota is composed of dynamic communities of microorganisms that communicate bidirectionally with the brain via cytokines, neurotransmitters, hormones, and secondary metabolites, known as the gut microbiota-brain axis. The gut microbiota-brain axis is suspected to be involved in the development of neurological diseases, including Alzheimer's disease (AD), Parkinson's disease, and Autism Spectrum Disorder. AD is an irreversible, neurodegenerative disease of the central nervous system (CNS), characterized by amyloid-β plaques, neurofibrillary tangles, and neuroinflammation. Microglia and astrocytes, the resident immune cells of the CNS, play an integral role in AD development, as neuroinflammation is a driving factor of disease severity. The gut microbiota-brain axis is a novel target for Alzheimer's disease therapeutics to modulate critical neuroimmune and metabolic pathways. Potential therapeutics include probiotics, prebiotics, fecal microbiota transplantation, and dietary intervention. This review summarizes our current understanding of the role of the gut microbiota-brain axis and neuroinflammation in the onset and development of Alzheimer's disease, limitations of current research, and potential for gut microbiota-brain axis targeted therapies.
Collapse
|
247
|
Microglia mediated neuroinflammation in autism spectrum disorder. J Psychiatr Res 2020; 130:167-176. [PMID: 32823050 DOI: 10.1016/j.jpsychires.2020.07.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/09/2020] [Accepted: 07/15/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Although the precise pathophysiologies underlying autism spectrum disorder (ASD) has not yet been fully clarified, growing evidence supports the involvement of neuroinflammation in the pathogenesis of this disorder, with microglia being particular relevance in the pathophysiologic processes. OBJECTIVE The present review aimed to systematically characterize existing literature regarding the role of microglia mediated neuroinflammation in the etiology of ASD. METHODS A systematic search was conducted for records indexed within Pubmed, EMBASE, or Web of Science to identify potentially eligible publications. Study selection and data extraction were performed by two authors, and the discrepancies in each step were settled through discussions. RESULTS A total of 14 studies comprising 1007 subjects met the eligibility criteria for this review, including 8 immunohistochemistry (IHC) studies, 5 genetic analysis studies, and 1 positron emission tomography (PET) studies. Although small in quantity, the included studies collectively pointed to a role of microglia mediated neuroinflammation in the pathogenesis of ASD. CONCLUSION Findings generated from this review consistently supported the involvement of neuroinflammation in the development of ASD, confirmed by the activation of microglia in different brain regions, involving increased cell number or cell density, morphological alterations, and phenotypic shifts.
Collapse
|
248
|
Gao MR, Wang M, Jia YY, Tian DD, Liu A, Wang WJ, Yang L, Chen JY, Yang Q, Liu R, Wu YM. Echinacoside protects dopaminergic neurons by inhibiting NLRP3/Caspase-1/IL-1β signaling pathway in MPTP-induced Parkinson’s disease model. Brain Res Bull 2020; 164:55-64. [DOI: 10.1016/j.brainresbull.2020.08.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/30/2022]
|
249
|
Haspula D, Clark MA. Cannabinoid Receptors: An Update on Cell Signaling, Pathophysiological Roles and Therapeutic Opportunities in Neurological, Cardiovascular, and Inflammatory Diseases. Int J Mol Sci 2020; 21:E7693. [PMID: 33080916 PMCID: PMC7590033 DOI: 10.3390/ijms21207693] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022] Open
Abstract
The identification of the human cannabinoid receptors and their roles in health and disease, has been one of the most significant biochemical and pharmacological advancements to have occurred in the past few decades. In spite of the major strides made in furthering endocannabinoid research, therapeutic exploitation of the endocannabinoid system has often been a challenging task. An impaired endocannabinoid tone often manifests as changes in expression and/or functions of type 1 and/or type 2 cannabinoid receptors. It becomes important to understand how alterations in cannabinoid receptor cellular signaling can lead to disruptions in major physiological and biological functions, as they are often associated with the pathogenesis of several neurological, cardiovascular, metabolic, and inflammatory diseases. This review focusses mostly on the pathophysiological roles of type 1 and type 2 cannabinoid receptors, and it attempts to integrate both cellular and physiological functions of the cannabinoid receptors. Apart from an updated review of pre-clinical and clinical studies, the adequacy/inadequacy of cannabinoid-based therapeutics in various pathological conditions is also highlighted. Finally, alternative strategies to modulate endocannabinoid tone, and future directions are also emphasized.
Collapse
Affiliation(s)
- Dhanush Haspula
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA;
| | - Michelle A. Clark
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| |
Collapse
|
250
|
Bjørklund G, Peana M, Maes M, Dadar M, Severin B. The glutathione system in Parkinson's disease and its progression. Neurosci Biobehav Rev 2020; 120:470-478. [PMID: 33068556 DOI: 10.1016/j.neubiorev.2020.10.004] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Redox dysfunctions and neuro-oxidative stress play a major role in the pathophysiology and progression of Parkinson's disease (PD). Glutathione (GSH) and the reduced/oxidized glutathione (GSH/GSSG) ratio are lowered in oxidative stress conditions and may lead to increased oxidative toxicity. GSH is involved not only in neuro-immune and neuro-oxidative processes, including thiol redox signaling, but also in cell proliferation and differentiation and in the regulation of cell death, including apoptotic pathways. Lowered GSH metabolism and a low GSH/GSSG ratio following oxidative stress are associated with mitochondrial dysfunctions and constitute a critical factor in the neuroinflammatory and neurodegenerative processes accompanying PD. This review provides indirect evidence that GSH redox signaling is associated with the pathophysiology of PD. Nevertheless, it has not been delineated whether GSH redox imbalances are a causative factor in PD or whether PD-associated pathways cause the GSH redox imbalances in PD. The results show that antioxidant approaches, including neuroprotective and anti-neuroinflammatory agents, which neutralize reactive oxygen species, may have therapeutic efficacy in the treatment of PD and its progression.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway.
| | | | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Impact Research Center, Deakin University, Geelong, Australia
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Beatrice Severin
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| |
Collapse
|