51
|
Watad A, McGonagle D, Anis S, Carmeli R, Cohen AD, Tsur AM, Ben-Shabat N, Lidar M, Amital H. TNF inhibitors have a protective role in the risk of dementia in patients with ankylosing spondylitis: Results from a nationwide study. Pharmacol Res 2022; 182:106325. [PMID: 35752359 DOI: 10.1016/j.phrs.2022.106325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/24/2022] [Accepted: 06/20/2022] [Indexed: 10/17/2022]
Abstract
OBJECTIVES Ankylosing spondylitis (AS) is a chronic progressive and debilitating form of arthritis with associated extra-articular features including uveitis, intestinal and lung apical inflammation and psoriasis. Putative associations between AS and neurologic disorders has been relatively overlooked. The purpose of this study is to assess the link between AS and major neurologic disorders and whether treatment with Tumor-Necrosis-Factor inhibitors (TNFi) has an impact on that association. METHODS A retrospective cross-sectional study was carried out based on the Clalit Health Services (CHS) computerized database. AS patients were compared to age- and gender-matched controls with respect to the proportion of Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, and multiple sclerosis (MS). The impact of AS therapy (biologic vs conventional therapy) was assessed as well. RESULTS 4,082 AS patients and 20,397 age- and gender-matched controls were identified. AS was associated with a higher prevalence of AD (odds-ratio(OR) 1.46 [95%Confidence-interval(CI) 1.13-1.87], p=0.003), epilepsy (OR 2.33 [95%CI 1.75-3.09] p<0.0001) and PD (OR 2.75 [95%CI 2.04-3.72], p<0.0001), whereas no statistically significant association was found for MS. Association with PD remained significant in the multivariate analysis (OR 1.49 [95%CI 1.05- 2.13],p=0.027). Within AS patients, the use of TNFi (OR 0.10 [95%CI 0.01-0.74], p=0.024) were associated with a lowered risk of developing AD. CONCLUSION AS is positively associated with AD, PD, and epilepsy but not MS. AS patients treated with TNFi have lower rates of AD.
Collapse
Affiliation(s)
- Abdulla Watad
- Department of Medicine 'B', Sheba Medical Centre, Tel-Hashomer, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Centre, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Israel; Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Institute of Molecular Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK.
| | - Dennis McGonagle
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Institute of Molecular Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
| | - Saar Anis
- Department of Neurology, Sheba Medical Center, Ramat Gan, Israel
| | - Reut Carmeli
- Department of Medicine 'B', Sheba Medical Centre, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Arnon D Cohen
- Chief Physician's Office, Clalit Health Services, Tel Aviv, Israel; Siaal Research Center for Family Medicine and Primary Care, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Avishai M Tsur
- Department of Medicine 'B', Sheba Medical Centre, Tel-Hashomer, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Centre, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Israel; Israel Defense Forces, Medical Corps, Tel Hashomer, Ramat Gan, Israel
| | - Niv Ben-Shabat
- Department of Medicine 'B', Sheba Medical Centre, Tel-Hashomer, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Centre, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Merav Lidar
- Rheumatology unit, Sheba Medical Centre, Ramat Gan, Israel
| | - Howard Amital
- Department of Medicine 'B', Sheba Medical Centre, Tel-Hashomer, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Centre, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Israel
| |
Collapse
|
52
|
Navarro-Hortal MD, Romero-Márquez JM, Osta S, Jiménez-Trigo V, Muñoz-Ollero P, Varela-López A. Natural Bioactive Products and Alzheimer’s Disease Pathology: Lessons from Caenorhabditis elegans Transgenic Models. Diseases 2022; 10:diseases10020028. [PMID: 35645249 PMCID: PMC9149938 DOI: 10.3390/diseases10020028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is an age-dependent, progressive disorder affecting millions of people. Currently, the therapeutics for AD only treat the symptoms. Although they have been used to discover new products of interest for this disease, mammalian models used to investigate the molecular determinants of this disease are often prohibitively expensive, time-consuming and very complex. On the other hand, cell cultures lack the organism complexity involved in AD. Given the highly conserved neurological pathways between mammals and invertebrates, Caenorhabditis elegans has emerged as a powerful tool for the investigation of the pathophysiology of human AD. Numerous models of both Tau- and Aβ-induced toxicity, the two prime components observed to correlate with AD pathology and the ease of performing RNA interference for any gene in the C. elegans genome, allow for the identification of multiple therapeutic targets. The effects of many natural products in main AD hallmarks using these models suggest promising health-promoting effects. However, the way in which they exert such effects is not entirely clear. One of the reasons is that various possible therapeutic targets have not been evaluated in many studies. The present review aims to explore shared therapeutical targets and the potential of each of them for AD treatment or prevention.
Collapse
|
53
|
Gnanaraj C, Sekar M, Fuloria S, Swain SS, Gan SH, Chidambaram K, Rani NNIM, Balan T, Stephenie S, Lum PT, Jeyabalan S, Begum MY, Chandramohan V, Thangavelu L, Subramaniyan V, Fuloria NK. In Silico Molecular Docking Analysis of Karanjin against Alzheimer's and Parkinson's Diseases as a Potential Natural Lead Molecule for New Drug Design, Development and Therapy. Molecules 2022; 27:2834. [PMID: 35566187 PMCID: PMC9100660 DOI: 10.3390/molecules27092834] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) and Alzheimer's disease (AD) are neurodegenerative disorders that have emerged as among the serious health problems of the 21st century. The medications currently available to treat AD and PD have limited efficacy and are associated with side effects. Natural products are one of the most vital and conservative sources of medicines for treating neurological problems. Karanjin is a furanoflavonoid, isolated mainly from Pongamia pinnata with several medicinal plants, and has been reported for numerous health benefits. However, the effect of karanjin on AD and PD has not yet been systematically investigated. To evaluate the neuroprotective effect of karanjin, extensive in silico studies starting with molecular docking against five putative targets for AD and four targets for PD were conducted. The findings were compared with three standard drugs using Auto Dock 4.1 and Molegro Virtual Docker software. Additionally, the physiochemical properties (Lipinski rule of five), drug-likeness and parameters including absorption, distribution, metabolism, elimination and toxicity (ADMET) profiles of karanjin were also studied. The molecular dynamics (MD) simulations were performed with two selective karanjin docking complexes to analyze the dynamic behaviors and binding free energy at 100 ns time scale. In addition, frontier molecular orbitals (FMOs) and density-functional theory (DFT) were also investigated from computational quantum mechanism perspectives using the Avogadro-ORCA 1.2.0 platform. Karanjin complies with all five of Lipinski's drug-likeness rules with suitable ADMET profiles for therapeutic use. The docking scores (kcal/mol) showed comparatively higher potency against AD and PD associated targets than currently used standard drugs. Overall, the potential binding affinity from molecular docking, static thermodynamics feature from MD-simulation and other multiparametric drug-ability profiles suggest that karanjin could be considered as a suitable therapeutic lead for AD and PD treatment. Furthermore, the present results were strongly correlated with the earlier study on karanjin in an Alzheimer's animal model. However, necessary in vivo studies, clinical trials, bioavailability, permeability and safe dose administration, etc. must be required to use karanjin as a potential drug against AD and PD treatment, where the in silico results are more helpful to accelerate the drug development.
Collapse
Affiliation(s)
- Charles Gnanaraj
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh 30450, Malaysia; (C.G.); (N.N.I.M.R.); (T.B.)
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh 30450, Malaysia;
| | - Shivkanya Fuloria
- Faculty of Pharmacy, Centre of Excellence for Biomaterials Engineering, AIMST University, Bedong 08100, Malaysia
| | - Shasank S. Swain
- Division of Microbiology and NCDs, ICMR-Regional Medical Research Centre, Bhubaneswar 751023, India;
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Bandar Sunway 47500, Malaysia;
| | - Kumarappan Chidambaram
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia;
| | - Nur Najihah Izzati Mat Rani
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh 30450, Malaysia; (C.G.); (N.N.I.M.R.); (T.B.)
| | - Tavamani Balan
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh 30450, Malaysia; (C.G.); (N.N.I.M.R.); (T.B.)
| | - Sarah Stephenie
- School of Biological Sciences, Faculty of Science and Technology, Quest International University Perak, Jalan Raja Permaisuri Bainun, Ipoh 30250, Malaysia;
| | - Pei Teng Lum
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh 30450, Malaysia;
| | - Srikanth Jeyabalan
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (DU), Porur, Chennai 600116, India;
| | - M. Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia;
| | - Vivek Chandramohan
- Department of Biotechnology, Siddaganga Institute of Technology, Tumakuru 572103, India;
| | - Lakshmi Thangavelu
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India;
| | - Vetriselvan Subramaniyan
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jalan SP 2, Bandar Saujana Putra, Jenjarom 42610, Malaysia;
| | - Neeraj Kumar Fuloria
- Faculty of Pharmacy, Centre of Excellence for Biomaterials Engineering, AIMST University, Bedong 08100, Malaysia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India;
| |
Collapse
|
54
|
Coupled Neural–Glial Dynamics and the Role of Astrocytes in Alzheimer’s Disease. MATHEMATICAL AND COMPUTATIONAL APPLICATIONS 2022. [DOI: 10.3390/mca27030033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Neurodegenerative diseases such as Alzheimer’s (AD) are associated with the propagation and aggregation of toxic proteins. In the case of AD, it was Alzheimer himself who showed the importance of both amyloid beta (Aβ) plaques and tau protein neurofibrillary tangles (NFTs) in what he called the “disease of forgetfulness”. The amyloid beta forms extracellular aggregates and plaques, whereas tau proteins are intracellular proteins that stabilize axons by cross-linking microtubules that can form largely messy tangles. On the other hand, astrocytes and microglial cells constantly clear these plaques and NFTs from the brain. Astrocytes transport nutrients from the blood to neurons. Activated astrocytes produce monocyte chemoattractant protein-1 (MCP-1), which attracts anti-inflammatory macrophages and clears Aβ. At the same time, the microglia cells are poorly phagocytic for Aβ compared to proinflammatory and anti-inflammatory macrophages. In addition to such distinctive neuropathological features of AD as amyloid beta and tau proteins, neuroinflammation has to be brought into the picture as well. Taking advantage of a coupled mathematical modelling framework, we formulate a network model, accounting for the coupling between neurons and astroglia and integrating all three main neuropathological features with the brain connectome data. We provide details on the coupled dynamics involving cytokines, astrocytes, and microglia. Further, we apply the tumour necrosis factor alpha (TNF-α) inhibitor and anti-Aβ drug and analyze their influence on the brain cells, suggesting conditions under which the drug can prevent cell damage. The important role of astrocytes and TNF-α inhibitors in AD pathophysiology is emphasized, along with potentially promising pathways for developing new AD therapies.
Collapse
|
55
|
Alzheimer Disease Occurs More Frequently In Patients With Inflammatory Bowel Disease: Insight From a Nationwide Study. J Clin Gastroenterol 2022; 57:501-507. [PMID: 35470286 DOI: 10.1097/mcg.0000000000001714] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 03/20/2022] [Indexed: 12/11/2022]
Abstract
Alzheimer disease (AD) affects 5 million Americans and early recognition improves cognitive function. Chronic inflammation and gut microbiome alteration are linked to cognitive decline which are common in inflammatory bowel disease (IBD). We investigated the association of IBD with development of AD. A commercial database (Explorys Inc., Cleveland, OH), an aggregate of electronic health records from 26 major US health care systems, was surveyed. Cohorts of patients with Systematized Nomenclature of Medicine-Clinical Terms (SNOMED-CT) diagnoses of Crohn's disease (CD), ulcerative colitis (UC), and AD were identified. IBD patients with new diagnosis of AD were characterized based on demographic and traditional AD risk factors and IBD-related features. Among 342,740 IBD patients in the database, AD developed in 5750 IBD patients (1.55%). After adjusting for traditional AD risk factors, IBD was identified as an independent risk factor for development of AD [odds ratio (OR)=2.30, 95% confidence interval (CI)=2.10-2.51]. IBD patients with AD were younger in comparison to AD patients without IBD. On sub-group analysis, patients with CD had higher odds of developing AD (adjusted OR=3.34, 95% CI=3.25-3.42) than UC (adjusted OR=1.09, 95% CI=1.06-1.14). Use of tumor necrosis factor (TNF-α) inhibitors in IBD was associated with significantly lower odds of developing AD in both CD and UC. In this population based study, IBD was independently associated with development of AD. Among IBD; the association was stronger in patients with CD in comparison with UC. Use of TNF-α inhibitors was associated with lower odds of developing AD.
