1
|
Saroj S, Kana Veedu A, Reddy U C, Venkatesan N, Verma AK, Kannoth Manheri M. Modulation of Doxorubicin-Induced ROS Accumulation in Cardiomyocytes Using Ibuprofen-Conjugated Synthetic Lipids as Carriers. ACS APPLIED BIO MATERIALS 2025. [PMID: 40408375 DOI: 10.1021/acsabm.4c01932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Conjugation of an NSAID such as ibuprofen to the head group of oxanorbornane-based lipids and the use of their aggregates as carriers for doxorubicin (Dox) are discussed here. These conjugates were characterized by various spectroscopic techniques, including 2D-NMR, and insights into their assembly were gathered through PXRD, AFM, SEM, DLS, and qNano techniques. Free lipids as well as their formulations (lipid:cholesterol:Dox in a 3:1.5:2 molar ratio) showed a high tendency to form solid lipid particles, which was verified by TEM analysis. The presence of the ibuprofen unit led to an increase in interlipid spacing and a characteristic change in their packing. Active loading through a pH gradient allowed us to achieve high drug entrapment and a controlled release profile. The formulation AT3.3, prepared by this method, showed a Dox entrapment of ∼90%, with a controlled release of ∼18% by the end of 24 h; only ∼66% of the entrapped Dox was released by the end of 5 days. Cytotoxicity studies in NIH3T3 cells and hemolytic assay results showed that these lipids and their formulations have a good safety profile. Results from flow cytometry experiments in A549 cells revealed that the formulation AT3.3 induces effects similar to free Dox, with cell cycle arrest predominantly at the S phase and G2/M phase. At the same time, the response from the blank formulation was comparable to that of the control. Confocal microscopy studies in NIH3T3 and A549 cells showed that free Dox gets localized mainly in the nucleus, while the use of the carrier (AT3.3) causes significant localization of the drug on the cytoplasmic side as well. ROS induction due to free Dox and its formulation (AT3.3) in cardiomyocytes and A549 cells was also compared, and the results showed a protective effect in cardiomyocytes when using this formulation.
Collapse
Affiliation(s)
- Soumya Saroj
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Akshaya Kana Veedu
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Chandrasekhar Reddy U
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Nalini Venkatesan
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600 036, India
| | - Abhishek K Verma
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | | |
Collapse
|
2
|
Casquero-Veiga M, Ceron C, Cortes-Canteli M. Alzheimer's disease and vascular biology - A focus on the procoagulant state. Curr Opin Cell Biol 2025; 95:102528. [PMID: 40347710 DOI: 10.1016/j.ceb.2025.102528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
Alzheimer's disease (AD) is characterized by a multifactorial pathophysiology. Beyond its classical hallmarks, growing evidence highlights vascular contributions, including hemostatic dysregulation and a prothrombotic state in AD. This review focuses on recent findings concerning two key blood clot components-fibrin(ogen) and platelets-and their roles in AD pathology, including fibrinogen's abnormal accumulation in the AD brain, its interaction with amyloid-β, together with the associated impacts on clot stability, vascular occlusion, and neuroinflammation; and the potential switch of platelets along the AD continuum from protective to deleterious. This review provides an update on the interplay between vascular dysfunction and AD, underscoring the need for comprehensive integrative research to address AD's complexity and advocating for personalized approaches to tackle this multifaceted disorder.
Collapse
Affiliation(s)
- Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Carlos Ceron
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta Cortes-Canteli
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Centro Internacional de Neurociencia Cajal - Consejo Superior de Investigaciones Científicas (CINC - CSIC), Madrid, Spain.
| |
Collapse
|
3
|
Zuo Z, Zhang H, Li Z, Qi F, Hu H, Yang J, Yao Z. Activation of Hippocampal Neuronal NADPH Oxidase NOX2 Promotes Depressive-Like Behaviour and Cognition Deficits in Chronic Restraint Stress Mouse Model. PHARMACOPSYCHIATRY 2025; 58:117-126. [PMID: 39547705 DOI: 10.1055/a-2429-4023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
BACKGROUND Nicotinamide adenosine dinucleotide phosphate oxidases (NOX) play important roles in mediating stress-induced depression. Three NOX isotypes are expressed mainly in the brain: NOX2, NOX3 and NOX4. In this study, the expression and cellular sources of these NOX isoforms was investigated in the context of stress-induced depression. METHODS Chronic restraint stress (CRS)-induced depressive-like behaviour and cognitive deficits were evaluated by tail suspension tests, forced swimming tests and the Morris water maze test. Hippocampal NOX expression was determined by immunofluorescence staining and western blotting. The hippocampal levels of the brain-derived neurotrophic factor (BDNF) mRNA were determined via quantitative real-time -polymerase chain reaction. Glucocorticoid levels in the hippocampus were measured using ELISA kits. RESULTS In the mouse CRS model, a significant increase in NOX2 expression was observed in the hippocampus, whereas no significant changes in NOX3 and NOX4 expression were detected. Next, NOX2 expression was primarily localised to neurons (NeuN+) but not microglia (Iba-1+) or astrocytes (GFAP+). Treatment with gp91ds-tat, a specific NOX2 inhibitor, effectively mitigated the behavioural deficits induced by CRS. The decreased expression of the BDNF mRNA in the hippocampus of CRS mice was restored upon gp91ds-tat treatment. A positive correlation was identified between neuronal NOX2 expression and serum glucocorticoid levels. CONCLUSIONS Our study indicated that neuronal NOX2 may be a critical mediator of depression-like behaviours and spatial cognitive deficits in mice subjected to CRS. Blockade of NOX2 signalling may be a promising therapeutic strategy for depression.
Collapse
Affiliation(s)
- Zejie Zuo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongyang Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhihui Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Fangfang Qi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Haojie Hu
- Department of Psychology, College of Arts and Sciences, New York University, NY, USA
| | - Junhua Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhibin Yao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Rola M, Zielonka J, Smulik-Izydorczyk R, Pięta J, Pierzchała K, Sikora A, Michalski R. Boronate-Based Bioactive Compounds Activated by Peroxynitrite and Hydrogen Peroxide. REDOX BIOCHEMISTRY AND CHEMISTRY 2024; 10:100040. [PMID: 39678628 PMCID: PMC11637410 DOI: 10.1016/j.rbc.2024.100040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Boronates react directly and stoichiometrically with peroxynitrite and hydrogen peroxide. For this reason, boronates have been widely used as peroxynitrite- and hydrogen peroxide-sensitive moieties in various donors of bioactive compounds. So far, numerous boronate-based prodrugs and theranostics have been developed, characterized, and used in biological research. Here, the kinetic aspects of their activation are discussed, and the potential benefits of modifying their original structure with a boronic or boronobenzyl moiety are described.
Collapse
Affiliation(s)
- Monika Rola
- Institute of Applied Radiation Chemistry, Department of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Renata Smulik-Izydorczyk
- Institute of Applied Radiation Chemistry, Department of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Jakub Pięta
- Institute of Applied Radiation Chemistry, Department of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Karolina Pierzchała
- Institute of Applied Radiation Chemistry, Department of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Adam Sikora
- Institute of Applied Radiation Chemistry, Department of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Radosław Michalski
- Institute of Applied Radiation Chemistry, Department of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| |
Collapse
|
5
|
Kim J, Moon JS. Molecular Roles of NADPH Oxidase-Mediated Oxidative Stress in Alzheimer's Disease: Isoform-Specific Contributions. Int J Mol Sci 2024; 25:12299. [PMID: 39596364 PMCID: PMC11594809 DOI: 10.3390/ijms252212299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Oxidative stress is linked to the pathogenesis of Alzheimer's disease (AD), a neurodegenerative disorder marked by memory impairment and cognitive decline. AD is characterized by the accumulation of amyloid-beta (Aβ) plaques and the formation of neurofibrillary tangles (NFTs) of hyperphosphorylated tau. AD is associated with an imbalance in redox states and excessive reactive oxygen species (ROS). Recent studies report that NADPH oxidase (NOX) enzymes are significant contributors to ROS generation in neurodegenerative diseases, including AD. NOX-derived ROS aggravates oxidative stress and neuroinflammation during AD. In this review, we provide the potential role of all NOX isoforms in AD pathogenesis and their respective structural involvement in AD progression, highlighting NOX enzymes as a strategic therapeutic target. A comprehensive understanding of NOX isoforms and their inhibitors could provide valuable insights into AD pathology and aid in the development of targeted treatments for AD.
Collapse
Affiliation(s)
- Junhyung Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea;
| | - Jong-Seok Moon
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea;
- Department of Pathology, College of Medicine, Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
6
|
Amelimojarad M, Amelimojarad M, Cui X. The emerging role of brain neuroinflammatory responses in Alzheimer's disease. Front Aging Neurosci 2024; 16:1391517. [PMID: 39021707 PMCID: PMC11253199 DOI: 10.3389/fnagi.2024.1391517] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
As the most common cause of dementia, Alzheimer's disease (AD) is characterized by neurodegeneration and synaptic loss with an increasing prevalence in the elderly. Increased inflammatory responses triggers brain cells to produce pro-inflammatory cytokines and accelerates the Aβ accumulation, tau protein hyper-phosphorylation leading to neurodegeneration. Therefore, in this paper, we discuss the current understanding of how inflammation affects brain activity to induce AD pathology, the inflammatory biomarkers and possible therapies that combat inflammation for AD.
Collapse
Affiliation(s)
| | | | - Xiaonan Cui
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Golzari-Sorkheh M, Liyanage I, Reed MA, Weaver DF. Alzheimer's Disease and COVID-19 Pathogenic Overlap: Implications for Drug Repurposing. Can J Neurol Sci 2024; 51:161-172. [PMID: 36991574 DOI: 10.1017/cjn.2023.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
As COVID-19 continues, a safe, cost-effective treatment strategy demands continued inquiry. Chronic neuroinflammatory disorders may appear to be of little relevance in this regard; often indolent and progressive disorders characterized by neuroinflammation (such as Alzheimer's disease (AD)) are fundamentally dissimilar in etiology and symptomology to COVID-19's rapid infectivity and pathology. However, the two disorders share extensive pathognomonic features, including at membrane, cytoplasmic, and extracellular levels, culminating in analogous immunogenic destruction of their respective organ parenchyma. We hypothesize that these mechanistic similarities may extent to therapeutic targets, namely that it is conceivable an agent against AD's immunopathy may have efficacy against COVID-19 and vice versa. It is notable that while extensively investigated, no agent has yet demonstrated significant therapeutic efficacy against AD's cognitive and memory declines. Yet this very failure has driven the development of numerous agents with strong mechanistic potential and clinical characteristics. Having already approved for clinical trials, these agents may be an expedient starting point in the urgent search for an effective COVID-19 therapy. Herein, we review the overlapping Alzheimer's/ COVID-19 targets and theorize several initial platforms.
Collapse
Affiliation(s)
| | - Imindu Liyanage
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Mark A Reed
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Donald F Weaver
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Zhang Z, Sun J, Li Y, Yang K, Wei G, Zhang S. Ameliorative effects of pine nut peptide-zinc chelate (Korean pine) on a mouse model of Alzheimer's disease. Exp Gerontol 2023; 183:112308. [PMID: 37821052 DOI: 10.1016/j.exger.2023.112308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/17/2023] [Accepted: 10/07/2023] [Indexed: 10/13/2023]
Abstract
In this study, 50 SD adult male mice were used to create an Alzheimer's disease model. The mice's learning and memory abilities were evaluated using an eight-arm radial maze experiment, and changes in body weight and food intake were noted. This helped to better validate the improvement of Alzheimer's disease caused by pine nut peptide-zinc chelate (Korean pine). For a more thorough investigation, mice's brains were dissected, Endogenous mercaptan antioxidants (enzymes), which are markers of brain tissue, were assessed, and mouse gut flora was analyzed. The findings demonstrated that pine nut peptide-zinc chelate (Korean pine) can improve learning and memory, stop brain aging and damage, and control gut flora in mice. It may exert its effects by ameliorating decreased AChE levels and increased ChAT levels in the central cholinergic system, endogenous thiol antioxidants (enzymes) in the cerebral cortex, and by controlling the bacterial flora in the gut.
