1
|
Rojo AI, Buttari B, Cadenas S, Carlos AR, Cuadrado A, Falcão AS, López MG, Georgiev MI, Grochot-Przeczek A, Gumeni S, Jimenez-Villegas J, Horbanczuk JO, Konu O, Lastres-Becker I, Levonen AL, Maksimova V, Michaeloudes C, Mihaylova LV, Mickael ME, Milisav I, Miova B, Rada P, Santos M, Seabra MC, Strac DS, Tenreiro S, Trougakos IP, Dinkova-Kostova AT. Model organisms for investigating the functional involvement of NRF2 in non-communicable diseases. Redox Biol 2025; 79:103464. [PMID: 39709790 PMCID: PMC11733061 DOI: 10.1016/j.redox.2024.103464] [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: 10/25/2024] [Revised: 11/26/2024] [Accepted: 12/08/2024] [Indexed: 12/24/2024] Open
Abstract
Non-communicable chronic diseases (NCDs) are most commonly characterized by age-related loss of homeostasis and/or by cumulative exposures to environmental factors, which lead to low-grade sustained generation of reactive oxygen species (ROS), chronic inflammation and metabolic imbalance. Nuclear factor erythroid 2-like 2 (NRF2) is a basic leucine-zipper transcription factor that regulates the cellular redox homeostasis. NRF2 controls the expression of more than 250 human genes that share in their regulatory regions a cis-acting enhancer termed the antioxidant response element (ARE). The products of these genes participate in numerous functions including biotransformation and redox homeostasis, lipid and iron metabolism, inflammation, proteostasis, as well as mitochondrial dynamics and energetics. Thus, it is possible that a single pharmacological NRF2 modulator might mitigate the effect of the main hallmarks of NCDs, including oxidative, proteostatic, inflammatory and/or metabolic stress. Research on model organisms has provided tremendous knowledge of the molecular mechanisms by which NRF2 affects NCDs pathogenesis. This review is a comprehensive summary of the most commonly used model organisms of NCDs in which NRF2 has been genetically or pharmacologically modulated, paving the way for drug development to combat NCDs. We discuss the validity and use of these models and identify future challenges.
Collapse
Affiliation(s)
- Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain.
| | - Brigitta Buttari
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161, Rome, Italy
| | - Susana Cadenas
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), Cantoblanco, Madrid, Spain
| | - Ana Rita Carlos
- CE3C-CHANGE, Department of Animal Biology, Faculty of Sciences, University of Lisbon, 1749-016, Lisbon, Portugal
| | - Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Ana Sofia Falcão
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Hospital Universitario de la Princesa, Madrid, Spain
| | - Milen I Georgiev
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000, Plovdiv, Bulgaria
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - José Jimenez-Villegas
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Jarosław Olav Horbanczuk
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, 36A Postępu, Jastrzębiec, 05-552, Poland
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; Department of Neuroscience, Bilkent University, Ankara, Turkey; UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Isabel Lastres-Becker
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute Teófilo Hernando for Drug Discovery, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Viktorija Maksimova
- Department of Applied Pharmacy, Division of Pharmacy, Faculty of Medical Sciences, Goce Delcev University, Stip, Krste Misirkov Str., No. 10-A, P.O. Box 201, 2000, Stip, Macedonia
| | | | - Liliya V Mihaylova
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000, Plovdiv, Bulgaria
| | - Michel Edwar Mickael
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, 36A Postępu, Jastrzębiec, 05-552, Poland
| | - Irina Milisav
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, 1000, Ljubljana, Slovenia; Laboratory of oxidative stress research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, 1000, Ljubljana, Slovenia
| | - Biljana Miova
- Department of Experimental Physiology and Biochemistry, Institute of Biology, Faculty of Natural Sciences and Mathematics, University "St Cyril and Methodius", Skopje, Macedonia
| | - Patricia Rada
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Marlene Santos
- REQUIMTE/LAQV, Escola Superior de Saúde (E2S), Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 400, 4200-072, Porto, Portugal; Molecular Oncology & Viral Pathology, IPO-Porto Research Center (CI-IPOP), Portuguese Institute of Oncology, 4200-072, Porto, Portugal
| | - Miguel C Seabra
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, 10 000, Zagreb, Croatia
| | - Sandra Tenreiro
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, UK; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Hiramatsu G, Mizutani R, Toume K, Inada Y, Sawahata M, Uta D, Komatsu K, Kume T. Preventive Effects of Psoraleae Semen Extracts on Cognitive Dysfunction in Alzheimer's Disease Model App NL-P-F Mice. Biol Pharm Bull 2025; 48:75-79. [PMID: 39894558 DOI: 10.1248/bpb.b24-00773] [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: 02/04/2025]
Abstract
Oxidative stress and neuroinflammation accompanied by microglial activation are increased in Alzheimer's disease (AD) and contribute to the pathogenesis of AD. Nuclear factor erythroid-derived 2-related factor 2 (Nrf2) is a master transcription factor that acts as an endogenous defense mechanism against oxidative stress and inflammation and is a potential target for preventing AD. Psoraleae Semen (PS) reportedly has antioxidant and anti-inflammatory effects. This study aimed to examine the effects of PS extract (PSE) on Nrf2 activation and prevention of cognitive dysfunction in AppNL-P-F AD model mice. The effects of PSE on antioxidant response element (ARE) activity and cytoprotection in PC12 cells and on microglial activation in BV-2 cells were evaluated. PSE showed high ARE activity and prevented 6-hydroxydopamine-induced cytotoxicity in PC12 cells. Moreover, PSE suppressed lipopolysaccharide-induced nitric oxide production in BV-2 cells. Oral administration of PSE prevented cognitive dysfunction in AppNL-P-F mice without affecting motor function. Our results support that PSE can contribute to the development of new preventive and therapeutic agents for AD focusing on Nrf2 activation.
Collapse
Affiliation(s)
- Genki Hiramatsu
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Reina Mizutani
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Kazufumi Toume
- Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Department of Natural Medicine, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kodo, Kyotanabe, Kyoto 610-0395, Japan
| | - Yosuke Inada
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Masahito Sawahata
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Daisuke Uta
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Katsuko Komatsu
- Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Toshiaki Kume
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| |
Collapse
|
3
|
Lv R, Zhao Y, Wang X, He Y, Dong N, Min X, Liu X, Yu Q, Yuan K, Yue H, Yin Q. GLP-1 analogue liraglutide attenuates CIH-induced cognitive deficits by inhibiting oxidative stress, neuroinflammation, and apoptosis via the Nrf2/HO-1 and MAPK/NF-κB signaling pathways. Int Immunopharmacol 2024; 142:113222. [PMID: 39321702 DOI: 10.1016/j.intimp.2024.113222] [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: 07/31/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Obstructive sleep apnea (OSA) is a common clinical condition linked to cognitive impairment, mainly characterized by chronic intermittent hypoxia (CIH). GLP-1 receptor agonist, known for promoting insulin secretion and reducing glucose levels, has demonstrated neuroprotective effects in various experimental models such as stroke, Alzheimer's disease, and Parkinson's disease. This study aims to investigate the potential role and mechanisms of the GLP-1 receptor agonist liraglutide in ameliorating OSA-induced cognitive deficits. CIH exposure, a well-established and mature OSA pathological model, was used both in vitro and in vivo. In vitro, CIH significantly activated oxidative stress, inflammation, and apoptosis in SH-SY5Y cells. Liraglutide enhanced the nuclear translocation of Nrf2, activating its downstream pathways, thereby mitigating CIH-induced injury in SH-SY5Y cells. Additionally, liraglutide modulated the MAPK/NF-κB signaling pathway, reducing the expression of inflammatory factors and proteins. In vivo, we subjected mice to an intermittent hypoxia incubator to mimic the pathogenesis of human OSA. The Morris water maze test revealed that CIH exposure substantially impaired spatial memory. Subsequent western blot analyses and histopathological examinations indicated that liraglutide could activate the Nrf2/HO-1 axis and inhibit the MAPK/NF-κB signaling pathway, thereby alleviating OSA-associated cognitive dysfunction in mice. These findings suggest that GLP-1 receptor agonists may offer a promising preventive strategy for OSA-associated cognitive impairment. By refining these findings, we provide new insights into GLP-1's protective mechanisms in combating cognitive deficits associated with CIH, underscoring its potential as a therapeutic agent for conditions linked to OSA.
Collapse
Affiliation(s)
- Renjun Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yan Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yao He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Na Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xiangzhen Min
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xueying Liu
- Jinan Third People's Hospital, Jinan, Shandong 250132, China
| | - Qin Yu
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No. 2018RU006), Peking University, Beijing, China
| | - Hongmei Yue
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China; Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China.
| |
Collapse
|
4
|
Su B, He Z, Liu J, Li M, Huang X. Mangiferin activates the nuclear factor erythroid 2-related factor pathway to protect SOD1-G93A induced NSC-34 motor neurons from oxidative stress and apoptosis. J Biochem Mol Toxicol 2024; 38:e23849. [PMID: 39264833 DOI: 10.1002/jbt.23849] [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: 02/19/2024] [Revised: 07/28/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024]
Abstract
One of the main factors in the pathophysiology of amyotrophic lateral sclerosis is oxidative stress. Mangiferin (MF), a natural plant polyphenol, has anti-inflammatory and antioxidant effects. The aim of our study was to investigate the protective effects and mechanisms of MF in the hSOD1-G93A ALS cell model. Our result revealed that MF treatment reduced the generation of reactive oxygen species (ROS) and malondialdehyde (MDA), decreased oxidative damage, and reduced apoptosis. Additionally, it was observed that MF significantly increased the synthesis of the antioxidant genes hemeoxygenase-1 and NAD(P)H: quinone oxidoreductase 1, which are downstream of the Nrf2 signaling pathway, and increased the expression and activation of nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 knockdown greatly promoted apoptosis, which was reversed by MF treatment. To summarize, MF promoted the Nrf2 pathway and scavenged MDA and ROS to protect the ALS cell model.