Collapse
|
56
|
The Memory Benefit to Aged APP/PS1 Mice from Long-Term Intranasal Treatment of Low-Dose THC. Int J Mol Sci 2022; 23:ijms23084253. [PMID: 35457070 PMCID: PMC9029288 DOI: 10.3390/ijms23084253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 12/19/2022] Open
Abstract
THC has been used as a promising treatment approach for neurological disorders, but the highly psychoactive effects have largely warned off many scientists from pursuing it further. We conducted an intranasal treatment using low-dose THC on 12-month-old APP/PS1 mice daily for 3 months to overcome any potential psychoactive response induced by the systemic delivery. Our results demonstrate that the THC nasal treatment at 0.002 and 0.02 mg/kg significantly slowed the memory decline compared to that in the vehicle-treated transgenic mouse control group. An enzyme-linked immunosorbent assay showed that the Aβ1–40 and 1–42 peptides decreased in the THC-treated groups. The Western blot data indicate that long-term low-dose THC intranasal administration promoted p-tau level reduction and mitochondrial function marker redistribution. The blood biochemical parameter data demonstrate some insignificant changes in cytokine, immunoglobulin, and immune cell profiles during intranasal THC treatment. Intranasal delivery is a non-invasive and convenient method that rapidly targets therapeutics to the brain, minimizing systemic exposure to avoid unwanted adverse effects. Our study provides new insights into the role of low-dose THC intranasal treatment as a pharmacological strategy to counteract alterations in Alzheimer’s disease-related cognitive performance.
Collapse
|
57
|
Dey M, Singh RK. Chronic oral exposure of aluminum chloride in rat modulates molecular and functional neurotoxic markers relevant to Alzheimer's disease. Toxicol Mech Methods 2022; 32:616-627. [PMID: 35341471 DOI: 10.1080/15376516.2022.2058898] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Aluminum is an environmentally abundant potential neurotoxic agent that may result in oxidative damage to a range of cellular biomarkers. The potential sources of aluminum accumulation in body include drinking water, food, medicines, vaccines, and aluminum cookware utensils etc. The accumulation of aluminum in brain is reported to be associated with cholinergic dysfunction, oxidative stress and neuronal damage, that may ultimately cause Alzheimer's disease. Since chronic exposure of aluminum leads to its accumulation in brain, so this study was done by a long-term (24 weeks) low dose (20 mg/kg) oral exposure of aluminum chloride in rats. In this chronic model, we have evaluated the major hallmarks of Alzheimer's disease including amyloid beta (Aβ1-42) and phosphorylated-tau (p231-tau) protein in brain tissue. Furthermore, we evaluated the level of acetyl cholinesterase activity, inflammatory cytokines such as TNF-α, IL-6 and IL-1β, and oxidative stress biomarkers in rat brain in this model. The neurobehavioral parameters were also assessed in animals by using spontaneous locomotor activity, passive avoidance, rotarod test and novel object recognition test to evaluate alteration in learning, memory and muscle co-ordination. We found that chronic oral exposure of aluminum chloride causes a significant increase in structural hallmarks such as Aβ1-42 and p231-tau levels along with proinflammatory cytokines (TNF-α and IL-6), oxidative stress, and a decrease in antioxidant markers such as GSH and catalase. in the brain tissue. These biomarkers significantly affected neurobehavioral parameters in animals. This study provides a mechanistic understanding of chronic aluminum-induced neuronal toxicity in brain with relevance to Alzheimer's disease.
Collapse
Affiliation(s)
- Mangaldeep Dey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli. Transit campus, Bijnour-sisendi road, Sarojini nagar, Lucknow-226002, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli. Transit campus, Bijnour-sisendi road, Sarojini nagar, Lucknow-226002, Uttar Pradesh, India
| |
Collapse
|
58
|
Rickman AD, Hilyard A, Heckmann BL. Dying by fire: noncanonical functions of autophagy proteins in neuroinflammation and neurodegeneration. Neural Regen Res 2022; 17:246-250. [PMID: 34269183 PMCID: PMC8463974 DOI: 10.4103/1673-5374.317958] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/04/2021] [Accepted: 03/30/2021] [Indexed: 11/04/2022] Open
Abstract
Neuroinflammation and neurodegeneration are key components in the establishment and progression of neurodegenerative diseases including Alzheimer's Disease (AD). Over the past decade increasing evidence is emerging for the use of components of the canonical autophagy machinery in pathways that are characterized by LC3 lipidation yet are distinct from traditional macro-autophagy. One such pathway that utilizes components of the autophagy machinery to target LC3 to endosomes, a process termed LC3-associated endocytosis (LANDO), has recently been identified and regulates neuroinflammation. Abrogation of LANDO in microglia cells results in a propensity for elevated neuroinflammatory cytokine production. Using the well-established 5xFAD model of AD to interrogate neuroinflammatory regulation, impairment of LANDO through deletion of a key upstream regulator Rubicon or other downstream autophagy components, exacerbated disease onset and severity, while deletion of microglial autophagy alone had no measurable effect. Mice presented with robust deposition of the neurotoxic AD protein β-amyloid (Aβ), microglial activation and inflammatory cytokine production, tau phosphorylation, and aggressive neurodegeneration culminating in severe memory impairment. LANDO-deficiency impaired recycling of receptors that recognize Aβ, including TLR4 and TREM2. LANDO-deficiency alone through deletion of the WD-domain of the autophagy protein ATG16L, revealed a role for LANDO in the spontaneous establishment of age-associated AD. LANDO-deficient mice aged to 2 years presented with advanced AD-like disease and pathology correlative to that observed in human AD patients. Together, these studies illustrate an important role for microglial LANDO in regulating CNS immune activation and protection against neurodegeneration. New evidence is emerging that demonstrates a putative linkage between pathways such as LANDO and cell death regulation via apoptosis and possibly necroptosis. Herein, we provide a review of the use of the autophagy machinery in non-canonical mechanisms that alter immune regulation and could have significant impact in furthering our understanding of not only CNS diseases like AD, but likely beyond.
Collapse
Affiliation(s)
- Alexis D. Rickman
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Addison Hilyard
- USF Health Byrd Alzheimer's Center and Neuroscience Institute, Morsani College of Medicine, Tampa, FL, USA
| | - Bradlee L. Heckmann
- USF Health Byrd Alzheimer's Center and Neuroscience Institute, Morsani College of Medicine, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
59
|
Metformin in Alzheimer’s disease: An overview of potential mechanisms, preclinical and clinical findings. Biochem Pharmacol 2022; 197:114945. [DOI: 10.1016/j.bcp.2022.114945] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 12/13/2022]
|
60
|
Zahedipour F, Hosseini SA, Henney NC, Barreto GE, Sahebkar A. Phytochemicals as inhibitors of tumor necrosis factor alpha and neuroinflammatory responses in neurodegenerative diseases. Neural Regen Res 2022; 17:1675-1684. [PMID: 35017414 PMCID: PMC8820712 DOI: 10.4103/1673-5374.332128] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
Abstract
Inflammatory processes and proinflammatory cytokines have a key role in the cellular processes of neurodegenerative diseases and are linked to the pathogenesis of functional and mental health disorders. Tumor necrosis factor alpha has been reported to play a major role in the central nervous system in Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis and many other neurodegenerative diseases. Therefore, a potent proinflammatory/proapoptotic tumor necrosis factor alpha could be a strong candidate for targeted therapy. Plant derivatives have now become promising candidates as therapeutic agents because of their antioxidant and chemical characteristics, and anti-inflammatory features. Recently, phytochemicals including flavonoids, terpenoids, alkaloids, and lignans have generated interest as tumor necrosis factor alpha inhibitor candidates for a number of diseases involving inflammation within the nervous system. In this review, we discuss how phytochemicals as tumor necrosis factor alpha inhibitors are a therapeutic strategy targeting neurodegeneration.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology, School of Medicine; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyede Atefe Hosseini
- Department of Medical Biotechnology, School of Medicine; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neil C Henney
- Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - George E Barreto
- Department of Biological Sciences; Health Research Institute, University of Limerick, Limerick, Ireland
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
61
|
Xiong W, Zhao X, Xu Q, Wei G, Zhang L, Fan Y, Wen L, Liu Y, Zhang T, Zhang L, Tong Y, Yin Q, Zhang TE, Yan Z. Qisheng Wan formula ameliorates cognitive impairment of Alzheimer's disease rat via inflammation inhibition and intestinal microbiota regulation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114598. [PMID: 34492320 DOI: 10.1016/j.jep.2021.114598] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Qisheng Wan formula (QWF) was first described in the book Sheng Ji Zong Lu in 1117. The book states that QWF can cure forgetfulness, improve the mind, and make people smart. Hence, QWF has been widely used to treat patients with forgetfulness or dementia. QWF, a classic Chinese formulation, comprises seven herbal drugs: the sclerotium of Poria cocos (Schw.) Wolf, bark of Cinnamomum cassia Presl, root of Polygala tenuifolia Willd., root and rhizome of Panax ginseng C. A. Mey., root of Asparagus cochinchinensis (Lour.) Merr., root and rhizome of Acorus tatarinowii Schott, and root bark of Lycium chinense Mill. AIM OF THE STUDY This study aimed to utilize modern pharmacological methods to evaluate the therapeutic effects and explore the underlying mechanism of QWF action on rats with Alzheimer's disease (AD). MATERIALS AND METHODS The chemical profile of QWF was characterized using ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. The AD rat model was established via a bilateral intraventricular injection of amyloid-β (1-42) (Aβ1-42). The rats were subsequently treated daily with QWF for 4 weeks. The Morris water maze test was performed to evaluate the cognition processes in the rats, whereas histological changes in the hippocampus were observed using hematoxylin and eosin staining. The expression levels of Aβ1-42, nuclear factor-kappa B (NF-κB), tumor necrosis factor (TNF)-α, and interleukin (IL)-6 in the hippocampus and colon were assessed. Moreover, the diversity and composition of the intestinal microbiota were analyzed using 16S rDNA gene sequencing. RESULTS One hundred and fourteen compounds were characterized in QWF. QWF significantly ameliorated the cognition processes and histopathological damages due to AD in rats by decreasing the deposition of Aβ1-42 and downregulating the expression of NF-κB, TNF-α, and IL-6. QWF also modulated changes in the diversity and composition of intestinal microbiota to suppress the relative abundance of inflammation-associated microbiota. CONCLUSION This study showed that QWF can suppress proinflammatory factors and modulate the intestinal microbiota in AD rats.
Collapse
Affiliation(s)
- Wei Xiong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Xiaoqin Zhao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Qing Xu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Guihua Wei
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Liudai Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Yuqing Fan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Lingmiao Wen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Yanjun Liu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Tinglan Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Li Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Yan Tong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Qiaozhi Yin
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.
| | - Tian-E Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.
| | - Zhiyong Yan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| |
Collapse
|
62
|
Pons V, Rivest S. Targeting Systemic Innate Immune Cells as a Therapeutic Avenue for Alzheimer Disease. Pharmacol Rev 2022; 74:1-17. [PMID: 34987086 DOI: 10.1124/pharmrev.121.000400] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is the first progressive neurodegenerative disease worldwide, and the disease is characterized by an accumulation of amyloid in the brain and neurovasculature that triggers cognitive decline and neuroinflammation. The innate immune system has a preponderant role in AD. The last decade, scientists focused their efforts on therapies aiming to modulate innate immunity. The latter is of great interest, since they participate to the inflammation and phagocytose the amyloid in the brain and blood vessels. We and others have developed pharmacological approaches to stimulate these cells using various ligands. These include toll-like receptor 4, macrophage colony stimulating factor, and more recently nucleotide-binding oligomerization domain-containing 2 receptors. This review will discuss the great potential to take advantage of the innate immune system to fight naturally against amyloid β accumulation and prevent its detrimental consequence on brain functions and its vascular system. SIGNIFICANCE STATEMENT: The focus on amyloid β removal from the perivascular space rather than targeting CNS plaque formation and clearance represents a new direction with a great potential. Small molecules able to act at the level of peripheral immunity would constitute a novel approach for tackling aberrant central nervous system biology, one of which we believe would have the potential of generating a lot of interest.