Collapse
Affiliation(s)
- Zhi Zhang
- College of Life Sciences, Northeast Forestry University
| | - Jiajia Sun
- College of Forestry, Northeast Forestry University.
| | - Yanxia Li
- Forestry Research Institute of Heilongjiang Province.
| | - Kexin Yang
- College of Forestry, Northeast Forestry University
| | - Gang Wei
- College of Forestry, Northeast Forestry University
| | - Shenglong Zhang
- Heilongjiang Guohong Energy Saving and Environmental Protection Co
| |
Collapse
|
9
|
Beura SK, Dhapola R, Panigrahi AR, Yadav P, Kumar R, Reddy DH, Singh SK. Antiplatelet drugs: Potential therapeutic options for the management of neurodegenerative diseases. Med Res Rev 2023; 43:1835-1877. [PMID: 37132460 DOI: 10.1002/med.21965] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/13/2023] [Accepted: 04/12/2023] [Indexed: 05/04/2023]
Abstract
The blood platelet plays an important role but often remains under-recognized in several vascular complications and associated diseases. Surprisingly, platelet hyperactivity and hyperaggregability have often been considered the critical risk factors for developing vascular dysfunctions in several neurodegenerative diseases (NDDs) like Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. In addition, platelet structural and functional impairments promote prothrombotic and proinflammatory environment that can aggravate the progression of several NDDs. These findings provide the rationale for using antiplatelet agents not only to prevent morbidity but also to reduce mortality caused by NDDs. Therefore, we thoroughly review the evidence supporting the potential pleiotropic effects of several novel classes of synthetic antiplatelet drugs, that is, cyclooxygenase inhibitors, adenosine diphosphate receptor antagonists, protease-activated receptor blockers, and glycoprotein IIb/IIIa receptor inhibitors in NDDs. Apart from this, the review also emphasizes the recent developments of selected natural antiplatelet phytochemicals belonging to key classes of plant-based bioactive compounds, including polyphenols, alkaloids, terpenoids, and flavonoids as potential therapeutic candidates in NDDs. We believe that the broad analysis of contemporary strategies and specific approaches for plausible therapeutic treatment for NDDs presented in this review could be helpful for further successful research in this area.
Collapse
Affiliation(s)
- Samir K Beura
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Rishika Dhapola
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Abhishek R Panigrahi
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Pooja Yadav
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Reetesh Kumar
- Department of Agricultural Sciences, Institute of Applied Sciences and Humanities, GLA University, Mathura, Uttar Pradesh, India
| | - Dibbanti H Reddy
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Sunil K Singh
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| |
Collapse
|
10
|
Ng PY, McNeely TL, Baker DJ. Untangling senescent and damage-associated microglia in the aging and diseased brain. FEBS J 2023; 290:1326-1339. [PMID: 34873840 PMCID: PMC9167891 DOI: 10.1111/febs.16315] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/16/2021] [Accepted: 12/06/2021] [Indexed: 01/10/2023]
Abstract
Microglial homeostasis has emerged as a critical mediator of health and disease in the central nervous system. In their neuroprotective role as the predominant immune cells of the brain, microglia surveil the microenvironment for debris and pathogens, while also promoting neurogenesis and performing maintenance on synapses. Chronological ageing, disease onset, or traumatic injury promotes irreparable damage or deregulated signaling to reinforce neurotoxic phenotypes in microglia. These insults may include cellular senescence, a stable growth arrest often accompanied by the production of a distinctive pro-inflammatory secretory phenotype, which may contribute to age- or disease-driven decline in neuronal health and cognition and is a potential novel therapeutic target. Despite this increased scrutiny, unanswered questions remain about what distinguishes senescent microglia and non-senescent microglia reacting to insults occurring in ageing, disease, and injury, and how central the development of senescence is in their pivot from guardian to assailant. To intelligently design future studies to untangle senescent microglia from other primed and reactionary states, specific criteria must be developed that define this population and allow for comparisons between different model systems. Comparing microglial activity seen in homeostasis, ageing, disease, and injury allows for a more coherent understanding of when and how senescent and other harmful microglial subpopulations should be targeted.
Collapse
Affiliation(s)
- Pei Y Ng
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Taylor L McNeely
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Darren J Baker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.,Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
11
|
Bubley A, Erofeev A, Gorelkin P, Beloglazkina E, Majouga A, Krasnovskaya O. Tacrine-Based Hybrids: Past, Present, and Future. Int J Mol Sci 2023; 24:ijms24021717. [PMID: 36675233 PMCID: PMC9863713 DOI: 10.3390/ijms24021717] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder which is characterized by β-amyloid (Aβ) aggregation, τ-hyperphosphorylation, and loss of cholinergic neurons. The other important hallmarks of AD are oxidative stress, metal dyshomeostasis, inflammation, and cell cycle dysregulation. Multiple therapeutic targets may be proposed for the development of anti-AD drugs, and the "one drug-multiple targets" strategy is of current interest. Tacrine (THA) was the first clinically approved cholinesterase (ChE) inhibitor, which was withdrawn due to high hepatotoxicity. However, its high potency in ChE inhibition, low molecular weight, and simple structure make THA a promising scaffold for developing multi-target agents. In this review, we summarized THA-based hybrids published from 2006 to 2022, thus providing an overview of strategies that have been used in drug design and approaches that have resulted in significant cognitive improvements and reduced hepatotoxicity.
Collapse
Affiliation(s)
- Anna Bubley
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1-3, Moscow 119991, Russia
| | - Alexaner Erofeev
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
| | - Peter Gorelkin
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
| | - Elena Beloglazkina
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1-3, Moscow 119991, Russia
| | - Alexander Majouga
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
| | - Olga Krasnovskaya
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1-3, Moscow 119991, Russia
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
- Correspondence:
| |
Collapse
|
12
|
Smith AN, Shaughness M, Collier S, Hopkins D, Byrnes KR. Therapeutic targeting of microglia mediated oxidative stress after neurotrauma. Front Med (Lausanne) 2022; 9:1034692. [PMID: 36405593 PMCID: PMC9671221 DOI: 10.3389/fmed.2022.1034692] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 10/06/2023] Open
Abstract
Inflammation is a primary component of the central nervous system injury response. Traumatic brain and spinal cord injury are characterized by a pronounced microglial response to damage, including alterations in microglial morphology and increased production of reactive oxygen species (ROS). The acute activity of microglia may be beneficial to recovery, but continued inflammation and ROS production is deleterious to the health and function of other cells. Microglial nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX), mitochondria, and changes in iron levels are three of the most common sources of ROS. All three play a significant role in post-traumatic brain and spinal cord injury ROS production and the resultant oxidative stress. This review will evaluate the current state of therapeutics used to target these avenues of microglia-mediated oxidative stress after injury and suggest avenues for future research.
Collapse
Affiliation(s)
- Austin N. Smith
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Michael Shaughness
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Sean Collier
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Deanna Hopkins
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Kimberly R. Byrnes
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
13
|
Araújo B, Caridade-Silva R, Soares-Guedes C, Martins-Macedo J, Gomes ED, Monteiro S, Teixeira FG. Neuroinflammation and Parkinson's Disease-From Neurodegeneration to Therapeutic Opportunities. Cells 2022; 11:cells11182908. [PMID: 36139483 PMCID: PMC9497016 DOI: 10.3390/cells11182908] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease (PD) is the second most prevalent neurodegenerative disorder worldwide. Clinically, it is characterized by a progressive degeneration of dopaminergic neurons (DAn), resulting in severe motor complications. Preclinical and clinical studies have indicated that neuroinflammation can play a role in PD pathophysiology, being associated with its onset and progression. Nevertheless, several key points concerning the neuroinflammatory process in PD remain to be answered. Bearing this in mind, in the present review, we cover the impact of neuroinflammation on PD by exploring the role of inflammatory cells (i.e., microglia and astrocytes) and the interconnections between the brain and the peripheral system. Furthermore, we discuss both the innate and adaptive immune responses regarding PD pathology and explore the gut–brain axis communication and its influence on the progression of the disease.
Collapse
Affiliation(s)
- Bruna Araújo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Rita Caridade-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Carla Soares-Guedes
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana Martins-Macedo
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Eduardo D. Gomes
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
| | - Fábio G. Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Correspondence:
| |
Collapse
|
14
|
Kang K, Jeong SY, Park K, Hahm MH, Kim J, Lee H, Kim C, Yun E, Han J, Yoon U, Lee S. Distinct cerebral cortical perfusion patterns in idiopathic normal-pressure hydrocephalus. Hum Brain Mapp 2022; 44:269-279. [PMID: 36102811 PMCID: PMC9783416 DOI: 10.1002/hbm.25974] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/29/2022] [Accepted: 05/12/2022] [Indexed: 02/05/2023] Open
Abstract
The aims of the study are to evaluate idiopathic normal-pressure hydrocephalus (INPH)-related cerebral blood flow (CBF) abnormalities and to investigate their relation to cortical thickness in INPH patients. We investigated cortical CBF utilizing surface-based early-phase 18 F-florbetaben (E-FBB) PET analysis in two groups: INPH patients and healthy controls. All 39 INPH patients and 20 healthy controls were imaged with MRI, including three-dimensional volumetric images, for automated surface-based cortical thickness analysis across the entire brain. A subgroup with 37 participants (22 INPH patients and 15 healthy controls) that also underwent 18 F-fluorodeoxyglucose (FDG) PET imaging was further analyzed. Compared with age- and gender-matched healthy controls, INPH patients showed statistically significant hyperperfusion in the high convexity of the frontal and parietal cortical regions. Importantly, within the INPH group, increased perfusion correlated with cortical thickening in these regions. Additionally, significant hypoperfusion mainly in the ventrolateral frontal cortex, supramarginal gyrus, and temporal cortical regions was observed in the INPH group relative to the control group. However, this hypoperfusion was not associated with cortical thinning. A subgroup analysis of participants that also underwent FDG PET imaging showed that increased (or decreased) cerebral perfusion was associated with increased (or decreased) glucose metabolism in INPH. A distinctive regional relationship between cerebral cortical perfusion and cortical thickness was shown in INPH patients. Our findings suggest distinct pathophysiologic mechanisms of hyperperfusion and hypoperfusion in INPH patients.
