251
|
van Onselen R, Downing TG. β- N-methylamino-L-alanine Inhibits Human Catalase Activity: Possible Implications for Neurodegenerative Disease Development. Int J Toxicol 2019; 38:129-134. [PMID: 30663459 DOI: 10.1177/1091581818821921] [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] [Indexed: 01/15/2023]
Abstract
The naturally produced, nonprotein amino acid β- N-methylamino-l-alanine (BMAA) has been proposed as a significant contributor to sporadic neurodegenerative disease development worldwide. However, the existing hypothesized mechanisms of toxicity do not adequately explain the role of BMAA in neurodegenerative disease development. There is evidence for BMAA-induced enzyme inhibition, but the effect of BMAA on human stress response enzymes has received little attention, despite the well-described role of oxidative stress in neurodegenerative disease development. The aim of this study was therefore to investigate the effect of BMAA on human catalase activity and compare it to the known inhibitor 3-amino-1,2,4-triazole. BMAA inhibited human erythrocyte catalase in a cell-free exposure to the same extent as the known inhibitor. Based on enzyme kinetics, the inhibition appears to be noncompetitive, possibly as a result of BMAA binding in the nicotinamide adenine dinucleotide phosphate (NADPH) binding site. BMAA-induced catalase inhibition was also observed in a human cell line culture. We therefore propose that BMAA-induced enzyme inhibition, specifically catalase inhibition, is a mechanism of toxicity that may contribute to the neurotoxicity of BMAA, further supporting the role of BMAA in neurodegenerative disease development.
Collapse
Affiliation(s)
- Rianita van Onselen
- 1 Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa
| | - Tim G Downing
- 1 Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa
| |
Collapse
|
252
|
Włodarek D. Role of Ketogenic Diets in Neurodegenerative Diseases (Alzheimer's Disease and Parkinson's Disease). Nutrients 2019; 11:nu11010169. [PMID: 30650523 PMCID: PMC6356942 DOI: 10.3390/nu11010169] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/31/2018] [Accepted: 01/09/2019] [Indexed: 12/29/2022] Open
Abstract
The goal of this review was to assess the effectiveness of ketogenic diets on the therapy of neurodegenerative diseases. The ketogenic diet is a low-carbohydrate and fat-rich diet. Its implementation has a fasting-like effect, which brings the body into a state of ketosis. The ketogenic diet has, for almost 100 years, been used in the therapy of drug-resistant epilepsy, but current studies indicate possible neuroprotective effects. Thus far, only a few studies have evaluated the role of the ketogenic diet in the prevention of Parkinson’s disease (PD) and Alzheimer’s disease (AD). Single studies with human participants have demonstrated a reduction of disease symptoms after application. The application of the ketogenic diet to elderly people, however, raises certain concerns. Persons with neurodegenerative diseases are at risk of malnutrition, while food intake reduction is associated with disease symptoms. In turn, the ketogenic diet leads to a reduced appetite; it is not attractive from an organoleptic point of view, and may be accompanied by side effects of the gastrointestinal system. All this may lead to further lowering of consumed food portions by elderly persons with neurodegenerative diseases and, in consequence, to further reduction in the supply of nutrients provided by the diet. Neither data on the long-term application of the ketogenic diet in patients with neurodegenerative disease or data on its effects on disease symptoms are available. Further research is needed to evaluate the suitability of the ketogenic diet in the therapy of AD- or PD-affected persons.
Collapse
Affiliation(s)
- Dariusz Włodarek
- Department of Dietetics, Faculty of Human Nutrition and Consumer Sciences, Warsaw University of Life Sciences (WULS-SGGW), 159c Nowoursynowska Str., 02-776 Warsaw, Poland.