Collapse
Affiliation(s)
- Vincent Pons
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Québec City, QC G1V 4G2, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Québec City, QC G1V 4G2, Canada
| |
Collapse
|
63
|
Duc Nguyen H, Pal Yu B, Hoang NHM, Jo WH, Young Chung H, Kim MS. Prolactin and Its Altered Action in Alzheimer's Disease and Parkinson's Disease. Neuroendocrinology 2022; 112:427-445. [PMID: 34126620 DOI: 10.1159/000517798] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/10/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Prolactin (PRL) is one of the most diverse pituitary hormones and is known to modulate normal neuronal function and neurodegenerative conditions. Many studies have described the influence that PRL has on the central nervous system and addressed its contribution to neurodegeneration, but little is known about the mechanisms responsible for the effects of PRL on neurodegenerative disorders, especially on Alzheimer's disease (AD) and Parkinson's disease (PD). SUMMARY We review and summarize the existing literature and current understanding of the roles of PRL on various PRL aspects of AD and PD. KEY MESSAGES In general, PRL is viewed as a promising molecule for the treatment of AD and PD. Modulation of PRL functions and targeting of immune mechanisms are needed to devise preventive or therapeutic strategies.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Byung Pal Yu
- Department of Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Ngoc Hong Minh Hoang
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Won Hee Jo
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Min-Sun Kim
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| |
Collapse
|
64
|
Deng SM, Chen CJ, Lin HL, Cheng IH. The beneficial effect of synbiotics consumption on Alzheimer's disease mouse model via reducing local and systemic inflammation. IUBMB Life 2021; 74:748-753. [PMID: 34962691 DOI: 10.1002/iub.2589] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 01/15/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that impairs multiple memory domains without an effective prevention or treatment approach. Amyloid plaque-induced neuroinflammation exacerbates neurodegeneration and cognitive impairment in AD. To reduce neuroinflammation, we applied prebiotics or synbiotics to modulate the gut-brain axis in the AD mouse model. AD-like deficits were reduced in mice treated with synbiotics, suggesting that dietary modulation of the gut-brain axis is a potential approach to delay AD progression.
Collapse
Affiliation(s)
- Shin-Meng Deng
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | | | | | - Irene H Cheng
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
65
|
Simultaneous administration of bromodomain and histone deacetylase I inhibitors alleviates cognition deficit in Alzheimer's model of rats. Brain Res Bull 2021; 179:49-56. [PMID: 34915044 DOI: 10.1016/j.brainresbull.2021.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Histone deacetylases (HDACs) target various genes responsible for cognitive functions. However, chromatin readers, particularly bromodomain-containing protein 4 (BRD4), are capable to change the final products of genes. The objective of this study was to evaluate the simultaneous effects of inhibition of HDACs and BRD4 on spatial and aversive memories impaired by amyloid β (Aβ) in a rat model of Alzheimer's disease (AD) considering CREB and TNF-α signaling. METHODS Forty male Wistar rats aged 3 months were randomly divided into five groups: saline +DMSO, Aβ+saline+DMSO, Aβ+JQ1, Aβ+MS-275, Aβ+JQ1+MS-275, and received the related treatments. MS-275, is the second generation of HDACs inhibitor, and JQ1 is a potent inhibitor of the BET family of bromodomain proteins in mammals. After the treatments, cognitive function was assessed by Morris water maze (MWM) and passive avoidance learning (PAL). The hippocampal level of mRNA for CREB and TNF-α, and also phosphorylated CREB were measured using real-time PCR and western blotting respectively. RESULTS Administration of JQ1 and MS-275, either separately or simultaneously, improved acquisition and retrieval of spatial and aversive memories as it was evident by decreased escape latency and increased time spent in the target quadrant (TTS) in Morris water maze (MWM), together with increase in step-through latency, but reduced time spent in the dark zone time in passive avoidance learning (PAL) compared with Aβ+saline+DMSO. Furthermore, there was a significant rise in the hippocampal level of CREB mRNA and phosphorylated CREB, but a reduction in TNF-α expression in comparison with Aβ + Saline. CONCLUSION Simultaneous administration of JQ1 and MS-275 improves acquisition and retrieval of both spatial and aversive memories partly via CREB and TNF-α signaling with no superiority to monotherapy.
Collapse
|
66
|
Taylor HBC, Jeans AF. Friend or Foe? The Varied Faces of Homeostatic Synaptic Plasticity in Neurodegenerative Disease. Front Cell Neurosci 2021; 15:782768. [PMID: 34955753 PMCID: PMC8702499 DOI: 10.3389/fncel.2021.782768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/19/2021] [Indexed: 11/28/2022] Open
Abstract
Homeostatic synaptic plasticity (HSP) regulates synaptic strength both pre- and postsynaptically to ensure stability and efficient information transfer in neural networks. A number of neurological diseases have been associated with deficits in HSP, particularly diseases characterised by episodic network instability such as migraine and epilepsy. Recently, it has become apparent that HSP also plays a role in many neurodegenerative diseases. In this mini review, we present an overview of the evidence linking HSP to each of the major neurodegenerative diseases, finding that HSP changes in each disease appear to belong to one of three broad functional categories: (1) deficits in HSP at degenerating synapses that contribute to pathogenesis or progression; (2) HSP induced in a heterosynaptic or cell non-autonomous manner to support the function of networks of which the degenerating synapses or cells are part; and (3) induction of HSP within the degenerating population of synapses to preserve function and to resist the impact of synapse loss. Understanding the varied manifestations of HSP in neurodegeneration will not only aid understanding mechanisms of disease but could also inspire much-needed novel approaches to therapy.
Collapse
Affiliation(s)
| | - Alexander F. Jeans
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
67
|
Kumar M, Bansal N. A Revisit to Etiopathogenesis and Therapeutic Strategies in Alzheimer's Disease. Curr Drug Targets 2021; 23:486-512. [PMID: 34792002 DOI: 10.2174/1389450122666211118125233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/05/2021] [Accepted: 09/13/2021] [Indexed: 11/22/2022]
Abstract
Dementia is a cluster of brain abnormalities that trigger progressive memory deficits and other cognitive abilities such as skills, language, or executive function. Alzheimer's disease (AD) is the foremost type of age-associated dementia that involves progressive neurodegeneration accompanied by profound cognitive deficits in advanced stages that severely hamper social or occupational abilities with or without the involvement of any other psychiatric condition. The last two decades witnessed a sharp increase (~123%) in mortality due to AD type dementia, typically owing to a very low disclosure rate (~45%) and hence, the prophylactic, as well as the therapeutic cure of AD, has been a huge challenge. Although understanding of AD pathogenesis has witnessed a remarkable growth (e.g., tauopathy, oxidative stress, lipid transport, glucose uptake, apoptosis, synaptic dysfunction, inflammation, and immune system), still a dearth of an effective therapeutic agent in the management of AD prompts the quest for newer pharmacological targets in the purview of its growing epidemiological status. Most of the current therapeutic strategies focus on modulation of a single target, e.g., inhibition of acetylcholinesterase, glutamate excitotoxicity (memantine), or nootropics (piracetam), even though AD is a multifaceted neurological disorder. There is an impedance urgency to find not only symptomatic but effective disease-modifying therapies. The present review focuses on the risk / protective factors and pathogenic mechanisms involved in AD. In addition to the existing symptomatic therapeutic approach, a diverse array of possible targets linked to pathogenic cascades have been re-investigated to envisage the pharmacotherapeutic strategies in AD.
Collapse
Affiliation(s)
- Manish Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Nitin Bansal
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University (CBLU), Bhiwani, Haryana 127021. India
| |
Collapse
|
68
|
Booth MJ, Kobayashi LC, Janevic MR, Clauw D, Piette JD. No increased risk of Alzheimer's disease among people with immune-mediated inflammatory diseases: findings from a longitudinal cohort study of U.S. older adults. BMC Rheumatol 2021; 5:48. [PMID: 34763722 PMCID: PMC8588609 DOI: 10.1186/s41927-021-00219-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/14/2021] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Immune-mediated inflammatory diseases (IMID) are characterized by systemic inflammation affecting the joints and bodily organs. Studies examining the association between individual IMIDs and the risk of Alzheimer's disease (AD) have yielded inconsistent findings. This study examines AD risk across a group of IMIDs in a large population-based sample of older adults. METHODS Data on a national sample of US adults over age 50 was drawn from the Health and Retirement Study (HRS) and linked Medicare claims from 2006 to 2014. IMIDs include rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, ulcerative colitis, and related conditions. We identified IMIDs from 2006 to 2009 Medicare claims using International Classification of Diseases (ICD9-CM) codes. The date of incident AD was derived from Chronic Conditions Warehouse (CCW) identifiers. We examined the risk of AD from 2009 to 2014 using Cox proportional hazards models, both unadjusted and adjusted for age, gender, education, race, and the genetic risk factor APOE-e4. RESULTS One hundred seventy-one (6.02%) of the 2842 total HRS respondents with Medicare coverage and genetic data were classified with IMIDs. Over the subsequent 6 years, 9.36% of IMID patients developed AD compared to 8.57% of controls (unadjusted hazard ratio (HR): 1.09, 95% CI .66-1.81, p = 0.74). Adjusted HR 1.27 (95% CI 0.76-2.12, p = 0.35). Age (HR for 10-year increment 3.56, p < .001), less than high school education (HR 1.70, p = .007), and APOE-e4 (HR 2.61, p < .001 for one or two copies), were also statistically significant predictors of AD. CONCLUSION HRS respondents with common IMIDs do not have increased risk of Alzheimer's disease over a 6-year period.
Collapse
Affiliation(s)
- Michael J Booth
- Department of Health Behavior and Health Education, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48130, USA.
| | - Lindsay C Kobayashi
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Mary R Janevic
- Department of Health Behavior and Health Education, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48130, USA
| | - Daniel Clauw
- Department of Anesthesiology, Rheumatology, Psychiatry, Michigan Medicine, Ann Arbor, MI, USA
| | - John D Piette
- Department of Health Behavior and Health Education, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48130, USA
- Department of Veterans Affairs Center for Clinical Management Research, Ann Arbor, MI, USA
| |
Collapse
|
69
|
Kaur D, Behl T, Chigurupati S, Sehgal A, Singh S, Sharma N, Badavath VN, Vargas-De-La-Cruz C, Bhatia S, Al-Harrasi A, Dey A, Aleya L, Bungau S. Deciphering the focal role of endostatin in Alzheimer's disease. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:61998-62011. [PMID: 34561808 DOI: 10.1007/s11356-021-16567-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/12/2021] [Indexed: 06/13/2023]
Abstract
Alzheimer's disease (AD) is a paramount chronic neurodegenerative condition that has been affecting elderly people since the 1900s. It causes memory loss, disorientation, and poor mental function. AD is considered to be one of the most serious problems that dementia sufferers face. Despite extensive investigation, the pathological origin of Alzheimer's disease remains a mystery. The amyloid cascade theory and the vascular hypothesis, which stresses the buildup of Aβ plaques, have dominated research into dementia and aging throughout history. However, research into this task failed to yield the long-awaited therapeutic miracle lead for Alzheimer's disease. Perhaps a hypothetical fragility in the context of Alzheimer's disease was regarded as a state distinct from aging in general, as suggested by the angiogenesis hypothesis, which suggests that old age is one state associated with upregulation of angiogenic growth factors, resulting in decreased microcirculation throughout the body. There has also been evidence that by controlling or inhibiting the components involved in the sequence of events that cause angiogenesis, there is a visible progression in AD patients. In Alzheimer's disease, one such antiangiogenic drug is being used.
Collapse
Affiliation(s)
- Dapinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India.