Collapse
Affiliation(s)
- Kyunghun Kang
- Department of Neurology, School of MedicineKyungpook National UniversityDaeguSouth Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, School of MedicineKyungpook National UniversityDaeguSouth Korea
| | - Ki‐Su Park
- Department of Neurosurgery, School of MedicineKyungpook National UniversityDaeguSouth Korea
| | - Myong Hun Hahm
- Department of Radiology, School of MedicineKyungpook National UniversityDaeguSouth Korea
| | - Jaeil Kim
- School of Computer Science and EngineeringKyungpook National UniversityDaeguSouth Korea
| | - Ho‐Won Lee
- Department of Neurology, School of MedicineKyungpook National UniversityDaeguSouth Korea,Brain Science and Engineering InstituteKyungpook National UniversityDaeguSouth Korea
| | - Chi‐Hun Kim
- Department of NeurologyHallym University Sacred Heart HospitalAnyangSouth Korea
| | - Eunkyeong Yun
- Department of Biomedical EngineeringDaegu Catholic UniversityGyeongsan‐siSouth Korea
| | - Jaehwan Han
- Department of Biomedical EngineeringDaegu Catholic UniversityGyeongsan‐siSouth Korea
| | - Uicheul Yoon
- Department of Biomedical EngineeringDaegu Catholic UniversityGyeongsan‐siSouth Korea
| | - Sang‐Woo Lee
- Department of Nuclear Medicine, School of MedicineKyungpook National UniversityDaeguSouth Korea
| |
Collapse
|
15
|
Al-Ghraiybah NF, Wang J, Alkhalifa AE, Roberts AB, Raj R, Yang E, Kaddoumi A. Glial Cell-Mediated Neuroinflammation in Alzheimer's Disease. Int J Mol Sci 2022; 23:10572. [PMID: 36142483 PMCID: PMC9502483 DOI: 10.3390/ijms231810572] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder; it is the most common cause of dementia and has no treatment. It is characterized by two pathological hallmarks, the extracellular deposits of amyloid beta (Aβ) and the intraneuronal deposits of Neurofibrillary tangles (NFTs). Yet, those two hallmarks do not explain the full pathology seen with AD, suggesting the involvement of other mechanisms. Neuroinflammation could offer another explanation for the progression of the disease. This review provides an overview of recent advances on the role of the immune cells' microglia and astrocytes in neuroinflammation. In AD, microglia and astrocytes become reactive by several mechanisms leading to the release of proinflammatory cytokines that cause further neuronal damage. We then provide updates on neuroinflammation diagnostic markers and investigational therapeutics currently in clinical trials to target neuroinflammation.
Collapse
Affiliation(s)
- Nour F. Al-Ghraiybah
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Junwei Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amer E. Alkhalifa
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Andrew B. Roberts
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Ruchika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Euitaek Yang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| |
Collapse
|
16
|
Targeting Microglia in Alzheimer’s Disease: From Molecular Mechanisms to Potential Therapeutic Targets for Small Molecules. Molecules 2022; 27:molecules27134124. [PMID: 35807370 PMCID: PMC9268715 DOI: 10.3390/molecules27134124] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a common, progressive, and devastating neurodegenerative disorder that mainly affects the elderly. Microglial dysregulation, amyloid-beta (Aβ) plaques, and intracellular neurofibrillary tangles play crucial roles in the pathogenesis of AD. In the brain, microglia play roles as immune cells to provide protection against virus injuries and diseases. They have significant contributions in the development of the brain, cognition, homeostasis of the brain, and plasticity. Multiple studies have confirmed that uncontrolled microglial function can result in impaired microglial mitophagy, induced Aβ accumulation and tau pathology, and a chronic neuroinflammatory environment. In the brain, most of the genes that are associated with AD risk are highly expressed by microglia. Although it was initially regarded that microglia reaction is incidental and induced by dystrophic neurites and Aβ plaques. Nonetheless, it has been reported by genome-wide association studies that most of the risk loci for AD are located in genes that are occasionally uniquely and highly expressed in microglia. This finding further suggests that microglia play significant roles in early AD stages and they be targeted for the development of novel therapeutics. In this review, we have summarized the molecular pathogenesis of AD, microglial activities in the adult brain, the role of microglia in the aging brain, and the role of microglia in AD. We have also particularly focused on the significance of targeting microglia for the treatment of AD.
Collapse
|
17
|
Ibuprofen Favors Binding of Amyloid-β Peptide to Its Depot, Serum Albumin. Int J Mol Sci 2022; 23:ijms23116168. [PMID: 35682848 PMCID: PMC9181795 DOI: 10.3390/ijms23116168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/24/2022] [Accepted: 05/28/2022] [Indexed: 12/15/2022] Open
Abstract
The deposition of amyloid-β peptide (Aβ) in the brain is a critical event in the progression of Alzheimer’s disease (AD). This Aβ deposition could be prevented by directed enhancement of Aβ binding to its natural depot, human serum albumin (HSA). Previously, we revealed that specific endogenous ligands of HSA improve its affinity to monomeric Aβ. We show here that an exogenous HSA ligand, ibuprofen (IBU), exerts the analogous effect. Plasmon resonance spectroscopy data evidence that a therapeutic IBU level increases HSA affinity to monomeric Aβ40/Aβ42 by a factor of 3–5. Using thioflavin T fluorescence assay and transmission electron microcopy, we show that IBU favors the suppression of Aβ40 fibrillation by HSA. Molecular docking data indicate partial overlap between the IBU/Aβ40-binding sites of HSA. The revealed enhancement of the HSA–Aβ interaction by IBU and the strengthened inhibition of Aβ fibrillation by HSA in the presence of IBU could contribute to the neuroprotective effects of the latter, previously observed in mouse and human studies of AD.
Collapse
|
18
|
Vav Proteins in Development of the Brain: A Potential Relationship to the Pathogenesis of Congenital Zika Syndrome? Viruses 2022; 14:v14020386. [PMID: 35215978 PMCID: PMC8874935 DOI: 10.3390/v14020386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 12/07/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy can result in a significant impact on the brain and eye of the developing fetus, termed congenital zika syndrome (CZS). At a morphological level, the main serious presentations of CZS are microcephaly and retinal scarring. At a cellular level, many cell types of the brain may be involved, but primarily neuronal progenitor cells (NPC) and developing neurons. Vav proteins have guanine exchange activity in converting GDP to GTP on proteins such as Rac1, Cdc42 and RhoA to stimulate intracellular signaling pathways. These signaling pathways are known to play important roles in maintaining the polarity and self-renewal of NPC pools by coordinating the formation of adherens junctions with cytoskeletal rearrangements. In developing neurons, these same pathways are adopted to control the formation and growth of neurites and mediate axonal guidance and targeting in the brain and retina. This review describes the role of Vavs in these processes and highlights the points of potential ZIKV interaction, such as (i) the binding and entry of ZIKV in cells via TAM receptors, which may activate Vav/Rac/RhoA signaling; (ii) the functional convergence of ZIKV NS2A with Vav in modulating adherens junctions; (iii) ZIKV NS4A/4B protein effects on PI3K/AKT in a regulatory loop via PPI3 to influence Vav/Rac1 signaling in neurite outgrowth; and (iv) the induction of SOCS1 and USP9X following ZIKV infection to regulate Vav protein degradation or activation, respectively, and impact Vav/Rac/RhoA signaling in NPC and neurons. Experiments to define these interactions will further our understanding of the molecular basis of CZS and potentially other developmental disorders stemming from in utero infections. Additionally, Vav/Rac/RhoA signaling pathways may present tractable targets for therapeutic intervention or molecular rationale for disease severity in CZS.
Collapse
|
19
|
Little K, Llorián-Salvador M, Scullion S, Hernández C, Simó-Servat O, Del Marco A, Bosma E, Vargas-Soria M, Carranza-Naval MJ, Van Bergen T, Galbiati S, Viganò I, Musi CA, Schlingemann R, Feyen J, Borsello T, Zerbini G, Klaassen I, Garcia-Alloza M, Simó R, Stitt AW. Common pathways in dementia and diabetic retinopathy: understanding the mechanisms of diabetes-related cognitive decline. Trends Endocrinol Metab 2022; 33:50-71. [PMID: 34794851 DOI: 10.1016/j.tem.2021.10.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/06/2021] [Accepted: 10/29/2021] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes (T2D) is associated with multiple comorbidities, including diabetic retinopathy (DR) and cognitive decline, and T2D patients have a significantly higher risk of developing Alzheimer's disease (AD). Both DR and AD are characterized by a number of pathological mechanisms that coalesce around the neurovascular unit, including neuroinflammation and degeneration, vascular degeneration, and glial activation. Chronic hyperglycemia and insulin resistance also play a significant role, leading to activation of pathological mechanisms such as increased oxidative stress and the accumulation of advanced glycation end-products (AGEs). Understanding these common pathways and the degree to which they occur simultaneously in the brain and retina during diabetes will provide avenues to identify T2D patients at risk of cognitive decline.
Collapse
Affiliation(s)
- Karis Little
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - María Llorián-Salvador
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Sarah Scullion
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Cristina Hernández
- Vall d'Hebron Research Institute and CIBERDEM (ISCIII), Barcelona, Spain
| | - Olga Simó-Servat
- Vall d'Hebron Research Institute and CIBERDEM (ISCIII), Barcelona, Spain
| | - Angel Del Marco
- Division of Physiology, School of Medicine, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
| | - Esmeralda Bosma
- Ocular Angiogenesis Group, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Vargas-Soria
- Division of Physiology, School of Medicine, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
| | - Maria Jose Carranza-Naval
- Division of Physiology, School of Medicine, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
| | | | - Silvia Galbiati
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Ilaria Viganò
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Clara Alice Musi
- Università Degli Studi di Milano and Istituto di Ricerche Farmacologiche Mario Negri- IRCCS, Milano, Italy
| | - Reiner Schlingemann
- Ocular Angiogenesis Group, University of Amsterdam, Amsterdam, The Netherlands; Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Lausanne, Switzerland
| | | | - Tiziana Borsello
- Università Degli Studi di Milano and Istituto di Ricerche Farmacologiche Mario Negri- IRCCS, Milano, Italy
| | - Gianpaolo Zerbini
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, University of Amsterdam, Amsterdam, The Netherlands
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
| | - Rafael Simó
- Vall d'Hebron Research Institute and CIBERDEM (ISCIII), Barcelona, Spain.
| | - Alan W Stitt
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK.
| | | |
Collapse
|
20
|
Zhang G, Wang Z, Hu H, Zhao M, Sun L. Microglia in Alzheimer's Disease: A Target for Therapeutic Intervention. Front Cell Neurosci 2021; 15:749587. [PMID: 34899188 PMCID: PMC8651709 DOI: 10.3389/fncel.2021.749587] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/28/2021] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most common types of age-related dementia worldwide. In addition to extracellular amyloid plaques and intracellular neurofibrillary tangles, dysregulated microglia also play deleterious roles in the AD pathogenesis. Numerous studies have demonstrated that unbridled microglial activity induces a chronic neuroinflammatory environment, promotes β-amyloid accumulation and tau pathology, and impairs microglia-associated mitophagy. Thus, targeting microglia may pave the way for new therapeutic interventions. This review provides a thorough overview of the pathophysiological role of the microglia in AD and illustrates the potential avenues for microglia-targeted therapies, including microglial modification, immunoreceptors, and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Guimei Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Zicheng Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Huiling Hu
- Department of Intensive Care Unit, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Meng Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
21
|
Sindona C, Schepici G, Contestabile V, Bramanti P, Mazzon E. NOX2 Activation in COVID-19: Possible Implications for Neurodegenerative Diseases. ACTA ACUST UNITED AC 2021; 57:medicina57060604. [PMID: 34208136 PMCID: PMC8230853 DOI: 10.3390/medicina57060604] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 12/11/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a rapidly spreading contagious infectious disease caused by the pathogen severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), that primarily affects the respiratory tract as well as the central nervous system (CNS). SARS-CoV-2 infection occurs through the interaction of the viral protein Spike with the angiotensin II receptor (ACE 2), leading to an increase of angiotensin II and activation of nicotinamide adenine dinucleotide phosphate oxidase2 (NOX2), resulting in the release of both reactive oxygen species (ROS) and inflammatory molecules. The purpose of the review is to explain that SARS-CoV-2 infection can determine neuroinflammation that induces NOX2 activation in microglia. To better understand the role of NOX2 in inflammation, an overview of its involvement in neurodegenerative diseases (NDs) such as Parkinson’s disease (PD), Alzheimer’s disease (AD), and amyotrophic lateral sclerosis (ALS) is provided. To write this manuscript, we performed a PubMed search to evaluate the possible relationship of SARS-CoV-2 infection in NOX2 activation in microglia, as well as the role of NOX2 in NDs. Several studies highlighted that NOX2 activation in microglia amplifies neuroinflammation. To date, there is no clinical treatment capable of counteracting its activation, however, NOX2 could be a promising pharmaceutical target useful for both the treatment and prevention of NDs and COVID-19 treatment.