| |
Collapse
|
253
|
Tan SH, Karri V, Tay NWR, Chang KH, Ah HY, Ng PQ, Ho HS, Keh HW, Candasamy M. Emerging pathways to neurodegeneration: Dissecting the critical molecular mechanisms in Alzheimer's disease, Parkinson's disease. Biomed Pharmacother 2019; 111:765-777. [PMID: 30612001 DOI: 10.1016/j.biopha.2018.12.101] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/18/2018] [Accepted: 12/23/2018] [Indexed: 01/02/2023] Open
Abstract
Neurodegenerative diseases are usually sporadic in nature and commonly influenced by a wide range of genetic, life style and environmental factors. A unifying feature of Alzheimer's disease (AD) and Parkinson's disease (PD) is the abnormal accumulation and processing of mutant or damaged intra and extracellular proteins; this leads to neuronal vulnerability and dysfunction in the brain. Through a detailed review of ubiquitin proteasome, mRNA splicing, mitochondrial dysfunction, and oxidative stress pathway interrelation on neurodegeneration can improve the understanding of the disease mechanism. The identified pathways common to AD and PD nominate promising new targets for further studies, and as well as biomarkers. These insights suggested would likely provide major stimuli for developing unified treatment approaches to combat neurodegeneration. More broadly, pathways can serve as vehicles for integrating findings from diverse studies of neurodegeneration. The evidence examined in this review provides a brief overview of the current literature on significant pathways in promoting in AD, PD. Additionally, these insights suggest that biomarkers and treatment strategies may require simultaneous targeting of multiple components.
Collapse
Affiliation(s)
- Sean Hong Tan
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Venkatanaidu Karri
- Department of Toxicogenomics, Faculty of Health, Medicines, Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Nicole Wuen Rong Tay
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Kuan Hui Chang
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Hui Yen Ah
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Phui Qi Ng
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Hui San Ho
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Hsiao Wai Keh
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
254
|
Liao W, Zheng Y, Fang W, Liao S, Xiong Y, Li Y, Xiao S, Zhang X, Liu J. Dual Specificity Phosphatase 6 Protects Neural Stem Cells from β-Amyloid-Induced Cytotoxicity through ERK1/2 Inactivation. Biomolecules 2018; 8:E181. [PMID: 30572643 PMCID: PMC6315916 DOI: 10.3390/biom8040181] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/09/2018] [Accepted: 12/11/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease with limited treatment options and no cure. Beta-amyloid (Aβ) is a hallmark of AD that has potent neurotoxicity in neural stem cells (NSCs). Dual specificity phosphatase 6 (DUSP6) is a member of the mitogen-activated protein kinases (MAPKs), which is involved in regulating various physiological and pathological processes. Whether DUSP6 has a protective effect on Aβ-induced NSC injury remains to be explored. C17.2 neural stem cells were transfected with DUSP6-overexpressed plasmid. NSCs with or without DUSP6 overexpression were administrated with Aβ25⁻35 at various concentrations (i.e., 0, 2.5, 5 μM). DUSP6 expression after Aβ treatment was detected by Real-Time Polymerase Chain Reaction (RT-PCR) and Western blot and cell vitality was examined by the CCK8 assay. The oxidative stress (intracellular reactive oxygen species (ROS) and malondialdehyde (MDA)), endoplasmic reticulum stress (ER calcium level) and mitochondrial dysfunction (cytochrome c homeostasis) were tested. The expression of p-ERK1/2 and ERK1/2 were assayed by Western blot. Our results showed that Aβ decreased the expression of DUSP6 in a dose-dependent manner. The overexpression of DUSP6 increased the cell vitality of NSCs after Aβ treatment. Oxidative stress, ER stress, and mitochondrial dysfunction induced by Aβ could be restored by DUSP6 overexpression. Additionally, the Aβ-induced ERK1/2 activation was reversed. In summary, DUSP6 might have a neuroprotective effect on Aβ-induced cytotoxicity, probably via ERK1/2 activation.