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, Saudi Arabia
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | | | - Celia Vargas-De-La-Cruz
- Faculty of Pharmacy and Biochemistry, Academic Department of Pharmacology, Bromatology and Toxicology, Centro Latinoamericano de Ensenanza e Investigacion en Bacteriologia Alimentaria, Universidad Nacinol Mayor de San Marcos, Lima, Peru
- E-Health Research Center, Universidad de Ciencias y Humanidades, Lima, Peru
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
- School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | - Simona Bungau
- Department of Pharmacy, Faculty of Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
70
|
Implications of Phosphoinositide 3-Kinase-Akt (PI3K-Akt) Pathway in the Pathogenesis of Alzheimer's Disease. Mol Neurobiol 2021; 59:354-385. [PMID: 34699027 DOI: 10.1007/s12035-021-02611-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/19/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the foremost type of dementia that afflicts considerable morbidity and mortality in aged population. Several transcription molecules, pathways, and molecular mechanisms such as oxidative stress, inflammation, autophagy, and immune system interact in a multifaceted way that disrupt physiological processes (cell growth, differentiation, survival, lipid and energy metabolism, endocytosis) leading to apoptosis, tauopathy, β-amyloidopathy, neuron, and synapse loss, which play an important role in AD pathophysiology. Despite of stupendous advancements in pathogenic mechanisms, treatment of AD is still a nightmare in the field of medicine. There is compelling urgency to find not only symptomatic but effective disease-modifying therapies. Recently, phosphoinositide 3-kinase (PI3K) and Akt are identified as a pathway triggered by diverse stimuli, including insulin, growth factors, cytokines, and cellular stress, that link amyloid-β, neurofibrillary tangles, and brain atrophy. The present review aims to explore and analyze the role of PI3K-Akt pathway in AD and agents which may modulate Akt and have therapeutic prospects in AD. The literature was researched using keywords "PI3K-Akt" and "Alzheimer's disease" from PubMed, Web of Science, Bentham, Science Direct, Springer Nature, Scopus, and Google Scholar databases including books. Articles published from 1992 to 2021 were prioritized and analyzed for their strengths and limitations, and most appropriate ones were selected for the purpose of review. PI3K-Akt pathway regulates various biological processes such as cell proliferation, motility, growth, survival, and metabolic functions, and inhibits many neurotoxic mechanisms. Furthermore, experimental data indicate that PI3K-Akt signaling might be an important therapeutic target in treatment of AD.
Collapse
|
71
|
Ghosh P, Singh R, Ganeshpurkar A, Pokle AV, Singh RB, Singh SK, Kumar A. Cellular and molecular influencers of neuroinflammation in Alzheimer's disease: Recent concepts & roles. Neurochem Int 2021; 151:105212. [PMID: 34656693 DOI: 10.1016/j.neuint.2021.105212] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/22/2021] [Accepted: 10/10/2021] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD), an extremely common neurodegenerative disorder of the older generation, is one of the leading causes of death globally. Besides the conventional hallmarks i.e. Amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs), neuroinflammation also serves as a major contributing factor in the pathogenesis of AD. There are mounting evidences to support the fundamental role of cellular (microglia, astrocytes, mast cells, and T-cells) and molecular (cytokines, chemokines, caspases, and complement proteins) influencers of neuroinflammation in producing/promoting neurodegeneration and dementia in AD. Genome-wide association studies (GWAS) have revealed the involvement of various single nucleotide polymorphisms (SNPs) of genes related to neuroinflammation with the risk of developing AD. Modulating the release of the neuroinflammatory molecules and targeting their relevant mechanisms may have beneficial effects on the onset, progress and severity of the disease. Here, we review the distinct role of various mediators and modulators of neuroinflammation that impact the pathogenesis and progression of AD as well as incite further research efforts for the treatment of AD through a neuroinflammatory approach.
Collapse
Affiliation(s)
- Powsali Ghosh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Singh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ankit Ganeshpurkar
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ankit Vyankatrao Pokle
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Bhushan Singh
- Institute of Pharmacy Harischandra PG College, Bawanbigha, Varanasi, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
72
|
Page MJ, Pretorius E. Platelet Behavior Contributes to Neuropathologies: A Focus on Alzheimer's and Parkinson's Disease. Semin Thromb Hemost 2021; 48:382-404. [PMID: 34624913 DOI: 10.1055/s-0041-1733960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The functions of platelets are broad. Platelets function in hemostasis and thrombosis, inflammation and immune responses, vascular regulation, and host defense against invading pathogens, among others. These actions are achieved through the release of a wide set of coagulative, vascular, inflammatory, and other factors as well as diverse cell surface receptors involved in the same activities. As active participants in these physiological processes, platelets become involved in signaling pathways and pathological reactions that contribute to diseases that are defined by inflammation (including by pathogen-derived stimuli), vascular dysfunction, and coagulation. These diseases include Alzheimer's and Parkinson's disease, the two most common neurodegenerative diseases. Despite their unique pathological and clinical features, significant shared pathological processes exist between these two conditions, particularly relating to a central inflammatory mechanism involving both neuroinflammation and inflammation in the systemic environment, but also neurovascular dysfunction and coagulopathy, processes which also share initiation factors and receptors. This triad of dysfunction-(neuro)inflammation, neurovascular dysfunction, and hypercoagulation-illustrates the important roles platelets play in neuropathology. Although some mechanisms are understudied in Alzheimer's and Parkinson's disease, a strong case can be made for the relevance of platelets in neurodegeneration-related processes.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| |
Collapse
|
73
|
Kern DM, Lovestone S, Cepeda MS. Treatment with TNF-α inhibitors versus methotrexate and the association with dementia and Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12163. [PMID: 34584936 PMCID: PMC8450793 DOI: 10.1002/trc2.12163] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Peripheral inhibition of tumor necrosis factor (TNF)-α, outside of the central nervous system, may result in clinical improvement of Alzheimer's disease (AD) outcomes. TNF-α inhibitors (TNFIs) are effective treatments for various autoimmune conditions and may be effective for preventing and/or treating AD. The objective of this study was to compare the risk of dementia and AD in patients initiating methotrexate versus those initiating TNFIs. METHODS Insurance claims data from databases of commercially insured and Medicare-eligible patients were used to estimate the risk of dementia and AD within patients with rheumatoid arthritis (RA) initiating a TNFI versus initiation of methotrexate. A sensitivity analysis included all patients without the RA diagnosis requirement. The at-risk period spanned from the index date until a diagnosis of the outcome, loss-to-follow-up, or receipt of the comparator drug. Patients were matched 1-to-1 using propensity scores. A Cox proportional hazards model was used to estimate the hazard ratio (HR). Negative controls were used to calibrate the results. RESULTS A total of 11,092 new TNFI patients and 44,023 new methotrexate patients were identified, and 8925 from each group were matched. The outcome of dementia occurred in 1.4% of patients in both groups. The calibrated results from the Cox regression found no difference between the two groups (commercially insured database: calibrated HR = 0.69, 95% confidence interval = 0.45 to 1.05; Medicare-only database: 1.14, 0.66 to 1.96). Results were similar in all sensitivity analyses: outcome of AD and including patients without RA. DISCUSSION No significant difference for the risk of dementia or AD was seen between patients initiating a TNFI versus methotrexate. Although this study cannot conclude whether use of TNFIs is protective against dementia and AD compared with receiving no treatment, there was no evidence that it is more protective than the active comparator methotrexate.
Collapse
Affiliation(s)
- David M. Kern
- Janssen Research & DevelopmentLLCTitusvilleNew JerseyUSA
| | - Simon Lovestone
- Janssen Research & DevelopmentNeuroscienceBeerse, TurnhoutsewegBelgium
| | | |
Collapse
|
74
|
Mohamed Asik R, Suganthy N, Aarifa MA, Kumar A, Szigeti K, Mathe D, Gulyás B, Archunan G, Padmanabhan P. Alzheimer's Disease: A Molecular View of β-Amyloid Induced Morbific Events. Biomedicines 2021; 9:biomedicines9091126. [PMID: 34572312 PMCID: PMC8468668 DOI: 10.3390/biomedicines9091126] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/22/2021] [Accepted: 08/27/2021] [Indexed: 12/26/2022] Open
Abstract
Amyloid-β (Aβ) is a dynamic peptide of Alzheimer’s disease (AD) which accelerates the disease progression. At the cell membrane and cell compartments, the amyloid precursor protein (APP) undergoes amyloidogenic cleavage by β- and γ-secretases and engenders the Aβ. In addition, externally produced Aβ gets inside the cells by receptors mediated internalization. An elevated amount of Aβ yields spontaneous aggregation which causes organelles impairment. Aβ stimulates the hyperphosphorylation of tau protein via acceleration by several kinases. Aβ travels to the mitochondria and interacts with its functional complexes, which impairs the mitochondrial function leading to the activation of apoptotic signaling cascade. Aβ disrupts the Ca2+ and protein homeostasis of the endoplasmic reticulum (ER) and Golgi complex (GC) that promotes the organelle stress and inhibits its stress recovery machinery such as unfolded protein response (UPR) and ER-associated degradation (ERAD). At lysosome, Aβ precedes autophagy dysfunction upon interacting with autophagy molecules. Interestingly, Aβ act as a transcription regulator as well as inhibits telomerase activity. Both Aβ and p-tau interaction with neuronal and glial receptors elevate the inflammatory molecules and persuade inflammation. Here, we have expounded the Aβ mediated events in the cells and its cosmopolitan role on neurodegeneration, and the current clinical status of anti-amyloid therapy.
Collapse
Affiliation(s)
- Rajmohamed Mohamed Asik
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (R.M.A.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India;
| | - Natarajan Suganthy
- Department of Nanoscience and Technology, Alagappa University, Karaikudi 630003, Tamil Nadu, India;
| | - Mohamed Asik Aarifa
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India;
| | - Arvind Kumar
- Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India;
| | - Krisztián Szigeti
- Department of Biophysics and Radiation Biology, Semmelweis University, 1094 Budapest, Hungary; (K.S.); (D.M.)
- CROmed Translational Research Centers, 1094 Budapest, Hungary
| | - Domokos Mathe
- Department of Biophysics and Radiation Biology, Semmelweis University, 1094 Budapest, Hungary; (K.S.); (D.M.)
- CROmed Translational Research Centers, 1094 Budapest, Hungary
- In Vivo Imaging Advanced Core Facility, Hungarian Center of Excellence for Molecular Medicine (HCEMM), 1094 Budapest, Hungary
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (R.M.A.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Department of Clinical Neuroscience, Karolinska Institute, 17176 Stockholm, Sweden
| | - Govindaraju Archunan
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India;
- Marudupandiyar College, Thanjavur 613403, Tamil Nadu, India
- Correspondence: (G.A.); (P.P.)
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (R.M.A.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Correspondence: (G.A.); (P.P.)
| |
Collapse
|
75
|
Reading CL, Ahlem CN, Murphy MF. NM101 Phase III study of NE3107 in Alzheimer's disease: rationale, design and therapeutic modulation of neuroinflammation and insulin resistance. Neurodegener Dis Manag 2021; 11:289-298. [PMID: 34251287 DOI: 10.2217/nmt-2021-0022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Recently, the roles of inflammation and insulin resistance in neurodegeneration have become better appreciated. NE3107, an oral small molecule, blood-brain permeable anti-inflammatory insulin sensitizer that binds extracellular signal-regulated kinase, has been shown to selectively inhibit inflammation-driven ERK- and NF-κB-stimulated inflammatory mediators, including TNF-α, without inhibiting their homeostatic functions. We describe the rationale and design of NM101, the first randomized, multicenter Phase III clinical study to examine the safety and efficacy of 30 week treatment with NE3107 versus placebo in elderly adults with mild-to-moderate Alzheimer's disease. Patients (316) will be randomized in a 1:1 ratio. The co-primary end points measure cognitive function (ADAS Cog12), and functional and behavioral characteristics (ADCS CGIC). Trial registration number: NCT04669028 (Clinicaltrials.gov).
Collapse
|
76
|
Dömling A, Li X. TNF-α: The shape of small molecules to come? Drug Discov Today 2021; 27:3-7. [PMID: 34229081 DOI: 10.1016/j.drudis.2021.06.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/11/2021] [Accepted: 06/30/2021] [Indexed: 11/16/2022]
Abstract
In 2020, the anti-tumor necrosis factor (TNF) monoclonal antibody Humira® generated US$165.8 billion in cumulative sales and snatched the crown for the industry's most successful drug from Lipitor (atorvastatin). TNF-α is a major component in beneficial and disease-related inflammation and TNF-α-inhibitor biologics have gained widespread use in autoimmune diseases, such as rheumatoid arthritis (RA). Many more diseases could benefit from TNF-α inhibitors, such as Alzheimer's disease (AD) or major depression. However, the nature of TNF-α-inhibitor biologics prohibits central nervous system (CNS) applications. Moreover, high drug production costs and pricing, together with antidrug immune reactions and insufficient patient coverage, argue for the development of small-molecule drugs. Recently, drug-like orally available small molecules were described with high activity in animal disease models with activities comparable to those of antibodies.