Collapse
Affiliation(s)
- Boyang Su
- Medical School of Chinese PLA, Beijing, China
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhengqing He
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jing Liu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mao Li
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xusheng Huang
- Medical School of Chinese PLA, Beijing, China
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
5
|
Zhang X, Zhou H, Liu H, Xu P. Role of Oxidative Stress in the Occurrence and Development of Cognitive Dysfunction in Patients with Obstructive Sleep Apnea Syndrome. Mol Neurobiol 2024; 61:5083-5101. [PMID: 38159196 DOI: 10.1007/s12035-023-03899-3] [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: 03/15/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
Obstructive sleep apnea syndrome (OSAS) causes recurrent apnea and intermittent hypoxia at night, leading to several complications such as cognitive dysfunction. However, the molecular mechanisms underlying cognitive dysfunction in OSAS are unclear, and oxidative stress mediated by intermittent hypoxia is an important mechanism. In addition, the improvement of cognitive dysfunction in patients with OSAS varies by different treatment regimens; among them, continuous positive airway pressure therapy (CPAP) is mostly recognized for improving cognitive dysfunction. In this review, we discuss the potential mechanisms of oxidative stress in OSAS, the common factors of affecting oxidative stress and the Links between oxidative stress and inflammation in OSAS, focusing on the potential links between oxidative stress and cognitive dysfunction in OSAS and the potential therapies for neurocognitive dysfunction in patients with OSAS mediated by oxidative stress. Therefore, further analysis on the relationship between oxidative stress and cognitive dysfunction in patients with OSAS will help to clarify the etiology and discover new treatment options, which will be of great significance for early clinical intervention.
Collapse
Affiliation(s)
- XiaoPing Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hongyan Zhou
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - HaiJun Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ping Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
6
|
Hussein RM, Kandeil MA, Soliman HM, El-Shahawy AA. Effect of quercetin-loaded poly (lactic-co-glycolic) acid nanoparticles on lipopolysaccharide-induced memory decline, oxidative stress, amyloidogenesis, neurotransmission, and Nrf2/HO-1 expression. Heliyon 2024; 10:e23527. [PMID: 38169932 PMCID: PMC10758873 DOI: 10.1016/j.heliyon.2023.e23527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/11/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Neuroinflammation contributes to the pathogenesis of several neurodegenerative disorders. This study examined the neuroprotective effect of quercetin (QUR)-loaded poly (lactic-co-glycolic) acid (PLGA) nanoparticles (QUR NANO) against the neurotoxicity induced by lipopolysaccharide (LPS) in mice. A QUR NANO formulation was prepared and characterized by differential scanning calorimetry, X-ray diffraction, entrapment efficiency (EE), high-resolution transmission electron microscopy, field emission scanning electron microscopy, and in vitro drug release profile. Levels of glutathione, malondialdehyde, catalase, inducible nitric oxide synthase (iNOS), amyloid beta 42 (Aβ42), β-secretase, gamma-aminobutyric acid (GABA), and acetylcholine esterase (AChE) were measured in the mouse brain tissues. The gene expression of nuclear factor erythroid-related factor 2 (Nrf-2) and heme oxygenase-1 (HO-1) were also determined. The prepared QUR NANO formulation showed 92.07 ± 3.21% EE and drug loading of 4.62 ± 0.55. It exhibited clusters of nano-spherical particles with smooth surface areas, and the loading process was confirmed. In vivo, the QUR NANO preserved the spatial memory of mice and protected the hippocampus from LPS-induced histological lesions. The QUR NANO significantly reduced the levels of malondialdehyde, iNOS, Aβ42, β-secretase, and AChE in brain tissue homogenates. Conversely, QUR NANO increased the glutathione, catalase, and GABA concentrations and upregulated the expression of Nrf-2 and HO-1 genes. Remarkably, the neuroprotective effect of QUR NANO was significantly greater than that of herbal QUR. In summary, the prepared QUR NANO formulation was efficient in mitigating LPS-induced neurotoxicity by reducing memory loss, oxidative stress, and amyloidogenesis while preserving neurotransmission and upregulating the expression of Nrf2 and HO-1 genes. This study addresses several key factors in neuroinflammatory disorders and explores the potential of QUR-loaded nanoparticles as a novel therapeutic approach to alleviate these factors.
Collapse
Affiliation(s)
- Rasha M. Hussein
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Mutah University, Al-Karak, Jordan
| | - Mohamed A. Kandeil
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Hatem M. Soliman
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Ahmed A.G. El-Shahawy
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Egypt
| |
Collapse
|
7
|
Li X, Quan M, Wei Y, Wang W, Xu L, Wang Q, Jia J. Critical thinking of Alzheimer's transgenic mouse model: current research and future perspective. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2711-2754. [PMID: 37480469 DOI: 10.1007/s11427-022-2357-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/23/2023] [Indexed: 07/24/2023]
Abstract
Transgenic models are useful tools for studying the pathogenesis of and drug development for Alzheimer's Disease (AD). AD models are constructed usually using overexpression or knock-in of multiple pathogenic gene mutations from familial AD. Each transgenic model has its unique behavioral and pathological features. This review summarizes the research progress of transgenic mouse models, and their progress in the unique mechanism of amyloid-β oligomers, including the first transgenic mouse model built in China based on a single gene mutation (PSEN1 V97L) found in Chinese familial AD. We further summarized the preclinical findings of drugs using the models, and their future application in exploring the upstream mechanisms and multitarget drug development in AD.
Collapse
Affiliation(s)
- Xinyue Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, 100053, China.
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
8
|
Tancheva L, Kalfin R, Minchev B, Uzunova D, Tasheva K, Tsvetanova E, Georgieva A, Alexandrova A, Stefanova M, Solak A, Lazarova M, Hodzhev Y, Grigorova V, Yarkov D, Petkova-Kirova P. Memory Recovery Effect of a New Bioactive Innovative Combination in Rats with Experimental Dementia. Antioxidants (Basel) 2023; 12:2050. [PMID: 38136170 PMCID: PMC10740861 DOI: 10.3390/antiox12122050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Alzheimer's disease manifests as a complex pathological condition, with neuroinflammation, oxidative stress and cholinergic dysfunction being a few of the many pathological changes. Due to the complexity of the disease, current therapeutic strategies aim at a multitargeted approach, often relying on a combination of substances with versatile and complementary effects. In the present study, a unique combination of α-lipoic acid, citicoline, extracts of leaves from olive tree and green tea, vitamin D3, selenium and an immune-supporting complex was tested in scopolamine-induced dementia in rats. Using behavioral and biochemical methods, we assessed the effects of the combination on learning and memory, and elucidated the mechanisms of these effects. Our results showed that, compared to its components, the experimental combination was most efficient in improving short- and long-term memory as assessed by the step-through method as well as spatial memory as assessed by T-maze and Barnes maze underlined by decreases in AChE activity (p < 0.05) and LPO (p < 0.001), increases in SOD activity in the cortex (p < 0.05) and increases in catalase (p < 0.05) and GPx (p < 0.01) activities and BDNF (p < 0.001) and pCREB (p < 0.05) levels in the hippocampus. No significant histopathological changes or blood parameter changes were detected, making the experimental combination an effective and safe candidate in a multitargeted treatment of AD.
Collapse
Affiliation(s)
- Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
- Department of Healthcare, South-West University “Neofit Rilski”, Ivan Mihailov Str. 66, 2700 Blagoevgrad, Bulgaria
| | - Borislav Minchev
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Diamara Uzunova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Krasimira Tasheva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 21, 1113 Sofia, Bulgaria;
| | - Elina Tsvetanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Almira Georgieva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Albena Alexandrova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
- National Sports Academy, Department of Physiology and Biochemistry, Acad. S. Mladenov Str. 21, 1700 Sofia, Bulgaria
| | - Miroslava Stefanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Ayten Solak
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
- Institute of Cryobiology and Food Technologies, Cherni Vrah Blvd 53, 1407 Sofia, Bulgaria
| | - Maria Lazarova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Yordan Hodzhev
- National Center of Infectious and Parasitic Diseases, Yanko Sakazov Blvd 26, 1504 Sofia, Bulgaria;
| | - Valya Grigorova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Dobri Yarkov
- Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Polina Petkova-Kirova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| |
Collapse
|
9
|
Cazzaro S, Woo JAA, Wang X, Liu T, Rego S, Kee TR, Koh Y, Vázquez-Rosa E, Pieper AA, Kang DE. Slingshot homolog-1-mediated Nrf2 sequestration tips the balance from neuroprotection to neurodegeneration in Alzheimer's disease. Proc Natl Acad Sci U S A 2023; 120:e2217128120. [PMID: 37463212 PMCID: PMC10374160 DOI: 10.1073/pnas.2217128120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 06/16/2023] [Indexed: 07/20/2023] Open
Abstract
Oxidative damage in the brain is one of the earliest drivers of pathology in Alzheimer's disease (AD) and related dementias, both preceding and exacerbating clinical symptoms. In response to oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) is normally activated to protect the brain from oxidative damage. However, Nrf2-mediated defense against oxidative stress declines in AD, rendering the brain increasingly vulnerable to oxidative damage. Although this phenomenon has long been recognized, its mechanistic basis has been a mystery. Here, we demonstrate through in vitro and in vivo models, as well as human AD brain tissue, that Slingshot homolog-1 (SSH1) drives this effect by acting as a counterweight to neuroprotective Nrf2 in response to oxidative stress and disease. Specifically, oxidative stress-activated SSH1 suppresses nuclear Nrf2 signaling by sequestering Nrf2 complexes on actin filaments and augmenting Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 interaction, independently of SSH1 phosphatase activity. We also show that Ssh1 elimination in AD models increases Nrf2 activation, which mitigates tau and amyloid-β accumulation and protects against oxidative injury, neuroinflammation, and neurodegeneration. Furthermore, loss of Ssh1 preserves normal synaptic function and transcriptomic patterns in tauP301S mice. Importantly, we also show that human AD brains exhibit highly elevated interactions of Nrf2 with both SSH1 and Keap1. Thus, we demonstrate here a unique mode of Nrf2 blockade that occurs through SSH1, which drives oxidative damage and ensuing pathogenesis in AD. Strategies to inhibit SSH1-mediated Nrf2 suppression while preserving normal SSH1 catalytic function may provide new neuroprotective therapies for AD and related dementias.