Collapse
|
22
|
Upadhyay A, Amanullah A, Joshi V, Dhiman R, Prajapati VK, Poluri KM, Mishra A. Ibuprofen-based advanced therapeutics: breaking the inflammatory link in cancer, neurodegeneration, and diseases. Drug Metab Rev 2021; 53:100-121. [PMID: 33820460 DOI: 10.1080/03602532.2021.1903488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ibuprofen is a classical nonsteroidal anti-inflammatory drug (NSAID) highly prescribed to reduce acute pain and inflammation under an array of conditions, including rheumatoid arthritis, osteoarthritis, dysmenorrhea, and gout. Ibuprofen acts as a potential inhibitor for cyclooxygenase enzymes (COX-1 and COX-2). In the past few decades, research on this small molecule has led to identifying other possible therapeutic benefits. Anti-tumorigenic and neuroprotective functions of Ibuprofen are majorly recognized in recent literature and need further consideration. Additionally, several other roles of this anti-inflammatory molecule have been discovered and subjected to experimental assessment in various diseases. However, the major challenge faced by Ibuprofen and other drugs of similar classes is their side effects, and tendency to cause gastrointestinal injury, generate cardiovascular risks, modulate hepatic and acute kidney diseases. Future research should also be conducted to deduce new methods and approaches of suppressing the unwanted toxic changes mediated by these drugs and develop new therapeutic avenues so that these small molecules continue to serve the purposes. This article primarily aims to develop a comprehensive and better understanding of Ibuprofen, its pharmacological features, therapeutic benefits, and possible but less understood medicinal properties apart from major challenges in its future application.KEY POINTSIbuprofen, an NSAID, is a classical anti-inflammatory therapeutic agent.Pro-apoptotic roles of NSAIDs have been explored in detail in the past, holding the key in anti-cancer therapies.Excessive and continuous use of NSAIDs may have several side effects and multiple organ damage.Hyperactivated Inflammation initiates multifold detrimental changes in multiple pathological conditions.Targeting inflammatory pathways hold the key to several therapeutic strategies against many diseases, including cancer, microbial infections, multiple sclerosis, and many other brain diseases.
Collapse
Affiliation(s)
- Arun Upadhyay
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Ayeman Amanullah
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Vibhuti Joshi
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Krishna Mohan Poluri
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| |
Collapse
|
23
|
Wickstead ES, Irving MA, Getting SJ, McArthur S. Exploiting formyl peptide receptor 2 to promote microglial resolution: a new approach to Alzheimer's disease treatment. FEBS J 2021; 289:1801-1822. [PMID: 33811735 DOI: 10.1111/febs.15861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/09/2021] [Accepted: 03/29/2021] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease and dementia are among the most significant current healthcare challenges given the rapidly growing elderly population, and the almost total lack of effective therapeutic interventions. Alzheimer's disease pathology has long been considered in terms of accumulation of amyloid beta and hyperphosphorylated tau, but the importance of neuroinflammation in driving disease has taken greater precedence over the last 15-20 years. Inflammatory activation of the primary brain immune cells, the microglia, has been implicated in Alzheimer's pathogenesis through genetic, preclinical, imaging and postmortem human studies, and strategies to regulate microglial activity may hold great promise for disease modification. Neuroinflammation is necessary for defence of the brain against pathogen invasion or damage but is normally self-limiting due to the engagement of endogenous pro-resolving circuitry that terminates inflammatory activity, a process that appears to fail in Alzheimer's disease. Here, we discuss the potential for a major regulator and promoter of resolution, the receptor FPR2, to restrain pro-inflammatory microglial activity, and propose that it may serve as a valuable target for therapeutic investigation in Alzheimer's disease.
Collapse
Affiliation(s)
| | - Murray A Irving
- Institute of Dentistry, Barts and the London School of Medicine & Dentistry, Blizard Institute, Queen Mary, University of London, UK
| | - Stephen J Getting
- College of Liberal Arts & Sciences, School of Life Sciences, University of Westminster, London, UK
| | - Simon McArthur
- Institute of Dentistry, Barts and the London School of Medicine & Dentistry, Blizard Institute, Queen Mary, University of London, UK
| |
Collapse
|
24
|
Nozari M, Nahavandi A, Zeinivand M, Eslami Gharaati M, Godarzi M, Ahmadi M, Jamali-Raeufy N. Ibuprofen Protection Against Restrained Chronic Stress-induced Depression in Male Rats. Basic Clin Neurosci 2021; 11:413-422. [PMID: 33613879 PMCID: PMC7878046 DOI: 10.32598/bcn.11.4.1775.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/10/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022] Open
Abstract
Introduction: Stress predisposes organisms to depression and cognitive impairments, and seems to interact with metabolic homeostasis. The inflammatory response and the upregulation of proinflammatory cytokines are some of the consequences related to chronic stress. In this study, we investigated the preventive effect of chronic administration of ibuprofen, as an inhibitor of cyclooxygenases, on the cognitive and behavioral alterations and the weight gain reduction induced by simultaneous chronic restraint stress in rats. Materials and Methods: Male Wistar rats were subjected to chronic restraint stress and injected daily with the variable doses of ibuprofen or vehicle, for 21 consecutive days. Then, all animals were tested with the forced swim test and passive avoidance conditioning. Also, the weight of the animals was recorded before and after the interventions. Ultimately, plasma interleukin 6 (IL-6) levels were measured. Results: Chronic stress increased depressive-like behaviors, impaired learning, and disrupted the normal weight gain. However, the animals that received the highest dose of ibuprofen showed less depressive-like behaviors, a better avoidance memory, and a higher weight gain. However, the level of plasma IL-6 did not differ significantly between the study groups. Conclusion: The administration of ibuprofen prevents the cognitive and behavioral consequences of chronic stress. During the recovery, the plasma levels of IL-6 were not elevated by stress, and the IL-6 levels did not predict the behavioral performance of the stressed animals. The exact mechanisms of the protective effects of ibuprofen against chronic stress need to be further investigated.
Collapse
Affiliation(s)
- Masoumeh Nozari
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Arezo Nahavandi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Motahareh Zeinivand
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Eslami Gharaati
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mina Godarzi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ahmadi
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nida Jamali-Raeufy
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Lebedev M, McEligot HA, Mutua VN, Walsh P, Carvallo Chaigneau FR, Gershwin LJ. Analysis of lung transcriptome in calves infected with Bovine Respiratory Syncytial Virus and treated with antiviral and/or cyclooxygenase inhibitor. PLoS One 2021; 16:e0246695. [PMID: 33600498 PMCID: PMC7891793 DOI: 10.1371/journal.pone.0246695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Bovine Respiratory Syncytial virus (BRSV) is one of the major infectious agents in the etiology of the bovine respiratory disease complex. BRSV causes a respiratory syndrome in calves, which is associated with severe bronchiolitis. In this study we describe the effect of treatment with antiviral fusion protein inhibitor (FPI) and ibuprofen, on gene expression in lung tissue of calves infected with BRSV. Calves infected with BRSV are an excellent model of human RSV in infants: we hypothesized that FPI in combination with ibuprofen would provide the best therapeutic intervention for both species. The following experimental treatment groups of BRSV infected calves were used: 1) ibuprofen day 3-10, 2) ibuprofen day 5-10, 3) placebo, 4) FPI day 5-10, 5) FPI and ibuprofen day 5-10, 6) FPI and ibuprofen day 3-10. All calves were infected with BRSV on day 0. Daily clinical evaluation with monitoring of virus shedding by qRT-PCR was conducted. On day10 lung tissue with lesions (LL) and non-lesional (LN) was collected at necropsy, total RNA extracted, and RNA sequencing performed. Differential gene expression analysis was conducted with Gene ontology (GO) and KEGG pathway enrichment analysis. The most significant differential gene expression in BRSV infected lung tissues was observed in the comparison of LL with LN; oxidative stress and cell damage was especially noticeable. Innate and adaptive immune functions were reduced in LL. As expected, combined treatment with FPI and Ibuprofen, when started early, made the most difference in gene expression patterns in comparison with placebo, especially in pathways related to the innate and adaptive immune response in both LL and LN. Ibuprofen, when used alone, negatively affected the antiviral response and caused higher virus loads as shown by increased viral shedding. In contrast, when used with FPI Ibuprofen enhanced the specific antiviral effect of FPI, due to its ability to reduce the damaging effect of prostanoids and oxidative stress.
Collapse
Affiliation(s)
- Maxim Lebedev
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Heather A. McEligot
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Victoria N. Mutua
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Paul Walsh
- Pediatric Emergency Medicine, Sutter Medical Center Sacramento, Sacramento, California, United States of America
| | - Francisco R. Carvallo Chaigneau
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech., Blacksburg, VA, United States of America
| | - Laurel J. Gershwin
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| |
Collapse
|
26
|
Potential Effects of Nutraceuticals in Retinopathy of Prematurity. Life (Basel) 2021; 11:life11020079. [PMID: 33499180 PMCID: PMC7912639 DOI: 10.3390/life11020079] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 02/07/2023] Open
Abstract
Retinopathy of prematurity (ROP), the most common cause of childhood blindness, is a hypoxia-induced eye disease characterized by retinal neovascularization. In the normal retina, a well-organized vascular network provides oxygen and nutrients as energy sources to maintain a normal visual function; however, it is disrupted when pathological angiogenesis is induced in ROP patients. Under hypoxia, inadequate oxygen and energy supply lead to oxidative stress and stimulate neovasculature formation as well as affecting the function of photoreceptors. In order to meet the metabolic needs in the developing retina, protection against abnormal vascular formation is one way to manage ROP. Although current treatments provide beneficial effects in reducing the severity of ROP, these invasive therapies may also induce life-long consequences such as systemic structural and functional complications as well as neurodevelopment disruption in the developing infants. Nutritional supplements for the newborns are a novel concept for restoring energy supply by protecting the retinal vasculature and may lead to better ROP management. Nutraceuticals are provided in a non-invasive manner without the developmental side effects associated with current treatments. These nutraceuticals have been investigated through various in vitro and in vivo methods and are indicated to protect retinal vasculature. Here, we reviewed and discussed how the use of these nutraceuticals may be beneficial in ROP prevention and management.