Collapse
Affiliation(s)
| | - Xin Li
- Drug Design Department, University of Groningen, the Netherlands
| |
Collapse
|
77
|
Kaur B, Mishra S, Kaur R, Kalotra S, Singh P. Rationally designed TNF-α inhibitors: Identification of promising cytotoxic agents. Bioorg Med Chem Lett 2021; 41:127982. [PMID: 33766762 DOI: 10.1016/j.bmcl.2021.127982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 11/17/2022]
Abstract
Design and synthesis of new indole derivatives as tumor growth inhibiting agents via inhibiting the TNF-α is described. The preliminary results showed the inhibition of LPS induced production of NO, TNF-α and IL-6 by these compounds out of which compounds 2d and 2g exhibited appreciable cytotoxicity against the 60 cell lines panel of human cancer. The rationale behind the design of the molecules and the results of their biological studies are presented. 2009 Elsevier Ltd. All rights reserved.
Collapse
Affiliation(s)
- Baljit Kaur
- Department of Chemistry, Centre for Advanced Studies, Guru Nanak Dev University, Amritsar 143005, India
| | - Sahil Mishra
- Department of Chemistry, Centre for Advanced Studies, Guru Nanak Dev University, Amritsar 143005, India
| | - Ramandeep Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, India
| | - Shikha Kalotra
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, India
| | - Palwinder Singh
- Department of Chemistry, Centre for Advanced Studies, Guru Nanak Dev University, Amritsar 143005, India.
| |
Collapse
|
78
|
Babić Leko M, Nikolac Perković M, Klepac N, Štrac DŠ, Borovečki F, Pivac N, Hof PR, Šimić G. IL-1β, IL-6, IL-10, and TNFα Single Nucleotide Polymorphisms in Human Influence the Susceptibility to Alzheimer's Disease Pathology. J Alzheimers Dis 2021; 75:1029-1047. [PMID: 32390629 DOI: 10.3233/jad-200056] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Neuroinflammation plays an important role in Alzheimer's disease (AD). During this process, activated microglia release pro-inflammatory cytokines such as interleukin (IL)-1α, IL-1β, IL-6, and tumor necrosis factor α (TNFα) that participate in neuron damage, but also anti-inflammatory cytokines (such as IL-10), which maintain homeostasis of immune response. Previous studies showed the association of IL-1α -889C/T (rs1800587), IL-1β-1473G/C (rs1143623), IL-6 -174C/G (rs1800795), IL-10 -1082G/A (rs1800896), and TNFα -308A/G (rs1800629) polymorphisms with AD. OBJECTIVE We aimed to investigate whether people with certain IL-1α, IL-1β, IL-6, IL-10, and TNFα genotypes in these polymorphisms are more prone to develop AD-related pathology, reflected by pathological levels of cerebrospinal fluid (CSF) AD biomarkers including amyloid-β1-42, total tau (t-tau), tau phosphorylated at Thr 181 (p-tau181), Ser 199 (p-tau199), and Thr 231 (p-tau231), and visinin-like protein 1 (VILIP-1). METHODS The study included 115 AD patients, 53 patients with mild cognitive impairment, and 11 healthy controls. The polymorphisms were determined using real-time polymerase chain reaction. Levels of CSF biomarkers were determined by enzyme-linked immunosorbent assay. RESULTS A significant increase in p-tau CSF levels was found in patients with the AA IL-10 -1082G/A and GG TNFα -308A/G genotypes, and in carriers of a G allele in IL-1β -1473C/G and IL-6 -174C/G polymorphisms. t-tau levels were increased in carriers of a G allele in IL-1β -1473C/G polymorphism. An increase in VILIP-1 levels was observed in patients with CG and GG IL-1β -1473C/G, GC IL-6 -174C/G, and GG TNFα -308A/G genotype. CONCLUSION These results suggest that persons carrying certain genotypes in IL10 (-1082G/A), IL1β (1473C/G), IL6 (-174C/G), and TNFIα (-308A/G) could be more vulnerable to development of neuroinflammation, and consequently of AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | | | - Nataša Klepac
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | | | - Fran Borovečki
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nela Pivac
- Department of Molecular Medicine, Institute Ruđer Bošković, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| |
Collapse
|
79
|
Neuroinflammation in Alzheimer's Disease. Biomedicines 2021; 9:biomedicines9050524. [PMID: 34067173 PMCID: PMC8150909 DOI: 10.3390/biomedicines9050524] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease associated with human aging. Ten percent of individuals over 65 years have AD and its prevalence continues to rise with increasing age. There are currently no effective disease modifying treatments for AD, resulting in increasingly large socioeconomic and personal costs. Increasing age is associated with an increase in low-grade chronic inflammation (inflammaging) that may contribute to the neurodegenerative process in AD. Although the exact mechanisms remain unclear, aberrant elevation of reactive oxygen and nitrogen species (RONS) levels from several endogenous and exogenous processes in the brain may not only affect cell signaling, but also trigger cellular senescence, inflammation, and pyroptosis. Moreover, a compromised immune privilege of the brain that allows the infiltration of peripheral immune cells and infectious agents may play a role. Additionally, meta-inflammation as well as gut microbiota dysbiosis may drive the neuroinflammatory process. Considering that inflammatory/immune pathways are dysregulated in parallel with cognitive dysfunction in AD, elucidating the relationship between the central nervous system and the immune system may facilitate the development of a safe and effective therapy for AD. We discuss some current ideas on processes in inflammaging that appear to drive the neurodegenerative process in AD and summarize details on a few immunomodulatory strategies being developed to selectively target the detrimental aspects of neuroinflammation without affecting defense mechanisms against pathogens and tissue damage.
Collapse
|
80
|
Uddin MS, Hasana S, Ahmad J, Hossain MF, Rahman MM, Behl T, Rauf A, Ahmad A, Hafeez A, Perveen A, Ashraf GM. Anti-Neuroinflammatory Potential of Polyphenols by Inhibiting NF-κB to Halt Alzheimer's Disease. Curr Pharm Des 2021; 27:402-414. [PMID: 33213314 DOI: 10.2174/1381612826666201118092422] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/01/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is an irrevocable chronic brain disorder featured by neuronal loss, microglial accumulation, and progressive cognitive impairment. The proper pathophysiology of this life-threatening disorder is not completely understood and no exact remedies have been found yet. Over the last few decades, research on AD has mainly highlighted pathomechanisms linked to a couple of the major pathological hallmarks, including extracellular senile plaques made of amyloid-β (Aβ) peptides, and intracellular neurofibrillary tangles (NFTs) made of tau proteins. Aβ can induce apoptosis, trigger an inflammatory response, and inhibit the synaptic plasticity of the hippocampus, which ultimately contributes to reducing cognitive functions and memory impairment. Recently, a third disease hallmark, the neuroinflammatory reaction that is mediated by cerebral innate immune cells, has become a spotlight in the current research area, assured by pre-clinical, clinical, and genetic investigations. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), a cytokine producer, is significantly associated with physiological inflammatory proceedings and thus shows a promising candidate for inflammation- based AD therapy. Recent data reveal that phytochemicals, mainly polyphenol compounds, exhibit potential neuroprotective functions and these may be considered as a vital resource for discovering several drug candidates against AD. Interestingly, phytochemicals can easily interfere with the signaling pathway of NF-κB. This review represents the anti-neuroinflammatory potential of polyphenols as inhibitors of NF-κB to combat AD pathogenesis.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Sharifa Hasana
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Jamil Ahmad
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | | | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Khyber Pakhtunkhwa, Pakistan
| | - Ausaf Ahmad
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Uttar Pradesh, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
81
|
Zhang L, Young JI, Gomez L, Silva TC, Schmidt MA, Cai J, Chen X, Martin ER, Wang L. Sex-specific DNA methylation differences in Alzheimer's disease pathology. Acta Neuropathol Commun 2021; 9:77. [PMID: 33902726 PMCID: PMC8074512 DOI: 10.1186/s40478-021-01177-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/10/2021] [Indexed: 12/14/2022] Open
Abstract
Sex is an important factor that contributes to the clinical and biological heterogeneities in Alzheimer's disease (AD), but the regulatory mechanisms underlying sex disparity in AD are still not well understood. DNA methylation is an important epigenetic modification that regulates gene transcription and is known to be involved in AD. We performed the first large-scale sex-specific meta-analysis of DNA methylation differences in AD neuropathology, by re-analyzing four recent epigenome-wide association studies totaling more than 1000 postmortem prefrontal cortex brain samples using a uniform analytical pipeline. For each cohort, we employed two complementary analytical strategies, a sex-stratified analysis that examined methylation-Braak stage associations in male and female samples separately, and a sex-by-Braak stage interaction analysis that compared the magnitude of these associations between different sexes. Our analysis uncovered 14 novel CpGs, mapped to genes such as TMEM39A and TNXB that are associated with the AD Braak stage in a sex-specific manner. TMEM39A is known to be involved in inflammation, dysregulated type I interferon responses, and other immune processes. TNXB encodes tenascin proteins, which are extracellular matrix glycoproteins demonstrated to modulate synaptic plasticity in the brain. Moreover, for many previously implicated genes in AD neuropathology, such as MBP and AZU1, our analysis provided the new insights that they were predominately driven by effects in only one sex. These sex-specific DNA methylation differences were enriched in divergent biological processes such as integrin activation in females and complement activation in males. Our study implicated multiple new loci and biological processes that affected AD neuropathology in a sex-specific manner.
Collapse
Affiliation(s)
- Lanyu Zhang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Juan I Young
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Lissette Gomez
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Tiago C Silva
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Michael A Schmidt
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Jesse Cai
- Brentwood High School, 5304 Murray Ln, Brentwood, TN, 37027, USA
| | - Xi Chen
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Eden R Martin
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Lily Wang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
82
|
Adetuyi BO, Farombi EO. 6-Gingerol, an active constituent of ginger, attenuates lipopolysaccharide-induced oxidation, inflammation, cognitive deficits, neuroplasticity, and amyloidogenesis in rat. J Food Biochem 2021; 45:e13660. [PMID: 33624846 DOI: 10.1111/jfbc.13660] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/05/2021] [Accepted: 01/31/2021] [Indexed: 01/23/2023]
Abstract
This study examined the protective effect of 6-Gingerol (6G) against lipopolysaccharide (LPS)-induced cognitive impairments, oxidative stress, neuroplasticity, amyloidogenesis, and inflammation. Male rats were allocated into six groups in this manner; Group I placed on normal saline only. Group II was treated for 7 days with LPS alone intraperitoneally at 250 µg/kg body weight (bw). Group III received 6G alone at 50 mg/kg bw orally for 14 days. Groups IV and V received 6G at 20 and 50 mg/kg bw for 7 days, respectively, and LPS for another 7 days to induce neurotoxicity. Group VI received 5 mg/kg bw of donepezil for 7 days and LPS for 7 days. Pretreatment with 20 and 50 mg/kg bw of 6G protected against LPS-mediated learning and memory function, and also locomotor and motor deficits. Besides, 20 and 50 mg/kg bw 6G mitigated LPS-induced alteration in markers of oxidative stress. Furthermore, induction of amyloidogenesis associated with disruption of histoarchitecture and high expression of interleukin 1β, inducible nitric oxide synthase, amyloid precursor protein (APP), β-secretase 1, and brain-derived neurotrophic factor by LPS was mitigated by the two doses of 6G in the rat hippocampus and cerebral cortex region of the brain. 6G pretreatment at the two doses mitigated LPS-mediated histopathological changes in the hippocampus and cerebral cortex of rats. Overall, our results demonstrate that the protective effect of 6G is mediated through the reversal of neurobehavioral deficit, oxidative stress, inflammation, and amyloidogenesis, thus making 6G a possible chemoprophylactic agent against brain injury as a result of LPS exposure. PRACTICAL APPLICATIONS: In the search for a holistic prevention of inflammation-associated neurodegeneration, nutraceuticals are becoming prominent. Hence, this study presents 6G, an active constituent of ginger, as a chemoprotective, antioxidant, and anti-inflammatory agent, which is able to ameliorate cognitive impairments, oxidative stress, neuroplasticity, amyloidogenesis, and inflammation in LPS-induced rat model of neuroinflammation.