Collapse
Affiliation(s)
- Sara Cazzaro
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Jung-A A. Woo
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Xinming Wang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Tian Liu
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Shanon Rego
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Teresa R. Kee
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Yeojung Koh
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Edwin Vázquez-Rosa
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Andrew A. Pieper
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH44106
- Brain Health Medicines, Center Harrington Discovery Institute, Cleveland, OH44106
| | - David E. Kang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH44106
| |
Collapse
|
10
|
Qiu X, Li L, Wei J, An X, Ampadu JA, Zheng W, Yu C, Peng C, Li X, Cai X. The protective role of Nrf2 on cognitive impairment in chronic intermittent hypoxia and sleep fragmentation mice. Int Immunopharmacol 2023; 116:109813. [DOI: 10.1016/j.intimp.2023.109813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/13/2023] [Accepted: 01/28/2023] [Indexed: 02/16/2023]
|
11
|
Baba MZ, Gomathy S, Wahedi U. Role of Nrf2 Pathway Activation in Neurological Disorder: A Brief Review. J Pharmacol Pharmacother 2022. [DOI: 10.1177/0976500x221128855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Oxidative stress plays a crucial role in the emergence of numerous neurodegenerative diseases, with protein accumulation and mitochondrial damage, which result in neurological disorders. To minimize oxidative stress, several defensive mechanisms protect nerve cells by releasing antioxidants such as nuclear erythroid factor2 (Nrf2)-Kelch-like ECH-associated protein1 (Keap1) signaling pathway activation has been proved to be a prospective treatment to reduce oxidative stress and neuroinflammation for protection of neurons in a variety of neurological disorders. In this review, we focus beneficial role of Nrf2 in Alzheimer’s and Parkinson’s diseases. Nrf2 is proved to be a master regulator of antioxidants by releasing over 250 cytoprotective genes aimed at oxidative stress and neuroinflammation. In animal studies Nrf2 activation is proved to improve autophagy, mitochondrial biogenesis, and Suppression of inflammatory cytokinin which protects neuronal cells and inhibit progressive neurodegeneration.
Collapse
Affiliation(s)
- Mohammad Zubair Baba
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ootacamund, Tamil Nadu, India
| | - S. Gomathy
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ootacamund, Tamil Nadu, India
| | - Umair Wahedi
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ootacamund, Tamil Nadu, India
| |
Collapse
|
12
|
Role of Nuclear Factor Erythroid 2 (Nrf2) in the Recovery of Long COVID-19 Using Natural Antioxidants: A Systematic Review. Antioxidants (Basel) 2022; 11:antiox11081551. [PMID: 36009268 PMCID: PMC9405009 DOI: 10.3390/antiox11081551] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/04/2022] [Accepted: 08/07/2022] [Indexed: 11/16/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease with approximately 517 million confirmed cases, with the average number of cases revealing that patients recover immediately without hospitalization. However, several other cases found that patients still experience various symptoms after 3–12 weeks, which is known as a long COVID syndrome. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can activate nuclear factor kappa beta (NF-κβ) and unbind the nuclear factor erythroid 2-related factor 2 (Nrf2) with Kelch-like ECH-associated protein 1 (Keap1), causing inhibition of Nrf2, which has an important role in antioxidant response and redox homeostasis. Disrupting the Keap1–Nrf2 pathway enhances Nrf2 activity, and has been identified as a vital approach for the prevention of oxidative stress and inflammation. Hence, natural antioxidants from various sources have been identified as a promising strategy to prevent oxidative stress, which plays a role in reducing the long COVID-19 symptoms. Oxygen-rich natural antioxidant compounds provide an effective Nrf2 activation effect that interact with the conserved amino acid residues in the Keap1-binding pocket, such as Ser602, Ser363, Ser508, and Ser555. In this review, the benefits of various natural antioxidant compounds that can modulate the Nrf2 signaling pathway, which is critical in reducing and curing long COVID-19, are highlighted and discussed.
Collapse
|
13
|
Shimizu S, Kasai S, Yamazaki H, Tatara Y, Mimura J, Engler MJ, Tanji K, Nikaido Y, Inoue T, Suganuma H, Wakabayashi K, Itoh K. Sulforaphane Increase Mitochondrial Biogenesis-Related Gene Expression in the Hippocampus and Suppresses Age-Related Cognitive Decline in Mice. Int J Mol Sci 2022; 23:ijms23158433. [PMID: 35955572 PMCID: PMC9369397 DOI: 10.3390/ijms23158433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/22/2022] Open
Abstract
Sulforaphane (SFN) is a potent activator of the transcriptional factor, Nuclear Factor Erythroid 2 (NF-E2)-Related factor 2 (NRF2). SFN and its precursor, glucoraphanin (sulforaphane glucosinolate, SGS), have been shown to ameliorate cognitive function in clinical trials and in vivo studies. However, the effects of SGS on age-related cognitive decline in Senescence-Accelerated Mouse Prone 8 (SAMP8) is unknown. In this study, we determined the preventive potential of SGS on age-related cognitive decline. One-month old SAMP8 mice or control SAM resistance 1 (SAMR1) mice were fed an ad libitum diet with or without SGS-containing broccoli sprout powder (0.3% w/w SGS in diet) until 13 months of age. SGS significantly improved long-term memory in SAMP8 at 12 months of age. Interestingly, SGS increased hippocampal mRNA and protein levels of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC1α) and mitochondrial transcription factor A (TFAM), which are master regulators of mitochondrial biogenesis, both in SAMR1 and SAMP8 at 13 months of age. Furthermore, mRNAs for nuclear respiratory factor-1 (NRF-1) and mitochondrial DNA-encoded respiratory complex enzymes, but not mitochondrial DNA itself, were increased by SGS in SAMP8 mice. These results suggest that SGS prevents age-related cognitive decline by maintaining mitochondrial function in senescence-accelerated mice.
Collapse
Affiliation(s)
- Sunao Shimizu
- Innovation Division, KAGOME Co., Ltd., 17 Nishitomiyama, Nasushiobara 329-2762, Tochigi, Japan; (S.S.); (T.I.); (H.S.)
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Shuya Kasai
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Hiromi Yamazaki
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Yota Tatara
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Junsei Mimura
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Máté János Engler
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Kunikazu Tanji
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (K.T.); (K.W.)
| | - Yoshikazu Nikaido
- Department of Metabolomics Innovation, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan
| | - Takuro Inoue
- Innovation Division, KAGOME Co., Ltd., 17 Nishitomiyama, Nasushiobara 329-2762, Tochigi, Japan; (S.S.); (T.I.); (H.S.)
| | - Hiroyuki Suganuma
- Innovation Division, KAGOME Co., Ltd., 17 Nishitomiyama, Nasushiobara 329-2762, Tochigi, Japan; (S.S.); (T.I.); (H.S.)
| | - Koichi Wakabayashi
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (K.T.); (K.W.)
| | - Ken Itoh
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
- Correspondence:
| |
Collapse
|
14
|
Microbial-derived metabolites as a risk factor of age-related cognitive decline and dementia. Mol Neurodegener 2022; 17:43. [PMID: 35715821 PMCID: PMC9204954 DOI: 10.1186/s13024-022-00548-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/30/2022] [Indexed: 02/06/2023] Open
Abstract
A consequence of our progressively ageing global population is the increasing prevalence of worldwide age-related cognitive decline and dementia. In the absence of effective therapeutic interventions, identifying risk factors associated with cognitive decline becomes increasingly vital. Novel perspectives suggest that a dynamic bidirectional communication system between the gut, its microbiome, and the central nervous system, commonly referred to as the microbiota-gut-brain axis, may be a contributing factor for cognitive health and disease. However, the exact mechanisms remain undefined. Microbial-derived metabolites produced in the gut can cross the intestinal epithelial barrier, enter systemic circulation and trigger physiological responses both directly and indirectly affecting the central nervous system and its functions. Dysregulation of this system (i.e., dysbiosis) can modulate cytotoxic metabolite production, promote neuroinflammation and negatively impact cognition. In this review, we explore critical connections between microbial-derived metabolites (secondary bile acids, trimethylamine-N-oxide (TMAO), tryptophan derivatives and others) and their influence upon cognitive function and neurodegenerative disorders, with a particular interest in their less-explored role as risk factors of cognitive decline.
Collapse
|
15
|
Li X, Ying H, Zhang Z, Yang Z, You C, Cai X, Lin Z, Xiao Y. Sulforaphane Attenuates Chronic Intermittent Hypoxia-Induced Brain Damage in Mice via Augmenting Nrf2 Nuclear Translocation and Autophagy. Front Cell Neurosci 2022; 16:827527. [PMID: 35401114 PMCID: PMC8986999 DOI: 10.3389/fncel.2022.827527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
Obstructive sleep apnea–hypopnea syndrome (OSAHS), typically characterized by chronic intermittent hypoxia (CIH), is associated with neurocognitive dysfunction in children. Sulforaphane (SFN), an activator of nuclear factor E2-related factor 2 (Nrf2), has been demonstrated to protect against oxidative stress in various diseases. However, the effect of SFN on OSAHS remains elusive. In this research, we investigated the neuroprotective role of SFN in CIH-induced cognitive dysfunction and underlying mechanisms of regulation of Nrf2 signaling pathway and autophagy. CIH exposures for 4 weeks in mice, modeling OSAHS, contributed to neurocognitive dysfunction, manifested as increased working memory errors (WMEs), reference memory errors (RMEs) and total memory errors (TEs) in the 8-arm radial maze test. The mice were intraperitoneally injected with SFN (0.5 mg/kg) 30 min before CIH exposure everyday. SFN treatment ameliorated neurocognitive dysfunction in CIH mice, which demonstrates less RME, WME, and TE. Also, SFN effectively alleviated apoptosis of hippocampal neurons following CIH by decreased TUNEL-positive cells, downregulated cleaved PARP, cleaved caspase 3, and upregulated Bcl-2. SFN protects hippocampal tissue from CIH-induced oxidative stress as evidenced by elevated superoxide dismutase (SOD) activities and reduced malondialdehyde (MDA). In addition, we found that SFN enhanced Nrf2 nuclear translocation to hold an antioxidative function on CIH-induced neuronal apoptosis in hippocampus. Meanwhile, SFN promoted autophagy activation, as shown by increased Beclin1, ATG5, and LC3II/LC3I. Overall, our findings indicated that SFN reduced the apoptosis of hippocampal neurons through antioxidant effect of Nrf2 and autophagy in CIH-induced brain damage, which highlights the potential of SFN as a novel therapy for OSAHS-related neurocognitive dysfunction.