Collapse
|
27
|
Teixeira-Santos L, Albino-Teixeira A, Pinho D. Neuroinflammation, oxidative stress and their interplay in neuropathic pain: Focus on specialized pro-resolving mediators and NADPH oxidase inhibitors as potential therapeutic strategies. Pharmacol Res 2020; 162:105280. [PMID: 33161139 DOI: 10.1016/j.phrs.2020.105280] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023]
Abstract
Neuropathic pain (NP) is a chronic condition that results from a lesion or disease of the nervous system, greatly impacting patients' quality of life. Current pharmacotherapy options deliver inadequate and/or insufficient responses and thus a significant unmet clinical need remains for alternative treatments in NP. Neuroinflammation, oxidative stress and their reciprocal relationship are critically involved in NP pathophysiology. In this context, new pharmacological approaches, aiming at enhancing the resolution phase of inflammation and/or restoring redox balance by targeting specific reactive oxygen species (ROS) sources, are emerging as potential therapeutic strategies for NP, with improved efficacy and safety profiles. Several reports have demonstrated that administration of exogenous specialized pro-resolving mediators (SPMs) ameliorates NP pathophysiology. Likewise, deletion or inhibition of the ROS-generating enzyme NADPH oxidase (NOX), particularly its isoforms 2 and 4, results in beneficial effects in NP models. Notably, SPMs also modulate oxidative stress and NOX also regulates neuroinflammation. By targeting neuroinflammatory and oxidative pathways, both SPMs analogues and isoform-specific NOX inhibitors are promising therapeutic strategies for NP.
Collapse
Affiliation(s)
- Luísa Teixeira-Santos
- Departamento de Biomedicina - Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Portugal; MedInUP - Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto, Portugal.
| | - António Albino-Teixeira
- Departamento de Biomedicina - Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Portugal; MedInUP - Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto, Portugal.
| | - Dora Pinho
- Departamento de Biomedicina - Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Portugal; MedInUP - Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto, Portugal.
| |
Collapse
|
28
|
Liu Z, Zhang B, Xia S, Fang L, Gou S. ROS-responsive and multifunctional anti-Alzheimer prodrugs: Tacrine-ibuprofen hybrids via a phenyl boronate linker. Eur J Med Chem 2020; 212:112997. [PMID: 33189440 DOI: 10.1016/j.ejmech.2020.112997] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
Current drugs available in clinic for Alzheimer's disease (AD) treatment can only alleviate disease symptoms without clearly curing or delaying the process of AD. And some AD drugs failed in Phase III clinical trials are only focused on targeting amyloid-β (Aβ). Therefore, an alternative strategy in AD drug design is meaningful to be involved in the multiple pathogenic factors which can affect each other at multiple levels. Herein, we report a series of ROS-responsive prodrugs based on multi-target-directed ligands (MTDLs) approach, which can specifically release tacrine derivatives and ibuprofen under oxidation of ROS and show acetylcholinesterase (AChE)-inhibiting, neuron-protective and anti-inflammatory effects in extracellular or intracellular assays. Related biological study illustrated that compound 22 was able to permeate blood-brain-barrier (BBB) showing little hepatotoxicity in comparison to tacrine. Besides, 22 hinted a therapeutic clue in AD-treatment by regulating proinflammatory factors (IL-1β and TNF-α) and apoptosis related proteins (Bax, Bcl-2 and cleaved caspase-3). Further spatial memory assays in Aβ-induced AD model showed that 22 enhanced the ability of learning and memory. Our study proves that the strategy of ROS-responsive prodrugs has promise for AD treatments in future and offers a way for AD drug development.
Collapse
Affiliation(s)
- Zhikun Liu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Bin Zhang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Shengjin Xia
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Lei Fang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | - Shaohua Gou
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
29
|
Ibuprofen and Severe Acute Respiratory Syndrome Coronavirus 2 Infection Are Synergistic on Cell Death. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2020. [DOI: 10.1097/ipc.0000000000000866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
30
|
Hedayati MA. Ibuprofen has Synergism with SARS-CoV-2 Infection. Open Microbiol J 2020. [DOI: 10.2174/1874285802014010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
31
|
Combined Biological Effects of N-Bromotaurine Analogs and Ibuprofen. Part II: Influence on a Local Defense System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019. [PMID: 31468465 DOI: 10.1007/978-981-13-8023-5_85] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
The stable N-bromotaurine analogs (N-dibromo-dimethyl taurine, N-monobromo-dimethyl taurine), and bromamine T (BAT) show anti-inflammatory and microbicidal properties. These bromamines are good candidates for a treatment of skin infectious/inflammatory diseases as local antiseptics. Ibuprofen, a non-steroidal anti-inflammatory drug (NSAID), is commonly used in various infectious/inflammatory diseases due to its analgesic and antipyretic therapeutic effects. However, systemic administration of ibuprofen may also result in adverse side effects. It has been reported that ibuprofen enhances serum levels of TNF-α and worsens secondary skin infections caused by invasive streptococci (S. pyogenes). Recently we have demonstrated that bromamines inhibit the stimulatory effect of ibuprofen on the production of inflammatory cytokines (TNF-α, IL-6). The aim of this study was to examine the combined antibacterial actions of ibuprofen and bromamines against S. pyogenes and their joint effect on the generation of reactive oxygen species (ROS) by activated neutrophils and macrophages. We have shown that the microbicidal activity of bromamines against S. pyogenes was not altered by ibuprofen. On the other hand, co-administration of ibuprofen and bromamines markedly decreased the generation of ROS by activated neutrophils and macrophages. Finally, we discuss how the antioxidant combined effect of bromamines and ibuprofen may affect a local defense system.
Collapse
|
32
|
Marra A, Naro A, Chillura A, Bramanti A, Maresca G, De Luca R, Manuli A, Bramanti P, Calabrò RS. Evaluating Peripersonal Space through the Functional Transcranial Doppler: Are We Paving the Way for Early Detecting Mild Cognitive Impairment to Dementia Conversion? J Alzheimers Dis 2019; 62:133-143. [PMID: 29439353 DOI: 10.3233/jad-170973] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Identifying the patients with mild cognitive impairment (MCI) who may develop dementia (MDC) is challenging. The study of peripersonal space (PPS) by using functional transcranial Doppler (fTCD) could be used for this purpose. OBJECTIVE To identify changes in cerebral blood flow (CBF) during motor tasks targeting PPS, which can predict MDC. METHODS We evaluated the changes in CBF in 22 patients with MCI and 23 with dementia [Alzheimer's disease (AD) and vascular dementia (VaD)] during a motor task (passive mobilization, motor imagery, and movement observation) in which the hand of the subject moved forward and backward the face. RESULTS CBF increased when the hand approached the face and decreased when the hand moved from the face in the healthy controls (HCs). CBF changed were detectable only in patients with MCI but not in those with the AD and those who were MDC after 8-month follow-up. On the other hand, the patients with VaD presented a paradoxical response to the motor task (i.e., a decrease of CBF rather than an increase, as observed in HCs and MCI). Therefore, we found a modulation of PPS-related CBF only in HCs and patients with stable MCI (at the 8-month follow-up). CONCLUSIONS fTCD may allow preliminarily differentiating and following-up the patients with MCI and MDC, thus allowing the physician to plan beforehand more individualized cognitive rehabilitative training.
Collapse
Affiliation(s)
- Angela Marra
- IRCCS centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | - Antonino Naro
- IRCCS centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Testing the Neuroprotective Properties of PCSO-524 ® Using a Neuronal Cell Cycle Suppression Assay. Mar Drugs 2019; 17:md17020079. [PMID: 30682813 PMCID: PMC6409808 DOI: 10.3390/md17020079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/20/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
Cell cycle reentry is a unified mechanism shared by several neurodegenerative diseases, including Alzheimer’s disease (AD) and Ataxia Telangiectasia (A-T). This phenotype is often related to neuroinflammation in the central nervous system. To mimic brain inflammation in vitro, we adopted the previously established method of using conditioned medium collected from activated THP-1 cells and applied it to both differentiated HT22 cells and primary neurons. Unscheduled cell cycle events were observed in both systems, indicating the potential of this approach as an in vitro model of neurodegenerative disease. We used this assay to measure the neuroprotective effects of New Zealand green-lipped mussel extract, PCSO-524®, to protect post-mitotic cells from cell cycle reentry. We found that, both in vitro and in an animal model, PCSO-524® displayed promising neuroprotective effects, and thus has potential to postpone or prevent the onset of neurodegenerative disease.
Collapse
|
34
|
Hui CW, Song X, Ma F, Shen X, Herrup K. Ibuprofen prevents progression of ataxia telangiectasia symptoms in ATM-deficient mice. J Neuroinflammation 2018; 15:308. [PMID: 30400801 PMCID: PMC6220455 DOI: 10.1186/s12974-018-1338-7] [Citation(s) in RCA: 19] [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: 01/06/2018] [Accepted: 10/18/2018] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Inflammation plays a critical role in accelerating the progression of neurodegenerative diseases, such as Alzheimer's disease (AD) and ataxia telangiectasia (A-T). In A-T mouse models, LPS-induced neuroinflammation advances the degenerative changes found in cerebellar Purkinje neurons both in vivo and in vitro. In the current study, we ask whether ibuprofen, a non-steroidal anti-inflammatory drug (NSAID), can have the opposite effect and delay the symptoms of the disease. METHODS We tested the beneficial effects of ibuprofen in both in vitro and in vivo models. Conditioned medium from LPS stimulated primary microglia (LM) applied to cultures of dissociated cortical neurons leads to numerous degenerative changes. Pretreatment of the neurons with ibuprofen, however, blocked this damage. Systemic injection of LPS into either adult wild-type or adult Atm-/- mice produced an immune challenge that triggered profound behavioral, biochemical, and histological effects. We used a 2-week ibuprofen pretreatment regimen to investigate whether these LPS effects could be blocked. We also treated young presymptomatic Atm-/- mice to determine if ibuprofen could delay the appearance of symptoms. RESULTS Adding ibuprofen directly to neuronal cultures significantly reduced LM-induced degeneration. Curiously, adding ibuprofen to the microglia cultures before the LPS challenge had little effect, thus implying a direct effect of the NSAID on the neuronal cultures. In vivo administration of ibuprofen to Atm-/- animals before a systemic LPS immune challenge suppressed cytological damage. The ibuprofen effects were widespread as microglial activation, p38 phosphorylation, DNA damage, and neuronal cell cycle reentry were all reduced. Unfortunately, ibuprofen only slightly improved the LPS-induced behavioral deficits. Yet, while the behavioral symptoms could not be reversed once they were established in adult Atm-/- animals, administration of ibuprofen to young mutant pups prevented their symptoms from appearing. CONCLUSION Inflammatory processes impact the normal progression of A-T implying that modulation of the immune system can have therapeutic benefit for both the behavioral and cellular symptoms of this neurodegenerative disease.
Collapse
Affiliation(s)
- Chin Wai Hui
- Division of Life Science and State Key Laboratory of Molecular Neurobiology, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Xuan Song
- Division of Life Science and State Key Laboratory of Molecular Neurobiology, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Fulin Ma
- Division of Life Science and State Key Laboratory of Molecular Neurobiology, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Xuting Shen
- Division of Life Science and State Key Laboratory of Molecular Neurobiology, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
- Present address: School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Karl Herrup
- Division of Life Science and State Key Laboratory of Molecular Neurobiology, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| |
Collapse
|
35
|
Tse KH, Cheng A, Ma F, Herrup K. DNA damage-associated oligodendrocyte degeneration precedes amyloid pathology and contributes to Alzheimer's disease and dementia. Alzheimers Dement 2018; 14:664-679. [PMID: 29328926 PMCID: PMC5938117 DOI: 10.1016/j.jalz.2017.11.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/18/2017] [Accepted: 11/28/2017] [Indexed: 11/20/2022]
Abstract
INTRODUCTION In looking for novel non-amyloid-based etiologies for Alzheimer's disease, we explore the hypothesis that age-related myelin loss is an attractive explanation for age-associated cognitive decline and dementia. METHODS We performed a meta-analysis of data in the National Alzheimer's Coordinating Center database accompanied by quantitative histopathology of myelin and oligodendrocytes (OLs) in frontal cortices of 24 clinically characterized individuals. Pathological findings were further validated in an Alzheimer's disease mouse model and in culture. RESULTS Myelin lesions increased with cognitive impairment in an amyloid-independent fashion with signs of degeneration appearing before neuronal loss. Myelinating OLs in the gray matter showed greater vulnerability than those in white matter, and the degenerative changes correlated with evidence of DNA damage. Similar results were found in myelinating OL cultures where DNA damage caused aberrant OL cell cycle re-entry and death. DISCUSSION We present the first comprehensive analysis of the cell biology of early myelin loss in sporadic Alzheimer's disease.