Collapse
Affiliation(s)
- Babatunde Oluwafemi Adetuyi
- Molecular Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ebenezer Olatunde Farombi
- Molecular Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
83
|
Gao H, Bigalke J, Jiang E, Fan Y, Chen B, Chen QH, Shan Z. TNFα Triggers an Augmented Inflammatory Response in Brain Neurons from Dahl Salt-Sensitive Rats Compared with Normal Sprague Dawley Rats. Cell Mol Neurobiol 2021; 42:1787-1800. [PMID: 33625627 PMCID: PMC8382783 DOI: 10.1007/s10571-021-01056-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/04/2021] [Indexed: 12/23/2022]
Abstract
Tumor Necrosis Factor (TNF)-α is a proinflammatory cytokine (PIC) and has been implicated in a variety of illness including cardiovascular disease. The current study investigated the inflammatory response trigged by TNFα in both cultured brain neurons and the hypothalamic paraventricular nucleus (PVN), a key cardiovascular relevant brain area, of the Sprague Dawley (SD) rats. Our results demonstrated that TNFα treatment induces a dose- and time-dependent increase in mRNA expression of PICs including Interleukin (IL)-1β and Interleukin-6 (IL6); chemokines including C-C Motif Chemokine Ligand 5 (CCL5) and C-C Motif Chemokine Ligand 12 (CCL12), inducible nitric oxide synthase (iNOS), as well as transcription factor NF-kB in cultured brain neurons from neonatal SD rats. Consistent with this finding, immunostaining shows that TNFα treatment increases immunoreactivity of IL1β, CCL5, iNOS and stimulates activation or expression of NF-kB, in both cultured brain neurons and the PVN of adult SD rats. We further compared mRNA expression of the aforementioned genes in basal level as well as in response to TNFα challenge between SD rats and Dahl Salt-sensitive (Dahl-S) rats, an animal model of salt-sensitive hypertension. Dahl-S brain neurons presented higher baseline levels as well as greater response to TNFα challenge in mRNA expression of CCL5, iNOS and IL1β. Furthermore, central administration of TNFα caused significant higher response in CCL12 in the PVN of Dahl-S rats. The increased inflammatory response to TNFα in Dahl-S rats may be indicative of an underlying mechanism for enhanced pressor reactivity to salt intake in the Dahl-S rat model.
Collapse
Affiliation(s)
- Huanjia Gao
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA.,The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, China
| | - Jeremy Bigalke
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA
| | - Enshe Jiang
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA.,Institute of Nursing and Health, Henan University, Henan, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, Henan University, Henan, China
| | - Yuanyuan Fan
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA.,School of Life Sciences, Henan University, Henan, China
| | - Bojun Chen
- Department of Emergency, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qing-Hui Chen
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA.,Health Research Institute, Michigan Technological University, Houghton, MI, 49931, USA
| | - Zhiying Shan
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA. .,Health Research Institute, Michigan Technological University, Houghton, MI, 49931, USA.
| |
Collapse
|
84
|
Kurach Ł, Kulczycka-Mamona S, Kowalczyk J, Skalicka-Woźniak K, Boguszewska-Czubara A, El Sayed N, Osmani M, Iwaniak K, Budzyńska B. Mechanisms of the Procognitive Effects of Xanthotoxin and Umbelliferone on LPS-Induced Amnesia in Mice. Int J Mol Sci 2021; 22:1779. [PMID: 33579030 PMCID: PMC7916770 DOI: 10.3390/ijms22041779] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/06/2021] [Accepted: 02/07/2021] [Indexed: 12/23/2022] Open
Abstract
Neuroinflammation plays an essential role in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease. Although coumarins have been shown to improve cognitive function in animal models and exert anti-inflammatory effects in cell cultures, the exact mechanism of their neuroprotective effects has not yet been fully elucidated. The present study aimed to investigate the neuroprotective effects of xanthotoxin (furanocoumarin) and umbelliferone (simple coumarin) in lipopolysaccharide-induced cognitive dysfunction in mice. For evaluation memory and learning processes, a passive avoidance test was used. Furthermore, acetylcholinesterase level and impact on the tumor necrosis factor α, interleukin 10 levels in the whole brain, and cyclooxygenase-II in hippocampus was established. Subchronic administration of both coumarins (15 mg/kg) enhanced the learning and memory function, but only the xanthotoxin improved cognitive processes impaired by lipopolysaccharide (0.8 mg/kg) administration. Behavioral results stay in line with acetylcholinesterase level in the brain. A statistically significant decrease in the level of tumor necrosis factor α and cyclooxygenase-II in lipopolysaccharide-treated rodents after coumarins' administration was observed. Together, our findings demonstrate that both coumarins improved cognitive functions, but only xanthotoxin significantly enhanced the learning and memory function and reduced the level of acetylcholinesterase in lipopolysaccharide-treated mice. This effect may suggest that only furanocoumarin-xanthotoxin attenuates neuroinflammation and enhances cholinergic neurotransmission, thus it can be a potential remedy with procognitive potential effective in treatment of neuroinflammatory disease.
Collapse
Affiliation(s)
- Łukasz Kurach
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, 4A Chodzki Str., 20-093 Lublin, Poland;
| | - Sylwia Kulczycka-Mamona
- Department of Applied Pharmacy, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland; (S.K.-M.); (J.K.); (K.I.)
| | - Joanna Kowalczyk
- Department of Applied Pharmacy, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland; (S.K.-M.); (J.K.); (K.I.)
| | - Krystyna Skalicka-Woźniak
- Independent Laboratory of Natural Products Chemistry, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland;
| | - Anna Boguszewska-Czubara
- Department of Medicinal Chemistry, Medical University of Lublin, 4A Chodzki Str., 20-093 Lublin, Poland;
| | - Nesrine El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt;
| | - Mitat Osmani
- Department of Pharmacy, University of Pristina, St. Bulevardi i Dëshmorëve, 10000 Pristina, Kosovo;
| | - Karol Iwaniak
- Department of Applied Pharmacy, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland; (S.K.-M.); (J.K.); (K.I.)
| | - Barbara Budzyńska
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, 4A Chodzki Str., 20-093 Lublin, Poland;
| |
Collapse
|
85
|
Thingore C, Kshirsagar V, Juvekar A. Amelioration of oxidative stress and neuroinflammation in lipopolysaccharide-induced memory impairment using Rosmarinic acid in mice. Metab Brain Dis 2021; 36:299-313. [PMID: 33068223 DOI: 10.1007/s11011-020-00629-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/11/2020] [Indexed: 12/13/2022]
Abstract
Oxidative stress plays a pivotal part in the manifestation of neuroinflammation, which further leads to neurodegenerative diseases like Alzheimer's disease (AD). Systemic administration of lipopolysaccharide (LPS) induces neuroinflammation resulting in memory impairment (MI) and cognitive decline. In this study, we evaluated whether prophylactic administration of Rosmarinic acid (RA), a naturally occurring compound, exerts a neuroprotective effect in LPS-induced MI and cognitive decline. Herein, Swiss albino mice were pre-treated with RA (0.5 mg/kg and 1 mg/kg i.p.) for 28 days and were intermittently exposed to LPS (0.25 mg/kg i.p.) for 7 days. LPS caused poor memory retention and increased cognitive decline in Morris water maze (MWM) and Y maze paradigms respectively. Additionally, LPS increased oxidative stress which was denoted by a decrease in superoxide dismutase (SOD) activity, decrease in reduced glutathione (GSH) levels, and increased lipid peroxidation in the brain. Imbalance in the cholinergic system was analyzed by measuring the acetylcholinesterase (AChE) activity. Pre-treatment with RA improved memory and behavioral disturbances by alleviating oxidative stress and AChE activity. LPS augmented levels of tumor necrosis factor (TNF-α), interleukin (IL)-6, caspase-3, and c-Jun. Pre-treatment with RA revitalized the elevated levels of proinflammatory cytokines and apoptotic proteins. In conclusion, this study showcases the amelioration of MI by RA in LPS-challenged memory and cognitive decline, which could be credited to its anti-oxidant effect, inhibitory effect on both proinflammatory cytokines and apoptotic regulators, and reduction in AChE activity.
Collapse
Affiliation(s)
- Chetan Thingore
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, Maharashtra, India
| | - Viplav Kshirsagar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, Maharashtra, India
| | - Archana Juvekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, Maharashtra, India.
| |
Collapse
|
86
|
Culibrk RA, Arabiyat AS, DeKalb CA, Hahn MS. Modeling Sympathetic Hyperactivity in Alzheimer's Related Bone Loss. J Alzheimers Dis 2021; 84:647-658. [PMID: 34569964 DOI: 10.3233/jad-215007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND A significant subset of patients with Alzheimer's disease (AD) exhibit low bone mineral density and are therefore more fracture-prone, relative to their similarly aged neurotypical counterparts. In addition to chronic immune hyperactivity, behavioral dysregulation of effector peripheral sympathetic neurons-which densely innervate bone and potently modulate bone remodeling-is implicated in this pathological bone reformation. OBJECTIVE Thus, there exists a pressing need for a robust in vitro model which allows interrogation of the paracrine interactions between the putative mediators of AD-related osteopenia: sympathetic neurons (SNs) and mesenchymal stem cells (MSCs). METHODS Toward this end, activated SN-like PC12 cells and bone marrow derived MSCs were cultured in poly(ethylene glycol) diacrylate (PEGDA) hydrogels in the presence or absence of the AD-relevant inflammatory cytokine tumor necrosis factor alpha (TNF-α) under mono- and co-culture conditions. RESULTS PC12s and MSCs exposed separately to TNF-α displayed increased expression of pro-inflammatory mediators and decreased osteopontin (OPN), respectively. These data indicate that TNF-α was capable of inducing a dysregulated state in both cell types consistent with AD. Co-culture of TNF-α-activated PC12s and MSCs further exacerbated pathological behaviors in both cell types. Specifically, PC12s displayed increased secretion of interleukin 6 relative to TNF-α stimulated monoculture controls. Similarly, MSCs demonstrated a further reduction in osteogenic capacity relative to TNF-α stimulated monoculture controls, as illustrated by a significant decrease in OPN and collagen type I alpha I chain. CONCLUSION Taken together, these data may indicate that dysregulated sympathetic activity may contribute to AD-related bone loss.
Collapse
Affiliation(s)
- Robert A Culibrk
- Hahn Tissue Lab, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Ahmad S Arabiyat
- Hahn Tissue Lab, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Carisa A DeKalb
- Hahn Tissue Lab, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Mariah S Hahn
- Hahn Tissue Lab, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| |
Collapse
|
87
|
Munafò A, Burgaletto C, Di Benedetto G, Di Mauro M, Di Mauro R, Bernardini R, Cantarella G. Repositioning of Immunomodulators: A Ray of Hope for Alzheimer's Disease? Front Neurosci 2020; 14:614643. [PMID: 33343293 PMCID: PMC7746859 DOI: 10.3389/fnins.2020.614643] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder characterized by cognitive decline and by the presence of amyloid β plaques and neurofibrillary tangles in the brain. Despite recent advances in understanding its pathophysiological mechanisms, to date, there are no disease-modifying therapeutic options, to slow or halt the evolution of neurodegenerative processes in AD. Current pharmacological treatments only transiently mitigate the severity of symptoms, with modest or null overall improvement. Emerging evidence supports the concept that AD is affected by the impaired ability of the immune system to restrain the brain's pathology. Deep understanding of the relationship between the nervous and the immune system may provide a novel arena to develop effective and safe drugs for AD treatment. Considering the crucial role of inflammatory/immune pathways in AD, here we discuss the current status of the immuno-oncological, immunomodulatory and anti-TNF-α drugs which are being used in preclinical studies or in ongoing clinical trials by means of the drug-repositioning approach.