Collapse
Affiliation(s)
- Xiucui Li
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huiya Ying
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zilong Zhang
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zijing Yang
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Cancan You
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiaohong Cai
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongdong Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanfeng Xiao
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
16
|
Alexander JF, Mahalingam R, Seua AV, Wu S, Arroyo LD, Hörbelt T, Schedlowski M, Blanco E, Kavelaars A, Heijnen CJ. Targeting the Meningeal Compartment to Resolve Chemobrain and Neuropathy via Nasal Delivery of Functionalized Mitochondria. Adv Healthc Mater 2022; 11:e2102153. [PMID: 35007407 PMCID: PMC9803615 DOI: 10.1002/adhm.202102153] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/03/2022] [Indexed: 01/03/2023]
Abstract
Cognitive deficits (chemobrain) and peripheral neuropathy occur in ∼75% of patients treated for cancer with chemotherapy and persist long-term in >30% of survivors. Without preventive or curative interventions and with increasing survivorship rates, the population debilitated by these neurotoxicities is rising. Platinum-based chemotherapeutics, including cisplatin, induce neuronal mitochondrial defects leading to chemobrain and neuropathic pain. This study investigates the capacity of nasally administered mesenchymal stem cell-derived mitochondria coated with dextran-triphenylphosphonium polymer (coated mitochondria) to reverse these neurotoxicities. Nasally administered coated mitochondria are rapidly detectable in macrophages in the brain meninges but do not reach the brain parenchyma. The coated mitochondria change expression of >2400 genes regulating immune, neuronal, endocrine and vascular pathways in the meninges of mice treated with cisplatin. Nasal administration of coated mitochondria reverses cisplatin-induced cognitive deficits and resolves neuropathic pain at a >55-times lower dose compared to uncoated mitochondria. Reversal of these neuropathologies is associated with resolution of cisplatin-induced deficits in myelination, synaptosomal mitochondrial integrity and neurogenesis. These findings demonstrate that nasally administered coated mitochondria promote resolution of chemobrain and peripheral neuropathy, thereby identifying a novel facile strategy for clinical application of mitochondrial donation and treating central and peripheral nervous system pathologies by targeting the brain meninges.
Collapse
Affiliation(s)
- Jenolyn F. Alexander
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States,Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, D-45147, Essen, Germany
| | - Rajasekaran Mahalingam
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Alexandre V. Seua
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Suhong Wu
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas, 77030, United States
| | - Luis D. Arroyo
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Tina Hörbelt
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, D-45147, Essen, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, D-45147, Essen, Germany
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas, 77030, United States
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Cobi J. Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States,Corresponding Author
| |
Collapse
|
17
|
Huang T, Tong H, Zhou H, Wang J, Hu L, Wang Y, Huang Z. ADSC-Exosomes Alleviate MTX-induced Rat Neuronal Damage by Activating Nrf2-ARE Pathway. J Mol Neurosci 2022; 72:1334-1344. [PMID: 35322376 PMCID: PMC9170627 DOI: 10.1007/s12031-022-01996-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 03/01/2022] [Indexed: 01/31/2023]
Abstract
The aim of this study was to analyze the efficacy and underlying mechanism of adipose-derived mesenchymal stem cell exosome (ADSC-exosomes)-mediated protection on methotrexate (MTX)-induced neuronal damage. We established a H2O2-induced oxidative stress model in vitro, as well as an MTX-induced neuronal damage rat model in vivo. We analyzed the effects of ADSC-exosomes on neuronal damage and Nrf2-ARE signaling pathway in rats and related mechanisms. The morphological and functional recovery of rat hippocampal neurons by ADSC-exosomes was examined by Nissl staining and modified neurological severity score (mNSS) score. The activation of Nrf2-ARE pathway effectively inhibited H2O2-induced oxidative stress. ADSC-exosomes treatment restored the activity of hippocampal neuronal cells, reduced ROS production, and inhibited hippocampal neuronal cells apoptosis. In in vivo experiments, ADSC-exosomes ameliorates MTX-induced hippocampal neuron damage by triggering Nrf2-ARE pathway, decreasing IL-6, IFN-, and TNF-a levels and TUNEL positive cells in hippocampus, and repairing hippocampal neuronal cell damage. ADSC-exosomes ameliorated MTX-induced neuronal damage and suppressed oxidative stress induced by neuronal damage through the activation of Nrf2-ARE signaling pathway.
Collapse
Affiliation(s)
- Tingting Huang
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongfei Tong
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haixia Zhou
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Juxiang Wang
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Linglong Hu
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yao Wang
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhen Huang
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
18
|
Gianferrara T, Cescon E, Grieco I, Spalluto G, Federico S. Glycogen Synthase Kinase 3β Involvement in Neuroinflammation and Neurodegenerative Diseases. Curr Med Chem 2022; 29:4631-4697. [PMID: 35170406 DOI: 10.2174/0929867329666220216113517] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/24/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND GSK-3β activity has been strictly related to neuroinflammation and neurodegeneration. Alzheimer's disease is the most studied neurodegenerative disease, but GSK-3β seems to be involved in almost all neurodegenerative diseases including Parkinson's disease, amyotrophic lateral sclerosis, frontotemporal dementia, Huntington's disease and the autoimmune disease multiple sclerosis. OBJECTIVE The aim of this review is to help researchers both working on this research topic or not to have a comprehensive overview on GSK-3β in the context of neuroinflammation and neurodegeneration. METHOD Literature has been searched using PubMed and SciFinder databases by inserting specific keywords. A total of more than 500 articles have been discussed. RESULTS First of all, the structure and regulation of the kinase were briefly discussed and then, specific GSK-3β implications in neuroinflammation and neurodegenerative diseases were illustrated also with the help of figures, to conclude with a comprehensive overview on the most important GSK-3β and multitarget inhibitors. For all discussed compounds, the structure and IC50 values at the target kinase have been reported. CONCLUSION GSK-3β is involved in several signaling pathways both in neurons as well as in glial cells and immune cells. The fine regulation and interconnection of all these pathways are at the base of the rationale use of GSK-3β inhibitors in neuroinflammation and neurodegeneration. In fact, some compounds are now under clinical trials. Despite this, pharmacodynamic and ADME/Tox profiles of the compounds were often not fully characterized and this is deleterious in such a complex system.
Collapse
Affiliation(s)
- Teresa Gianferrara
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Eleonora Cescon
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Ilenia Grieco
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Giampiero Spalluto
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Stephanie Federico
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
19
|
Ren J, Zhang S, Wang X, Deng Y, Zhao Y, Xiao Y, Liu J, Chu L, Qi X. MEF2C ameliorates learning, memory, and molecular pathological changes in Alzheimer’s disease in vivo and in vitro. Acta Biochim Biophys Sin (Shanghai) 2021; 54:77-90. [PMID: 35130621 PMCID: PMC9909301 DOI: 10.3724/abbs.2021012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Myocyte enhancer factor 2C (MEF2C) is highly expressed in the nervous system, and regulates neuro-development, synaptic plasticity, and inflammation. However, its mechanism in Alzheimer's disease (AD) is underestimated. In this study, the role and mechanism of MEF2C were investigated in the brain tissue specimens from patients with AD, APPswe/PSEN1dE9 double transgenic (APP/PS1_DT) mice, and SH-SY5Y cells treated with β-amyloid peptide (Aβ). The results indicated that the expression of MEF2C is significantly reduced, and the expression of MEF2C/Aβ in different parts of brain is negatively correlated in patients with AD. Knockdown of MEF2C promotes cell apoptosis and the level of β-amyloid precursor protein cleaving enzyme 1 (BACE) but reduces BACE2 expression. In addition, knockdown of enhances the generation and aggregation of Aβ in the cortex of APP/PS1_DT mice, reduces the expression of synaptic proteins, exacerbates the ability of learning and memory of APP/PS1_DT mice, damages the structure of mitochondria, increases the oxidative stress (OS) level, and inhibits the expression levels of members of the Nrf2-ARE signal pathway. In summary, inhibition of MEF2C exacerbates the toxic effect of Aβ and , damages synaptic plasticity, reduces the ability of learning and memory of APP/PS1 mice, and increases the level of OS via the Nrf2-ARE signal pathway.
Collapse
Affiliation(s)
- Jiamou Ren
- Key Laboratory of Endemic and Ethnic DiseasesMinistry of Education & Key Laboratory of Medical Molecular Biology of Guizhou ProvinceGuizhou Medical UniversityGuiyang550004China,Department of Laboratory Medicinethe 4th People′s Hospital of GuiyangGuiyang550004China
| | - Shuli Zhang
- Chinese People′s Liberation ArmySecret Service Center Sanatorium of XiamenXiamen361000China
| | - Xiaoling Wang
- Key Laboratory of Endemic and Ethnic DiseasesMinistry of Education & Key Laboratory of Medical Molecular Biology of Guizhou ProvinceGuizhou Medical UniversityGuiyang550004China
| | - Yuxin Deng
- Key Laboratory of Endemic and Ethnic DiseasesMinistry of Education & Key Laboratory of Medical Molecular Biology of Guizhou ProvinceGuizhou Medical UniversityGuiyang550004China
| | - Yi Zhao
- Key Laboratory of Endemic and Ethnic DiseasesMinistry of Education & Key Laboratory of Medical Molecular Biology of Guizhou ProvinceGuizhou Medical UniversityGuiyang550004China
| | - Yan Xiao
- Key Laboratory of Endemic and Ethnic DiseasesMinistry of Education & Key Laboratory of Medical Molecular Biology of Guizhou ProvinceGuizhou Medical UniversityGuiyang550004China
| | - Jian Liu
- Department of NeurosurgeryAffiliated Hospital of Guizhou Medical UniversityGuiyang550004China
| | - Liangzhao Chu
- Department of NeurosurgeryAffiliated Hospital of Guizhou Medical UniversityGuiyang550004China,Correspondence address. Tel: +86-851-86752814; E-mail: (X.Q.) / E-mail: (L.C.)@qq.com
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic DiseasesMinistry of Education & Key Laboratory of Medical Molecular Biology of Guizhou ProvinceGuizhou Medical UniversityGuiyang550004China,Translational Medicine Research CenterGuizhou Medical UniversityGuiyang550004China,Correspondence address. Tel: +86-851-86752814; E-mail: (X.Q.) / E-mail: (L.C.)@qq.com
| |
Collapse
|
20
|
Cao Y, Tan X, Lu Q, Huang K, Tang X, He Z. miR-590-3 and SP1 Promote Neuronal Apoptosis in Patients with Alzheimer's Disease via AMPK Signaling Pathway. CONTRAST MEDIA & MOLECULAR IMAGING 2021; 2021:6010362. [PMID: 34992508 PMCID: PMC8695010 DOI: 10.1155/2021/6010362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/22/2021] [Accepted: 12/02/2021] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurological degenerative illness with a hidden onset. Its pathogenesis is complicated, although with molecular biology research on cancer and targeted research on pathogenic mechanisms, good progress has not yet been made. Therefore, this work built a multifactor-driven neuronal apoptosis dysfunction module for the purpose of probing its underlying pathogenic mechanisms. We performed differential expression analysis, coexpression analysis, enrichment analysis, and hypergeometric tests to calculate the underlying regulatory effects of multifactors on the modules by the way of the whole gene expression profile of AD and identify a series of ncRNA (miR-320a) and TF (NFKB1). Additionally, we screened 10 modules corresponding to the Hub gene, which tend to regulate the physiological progress of inflammation, regulation of autophagy, cerebral cortex neuron differentiation, glial cell apoptotic, and so on. Meanwhile, Alzheimer's disease is triggered by signaling pathways such as the MPK signaling pathway. In this study, a dysfunction module is utilized to verify that miR-590-3 and SP1 motility factors can regulate neurons in Alzheimer's disease through the MPK signaling pathway, not only providing new insights into the pathogenesis of Alzheimer's disease but also laying a solid theoretical foundation for the biologists to further cure Alzheimer's disease.