Collapse
Affiliation(s)
- Kai-Hei Tse
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Aifang Cheng
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Fulin Ma
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Karl Herrup
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| |
Collapse
|
36
|
Modjtahedi BS, Fong DS, Jorgenson E, Van Den Eeden SK, Quinn V, Slezak JM. The Relationship Between Nonsteroidal Anti-inflammatory Drug Use and Age-related Macular Degeneration. Am J Ophthalmol 2018; 188:111-122. [PMID: 29360460 DOI: 10.1016/j.ajo.2018.01.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE To describe the relationship between the incidence of age-related macular degeneration (AMD) and nonsteroidal anti-inflammatory drug (NSAIDs) use. DESIGN Prospective cohort study. METHODS This study consisted of participants in the California Men's Health Study. Those who completed surveys in 2002-2003 and 2006 were included. Men who self-reported use of aspirin, ibuprofen, naproxen, valdecoxib, celecoxib, and/or rofecoxib at least 3 days per week were considered NSAID users. Patients were categorized as non-users, former users, new users, or longer-term users based on survey responses. NSAID use was also categorized by type: any NSAIDs, aspirin, and/or non-aspirin NSAIDs. Age, race/ethnicity, smoking status, education, income, alcohol use, and Charlson comorbidity index score were included in the multivariate analysis as risk factors for AMD. RESULTS A total of 51 371 men were included. Average follow-up time was 7.4 years. There were 292 (0.6%) and 1536 (3%) cases of exudative and nonexudative AMD, respectively. Longer-term use of any NSAID was associated with lower risk of exudative AMD (hazard ratio [HR] 0.69, 95% confidence interval [CI] 0.50-0.96, P = .029). New users of any NSAIDs (HR = 0.79, 95% CI 0.68-0.93, P = .0039) and aspirin (HR = 0.82, 95% CI 0.70-0.97, P = .018) had a lower risk of nonexudative AMD, although this trend did not persist in longer-term users. The relationship between exudative or nonexudative AMD and the remaining categories of NSAID use were not significant. CONCLUSION The overall impact of NSAIDs on AMD incidence is small; however, the lower risk of exudative AMD in longer-term NSAID users may point to a protective effect and deserves further study as a possible mechanism to modulate disease risk.
Collapse
Affiliation(s)
- Bobeck S Modjtahedi
- Department of Ophthalmology, Southern California Permanente Medical Group, Baldwin Park, California; Eye Monitoring Center, Kaiser Permanente Southern California, Baldwin Park, California.
| | - Donald S Fong
- Department of Ophthalmology, Southern California Permanente Medical Group, Baldwin Park, California; Eye Monitoring Center, Kaiser Permanente Southern California, Baldwin Park, California; Department of Research and Evaluation, Southern California Permanente Medical Group, Pasadena, California
| | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | | | - Virginia Quinn
- Department of Research and Evaluation, Southern California Permanente Medical Group, Pasadena, California
| | - Jeffrey M Slezak
- Department of Research and Evaluation, Southern California Permanente Medical Group, Pasadena, California
| |
Collapse
|
37
|
Ravelli KG, Rosário BDA, Vasconcelos AR, Scavone C, Camarini R, Hernandes MS, Britto LR. NADPH oxidase contributes to streptozotocin-induced neurodegeneration. Neuroscience 2017; 358:227-237. [PMID: 28687315 DOI: 10.1016/j.neuroscience.2017.06.050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/12/2017] [Accepted: 06/26/2017] [Indexed: 12/26/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the progressive loss of memory. The neurodegeneration induced by AD has been linked to oxidative damage. However, little is known about the involvement of NADPH oxidase 2 (Nox2), a multisubunit enzyme that catalyzes the reduction of oxygen to produce reactive oxygen species, in the pathogenesis of AD. The main purpose of this study was to investigate the involvement of Nox2 in memory, in AD-related brain abnormalities, oxidative damage, inflammation and neuronal death in the hippocampus in the streptozotocin (STZ)-induced AD-like state by comparing the effects of that drug on mice lacking gp91phox-/- and wild-type (Wt) mice. Nox2 gene expression was found increased in Wt mice after STZ injection. In object recognition test, Wt mice injected with STZ presented impairment in short- and long-term memory, which was not observed following Nox2 deletion. STZ treatment induced increased phosphorylation of Tau and increased amyloid-β, apoptosis-inducing factor (AIF) and astrocyte and microglial markers expression in Wt mice but not in gp91phox-/-. STZ treatment increased oxidative damage and pro-inflammatory cytokines' release in Wt mice, which was not observed in gp91phox-/- mice. Nox2 deletion had a positive effect on the IL-10 baseline production, suggesting that this cytokine might contribute to the neuroprotection mechanism against STZ-induced neurodegeneration. In summary, our data suggest that the Nox2-dependent reactive oxygen species (ROS) generation contributes to the STZ-induced AD-like state.
Collapse
Affiliation(s)
| | | | | | | | - Rosana Camarini
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
| | - Marina S Hernandes
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States.
| | - Luiz Roberto Britto
- Department of Physiology and Biophysics, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Tse KH, Herrup K. Re-imagining Alzheimer's disease - the diminishing importance of amyloid and a glimpse of what lies ahead. J Neurochem 2017; 143:432-444. [PMID: 28547865 DOI: 10.1111/jnc.14079] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/13/2017] [Accepted: 05/23/2017] [Indexed: 12/13/2022]
Abstract
Many have criticized the amyloid cascade hypothesis of Alzheimer's disease for its inconsistencies and failures to either accurately predict disease symptoms or guide the development of productive therapies. In addition to criticisms, however, we believe that the field would benefit from having alternative narratives and disease models that can either replace or function alongside of an amyloid-centric view of Alzheimer's. This review is an attempt to meet that need. We offer three experimentally verified amyloid-independent mechanisms, each of which plausibly contributes substantially to the aetiology of Alzheimer's disease: loss of DNA integrity, faulty cell cycle regulation, regression of myelination. We outline the ways in which the failure of each can contribute to AD initiation and progression, and review how, acting alone or in combination with each other, they are sufficient for explaining the full range of AD pathologies. Yet, these three alternatives represent only a few of the many non-amyloid mechanisms that can explain AD pathogenesis. Therefore instead of proposing a single 'alternative hypothesis' to the amyloid cascade theory, sporadic AD is pictured as the result of independent yet intersecting age-related pathologies that afflict the ageing human brain. This article is part of the series "Beyond Amyloid". Cover Image for this issue: doi. 10.1111/jnc.13823.
Collapse
Affiliation(s)
- Kai-Hei Tse
- Division of Life Science and the State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Karl Herrup
- Division of Life Science and the State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| |
Collapse
|
39
|
Perazzo J, Castanho MARB, Sá Santos S. Pharmacological Potential of the Endogenous Dipeptide Kyotorphin and Selected Derivatives. Front Pharmacol 2017; 7:530. [PMID: 28127286 PMCID: PMC5226936 DOI: 10.3389/fphar.2016.00530] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/20/2016] [Indexed: 12/27/2022] Open
Abstract
The endogenous peptide kyotorphin (KTP) has been extensively studied since it was discovered in 1979. The dipeptide is distributed unevenly over the brain but the majority is concentrated in the cerebral cortex. The putative KTP receptor has not been identified yet. As many other neuropeptides, KTP clearance is mediated by extracellular peptidases and peptide transporters. From the wide spectrum of biological activity of KTP, analgesia was by far the most studied. The mechanism of action is still unclear, but researchers agree that KTP induces Met-enkephalins release. More recently, KTP was proposed as biomarker of Alzheimer disease. Despite all that, KTP limited pharmacological value prompted researchers to develop derivatives more lipophilic and therefore more prone to cross the blood–brain barrier (BBB), and also more resistant to enzymatic degradation. Conjugation of KTP with functional molecules, such as ibuprofen, generated a new class of compounds with additional biological properties. Moreover, the safety profile of these derivatives compared to opioids and their efficacy as neuroprotective agents greatly increases their pharmacological value.
Collapse
Affiliation(s)
- Juliana Perazzo
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa Lisboa, Portugal
| | - Miguel A R B Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa Lisboa, Portugal
| | - Sónia Sá Santos
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa Lisboa, Portugal
| |
Collapse
|
40
|
Márquez Loza A, Elias V, Wong CP, Ho E, Bermudez M, Magnusson KR. Effects of ibuprofen on cognition and NMDA receptor subunit expression across aging. Neuroscience 2017; 344:276-292. [PMID: 28057539 DOI: 10.1016/j.neuroscience.2016.12.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/13/2016] [Accepted: 12/22/2016] [Indexed: 11/28/2022]
Abstract
Age-related declines in long- and short-term memory show relationships to decreases in N-methyl-d-aspartate (NMDA) receptor expression, which may involve inflammation. This study was designed to determine effects of an anti-inflammatory drug, ibuprofen, on cognitive function and NMDA receptor expression across aging. Male C57BL/6 mice (ages 5, 14, 20, and 26months) were fed ibuprofen (375ppm) in NIH31 diet or diet alone for 6weeks prior to testing. Behavioral testing using the Morris water maze showed that older mice performed significantly worse than younger in spatial long-term memory, reversal, and short-term memory tasks. Ibuprofen enhanced overall performance in the short-term memory task, but this appeared to be more related to improved executive function than memory. Ibuprofen induced significant decreases over all ages in the mRNA densities for GluN2B subunit, all GluN1 splice variants, and GluN1-1 splice forms in the frontal cortex and in protein expression of GluN2A, GluN2B and GluN1 C2' cassettes in the hippocampus. GluN1-3 splice form mRNA and C2' cassette protein were significantly increased across ages in frontal lobes of ibuprofen-treated mice. Ibuprofen did not alter expression of pro-inflammatory cytokines IL-1β and TNFα, but did reduce the area of reactive astrocyte immunostaining in frontal cortex of aged mice. Enhancement in executive function showed a relationship to increased GluN1-3 mRNA and decreased gliosis. These findings suggest that inflammation may play a role in executive function declines in aged animals, but other effects of ibuprofen on NMDA receptors appeared to be unrelated to aging or inflammation.