Collapse
Affiliation(s)
- Wang Liao
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China.
| | - Yuqiu Zheng
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China.
| | - Wenli Fang
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China.
| | - Shaowei Liao
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China.
| | - Ying Xiong
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China.
| | - Yi Li
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China.
| | - Songhua Xiao
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China.
| | - Xingcai Zhang
- John A Paulson School of Engineering and Applied Science, Harvard University, Cambridge, MA 02138, USA.
| | - Jun Liu
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
255
|
Kausar S, Wang F, Cui H. The Role of Mitochondria in Reactive Oxygen Species Generation and Its Implications for Neurodegenerative Diseases. Cells 2018; 7:cells7120274. [PMID: 30563029 PMCID: PMC6316843 DOI: 10.3390/cells7120274] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/07/2018] [Accepted: 12/14/2018] [Indexed: 12/21/2022] Open
Abstract
Mitochondria are dynamic cellular organelles that consistently migrate, fuse, and divide to modulate their number, size, and shape. In addition, they produce ATP, reactive oxygen species, and also have a biological role in antioxidant activities and Ca2+ buffering. Mitochondria are thought to play a crucial biological role in most neurodegenerative disorders. Neurons, being high-energy-demanding cells, are closely related to the maintenance, dynamics, and functions of mitochondria. Thus, impairment of mitochondrial activities is associated with neurodegenerative diseases, pointing to the significance of mitochondrial functions in normal cell physiology. In recent years, considerable progress has been made in our knowledge of mitochondrial functions, which has raised interest in defining the involvement of mitochondrial dysfunction in neurodegenerative diseases. Here, we summarize the existing knowledge of the mitochondrial function in reactive oxygen species generation and its involvement in the development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Saima Kausar
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing 400716, China.
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China.
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China.
| | - Feng Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing 400716, China.
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China.
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China.
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing 400716, China.
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China.
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China.
| |
Collapse
|
256
|
Budziosz J, Stanek A, Sieroń A, Witkoś J, Cholewka A, Sieroń K. Effects of Low-Frequency Electromagnetic Field on Oxidative Stress in Selected Structures of the Central Nervous System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1427412. [PMID: 30647806 PMCID: PMC6311791 DOI: 10.1155/2018/1427412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/07/2018] [Accepted: 11/14/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The aim of the study was to evaluate the effects of a 28-day exposure to a 50 Hz electromagnetic field of 10 kV/m on the oxidative stress in selected rat central nervous system (CNS) structures. MATERIAL AND METHODS Twenty male Wistar rats served as experimental subjects. Ten rats were exposed to an electromagnetic field with a frequency of 50 Hz, intensity of 10 kV/m, and magnetic induction of 4.3 pT for 22 hours a day. The control group of ten rats was subject to sham exposure. Homogenates of the frontal cortex, hippocampus, brainstem, hypothalamus, striatum, and cerebellum were evaluated for selected parameters of oxidative stress. RESULTS Following the four-week exposure to a low-frequency electromagnetic field, the mean malondialdehyde levels and total oxidant status of CNS structures did not differ significantly between the experimental and control groups. However, the activities of antioxidant enzymes in brain structure homogenates were decreased except for frontal cortex catalase, glutathione peroxidase, and hippocampal glutathione reductase. The low-frequency electromagnetic field had no effect on the nonenzymatic antioxidant system of the examined brain structures except for the frontal cortex. CONCLUSION The four-week exposure of male rats to a low-frequency electromagnetic field did not affect oxidative stress in the investigated brain structures.