Collapse
Affiliation(s)
- Yanqun Cao
- Shaoyang University Basic Medical College, Shaoyang 422000, Hunan Province, China
| | - Xiangxiang Tan
- Shaoyang University School of Nursing, Shaoyang 422000, Hunan Province, China
| | - Quzhe Lu
- Shaoyang University Basic Medical College, Shaoyang 422000, Hunan Province, China
| | - Kai Huang
- Shaoyang University Basic Medical College, Shaoyang 422000, Hunan Province, China
| | - Xiaoer Tang
- Shaoyang University Basic Medical College, Shaoyang 422000, Hunan Province, China
| | - Zhiming He
- Shaoyang University Basic Medical College, Shaoyang 422000, Hunan Province, China
| |
Collapse
|
21
|
Posttranscriptional regulation of Nrf2 through miRNAs and their role in Alzheimer's disease. Pharmacol Res 2021; 175:106018. [PMID: 34863823 DOI: 10.1016/j.phrs.2021.106018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022]
Abstract
The nuclear factor erythroid-derived 2-related factor 2 (NFE2L2/Nrf2) is a pivotal facilitator of cytoprotective responses against the oxidative/electrophilic insults. Upon activation, Nrf2 induces transcription of a wide range of cytoprotective genes having antioxidant response element (ARE) in their promoter region. Dysfunction in Nrf2 signaling has been linked to the pathogenesis of AD and several studies have suggested that boosting Nrf2 expression/activity by genetic or pharmacological approaches is beneficial in AD. Among the diverse mechanisms that regulate the Nrf2 signaling, miRNAs-mediated regulation of Nrf2 has gained much attention in recent years. Several miRNAs have been reported to directly repress the post-transcriptional expression of Nrf2 and thereby negatively regulate the Nrf2-dependent cellular cytoprotective response in AD. Moreover, several Nrf2 targeting miRNAs are misregulated in AD brains. This review is focused on the role of misregulated miRNAs that directly target Nrf2, in AD pathophysiology. Here, alongside a general description of functional interactions between miRNAs and Nrf2, we have reviewed the evidence indicating the possible role of these miRNAs in AD pathogenesis.
Collapse
|
22
|
Role of Nrf2 in Synaptic Plasticity and Memory in Alzheimer's Disease. Cells 2021; 10:cells10081884. [PMID: 34440653 PMCID: PMC8391447 DOI: 10.3390/cells10081884] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important transcription factor that reduces oxidative stress. When reactive oxygen species (ROS) or reactive nitrogen species (RNS) are detected, Nrf2 translocates from the cytoplasm into the nucleus and binds to the antioxidant response element (ARE), which regulates the expression of antioxidant and anti-inflammatory genes. Nrf2 impairments are observed in the majority of neurodegenerative disorders, including Alzheimer’s disease (AD). The classic hallmarks of AD include β-amyloid (Aβ) plaques, and neurofibrillary tangles (NFTs). Oxidative stress is observed early in AD and is a novel therapeutic target for the treatment of AD. The nuclear translocation of Nrf2 is impaired in AD compared to controls. Increased oxidative stress is associated with impaired memory and synaptic plasticity. The administration of Nrf2 activators reverses memory and synaptic plasticity impairments in rodent models of AD. Therefore, Nrf2 activators are a potential novel therapeutic for neurodegenerative disorders including AD.
Collapse
|
23
|
Bhandari R, Khanna G, Kaushik D, Kuhad A. Divulging the Intricacies of Crosstalk Between NF-Kb and Nrf2-Keap1 Pathway in Neurological Complications of COVID-19. Mol Neurobiol 2021; 58:3347-3361. [PMID: 33683626 PMCID: PMC7938034 DOI: 10.1007/s12035-021-02344-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/25/2021] [Indexed: 01/06/2023]
Abstract
The severity of COVID-19 infection is surging day by day. With the cases increasing daily, it is becoming more and more essential to understand the pathogenic mechanisms underlying the severity of the disease. It is now well known that the infection manifests itself primarily as respiratory, but the involvement of the other organ systems has now been documented in many studies. SARS-CoV-2 can invade the nervous system by a multitude of proposed mechanisms that have been discussed in this review. NF-κB and Nrf2 are transcription factors that regulate genes responsible for inflammatory and anti-oxidant response respectively. Specific focus in this review has been given to NF-κB and Nrf2 pathways that are involved in the cytokine storm and oxidative stress that are the hallmarks of COVID-19. As the immune injury is an important mechanism of neuro-invasion and neuroinflammation, there is the possible involvement of these two pathways in the neurological complications. The crosstalk mechanisms of these signaling pathways have also been discussed. Immuno-modulators both synthetic and natural are promising candidates in catering to the pathologies targeted in the aforementioned pathways.
Collapse
Affiliation(s)
- Ranjana Bhandari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India.
| | - Garima Khanna
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Dhriti Kaushik
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India.
| |
Collapse
|
24
|
Jankowska A, Satała G, Bojarski AJ, Pawłowski M, Chłoń-Rzepa G. Multifunctional Ligands with Glycogen Synthase Kinase 3 Inhibitory Activity as a New Direction in Drug Research for Alzheimer's Disease. Curr Med Chem 2021; 28:1731-1745. [PMID: 32338201 DOI: 10.2174/0929867327666200427100453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) belongs to the most common forms of dementia that causes a progressive loss of brain cells and leads to memory impairment and decline of other thinking skills. There is yet no effective treatment for AD; hence, the search for new drugs that could improve memory and other cognitive functions is one of the hot research topics worldwide. Scientific efforts are also directed toward combating behavioral and psychological symptoms of dementia, which are an integral part of the disease. Several studies have indicated that glycogen synthase kinase 3 beta (GSK3β) plays a crucial role in the pathogenesis of AD. Moreover, GSK3β inhibition provided beneficial effects on memory improvement in multiple animal models of AD. The present review aimed to update the most recent reports on the discovery of novel multifunctional ligands with GSK3β inhibitory activity as potential drugs for the symptomatic and disease-modifying therapy of AD. Compounds with GSK3β inhibitory activity seem to be an effective pharmacological approach for treating the causes and symptoms of AD as they reduced neuroinflammation and pathological hallmarks in animal models of AD and provided relief from cognitive and neuropsychiatric symptoms. These compounds have the potential to be used as drugs for the treatment of AD, but their precise pharmacological, pharmacokinetic, toxicological and clinical profiles need to be defined.
Collapse
Affiliation(s)
- Agnieszka Jankowska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Grzegorz Satała
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - Andrzej J Bojarski
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - Maciej Pawłowski
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, 30-688 Krakow, Poland
| | - GraŻyna Chłoń-Rzepa
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, 30-688 Krakow, Poland
| |
Collapse
|
25
|
Wang J, Ding Y, Zhuang L, Wang Z, Xiao W, Zhu J. Ginkgolide B‑induced AMPK pathway activation protects astrocytes by regulating endoplasmic reticulum stress, oxidative stress and energy metabolism induced by Aβ1‑42. Mol Med Rep 2021; 23:457. [PMID: 33880582 PMCID: PMC8072312 DOI: 10.3892/mmr.2021.12096] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Ginkgolide B (GB), the diterpenoid lactone compound isolated from the extracts of Ginkgo biloba leaves, significantly improves cognitive impairment, but its potential pharmacological effect on astrocytes induced by β-amyloid (Aβ)1-42 remains to be elucidated. The present study aimed to investigate the protective effect and mechanism of GB on astrocytes with Aβ1-42-induced apoptosis in Alzheimer's disease (AD). Astrocytes obtained from Sprague Dawley rats were randomly divided into control, Aβ, GB and GB + compound C groups. Cell viability and apoptosis were analyzed using Cell Counting Kit-8 and flow cytometry assays, respectively. Protein and mRNA expression levels were analyzed using western blotting and reverse transcription-quantitative PCR, respectively. The levels of superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), reactive oxygen species (ROS) and ATP were determined using the corresponding commercial kits. The findings revealed that GB attenuated Aβ1-42-induced apoptosis and the 5′ adenosine monophosphate- activated protein kinase (AMPK) inhibitor compound C reversed the protective effects of GB. In addition, GB reversed Aβ1-42-induced oxidative damage and energy metabolism disorders, including decreases in the levels of SOD, GSH-Px and ATP and increased the levels of MDA and ROS in astrocytes, while compound C reversed the anti-oxidative effect and the involvement of GB in maintaining energy metabolism in astrocytes. Finally, GB decreased the expression levels of the endoplasmic reticulum stress (ERS) proteins and the apoptotic protein CHOP and increased both mRNA and protein expression of the components of the energy metabolism-related AMPK/peroxisome proliferator-activated receptor γ coactivator 1α/peroxisome proliferator-activated receptor α and anti-oxidation-related nuclear respiratory factor 2/heme oxygenase 1/NAD(P)H dehydrogenase (quinone 1) pathways and downregulated the expression of β-secretase 1. However, compound C could antagonize these effects. In conclusion, the findings demonstrated that GB protected against Aβ1-42-induced apoptosis by inhibiting ERS, oxidative stress, energy metabolism disorders and Aβ1-42 production probably by activating AMPK signaling pathways. The findings provided an innovative insight into the treatment using GB as a therapeutic in Aβ1-42-related AD.