Collapse
Affiliation(s)
- Alejandra Márquez Loza
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| | - Valerie Elias
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| | - Carmen P Wong
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | - Emily Ho
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA; School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | - Michelle Bermudez
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| | - Kathy R Magnusson
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
41
|
Proshkina E, Lashmanova E, Dobrovolskaya E, Zemskaya N, Kudryavtseva A, Shaposhnikov M, Moskalev A. Geroprotective and Radioprotective Activity of Quercetin, (-)-Epicatechin, and Ibuprofen in Drosophila melanogaster. Front Pharmacol 2016; 7:505. [PMID: 28066251 PMCID: PMC5179547 DOI: 10.3389/fphar.2016.00505] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 12/07/2016] [Indexed: 12/15/2022] Open
Abstract
The modulation of longevity genes and aging-associated signaling pathways using pharmacological agents is one of the potential ways to prolong the lifespan and increase the vitality of an organism. Phytochemicals flavonoids and non-steroidal anti-inflammatory drugs have a large potential as geroprotectors. The goal of the present study was to investigate the effects of long-term and short-term consumption of quercetin, (-)-epicatechin, and ibuprofen on the lifespan, resistance to stress factors (paraquat, hyperthermia, γ-radiation, and starvation), as well as age-dependent physiological parameters (locomotor activity and fecundity) of Drosophila melanogaster. The long-term treatment with quercetin and (-)-epicatechin didn't change or decreased the lifespan of males and females. In contrast, the short-term treatment with flavonoids had a beneficial effect and stimulated the resistance to paraquat and acute γ-irradiation. The short-term ibuprofen consumption had a positive effect on the lifespan of females when it was carried out at the middle age (30–40 days), and to the survival of flies under conditions of oxidative and genotoxic stresses. However, it didn't change the lifespan of males and females after the treatment during first 10 days of an imago life. Additionally, quercetin, (-)-epicatechin, and ibuprofen decreased the spontaneous locomotor activity of males, but had no effect of stimulated the physical activity and fecundity of females. Revealed quercetin, (-)-epicatechin, and ibuprofen activity can be associated with the stimulation of stress response mechanisms through the activation of pro-longevity pathways, or the induction of hormesis.
Collapse
Affiliation(s)
- Ekaterina Proshkina
- Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences (RAS) Syktyvkar, Russia
| | - Ekaterina Lashmanova
- Laboratory of Genetics of Aging and Longevity, Moscow Institute of Physics and Technology (MIPT) Dolgoprudny, Russia
| | - Eugenia Dobrovolskaya
- Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences (RAS) Syktyvkar, Russia
| | - Nadezhda Zemskaya
- Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences (RAS)Syktyvkar, Russia; Department of Ecology, Institute of Natural Sciences, Syktyvkar State UniversitySyktyvkar, Russia
| | - Anna Kudryavtseva
- Engelhardt Institute of Molecular Biology (EIMB), Russian Academy of Sciences (RAS) Moscow, Russia
| | - Mikhail Shaposhnikov
- Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences (RAS)Syktyvkar, Russia; Department of Ecology, Institute of Natural Sciences, Syktyvkar State UniversitySyktyvkar, Russia
| | - Alexey Moskalev
- Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences (RAS)Syktyvkar, Russia; Laboratory of Genetics of Aging and Longevity, Moscow Institute of Physics and Technology (MIPT)Dolgoprudny, Russia; Department of Ecology, Institute of Natural Sciences, Syktyvkar State UniversitySyktyvkar, Russia; Engelhardt Institute of Molecular Biology (EIMB), Russian Academy of Sciences (RAS)Moscow, Russia
| |
Collapse
|
42
|
Current pharmacotherapy and putative disease-modifying therapy for Alzheimer's disease. Neurol Sci 2016; 37:1403-35. [PMID: 27250365 DOI: 10.1007/s10072-016-2625-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/24/2016] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease of the central nervous system correlated with the progressive loss of cognition and memory. β-Amyloid plaques, neurofibrillary tangles and the deficiency in cholinergic neurotransmission constitute the major hallmarks of the AD. Two major hypotheses have been implicated in the pathogenesis of AD namely the cholinergic hypothesis which ascribed the clinical features of dementia to the deficit cholinergic neurotransmission and the amyloid cascade hypothesis which emphasized on the deposition of insoluble peptides formed due to the faulty cleavage of the amyloid precursor protein. Current pharmacotherapy includes mainly the acetylcholinesterase inhibitors and N-methyl-D-aspartate receptor agonist which offer symptomatic therapy and does not address the underlying cause of the disease. The disease-modifying therapy has garnered a lot of research interest for the development of effective pharmacotherapy for AD. β and γ-Secretase constitute attractive targets that are focussed in the disease-modifying approach. Potentiation of α-secretase also seems to be a promising approach towards the development of an effective anti-Alzheimer therapy. Additionally, the ameliorative agents that prevent aggregation of amyloid peptide and also the ones that modulate inflammation and oxidative damage associated with the disease are focussed upon. Development in the area of the vaccines is in progress to combat the characteristic hallmarks of the disease. Use of cholesterol-lowering agents also is a fruitful strategy for the alleviation of the disease as a close association between the cholesterol and AD has been cited. The present review underlines the major therapeutic strategies for AD with focus on the new developments that are on their way to amend the current therapeutic scenario of the disease.
Collapse
|
43
|
Woodling NS, Colas D, Wang Q, Minhas P, Panchal M, Liang X, Mhatre SD, Brown H, Ko N, Zagol-Ikapitte I, van der Hart M, Khroyan TV, Chuluun B, Priyam PG, Milne GL, Rassoulpour A, Boutaud O, Manning-Boğ AB, Heller HC, Andreasson KI. Cyclooxygenase inhibition targets neurons to prevent early behavioural decline in Alzheimer's disease model mice. Brain 2016; 139:2063-81. [PMID: 27190010 DOI: 10.1093/brain/aww117] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/31/2016] [Indexed: 01/22/2023] Open
Abstract
Identifying preventive targets for Alzheimer's disease is a central challenge of modern medicine. Non-steroidal anti-inflammatory drugs, which inhibit the cyclooxygenase enzymes COX-1 and COX-2, reduce the risk of developing Alzheimer's disease in normal ageing populations. This preventive effect coincides with an extended preclinical phase that spans years to decades before onset of cognitive decline. In the brain, COX-2 is induced in neurons in response to excitatory synaptic activity and in glial cells in response to inflammation. To identify mechanisms underlying prevention of cognitive decline by anti-inflammatory drugs, we first identified an early object memory deficit in APPSwe-PS1ΔE9 mice that preceded previously identified spatial memory deficits in this model. We modelled prevention of this memory deficit with ibuprofen, and found that ibuprofen prevented memory impairment without producing any measurable changes in amyloid-β accumulation or glial inflammation. Instead, ibuprofen modulated hippocampal gene expression in pathways involved in neuronal plasticity and increased levels of norepinephrine and dopamine. The gene most highly downregulated by ibuprofen was neuronal tryptophan 2,3-dioxygenase (Tdo2), which encodes an enzyme that metabolizes tryptophan to kynurenine. TDO2 expression was increased by neuronal COX-2 activity, and overexpression of hippocampal TDO2 produced behavioural deficits. Moreover, pharmacological TDO2 inhibition prevented behavioural deficits in APPSwe-PS1ΔE9 mice. Taken together, these data demonstrate broad effects of cyclooxygenase inhibition on multiple neuronal pathways that counteract the neurotoxic effects of early accumulating amyloid-β oligomers.
Collapse
Affiliation(s)
- Nathaniel S Woodling
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA 2 Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Damien Colas
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Qian Wang
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paras Minhas
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maharshi Panchal
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xibin Liang
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Siddhita D Mhatre
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Holden Brown
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA 4 Brains On-line LLC, South San Francisco, CA, USA
| | - Novie Ko
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Irene Zagol-Ikapitte
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marieke van der Hart
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taline V Khroyan
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bayarsaikhan Chuluun
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Prachi G Priyam
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ginger L Milne
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arash Rassoulpour
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Olivier Boutaud
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amy B Manning-Boğ
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - H Craig Heller
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Katrin I Andreasson
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
44
|
Woodling NS, Andreasson KI. Untangling the Web: Toxic and Protective Effects of Neuroinflammation and PGE2 Signaling in Alzheimer's Disease. ACS Chem Neurosci 2016; 7:454-63. [PMID: 26979823 DOI: 10.1021/acschemneuro.6b00016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The neuroinflammatory response has received increasing attention as a key factor in the pathogenesis of Alzheimer's disease (AD). Microglia, the innate immune cells and resident phagocytes of the brain, respond to accumulating Aβ peptides by generating a nonresolving inflammatory response. While this response can clear Aβ peptides from the nervous system in some settings, its failure to do so in AD accelerates synaptic injury, neuronal loss, and cognitive decline. The complex molecular components of this response are beginning to be unraveled, with identification of both damaging and protective roles for individual components of the neuroinflammatory response. Even within one molecular pathway, contrasting effects are often present. As one example, recent studies of the inflammatory cyclooxygenase-prostaglandin pathway have revealed both beneficial and detrimental effects dependent on the disease context, cell type, and downstream signaling pathway. Nonsteroidal anti-inflammatory drugs (NSAIDs), which inhibit cyclooxygenases, are associated with reduced AD risk when taken by cognitively normal populations, but additional clinical and mouse model studies have added complexities and caveats to this finding. Downstream of cyclooxygenase activity, prostaglandin E2 signaling exerts both damaging pro-inflammatory and protective anti-inflammatory effects through actions of specific E-prostanoid G-protein coupled receptors on specific cell types. These complexities underscore the need for careful study of individual components of the neuroinflammatory response to better understand their contribution to AD pathogenesis and progression.
Collapse
Affiliation(s)
- Nathaniel S. Woodling
- Department of Neurology and
Neurological Sciences, Stanford University School of Medicine, 1201
Welch Road, Stanford, California 94305, United States
| | - Katrin I. Andreasson
- Department of Neurology and
Neurological Sciences, Stanford University School of Medicine, 1201
Welch Road, Stanford, California 94305, United States
| |
Collapse
|
45
|
LIU YAOWU, ZHU XIA, CHENG YAQIN, LU QIAN, ZHANG FAN, GUO HAO, YIN XIAOXING. Ibuprofen attenuates nephropathy in streptozotocin-induced diabetic rats. Mol Med Rep 2016; 13:5326-34. [DOI: 10.3892/mmr.2016.5150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 01/08/2016] [Indexed: 11/05/2022] Open
|
46
|
Dey A, Kang X, Qiu J, Du Y, Jiang J. Anti-Inflammatory Small Molecules To Treat Seizures and Epilepsy: From Bench to Bedside. Trends Pharmacol Sci 2016; 37:463-484. [PMID: 27062228 DOI: 10.1016/j.tips.2016.03.001] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/03/2016] [Accepted: 03/07/2016] [Indexed: 12/22/2022]
Abstract
As a crucial component of brain innate immunity, neuroinflammation initially contributes to neuronal tissue repair and maintenance. However, chronic inflammatory processes within the brain and associated blood-brain barrier (BBB) impairment often cause neurotoxicity and hyperexcitability. Mounting evidence points to a mutual facilitation between inflammation and epilepsy, suggesting that blocking the undesired inflammatory signaling within the brain might provide novel strategies to treat seizures and epilepsy. Neuroinflammation is primarily characterized by the upregulation of proinflammatory mediators in epileptogenic foci, among which cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2), interleukin-1β (IL-1β), transforming growth factor-β (TGF-β), toll-like receptor 4 (TLR4), high-mobility group box 1 (HMGB1), and tumor necrosis factor-α (TNF-α) have been extensively studied. Small molecules that specifically target these key proinflammatory perpetrators have been evaluated for antiepileptic and antiepileptogenic effects in animal models. These important preclinical studies provide new insights into the regulation of inflammation in epileptic brains and guide drug discovery efforts aimed at developing novel anti-inflammatory therapies for seizures and epilepsy.