Collapse
Affiliation(s)
- Antonio Munafò
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Chiara Burgaletto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Giulia Di Benedetto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Marco Di Mauro
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Rosaria Di Mauro
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.,Unit of Clinical Toxicology, University Hospital, University of Catania, Catania, Italy
| | - Giuseppina Cantarella
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| |
Collapse
|
88
|
Rhea EM, Logsdon AF, Banks WA, Erickson ME. Intranasal Delivery: Effects on the Neuroimmune Axes and Treatment of Neuroinflammation. Pharmaceutics 2020; 12:pharmaceutics12111120. [PMID: 33233734 PMCID: PMC7699866 DOI: 10.3390/pharmaceutics12111120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/02/2023] Open
Abstract
This review highlights the pre-clinical and clinical work performed to use intranasal delivery of various compounds from growth factors to stem cells to reduce neuroimmune interactions. We introduce the concept of intranasal (IN) delivery and the variations of this delivery method based on the model used (i.e., rodents, non-human primates, and humans). We summarize the literature available on IN delivery of growth factors, vitamins and metabolites, cytokines, immunosuppressants, exosomes, and lastly stem cells. We focus on the improvement of neuroimmune interactions, such as the activation of resident central nervous system (CNS) immune cells, expression or release of cytokines, and detrimental effects of signaling processes. We highlight common diseases that are linked to dysregulations in neuroimmune interactions, such as Alzheimer's disease, Parkinson's disease, stroke, multiple sclerosis, and traumatic brain injury.
Collapse
Affiliation(s)
- Elizabeth M. Rhea
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; (A.F.L.); (W.A.B.); (M.E.E.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- Correspondence: ; Tel.: +1-206-764-2938
| | - Aric F. Logsdon
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; (A.F.L.); (W.A.B.); (M.E.E.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - William A. Banks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; (A.F.L.); (W.A.B.); (M.E.E.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Michelle E. Erickson
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; (A.F.L.); (W.A.B.); (M.E.E.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
89
|
Heo CE, Han JY, Lim S, Lee J, Im D, Lee MJ, Kim YK, Kim HI. ATP Kinetically Modulates Pathogenic Tau Fibrillations. ACS Chem Neurosci 2020; 11:3144-3152. [PMID: 32915536 DOI: 10.1021/acschemneuro.0c00479] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Advanced understanding of Alzheimer's disease (AD) and several tauopathies over the past decades indicates the pathological importance of tau aggregation in these diseases. Herein, we demonstrated that adenosine triphosphate (ATP), a highly charged anionic molecule found abundantly in the cytosol of cells, catalyzes fibrillation of tau as well as human islet amyloid polypeptide, a representative of basic intrinsically disordered proteins. Our results showed that ATP attracts multiple lysine residues of the four-repeat domain of tau (K18) via supramolecular complexation, thereby forming dimers that are converted to nuclei and accelerate fibril elongation. However, ATP was not directly incorporated into the K18 fibrils, suggesting that ATP plays the role of a catalyst, rather than a reactant, during K18 fibrillation. We also characterized the correlation between ATP dyshomeostasis and tau aggregation in the cellular environment. Our multiple biophysical approaches, including native mass spectrometry (MS), small-angle X-ray scattering (SAXS), and molecular dynamics (MD) simulation, provided insights into the molecular-level influence of ATP on the structural changes and fibrillation of tau.
Collapse
Affiliation(s)
- Chae Eun Heo
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
- Center for Proteogenomics Research, Korea University, Seoul 02841, Republic of Korea
| | - Jong Yoon Han
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
- Center for Proteogenomics Research, Korea University, Seoul 02841, Republic of Korea
| | - Sungsu Lim
- Brain Science Institute, Center for Neuromedicine, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jeeyoung Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Dongjoon Im
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
- Center for Proteogenomics Research, Korea University, Seoul 02841, Republic of Korea
| | - Min Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Yun Kyung Kim
- Brain Science Institute, Center for Neuromedicine, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Biological Chemistry, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hugh I. Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
- Center for Proteogenomics Research, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
90
|
Darvishi N, Yousefinejad V, Akbari ME, Abdi M, Moradi N, Darvishi S, Mehrabi Y, Ghaderi E, Jamshidi-Naaeini Y, Ghaderi B, Davoodi SH. Antioxidant and anti-inflammatory effects of oral propolis in patients with breast cancer treated with chemotherapy: a Randomized controlled trial. J Herb Med 2020. [DOI: 10.1016/j.hermed.2020.100385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
91
|
Jackson JM, Bay AA, Barter JD, Ni L, Caudle WM, Serra MC, Wharton W, Hackney ME. The Role of Nutrition and Inflammation on Cognition in a High-Risk Group for Alzheimer's Disease. J Alzheimers Dis Rep 2020; 4:345-352. [PMID: 33024941 PMCID: PMC7504978 DOI: 10.3233/adr-200224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background: Alzheimer’s disease (AD) is a prevalent neurodegenerative disease. Treatments are necessary to target people at high risk for AD. Inflammation, particularly tumor necrosis factor alpha (TNFα), appears to be an important marker associated with the development of AD pathophysiology. Consuming a high-fat diet induces tissue expression of TNFα. Objective: This study investigates the relationship between nutrition, circulating inflammation, and cognition in African American women (age: M = 59.5 (±8.20) [42–73] years) at risk for developing AD. Methods: Participants were split into high-fat and low-fat groups based on total dietary fat consumption self-reported on the Lower Mississippi Delta Nutrition Intervention Research Initiative Food Frequency Questionnaire (Delta NIRI FFQ). Results: A high-fat diet was associated with increased blood serum TNFα (p = 0.02) compared to the low-fat diet. In addition, global cognition scores were 9.0% better in those who consumed a higher fat diet (p = 0.04). No significant differences across groups were noted for executive function, dual-tasking, and visuospatial performance. Conclusion: These results indicate that there may be multiple biological pathways involved in AD development, suggesting the need for more holistic approaches to mitigate AD-development risk.
Collapse
Affiliation(s)
- Jordan M Jackson
- Emory University's Rollins School of Public Health, Gangarosa Department of Environmental Health, Atlanta, GA, USA
| | - Allison A Bay
- Emory University School of Medicine Department of Medicine, Division of General Medicine and Geriatrics, Atlanta, GA, USA
| | - Jolie Denise Barter
- Emory University School of Medicine Department of Medicine, Division of General Medicine and Geriatrics, Atlanta, GA, USA
| | - Liang Ni
- Emory University School of Medicine Department of Medicine, Division of General Medicine and Geriatrics, Atlanta, GA, USA
| | - William Michael Caudle
- Emory University's Rollins School of Public Health, Gangarosa Department of Environmental Health, Atlanta, GA, USA.,Emory Center for Neurodegenerative Disease, Atlanta, GA, USA
| | - Monica C Serra
- Division of Geriatrics, Gerontology & Palliative Medicine, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,San Antonio Geriatrics Research, Education & Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, USA
| | | | - Madeleine E Hackney
- Emory University School of Medicine Department of Medicine, Division of General Medicine and Geriatrics, Atlanta, GA, USA.,Emory University School of Nursing, Atlanta, GA, USA.,Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur, GA, USA.,Emory University School of Medicine, Department of Rehabilitation Medicine, Atlanta, GA, USA
| |
Collapse
|
92
|
Guarnieri G, Sarchielli E, Comeglio P, Herrera-Puerta E, Piaceri I, Nacmias B, Benelli M, Kelsey G, Maggi M, Gallina P, Vannelli GB, Morelli A. Tumor Necrosis Factor α Influences Phenotypic Plasticity and Promotes Epigenetic Changes in Human Basal Forebrain Cholinergic Neuroblasts. Int J Mol Sci 2020; 21:E6128. [PMID: 32854421 PMCID: PMC7504606 DOI: 10.3390/ijms21176128] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 01/08/2023] Open
Abstract
TNFα is the main proinflammatory cytokine implicated in the pathogenesis of neurodegenerative disorders, but it also modulates physiological functions in both the developing and adult brain. In this study, we investigated a potential direct role of TNFα in determining phenotypic changes of a recently established cellular model of human basal forebrain cholinergic neuroblasts isolated from the nucleus basalis of Meynert (hfNBMs). Exposing hfNBMs to TNFα reduced the expression of immature markers, such as nestin and β-tubulin III, and inhibited primary cilium formation. On the contrary, TNFα increased the expression of TNFα receptor TNFR2 and the mature neuron marker MAP2, also promoting neurite elongation. Moreover, TNFα affected nerve growth factor receptor expression. We also found that TNFα induced the expression of DNA-methylation enzymes and, accordingly, downregulated genes involved in neuronal development through epigenetic mechanisms, as demonstrated by methylome analysis. In summary, TNFα showed a dual role on hfNBMs phenotypic plasticity, exerting a negative influence on neurogenesis despite a positive effect on differentiation, through mechanisms that remain to be elucidated. Our results help to clarify the complexity of TNFα effects in human neurons and suggest that manipulation of TNFα signaling could provide a potential therapeutic approach against neurodegenerative disorders.
Collapse
Affiliation(s)
- Giulia Guarnieri
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.S.); (G.B.V.)
| | - Erica Sarchielli
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.S.); (G.B.V.)
| | - Paolo Comeglio
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | | | - Irene Piaceri
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50134 Florence, Italy; (I.P.); (B.N.)
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50134 Florence, Italy; (I.P.); (B.N.)
| | - Matteo Benelli
- Bioinformatics Unit, Hospital of Prato, Azienda USL Toscana Centro, 50122 Prato, Italy;
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK;
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 1TN, UK
| | - Mario Maggi
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Pasquale Gallina
- Neurosurgical Unit, Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, 50134 Florence, Italy;
| | - Gabriella Barbara Vannelli
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.S.); (G.B.V.)
| | - Annamaria Morelli
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.S.); (G.B.V.)
| |
Collapse
|
93
|
Ponvilawan B, Charoenngam N, Rittiphairoj T, Ungprasert P. Ankylosing spondylitis is associated with an increased risk of dementia: A systematic review and meta-analysis. Int J Rheum Dis 2020; 23:1452-1459. [PMID: 32715652 DOI: 10.1111/1756-185x.13920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/25/2020] [Accepted: 07/01/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVES To investigate the relationship between ankylosing spondylitis (AS) and risk of dementia. METHODS Potentially eligible studies that compared the risk of developing dementia between patients with AS and individuals without AS were identified from MEDLINE and EMBASE databases from inception to May 2019 using a search strategy that comprised terms for "dementia" and "ankylosing spondylitis". Eligible studies could be either cohort studies or case-control studies. For cohort design, eligible studies included patients with AS and comparators without AS and followed them for incident dementia. For case-control design, eligible studies included cases with dementia and controls without dementia and explored prior history of AS in both groups. Effect estimate and standard error from each study were extracted and combined together using the random effect, generic inverse variance. Funnel plot was used to assess for publication bias. RESULTS A total of 7091 articles were identified using the aforementioned search strategy. After 2 rounds of independent review by 3 investigators, 4 studies fulfilled the inclusion criteria and were included into the meta-analysis. The risk of developing dementia was significantly higher among patients with AS than individuals without AS with the pooled relative risk of 1.19 (95% CI 1.01-1.41; I2 = 76%). The funnel plot was relatively symmetric and was not suggestive of presence of publication bias. CONCLUSION A significantly 1.2 fold higher risk of developing dementia among patients with AS was demonstrated by this systematic review and meta-analysis.