Collapse
Affiliation(s)
- Jing Wang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P.R. China
| | - Yan Ding
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P.R. China
| | - Linwu Zhuang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P.R. China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, Jiangsu 222000, P.R. China
| | - Wei Xiao
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, Jiangsu 222000, P.R. China
| | - Jingbo Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P.R. China
| |
Collapse
|
26
|
Qu Y, Wang W, Chen T, Yang Y, Zhang Y, Wang D. The neuroprotection of deproteinized calf blood extractives injection against Alzheimer's disease via regulation of Nrf-2 signaling. Aging (Albany NY) 2021; 13:11150-11169. [PMID: 33819182 PMCID: PMC8109110 DOI: 10.18632/aging.202776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 02/12/2021] [Indexed: 12/29/2022]
Abstract
Alzheimer’s disease (AD) is characterized by cognitive decline due to the accumulation of extracellular β-amyloid (Aβ) plaques and neurofibrillary tangles in the brain, which impair glutamate (Glu) metabolism. Deproteinized Calf Blood Extractive Injection (DCBEI) is a biopharmaceutical that contains 17 types of amino acids and 5 types of nucleotides. In this study, we found that DCBEI pretreatment reduced L-Glu-dependent neuroexcitation toxicity by maintaining normal mitochondrial function in HT22 cells. DCBEI treatment also reduced the expression of pro-apoptosis proteins and increased the expression of anti-apoptosis proteins. Furthermore, DCBEI attenuated AD-like behaviors (detected via the Morris water maze test) in B6C3-Tg (APPswePSEN1dE9)/Nju double transgenic (APP/PS1) mice; this effect was associated with a reduction in the amount of Aβ and neurofibrillary tangle deposition and the concomitant reduction of phospho-Tau in the hippocampus. Metabonomic profiling revealed that DCBEI regulated the level of neurotransmitters in the hippocampus of APP/PS1 mice. Label-free proteomics revealed that DCBEI regulated the expression of Nrf-2 and its downstream targets, as well as the levels of phospho-protein kinase B and mitogen-activated protein kinase. Together, these data show that DCBEI can ameliorate AD symptoms by upregulating Nrf2-mediated antioxidative pathways and thus preventing mitochondrial apoptosis.
Collapse
Affiliation(s)
- Yidi Qu
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenqi Wang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Tianrui Chen
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yumin Yang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yizhi Zhang
- Department of Neurology, The Second Hospital of Jilin University, Jilin University, Changchun 130041, China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
27
|
Xu D, Zhou C, Lin J, Cai W, Lin W. Dexmedetomidine provides protection to neurons against OGD/R-induced oxidative stress and neuronal apoptosis. Toxicol Mech Methods 2021; 31:374-382. [PMID: 33648426 DOI: 10.1080/15376516.2021.1888363] [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] [Indexed: 12/17/2022]
Abstract
Dexmedetomidine, a potent α2-adrenoceptor (α2-AR) agonist, is extensively used in the operating room (OR) and intensive care unit (ICU) and has been applied for the treatment of several diseases. Western blotting has been routinely used to investigate the protein levels of α-adrenergic receptor (α-AR), apoptosis related proteins (Bcl-2, Bax and Cleaved Caspase 3) and a range of proteins associated with the Nrf2/ARE pathway (Nrf2, HO-1, NQO-1, SOD) in neurons. The CCK-8 assay was used to determine cell survival rates while the Co-IP assay was used to investigate the binding ability between α2-AR and Nrf2. The TUNEL assay was used to detect cell apoptosis in neurons. OGD/R treatment reduced the level of α2-AR protein in neurons and reduced neuronal survival in a time-dependent manner. However, treatment with dexmedetomidine led to the increased protein expression of α2-AR in OGD/R-treated neurons and enhanced survival rate of OGD/R-treated neurons. From a mechanistic point-of-view, Nrf2 can effectively bind with α2-AR. Silencing Nrf2 reversed the effects of dexmedetomidine on cell viability, oxidative stress, and neuronal apoptosis in OGD/R-treated neurons. The activation of α2-AR by dexmedetomidine had a protective effect in neurons against OGD/R-triggered oxidative stress and neuronal apoptosis by modulating the Nrf2/ARE pathway.
Collapse
Affiliation(s)
- Deming Xu
- Department of Anesthesiology, The Affiliated Hospital (Group) of Putian University, Putian City, China
| | - Changbi Zhou
- Department of Anesthesiology, The Affiliated Hospital (Group) of Putian University, Putian City, China
| | - Juanyun Lin
- Department of Anesthesiology, The Affiliated Hospital (Group) of Putian University, Putian City, China
| | - Wenhui Cai
- Department of Anesthesiology, The Affiliated Hospital (Group) of Putian University, Putian City, China
| | - Wei Lin
- Department of General Surgery, The Affiliated Hospital (Group) of Putian University, Putian City, China
| |
Collapse
|
28
|
Hedrich WD, Wang H. Friend or Foe: Xenobiotic Activation of Nrf2 in Disease Control and Cardioprotection. Pharm Res 2021; 38:213-241. [PMID: 33619640 DOI: 10.1007/s11095-021-02997-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/08/2020] [Indexed: 12/30/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that governs a highly conserved pathway central to the protection of cells against various oxidative stresses. However, the biological impact of xenobiotic intervention of Nrf2 in physiological and pathophysiological conditions remains debatable. Activation of Nrf2 in cancer cells has been shown to elevate drug resistance and increase cell survival and proliferation, while inhibition of Nrf2 sensitizes cancer cells to drug treatment. On the other hand, activation of Nrf2 in normal healthy cells has been explored as a rather successful strategy for cancer chemoprevention. Selective activation of Nrf2 in off-target cells has recently been investigated as an approach for protecting off-target tissues from untoward drug toxicity. Specifically, induction of antioxidant response element genes via Nrf2 activation in cardiac cells is being explored as a means to limit the well-documented cardiotoxicity accompanied by cancer treatment with commonly prescribed anthracycline drugs. In addition to cancers, Nrf2 has been implicated in many other diseases including Alzheimer's and Parkinson's Diseases, diabetes, and cardiovascular disease. In this review, we discuss the roles of Nrf2 and its downstream target genes in the treatment of various diseases, and its recently explored potential for increasing the benefit: risk ratio of commonly utilized cancer treatments.
Collapse
Affiliation(s)
- William D Hedrich
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland, 21201, USA.,Bristol-Myers Squibb Company, Pharmaceutical Candidate Optimization, Metabolism and Pharmacokinetics, Rt. 206 and Province Line Road, Princeton, New Jersey, 08543, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland, 21201, USA.
| |
Collapse
|
29
|
Fu Y, Jia J. Isoliquiritigenin Confers Neuroprotection and Alleviates Amyloid-β42-Induced Neuroinflammation in Microglia by Regulating the Nrf2/NF-κB Signaling. Front Neurosci 2021; 15:638772. [PMID: 33642990 PMCID: PMC7904903 DOI: 10.3389/fnins.2021.638772] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Background Neuroinflammation and oxidative stress are two major pathological characteristics of Alzheimer's disease (AD). Amyloid-β oligomers (AβO), a toxic form of Aβ, promote the neuroinflammation and oxidative stress in the development of AD. Isoliquiritigenin (ISL), a natural flavonoid isolated from the root of liquorice, has been shown to exert inhibitory effects on inflammatory response and oxidative stress. Objectives The main purpose of this study is to assess the influence of ISL on inflammatory response and oxidative stress in BV2 cells stimulated with AβO, and to explore the underlying molecular mechanisms. Methods 3-(4,5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H- tetrazolium bromide (MTT) and lactate dehydrogenase (LDH) cytotoxicity assays were used to assess the toxic or protective effects of ISL. The expression levels of interleukin-1β, interleukin-6, and tumor necrosis factor-α were assessed by quantitative real-time polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assays. Morphological changes in BV2 cells were assessed by immunofluorescence method. Nitric oxide (NO) assay kit was used to determinate the NO production. Western blot, qRT-PCR and immunofluorescence were used to explore the underlying molecular mechanisms. Results ISL treatment reduced the production of inflammatory cytokines and NO, and alleviated the morphological changes in BV2 cells induced by AβO. ISL treatment further protected N2a cells from the toxic medium of AβO-stimulated BV2 cells. ISL activated nuclear factor erythroid-2 related factor 2 (Nrf2) signaling and suppressed nuclear factor-κB (NF-κB) signaling in BV2 cells. Conclusion ISL suppresses AβO-induced inflammation and oxidative stress in BV2 cells via the regulation of Nrf2/NF-κB signaling. Therefore, ISL indirectly protects neurons from the damage of toxic conditioned media.
Collapse
Affiliation(s)
- Yue Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, Beijing, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| |
Collapse
|
30
|
Protective Effect of Biobran/MGN-3 against Sporadic Alzheimer's Disease Mouse Model: Possible Role of Oxidative Stress and Apoptotic Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8845064. [PMID: 33574982 PMCID: PMC7857904 DOI: 10.1155/2021/8845064] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/14/2020] [Accepted: 01/03/2021] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is a debilitating and irreversible brain disease that affects an increasing number of aged individuals, mandating the development of protective nutraceuticals. Biobran/MGN-3, an arabinoxylan from rice bran, has potent antioxidant, antiaging, and immunomodulatory effects. The aim of the present study was to investigate the protective effect of Biobran against sporadic Alzheimer's disease (SAD). SAD was induced in mice via intracerebroventricular injection of streptozotocin (STZ) (3 mg/kg). STZ-treated mice were administered with Biobran for 21 days. The effects of Biobran on memory and learning were measured via the Morris water maze, novel object recognition, and Y-maze tests. Biomarkers for apoptosis, oxidative stress, and amyloidogenesis were measured using ELISA and western blot analysis. Histopathological examination was performed to confirm neuronal damage and amyloid-beta deposition. Biobran reversed the spatial memory deficit in SAD-induced mice, and it increased the expression of glutathione, reduced malondialdehyde, decreased IL-6, decreased intercellular adhesion molecule-1 (ICAM-1), and significantly increased nuclear factor erythroid 2-related factor 2 (Nrf2) and antioxidant response element (ARE). Moreover, Biobran exerted a protective effect against amyloid-beta-induced apoptosis via the suppression of both cleaved caspase-3 and the proapoptotic protein Bax and via the upregulation of the antiapoptotic protein Bcl-2. Furthermore, it reduced the expression of forkhead box class O proteins. It could be concluded from this study that Biobran may be a useful nutritional antioxidant agent for protection against SAD through its activation of the gene expression of Nrf2/ARE, which in turn modulates the apoptotic and amyloidogenic pathways.
Collapse
|
31
|
Owens LV, Benedetto A, Dawson N, Gaffney CJ, Parkin ET. Gene therapy-mediated enhancement of protective protein expression for the treatment of Alzheimer's disease. Brain Res 2021; 1753:147264. [PMID: 33422539 DOI: 10.1016/j.brainres.2020.147264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/22/2020] [Accepted: 12/20/2020] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is the leading form of dementia but lacks curative treatments. Current understanding of AD aetiology attributes the development of the disease to the misfolding of two proteins; amyloid-β (Aβ) and hyperphosphorylated tau, with their pathological accumulation leading to concomitant oxidative stress, neuroinflammation, and neuronal death. These processes are regulated at multiple levels to maintain homeostasis and avert disease. However, many of the relevant regulatory proteins appear to be downregulated in the AD-afflicted brain. Enhancement/restoration of these 'protective' proteins, therefore, represents an attractive therapeutic avenue. Gene therapy is a desirable means of achieving this because it is not associated with the side-effects linked to systemic protein administration, and sustained protein expression virtually eliminates compliance issues. The current article represents a focused and succinct review of the better established 'protective' protein targets for gene therapy enhancement/restoration rather than being designed as an exhaustive review incorporating less validated protein subjects. In addition, we will discuss how the risks associated with uncontrolled or irreversible gene expression might be mitigated through combining neuronal-specific promoters, inducible expression systems and localised injections. Whilst many of the gene therapy targets reviewed herein are yet to enter clinical trials, preclinical testing has thus far demonstrated encouraging potential for the gene therapy-based treatment of AD.