Collapse
Affiliation(s)
- Avijit Dey
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH 45267-0514, USA
| | - Xu Kang
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH 45267-0514, USA
| | - Jiange Qiu
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH 45267-0514, USA
| | - Yifeng Du
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH 45267-0514, USA
| | - Jianxiong Jiang
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH 45267-0514, USA.
| |
Collapse
|
47
|
Sá Santos S, Santos SM, Pinto ART, Ramu VG, Heras M, Bardaji E, Tavares I, Castanho MARB. Amidated and Ibuprofen-Conjugated Kyotorphins Promote Neuronal Rescue and Memory Recovery in Cerebral Hypoperfusion Dementia Model. Front Aging Neurosci 2016; 8:1. [PMID: 26858637 PMCID: PMC4726799 DOI: 10.3389/fnagi.2016.00001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 01/04/2016] [Indexed: 12/25/2022] Open
Abstract
Chronic brain ischemia is a prominent risk factor for neurological dysfunction and progression for dementias, including Alzheimer’s disease (AD). In rats, permanent bilateral common carotid artery occlusion (2VO) causes a progressive neurodegeneration in the hippocampus, learning deficits and memory loss as it occurs in AD. Kyotorphin (KTP) is an endogenous antinociceptive dipeptide whose role as neuromodulator/neuroprotector has been suggested. Recently, we designed two analgesic KTP-derivatives, KTP-amide (KTP–NH2) and KTP–NH2 linked to ibuprofen (IbKTP–NH2) to improve KTP brain targeting. This study investigated the effects of KTP-derivatives on cognitive/behavioral functions (motor/spatial memory/nociception) and hippocampal pathology of female rats in chronic cerebral hypoperfusion (2VO-rat model). 2VO-animals were treated with KTP–NH2 or IbKTP–NH2 for 7 days at weeks 2 and 5 post-surgery. After behavioral testing (week 6), coronal sections of hippocampus were H&E-stained or immunolabeled for the cellular markers GFAP (astrocytes) and NFL (neurons). Our findings show that KTP-derivatives, mainly IbKTP–NH2, enhanced cognitive impairment of 2VO-animals and prevented neuronal damage in hippocampal CA1 subfield, suggesting their potential usefulness for the treatment of dementia.
Collapse
Affiliation(s)
- Sónia Sá Santos
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa Lisboa, Portugal
| | - Sara M Santos
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa Lisboa, Portugal
| | - Antónia R T Pinto
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa Lisboa, Portugal
| | - Vasanthakumar G Ramu
- Laboratori d'Innovació en Processos i Productes de Síntesi Orgànica, Departament de Química, Universitat de Girona Girona, Spain
| | - Montserrat Heras
- Laboratori d'Innovació en Processos i Productes de Síntesi Orgànica, Departament de Química, Universitat de Girona Girona, Spain
| | - Eduard Bardaji
- Laboratori d'Innovació en Processos i Productes de Síntesi Orgànica, Departament de Química, Universitat de Girona Girona, Spain
| | - Isaura Tavares
- Departamento de Biologia Experimental, Faculdade de Medicina do PortoPorto, Portugal; Instituto de Biologia Molecular e CelularPorto, Portugal; i3S - Instituto de Inovação e Investigação em Saúde, Universidade do PortoPorto, Portugal
| | | |
Collapse
|
48
|
Peters DG, Connor JR, Meadowcroft MD. The relationship between iron dyshomeostasis and amyloidogenesis in Alzheimer's disease: Two sides of the same coin. Neurobiol Dis 2015; 81:49-65. [PMID: 26303889 DOI: 10.1016/j.nbd.2015.08.007] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 08/04/2015] [Accepted: 08/12/2015] [Indexed: 12/21/2022] Open
Abstract
The dysregulation of iron metabolism in Alzheimer's disease is not accounted for in the current framework of the amyloid cascade hypothesis. Accumulating evidence suggests that impaired iron homeostasis is an early event in Alzheimer's disease progression. Iron dyshomeostasis leads to a loss of function in several enzymes requiring iron as a cofactor, the formation of toxic oxidative species, and the elevated production of beta-amyloid proteins. Several common genetic polymorphisms that cause increased iron levels and dyshomeostasis have been associated with Alzheimer's disease but the pathoetiology is not well understood. A full picture is necessary to explain how heterogeneous circumstances lead to iron loading and amyloid deposition. There is evidence to support a causative interplay between the concerted loss of iron homeostasis and amyloid plaque formation. We hypothesize that iron misregulation and beta-amyloid plaque pathology are synergistic in the process of neurodegeneration and ultimately cause a downward cascade of events that spiral into the manifestation of Alzheimer's disease. In this review, we amalgamate recent findings of brain iron metabolism in healthy versus Alzheimer's disease brains and consider unique mechanisms of iron transport in different brain cells as well as how disturbances in iron regulation lead to disease etiology and propagate Alzheimer's pathology.
Collapse
Affiliation(s)
- Douglas G Peters
- Department of Neurosurgery, The Pennsylvania State University, College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, USA; Department of Neural and Behavioral Sciences, The Pennsylvania State University, College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, USA
| | - James R Connor
- Department of Neurosurgery, The Pennsylvania State University, College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Mark D Meadowcroft
- Department of Neurosurgery, The Pennsylvania State University, College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, USA; Department of Radiology, The Center for NMR Research, The Pennsylvania State University, College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
49
|
Ramirez A, van der Flier WM, Herold C, Ramonet D, Heilmann S, Lewczuk P, Popp J, Lacour A, Drichel D, Louwersheimer E, Kummer MP, Cruchaga C, Hoffmann P, Teunissen C, Holstege H, Kornhuber J, Peters O, Naj AC, Chouraki V, Bellenguez C, Gerrish A, International Genomics of Alzheimer's Project (IGAP), Alzheimer's Disease Neuroimaging Initiative (ADNI), Heun R, Frölich L, Hüll M, Buscemi L, Herms S, Kölsch H, Scheltens P, Breteler MM, Rüther E, Wiltfang J, Goate A, Jessen F, Maier W, Heneka MT, Becker T, Nöthen MM. SUCLG2 identified as both a determinator of CSF Aβ1-42 levels and an attenuator of cognitive decline in Alzheimer's disease. Hum Mol Genet 2014; 23:6644-58. [PMID: 25027320 PMCID: PMC4240204 DOI: 10.1093/hmg/ddu372] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 06/27/2014] [Accepted: 07/09/2014] [Indexed: 01/17/2023] Open
Abstract
Cerebrospinal fluid amyloid-beta 1-42 (Aβ1-42) and phosphorylated Tau at position 181 (pTau181) are biomarkers of Alzheimer's disease (AD). We performed an analysis and meta-analysis of genome-wide association study data on Aβ1-42 and pTau181 in AD dementia patients followed by independent replication. An association was found between Aβ1-42 level and a single-nucleotide polymorphism in SUCLG2 (rs62256378) (P = 2.5×10(-12)). An interaction between APOE genotype and rs62256378 was detected (P = 9.5 × 10(-5)), with the strongest effect being observed in APOE-ε4 noncarriers. Clinically, rs62256378 was associated with rate of cognitive decline in AD dementia patients (P = 3.1 × 10(-3)). Functional microglia experiments showed that SUCLG2 was involved in clearance of Aβ1-42.
Collapse
Affiliation(s)
- Alfredo Ramirez
- Department of Psychiatry and Psychotherapy, Institute of Human Genetics,
| | - Wiesje M van der Flier
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, 1081 HZ, Amsterdam, The Netherlands, Department of Epidemiology & Biostatistics, VU University Medical Center, 1007 MB, Amsterdam, The Netherlands
| | - Christine Herold
- German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | | | - Stefanie Heilmann
- Institute of Human Genetics, Department of Genomics, Life & Brain Center
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | | | - André Lacour
- German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Dmitriy Drichel
- German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Eva Louwersheimer
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, 1081 HZ, Amsterdam, The Netherlands, Department of Epidemiology & Biostatistics, VU University Medical Center, 1007 MB, Amsterdam, The Netherlands
| | - Markus P Kummer
- Clinical Neuroscience Unit, Department of Neurology, German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Carlos Cruchaga
- Department of Psychiatry, Hope Center for Neurological Disorders, School of Medicine
| | - Per Hoffmann
- Institute of Human Genetics, Department of Genomics, Life & Brain Center, Division of Medical Genetics, University Hospital and Department of Biomedicine, University of Basel, CH-4058, Basel, Switzerland
| | - Charlotte Teunissen
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, 1081 HZ, Amsterdam, The Netherlands, Department of Epidemiology & Biostatistics, VU University Medical Center, 1007 MB, Amsterdam, The Netherlands
| | - Henne Holstege
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, 1081 HZ, Amsterdam, The Netherlands, Department of Epidemiology & Biostatistics, VU University Medical Center, 1007 MB, Amsterdam, The Netherlands
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Oliver Peters
- Department of Psychiatry, Charité, 14050, Berlin, Germany
| | - Adam C Naj
- Center for Clinical Epidemiology & Biostatistics, University of Pennsylvania, PA 19104, Philadelphia, USA
| | - Vincent Chouraki
- Department of Neurology, Boston University School of Medicine, MA 02118, Boston, USA, The Framingham Heart Study, MA 01702, Framingham, USA
| | - Céline Bellenguez
- Inserm, U744, Lille 59000, France, Université Lille 2, Lille 59000, France, Institut Pasteur de Lille, Lille 59000, France
| | - Amy Gerrish
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | | | | | | | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
| | - Michael Hüll
- Centre for Geriatric Medicine and Section of Gerontopsychiatry and Neuropsychology, Medical School, University of Freiburg, 79106, Freiburg, Germany
| | - Lara Buscemi
- Department of Fundamental Neurosciences, UNIL, 1005 Lausanne, Switzerland and
| | - Stefan Herms
- Institute of Human Genetics, Department of Genomics, Life & Brain Center, Division of Medical Genetics, University Hospital and Department of Biomedicine, University of Basel, CH-4058, Basel, Switzerland
| | | | - Philip Scheltens
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, 1081 HZ, Amsterdam, The Netherlands, Department of Epidemiology & Biostatistics, VU University Medical Center, 1007 MB, Amsterdam, The Netherlands
| | - Monique M Breteler
- German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Eckart Rüther
- Department of Psychiatry and Psychotherapy, University of Göttingen, 37075 Göttingen, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University of Göttingen, 37075 Göttingen, Germany
| | - Alison Goate
- Department of Psychiatry, Department of Genetics, Washington University, St. Louis, MO 63110, USA
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Michael T Heneka
- Clinical Neuroscience Unit, Department of Neurology, German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Tim Becker
- Institute for Medical Biometry, Informatics, and Epidemiology, University of Bonn, 53127, Bonn, Germany, German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, Department of Genomics, Life & Brain Center
| |
Collapse
|
50
|
Humanin attenuates Alzheimer-like cognitive deficits and pathological changes induced by amyloid β-peptide in rats. Neurosci Bull 2014; 30:923-935. [PMID: 25391447 DOI: 10.1007/s12264-014-1479-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/12/2014] [Indexed: 12/20/2022] Open
Abstract
Amyloid β-peptide (Aβ) has been implicated as a key molecule in the neurodegenerative cascades of Alzheimer's disease (AD). Humanin (HN) is a secretory peptide that inhibits the neurotoxicity of Aβ. However, the mechanism(s) by which HN exerts its neuroprotection against Aβ-induced AD-like pathological changes and memory deficits are yet to be completely defined. In the present study, we provided evidence that treatment of rats with HN increases the number of dendritic branches and the density of dendritic spines, and upregulates pre- and post-synaptic protein levels; these effects lead to enhanced long-term potentiation and amelioration of the memory deficits induced by Aβ(1-42). HN also attenuated Aβ(1-42)-induced tau hyperphosphorylation, apparently by inhibiting the phosphorylation of Tyr307 on the inhibitory protein phosphatase-2A (PP2A) catalytic subunit and thereby activating PP2A. HN also inhibited apoptosis and reduced the oxidative stress induced by Aβ(1-42). These findings provide novel mechanisms of action for the ability of HN to protect against Aβ(1-42)-induced AD-like pathological changes and memory deficits.
Collapse
|