Collapse
Affiliation(s)
- Ben Ponvilawan
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nipith Charoenngam
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thanitsara Rittiphairoj
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Patompong Ungprasert
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
94
|
Min SK, Kim JE, Hong JM, Yim JH, Park H, Youn UJ, Han SJ, Kim IC. Anti-inflammatory Effects of Lecania gerlachei Extract Collected from the Antarctic King Sejong Island. BIOTECHNOL BIOPROC E 2020. [DOI: 10.1007/s12257-019-0371-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
95
|
Sharma V, Kaur A, Singh TG. Counteracting role of nuclear factor erythroid 2-related factor 2 pathway in Alzheimer's disease. Biomed Pharmacother 2020; 129:110373. [PMID: 32603894 DOI: 10.1016/j.biopha.2020.110373] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/30/2022] Open
Abstract
A salient pathological features in Alzheimer's disease includes redox impairment and neuroinflammation. Nuclear factor erythroid 2-related factor 2 (Nrf2) and Nuclear factor kappa B (NF-ҡB) are the two key transcription factors that regulate cellular responses to redox impairment and neuroinflammation respectively. An effective way to confer neuroprotection in central nervous system (CNS) is the activation of a transcription factor i.e Nuclear factor erythroid 2-related factor 2 (Nrf2). An enhancer element known as Antioxidant Response Element (ARE) mediates the expression of phase II detoxification enzymes. Nrf2 is a nuclear transcription factor that binds to ARE thereby transcribing expression of several antioxidant genes. Kelch ECH associating protein-1 (Keap1), a culin 3-based E3 ligase, polyubiquitinates Nrf2 and targets it for its degradation. Disruption in the interaction between Keap1/Nrf2 can increase the brain's endogenous antioxidant capacity and thereby responsible for cell defence against oxidative stress and neuroinflammation in Alzheimer's disease (AD). The current review discusses about Keap1-Nrf2-ARE structure and function with special emphasis on the various pathways involved in positive and negative modulation of Nrf2, namely Phosphoinositide 3- kinase (PI3K), Glycogen synthase kinase-3β (GSK-3β), Nuclear factor kappa-b (NF-ҡb), Janus kinase/signal transducer and activator of transcription (JAK-STAT),Tumour Necrosis Factor- α (TNF-α), p38Mitogen-activated protein kinases (p38MAPK), Cyclic AMP response element binding protein (CREB) and intrinsic & extrinsic apoptotic pathway. Furthermore, this review highlights the miscellaneous Nrf2 activators as promising therapeutic agents for slowingdown the progression of AD.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | |
Collapse
|
96
|
Kim S, Nam Y, Kim C, Lee H, Hong S, Kim HS, Shin SJ, Park YH, Mai HN, Oh SM, Kim KS, Yoo DH, Chung WK, Chung H, Moon M. Neuroprotective and Anti-Inflammatory Effects of Low-Moderate Dose Ionizing Radiation in Models of Alzheimer's Disease. Int J Mol Sci 2020; 21:E3678. [PMID: 32456197 PMCID: PMC7279400 DOI: 10.3390/ijms21103678] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. The neuropathological features of AD include amyloid-β (Aβ) deposition and hyperphosphorylated tau accumulation. Although several clinical trials have been conducted to identify a cure for AD, no effective drug or treatment has been identified thus far. Recently, the potential use of non-pharmacological interventions to prevent or treat AD has gained attention. Low-dose ionizing radiation (LDIR) is a non-pharmacological intervention which is currently being evaluated in clinical trials for AD patients. However, the mechanisms underlying the therapeutic effects of LDIR therapy have not yet been established. In this study, we examined the effect of LDIR on Aβ accumulation and Aβ-mediated pathology. To investigate the short-term effects of low-moderate dose ionizing radiation (LMDIR), a total of 9 Gy (1.8 Gy per fraction for five times) were radiated to 4-month-old 5XFAD mice, an Aβ-overexpressing transgenic mouse model of AD, and then sacrificed at 4 days after last exposure to LMDIR. Comparing sham-exposed and LMDIR-exposed 5XFAD mice indicated that short-term exposure to LMDIR did not affect Aβ accumulation in the brain, but significantly ameliorated synaptic degeneration, neuronal loss, and neuroinflammation in the hippocampal formation and cerebral cortex. In addition, a direct neuroprotective effect was confirmed in SH-SY5Y neuronal cells treated with Aβ1-42 (2 μM) after single irradiation (1 Gy). In BV-2 microglial cells exposed to Aβ and/or LMDIR, LMDIR therapy significantly inhibited the production of pro-inflammatory molecules and activation of the nuclear factor-kappa B (NF-κB) pathway. These results indicate that LMDIR directly ameliorated neurodegeneration and neuroinflammation in vivo and in vitro. Collectively, our findings suggest that the therapeutic benefits of LMDIR in AD may be mediated by its neuroprotective and anti-inflammatory effects.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Chanyang Kim
- Department of Core Research Laboratory, Medical Science Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea;
| | - Hyewon Lee
- Department of Occupational Therapy, Konyang University, Daejeon 35365, Korea; (H.L.); (D.-H.Y.)
| | - Seojin Hong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Hyeon Soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Han Ngoc Mai
- Department of Radiation Oncology, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea;
| | - Sang-Muk Oh
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Kyoung Soo Kim
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University School of Medicine, Seoul 02447, Korea;
| | - Doo-Han Yoo
- Department of Occupational Therapy, Konyang University, Daejeon 35365, Korea; (H.L.); (D.-H.Y.)
| | - Weon Kuu Chung
- Department of Radiation Oncology, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea;
| | - Hyunju Chung
- Department of Core Research Laboratory, Medical Science Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea;
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| |
Collapse
|
97
|
Sasayama D, Hattori K, Yokota Y, Matsumura R, Teraishi T, Yoshida S, Kunugi H. Increased apolipoprotein E and decreased TNF-α in the cerebrospinal fluid of nondemented APOE-ε4 carriers. Neuropsychopharmacol Rep 2020; 40:201-205. [PMID: 32426945 PMCID: PMC7722685 DOI: 10.1002/npr2.12110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/30/2020] [Accepted: 04/10/2020] [Indexed: 01/12/2023] Open
Abstract
Aim The ε4 allele of apolipoprotein E gene (APOE) is a well‐known risk factor of late‐onset Alzheimer's disease. However, little is known why this variant confers a risk for Alzheimer's disease. The aim of this study was to examine the influence of the APOE genotype on cerebrospinal fluid (CSF) protein levels. Methods The present study performed a secondary analysis on our previously generated database to compare the CSF levels of 1128 proteins between APOE‐ε4 carriers (28 subjects) and noncarriers (104 subjects). All subjects were physically healthy Japanese individuals without dementia. Results CSF levels of apoE2, apoE3, and apoE4 were significantly higher (all nominal P < 10 × 10−5, false discovery rate < 0.001) and those of tumor necrosis factor‐α (TNF‐α) were significantly lower (nominal P = 1.39 × 10−6, false discovery rate < 0.001) in APOE‐ε4 carriers than in noncarriers. No significant correlation was observed between the CSF levels of TNF‐α and any of the apoE proteins. Conclusions Our findings indicate the possible roles of apoE and TNF‐α in the pathogenesis of APOE‐ε4‐associated Alzheimer's disease. The CSF levels of apoE2, apoE3, and apoE4 were higher in APOE‐ε4 carriers. The CSF levels of TNF‐α were significantly lower in APOE‐ε4 carriers. Our findings indicate the possible roles of apoE and TNF‐α in the pathogenesis of APOE‐ε4‐associated Alzheimer's disease.![]()
Collapse
Affiliation(s)
- Daimei Sasayama
- Department of Mental Disorder Research, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan.,Department of Psychiatry, Shinshu University School of Medicine, Matsumoto, Japan.,Child and Adolescent Developmental Psychiatry, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kotaro Hattori
- Department of Mental Disorder Research, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan.,Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Yuuki Yokota
- Department of Mental Disorder Research, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan.,Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Ryo Matsumura
- Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Toshiya Teraishi
- Department of Mental Disorder Research, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan
| | - Sumiko Yoshida
- Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Japan.,Department of Psychiatry, National Center of Neurology and Psychiatry, National Center Hospital, Kodaira, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan.,Department of Psychiatry, Teikyo University School of Medicine, Itabashi-ku, Japan
| |
Collapse
|
98
|
Pak VM, Onen SH, Bliwise DL, Kutner NG, Russell KL, Onen F. Sleep Disturbances in MCI and AD: Neuroinflammation as a Possible Mediating Pathway. Front Aging Neurosci 2020; 12:69. [PMID: 32457592 PMCID: PMC7227443 DOI: 10.3389/fnagi.2020.00069] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/26/2020] [Indexed: 12/11/2022] Open
Abstract
Mild cognitive impairment (MCI) and Alzheimer's disease (AD) affect a high proportion of the elderly population with an increasing prevalence. Sleep disturbances are frequent in those with MCI and AD. This review summarizes existing research on sleep disturbances and neuroinflammation in MCI and AD. Although strong evidence supports various pathways linking sleep and AD pathology, the temporal direction of this central relationship is not yet known. Improved understanding of sleep disturbance and neuroinflammation in MCI and AD may aid in the identification of targets for their prevention.
Collapse
Affiliation(s)
- Victoria M. Pak
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - S.-Hakki Onen
- Centre de Sommeil, Hôpital de la Croix-Rousse, Lyon, France
- INSERM U128, Université de Lyon, Lyon, France
| | - Donald L. Bliwise
- Department of Neurology, Emory University, Atlanta, GA, United States
| | - Nancy G. Kutner
- Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States
| | - Katherine L. Russell
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - Fannie Onen
- CHU Bichat–Claude-Bernard, AP-HP, Service de Gériatrie, Paris, France
- CESP & INSERM 1178 Université Paris Sud, Paris, France
| |
Collapse
|
99
|
Pardridge WM. The Isolated Brain Microvessel: A Versatile Experimental Model of the Blood-Brain Barrier. Front Physiol 2020; 11:398. [PMID: 32457645 PMCID: PMC7221163 DOI: 10.3389/fphys.2020.00398] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022] Open
Abstract
A versatile experimental model for the investigation of the blood-brain barrier (BBB), including the neuro-vascular unit, is the isolated brain microvessel preparation. Brain microvessels are primarily comprised of endothelial cells, but also include pericytes, pre-capillary arteriolar smooth muscle cells, astrocyte foot processes, and occasional nerve endings. These microvessels can be isolated from brain with a 3 h procedure, and the microvessels are free of brain parenchyma. Brain microvessels have been isolated from fresh animal brain, fresh human brain obtained at neurosurgery, as well as fresh or frozen autopsy human brain. Brain microvessels are the starting point for isolation of brain microvessel RNA, which then enables the production of BBB cDNA libraries and a genomics analysis of the brain microvasculature. Brain microvessels, combined with quantitative targeted absolute proteomics, allow for the quantitation of specific transporters or receptors expressed at the brain microvasculature. Brain microvessels, combined with specific antibodies and immune labeling of isolated capillaries, allow for the cellular location of proteins expressed within the neuro-vascular unit. Isolated brain microvessels can be used as an “in vitro” preparation of the BBB for the study of the kinetic parameters of BBB carrier-mediated transport (CMT) systems, or for the determination of dissociation constants of peptide binding to BBB receptor-mediated transport (RMT) systems expressed at either the animal or the human BBB. This review will discuss how the isolated brain microvessel model system has advanced our understanding of the organization and functional properties of the BBB, and highlight recent renewed interest in this 50 year old model of the BBB.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
100
|
Li Y, Ning L, Yin Y, Wang R, Zhang Z, Hao L, Wang B, Zhao X, Yang X, Yin L, Wu S, Guo D, Zhang C. Age-related shifts in gut microbiota contribute to cognitive decline in aged rats. Aging (Albany NY) 2020; 12:7801-7817. [PMID: 32357144 PMCID: PMC7244050 DOI: 10.18632/aging.103093] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/24/2020] [Indexed: 05/15/2023]
Abstract
Cognitive function declines during the aging process, meanwhile, gut microbiota of the elderly changed significantly. Although previous studies have reported the effect of gut microbiota on learning and memory, all the reports were based on various artificial interventions to change the gut microbiota without involvement of aging biological characteristics. Here, we investigated the effect of aged gut microbiota on cognitive function by using fecal microbiota transplantation (FMT) from aged to young rats. Results showed that FMT impaired cognitive behavior in young recipient rats; decreased the regional homogeneity in medial prefrontal cortex and hippocampus; changed synaptic structures and decreased dendritic spines; reduced expression of brain-derived neurotrophic factor (BDNF), N-methyl-D-aspartate receptor NR1 subunit, and synaptophysin; increased expression of advanced glycation end products (AGEs) and receptor for AGEs (RAGE). All these behavioral, brain structural and functional alterations induced by FMT reflected cognitive decline. In addition, FMT increased levels of pro-inflammatory cytokines and oxidative stress in young rats, indicating that inflammation and oxidative stress may underlie gut-related cognitive decline in aging. This study provides direct evidence for the contribution of gut microbiota to the cognitive decline during normal aging and suggests that restoring microbiota homeostasis in the elderly may improve cognitive function.
Collapse
Affiliation(s)
- Yanli Li
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Li Ning
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Yiru Yin
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Rui Wang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Zhiyong Zhang
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Lijun Hao
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Bin Wang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan 030024, Shanxi, P.R. China
| | - Xin Zhao
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Xiaorong Yang
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Litian Yin
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Shufen Wu
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Dawei Guo
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Ce Zhang
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| |
Collapse
|