Collapse
Affiliation(s)
- Lauren V Owens
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Alexandre Benedetto
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Neil Dawson
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Christopher J Gaffney
- Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Edward T Parkin
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK.
| |
Collapse
|
32
|
Zhang L, Zhou Q, Zhou CL. RTA-408 protects against propofol-induced cognitive impairment in neonatal mice via the activation of Nrf2 and the inhibition of NF-κB p65 nuclear translocation. Brain Behav 2021; 11:e01918. [PMID: 33295701 PMCID: PMC7821557 DOI: 10.1002/brb3.1918] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To explore the effect of RTA-408 on the propofol-induced cognitive impairment of neonatal mice via regulating Nrf2 and NF-κB p65 nuclear translocation. METHODS C57BL/6 neonatal mice were randomized into intralipid, propofol, vehicle + propofol, and RTA-408 + propofol groups. The learning and memory ability was inspected by Morries water maze (MWM) test. TUNEL staining was performed to examine the apoptosis of neurons in hippocampus. The gene and protein expressions in hippocampus were detected by immunohistochemistry, qRT-PCR, or Western blotting. The activities of glutathione peroxidase (GPx), superoxide dismutase (SOD), and catalase (CAT) were tested by the corresponding kits. RESULTS Propofol prolonged escape latency of mice, decreased the times of crossing the platform, and shortened the time of staying in the target quadrant, while RTA-408 treatment improved the above-mentioned situation. Besides, Nrf2 protein in hippocampus of mice induced by propofol was decreased with the increased NF-κB p65 nuclear translocation, which was reversed by RTA-408. Meanwhile, RTA-408 decreased the apoptosis of neurons accompanying with the down-regulation of Caspase-3 and the up-regulations of neuronal-specific nuclear protein (NeuN), microtubule-associated protein 2 (Map2), Ca2+ /Calmodulin-dependent Protein Kinase II (CaMKII), and parvalbumin (PV) immunostaining in hippocampus. Besides, propofol-induced high levels of proinflammatory cytokines and antioxidase activities in hippocampus were reduced by RTA-408. CONCLUSION RTA-408 improved propofol-induced cognitive impairment in neonatal mice via enhancing survival of neurons, reducing the apoptosis of hippocampal neurons, mitigating the inflammation and oxidative stress, which may be correlated with the activation of Nrf2 and the inhibition of NF-κB p65 nuclear translocation.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Anesthesiology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - Qian Zhou
- Department of Anesthesiology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - Chun-Li Zhou
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
33
|
DOVINOVA I, KVANDOVA M, BALIS P, GRESOVA L, MAJZUNOVA M, HORAKOVA L, CHAN J, BARANCIK M. The Role of Nrf2 and PPARγ in the Improvement of Oxidative Stress in Hypertension and Cardiovascular Diseases. Physiol Res 2020. [DOI: 10.33549/physiolres.934612] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species are an important element of redox regulation in cells and tissues. During physiological processes, molecules undergo chemical changes caused by reduction and oxidation reactions. Free radicals are involved in interactions with other molecules, leading to oxidative stress. Oxidative stress works two ways depending on the levels of oxidizing agents and products. Excessive action of oxidizing agents damages biomolecules, while a moderate physiological level of oxidative stress (oxidative eustress) is necessary to control life processes through redox signaling required for normal cellular operation. High levels of reactive oxygen species (ROS) mediate pathological changes. Oxidative stress helps to regulate cellular phenotypes in physiological and pathological conditions. Nrf2 (nuclear factor erythroid 2-related factor 2, NFE2L2) transcription factor functions as a target nuclear receptor against oxidative stress and is a key factor in redox regulation in hypertension and cardiovascular disease. Nrf2 mediates transcriptional regulation of a variety of target genes. The Keap1-Nrf2-ARE system regulates many detoxification and antioxidant enzymes in cells after the exposure to reactive oxygen species and electrophiles. Activation of Nrf2/ARE signaling is differentially regulated during acute and chronic stress. Keap1 normally maintains Nrf2 in the cytosol and stimulates its degradation through ubiquitination. During acute oxidative stress, oxidized molecules modify the interaction of Nrf2 and Keap1, when Nrf2 is released from the cytoplasm into the nucleus where it binds to the antioxidant response element (ARE). This triggers the expression of antioxidant and detoxification genes. The consequence of long-term chronic oxidative stress is activation of glycogen synthase kinase 3β (GSK-3β) inhibiting Nrf2 activity and function. PPARγ (peroxisome proliferator-activated receptor gamma) is a nuclear receptor playing an important role in the management of cardiovascular diseases, hypertension and metabolic syndrome. PPARγ targeting of genes with peroxisome proliferator response element (PPRE) has led to the identification of several genes involved in lipid metabolism or oxidative stress. PPARγ stimulation is triggered by endogenous and exogenous ligands – agonists and it is involved in the activation of several cellular signaling pathways involved in oxidative stress response, such as the PI3K/Akt/NOS pathway. Nrf2 and PPARγ are linked together with their several activators and Nrf2/ARE and PPARγ/PPRE pathways can control several types of diseases.
Collapse
Affiliation(s)
- I DOVINOVA
- Centre of Experimental Medicine, Slovak Academy Of Sciences, Bratislava, Slovak Republic
| | - M KVANDOVA
- Center for Cardiology, Cardiology I, Medical Center of the Johannes Gutenberg-Universität Mainz, Germany
| | - P BALIS
- Centre of Experimental Medicine, Slovak Academy Of Sciences, Bratislava, Slovak Republic
| | - L GRESOVA
- Centre of Experimental Medicine, Slovak Academy Of Sciences, Bratislava, Slovak Republic
| | - M MAJZUNOVA
- Centre of Experimental Medicine, Slovak Academy Of Sciences, Bratislava, Slovak Republic
| | - L HORAKOVA
- Centre of Experimental Medicine, Slovak Academy Of Sciences, Bratislava, Slovak Republic
| | - J CHAN
- Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - M BARANCIK
- Centre of Experimental Medicine, Slovak Academy Of Sciences, Bratislava, Slovak Republic
| |
Collapse
|
34
|
Protective effects of Clostridium butyricum against oxidative stress induced by food processing and lipid-derived aldehydes in Caco-2 cells. Appl Microbiol Biotechnol 2020; 104:9343-9361. [PMID: 32965561 DOI: 10.1007/s00253-020-10896-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/18/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023]
Abstract
The human body is almost always facing the oxidative stress caused by foodborne aldehydes such as glyoxal (GO) and methylglyoxal (MGO), 4-hydroxyhexenal (HHE), and 4-hydroxynonenal (HNE). When these aldehydes build up, they can cause a range of harm. However, a probiotic, Clostridium butyricum, can increase nuclear factor erythroid-2 related factor 2 (Nrf2) and may have the potential to relieve oxidative stress. If C. butyricum is indeed resistant to aldehydes, the advantages (accessibility, convenience, and safety) will be of great significance compared with drugs. Unfortunately, whether C. butyricum can play a role in alleviating toxic effects of foodborne aldehydes in the intestine (the first line of defense against food-derived toxin) was unclear. To investigate these, we measured the viability, ROS, autophagy, and inflammatory cytokine expression of Caco-2 which were co-cultured with C. butyricum and stimulated by the four aldehydes via Nrf2 pathway (Staphylococcus aureus and Enterococcus faecium as controls). Then, we explored the link among C. butyricum, NLRP6, and Nrf2 signaling pathways when facing the stimuli. In the present study, we demonstrated that Clostridium butyricum relieved the oxidative stress induced by the aldehydes in Caco-2. Most interestingly, we found a "complementary" relationship between NLRP6 and Nrf2 in C. butyricum treatment under aldehyde stress. Our research not only makes a contribution to the popularization of C. butyricum as a probiotic-rich food instead of medicines but also sheds new light on the application of subsequent microecological formulation of C. butyricum. KEY POINTS: • The adverse effects are caused in a dose-dependent manner by foodborne aldehydes. • Clostridium butyricum can significantly ameliorate oxidative stress. • There is a "complementary" relationship between the NLRP6 and Nrf2 signaling pathways. • Using Clostridium butyricum foods to alleviate oxidative stress shows great prospects.
Collapse
|
35
|
Lanzillotta C, Zuliani I, Tramutola A, Barone E, Blarzino C, Folgiero V, Caforio M, Valentini D, Villani A, Locatelli F, Butterfield DA, Head E, Perluigi M, Abisambra JF, Di Domenico F. Chronic PERK induction promotes Alzheimer-like neuropathology in Down syndrome: Insights for therapeutic intervention. Prog Neurobiol 2020; 196:101892. [PMID: 32795489 DOI: 10.1016/j.pneurobio.2020.101892] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/30/2020] [Accepted: 08/02/2020] [Indexed: 12/11/2022]
Abstract
A major challenge in neurobiology is the identification of the mechanisms by which protein misfolding leads to cellular toxicity. Many neurodegenerative disorders, in which aberrant protein conformers aggregate into pathological inclusions, present the chronic activation of the PERK branch of the unfolded protein response. The adaptive effects of the PERK pathway include reduction of translation by transient inhibition of eIF2α and antioxidant protein production via induction of Nrf2 transcription factor. In contrast, PERK prolonged activation leads to sustained reduction in protein synthesis and induction of cell death pathways. To further investigate the role of the PERK pathway in neurodegenerative disorders, we focused on Down syndrome (DS), in which aging confers a high risk of Alzheimer disease (AD). By investigating human DS frontal cortices, we found early and sustained PERK activation associated with the induction of eIF2α and ATF4 downstream signals. We also observed that the Nrf2 response is uncoupled from PERK and its antioxidant effects are repressed in a mechanism implicating the transcription repressor Bach1. The pharmacological inhibition of PERK in DS mice reduced eIF2α-related translational repression and promoted Nrf2 nuclear translocation, favoring the rescue of Nrf2/Bach1 imbalance. The further analysis of peripheral cells from living DS individuals provided strong support of the pathological link between PERK and trisomy 21. Our results suggest that failure to regulate the PERK pathway is a peculiar characteristic of DS pathology and it may represent an essential step to promote cellular dysfunction, which actively contributes in the brain to the early development of AD.
Collapse
Affiliation(s)
- Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Ilaria Zuliani
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Carla Blarzino
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Valentina Folgiero
- Department of Pediatric Hematology/Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
| | - Matteo Caforio
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy; Department of Pediatric Hematology/Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
| | - Diletta Valentini
- Pediatric and Infectious Disease Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Alberto Villani
- Pediatric and Infectious Disease Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy; Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, USA
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Jose F Abisambra
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
36
|
Xiang J, Cao K, Dong YT, Xu Y, Li Y, Song H, Zeng XX, Ran LY, Hong W, Guan ZZ. Lithium chloride reduced the level of oxidative stress in brains and serums of APP/PS1 double transgenic mice via the regulation of GSK3β/Nrf2/HO-1 pathway. Int J Neurosci 2019; 130:564-573. [DOI: 10.1080/00207454.2019.1688808] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Jie Xiang
- Department of Pathology at Guizhou Medical University and Pathological Department at the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Kun Cao
- Department of Pathology at Guizhou Medical University and Pathological Department at the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Yang-Ting Dong
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, Guizhou, People’s Republic of China
| | - Yi Xu
- Department of Pathology at Guizhou Medical University and Pathological Department at the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Yi Li
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, Guizhou, People’s Republic of China
| | - Hui Song
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, Guizhou, People’s Republic of China
| | - Xiao-Xiao Zeng
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, Guizhou, People’s Republic of China
| | - Long-Yan Ran
- Department of Pathology at Guizhou Medical University and Pathological Department at the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Wei Hong
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, Guizhou, People’s Republic of China
| | - Zhi-Zhong Guan
- Department of Pathology at Guizhou Medical University and Pathological Department at the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, Guizhou, People’s Republic of China
| |
Collapse
|
37
|
Mitochondrial Dysfunction and Alpha-Lipoic Acid: Beneficial or Harmful in Alzheimer's Disease? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8409329. [PMID: 31885820 PMCID: PMC6914903 DOI: 10.1155/2019/8409329] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/30/2019] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterised by impairments in the cognitive domains associated with orientation, recording, and memory. This pathology results from an abnormal deposition of the β-amyloid (Aβ) peptide and the intracellular accumulation of neurofibrillary tangles. Mitochondrial dysfunctions play an important role in the pathogenesis of AD, due to disturbances in the bioenergetic properties of cells. To date, the usual therapeutic drugs are limited because of the diversity of cellular routes in AD and the toxic potential of these agents. In this context, alpha-lipoic acid (α-LA) is a well-known fatty acid used as a supplement in several health conditions and diseases, such as periphery neuropathies and neurodegenerative disorders. It is produced in several cell types, eukaryotes, and prokaryotes, showing antioxidant and anti-inflammatory properties. α-LA acts as an enzymatic cofactor able to regulate metabolism, energy production, and mitochondrial biogenesis. In addition, the antioxidant capacity of α-LA is associated with two thiol groups that can be oxidised or reduced, prevent excess free radical formation, and act on improvement of mitochondrial performance. Moreover, α-LA has mechanisms of epigenetic regulation in genes related to the expression of various inflammatory mediators, such PGE2, COX-2, iNOS, TNF-α, IL-1β, and IL-6. Regarding the pharmacokinetic profile, α-LA has rapid uptake and low bioavailability and the metabolism is primarily hepatic. However, α-LA has low risk in prolonged use, although its therapeutic potential, interactions with other substances, and adverse reactions have not been well established in clinical trials with populations at higher risk for diseases of aging. Thus, this review aimed to describe the pharmacokinetic profile, bioavailability, therapeutic efficacy, safety, and effects of combined use with centrally acting drugs, as well as report in vitro and in vivo studies that demonstrate the mitochondrial mechanisms of α-LA involved in AD protection.
Collapse
|
38
|
Zhang L, Yu J, Ye M, Zhao H. Upregulation of CKIP- 1 inhibits high-glucose induced inflammation and oxidative stress in HRECs and attenuates diabetic retinopathy by modulating Nrf2/ ARE signaling pathway: an in vitro study. Cell Biosci 2019; 9:67. [PMID: 31462987 PMCID: PMC6708125 DOI: 10.1186/s13578-019-0331-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The aim of this study was to investigate the underlying mechanisms of diabetic retinopathy (DR) development. METHODS Real-Time qPCR was used to detect Casein kinase 2 interacting protein-1 (CKIP-1) and Nuclear factor E2-related factor 2 (Nrf2) mRNA levels. Western Blot was employed to detect protein levels. Malondialdehyde (MDA) assay kit, superoxide dismutase (SOD) kit and glutathione peroxidase (GSH-Px) kit were used to evaluate oxidative stress in high-glucose treated human retinal endothelial cells (HRECs). Calcein-AM/propidium iodide (PI) double stain kit was employed to detect cell apoptosis. Enzyme-linked ImmunoSorbent Assay (ELISA) was used to detect inflammation associated cytokines secretion. Co-immunoprecipitation (CO-IP) was performed to investigate the interactions between CKIP-1 and Nrf2. Luciferase reporter gene system was used to detect the transcriptional activity of Nrf2. RESULTS CKIP-1 was significantly downregulated in either DR tissues or high-glucose treated HRECs comparing to the Control groups. Besides, high-glucose (25 mM) inhibited HRECs viability and induced oxidative stress, inflammation associated cytokines (TNF-α, IL-6 and IL-1β) secretion and cell apoptosis, which were all reversed by synergistically overexpressing CKIP-1 and aggravated by knocking down CKIP-1. Of note, we found that overexpressed CKIP-1 activated Nrf2/ARE signaling pathway and increased its downstream targets including HO-1, NQO-1, γGCS and SOD in high-glucose treated HRECs. Further results also showed that CKIP-1 regulated cell viability, oxidative stress, inflammation and apoptosis in high-glucose treated HRECs by activating Nrf2/ARE signaling pathway. CONCLUSION We concluded that overexpressed CKIP-1 alleviated DR progression by activating Nrf2/ARE signaling pathway.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Ophthalmology, Zhejiang Provincial People’s Hospital, No.158, Shangtang Road, Xiacheng District, Hangzhou, 310014 Zhejiang China
- Department of Ophthalmology, People’s Hospital of Hangzhou Medical College, No.128, ShangTang Road, XiaCheng District, Hangzhou, 310014 Zhejiang China
| | - Jie Yu
- Department of Ophthalmology, Zhejiang Provincial People’s Hospital, No.158, Shangtang Road, Xiacheng District, Hangzhou, 310014 Zhejiang China
- Department of Ophthalmology, People’s Hospital of Hangzhou Medical College, No.128, ShangTang Road, XiaCheng District, Hangzhou, 310014 Zhejiang China
| | - Mingxia Ye
- Department of Ophthalmology, Zhejiang Provincial People’s Hospital, No.158, Shangtang Road, Xiacheng District, Hangzhou, 310014 Zhejiang China
- Department of Ophthalmology, People’s Hospital of Hangzhou Medical College, No.128, ShangTang Road, XiaCheng District, Hangzhou, 310014 Zhejiang China
| | - Hailan Zhao
- Department of Ophthalmology, Zhejiang Provincial People’s Hospital, No.158, Shangtang Road, Xiacheng District, Hangzhou, 310014 Zhejiang China
- Department of Ophthalmology, People’s Hospital of Hangzhou Medical College, No.128, ShangTang Road, XiaCheng District, Hangzhou, 310014 Zhejiang China
| |
Collapse
|
39
|
Morroni F, Sita G, Graziosi A, Ravegnini G, Molteni R, Paladini MS, Dias KST, dos Santos AF, Viegas C, Camps I, Pruccoli L, Tarozzi A, Hrelia P. PQM130, a Novel Feruloyl-Donepezil Hybrid Compound, Effectively Ameliorates the Cognitive Impairments and Pathology in a Mouse Model of Alzheimer's Disease. Front Pharmacol 2019; 10:658. [PMID: 31244664 PMCID: PMC6581760 DOI: 10.3389/fphar.2019.00658] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/21/2019] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is the most frequent type of dementia in older people. The complex nature of AD calls for the development of multitarget agents addressing key pathogenic processes. Donepezil, an acetylcholinesterase inhibitor, is a first-line acetylcholinesterase inhibitor used for the treatment of AD. Although several studies have demonstrated the symptomatic efficacy of donepezil treatment in AD patients, the possible effects of donepezil on the AD process are not yet known. In this study, a novel feruloyl-donepezil hybrid compound (PQM130) was synthesized and evaluated as a multitarget drug candidate against the neurotoxicity induced by Aβ1-42 oligomer (AβO) injection in mice. Interestingly, PQM130 had already shown anti-inflammatory activity in different in vivo models and neuroprotective activity in human neuronal cells. The intracerebroventricular (i.c.v.) injection of AβO in mice caused the increase of memory impairment, oxidative stress, neurodegeneration, and neuroinflammation. Instead, PQM130 (0.5-1 mg/kg) treatment after the i.c.v. AβO injection reduced oxidative damage and neuroinflammation and induced cell survival and protein synthesis through the modulation of glycogen synthase kinase 3β (GSK3β) and extracellular signal-regulated kinases (ERK1/2). Moreover, PQM130 increased brain plasticity and protected mice against the decline in spatial cognition. Even more interesting is that PQM130 modulated different pathways compared to donepezil, and it is much more effective in counteracting AβO damage. Therefore, our findings highlighted that PQM130 is a potent multi-functional agent against AD and could act as a promising neuroprotective compound for anti-AD drug development.
Collapse
Affiliation(s)
- Fabiana Morroni
- Department of Pharmacy and BioTechnology–FaBiT, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Giulia Sita
- Department of Pharmacy and BioTechnology–FaBiT, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Agnese Graziosi
- Department of Pharmacy and BioTechnology–FaBiT, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Gloria Ravegnini
- Department of Pharmacy and BioTechnology–FaBiT, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Maria Serena Paladini
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | | | | | - Claudio Viegas
- Institute of Chemistry, Federal University of Alfenas, Alfenas, MG, Brazil
| | - Ihosvany Camps
- Institute of Exact Sciences, Federal University of Alfenas, Alfenas, MG, Brazil
| | - Letizia Pruccoli
- Department for Life Quality Studies-QuVi, Alma Mater Studiorum-University of Bologna, Rimini, Italy
| | - Andrea Tarozzi
- Department for Life Quality Studies-QuVi, Alma Mater Studiorum-University of Bologna, Rimini, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and BioTechnology–FaBiT, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| |
Collapse
|