Collapse
Affiliation(s)
- Jan Budziosz
- School of Health Sciences in Katowice, Department of Physical Medicine, Chair of Physiotherapy, Medical University of Silesia, Medyków Street 12, 40-752 Katowice, Poland
| | - Agata Stanek
- School of Medicine with the Division of Dentistry in Zabrze, Department of Internal Medicine, Angiology and Physical Medicine, Medical University of Silesia, Batorego Street 15, 41-902 Bytom, Poland
| | - Aleksander Sieroń
- School of Medicine with the Division of Dentistry in Zabrze, Department of Internal Medicine, Angiology and Physical Medicine, Medical University of Silesia, Batorego Street 15, 41-902 Bytom, Poland
| | - Joanna Witkoś
- School of Health Sciences in Katowice, Department of Physical Medicine, Chair of Physiotherapy, Medical University of Silesia, Medyków Street 12, 40-752 Katowice, Poland
| | - Armand Cholewka
- Department of Medical Physics, Chełkowski Institute of Physics, University of Silesia, 4 Uniwersytecka Street, 40-007 Katowice, Poland
| | - Karolina Sieroń
- School of Health Sciences in Katowice, Department of Physical Medicine, Chair of Physiotherapy, Medical University of Silesia, Medyków Street 12, 40-752 Katowice, Poland
| |
Collapse
|
257
|
Issitt T, Bosseboeuf E, De Winter N, Dufton N, Gestri G, Senatore V, Chikh A, Randi AM, Raimondi C. Neuropilin-1 Controls Endothelial Homeostasis by Regulating Mitochondrial Function and Iron-Dependent Oxidative Stress. iScience 2018; 11:205-223. [PMID: 30623799 PMCID: PMC6327076 DOI: 10.1016/j.isci.2018.12.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/24/2018] [Accepted: 12/04/2018] [Indexed: 01/13/2023] Open
Abstract
The transmembrane protein neuropilin-1 (NRP1) promotes vascular endothelial growth factor (VEGF) and extracellular matrix signaling in endothelial cells (ECs). Although it is established that NRP1 is essential for angiogenesis, little is known about its role in EC homeostasis. Here, we report that NRP1 promotes mitochondrial function in ECs by preventing iron accumulation and iron-induced oxidative stress through a VEGF-independent mechanism in non-angiogenic ECs. Furthermore, NRP1-deficient ECs have reduced growth and show the hallmarks of cellular senescence. We show that a subcellular pool of NRP1 localizes in mitochondria and interacts with the mitochondrial transporter ATP-binding cassette B8 (ABCB8). NRP1 loss reduces ABCB8 levels, resulting in iron accumulation, iron-induced mitochondrial superoxide production, and iron-dependent EC senescence. Treatment of NRP1-deficient ECs with the mitochondria-targeted antioxidant compound mitoTEMPO or with the iron chelator deferoxamine restores mitochondrial activity, inhibits superoxide production, and protects from cellular senescence. This finding identifies an unexpected role of NRP1 in EC homeostasis. A subcellular pool of NRP1 localizes in the mitochondria of endothelial cells (ECs) NRP1 regulates mitochondrial function via ABCB8 transporter NRP1 loss induces iron accumulation and iron-dependent oxidative stress in ECs NRP1 protects ECs from iron-dependent premature cellular senescence
Collapse
Affiliation(s)
- Theo Issitt
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Emy Bosseboeuf
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Natasha De Winter
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Neil Dufton
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Gaia Gestri
- Division of Biosciences, Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Valentina Senatore
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Anissa Chikh
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Anna M Randi
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Claudio Raimondi
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
258
|
Sahiner UM, Birben E, Erzurum S, Sackesen C, Kalayci Ö. Oxidative stress in asthma: Part of the puzzle. Pediatr Allergy Immunol 2018; 29:789-800. [PMID: 30069955 DOI: 10.1111/pai.12965] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/08/2018] [Accepted: 07/23/2018] [Indexed: 01/17/2023]
Abstract
An imbalance between the production of reactive oxygen species and the capacity of antioxidant defense mechanisms favoring oxidants is called oxidative stress and is implicated in asthmatic inflammation and severity. Major reactive oxygen species that are formed endogenously include hydrogen peroxide, superoxide anion, hydroxyl radical, and hypohalite radical; and the major antioxidants that fight against the endogenous and environmental oxidants are superoxide dismutase, catalase, and glutathione. Despite the well-known presence of oxidative stress in asthma, studies that target oxidative burden using a variety of nutritional, pharmacological, and environmental approaches have generally been disappointing. In this review, we summarize the current knowledge on oxidative stress and antioxidant imbalance in asthma. In addition, we focus on possible biomarkers of oxidative stress in asthma and on current and future treatment strategies using the modulation of oxidative stress to treat asthma patients.
Collapse
Affiliation(s)
- Umit M Sahiner
- Department of Pediatric Allergy and Asthma, Hacettepe University School of Medicine, Ankara, Turkey
| | - Esra Birben
- Department of Pediatric Allergy and Asthma, Hacettepe University School of Medicine, Ankara, Turkey
| | - Serpil Erzurum
- Department of Pathobiology, Cleveland Clinic, Lerner Research Institute, and the Respiratory Institute, Cleveland, Ohio
| | - Cansin Sackesen
- Department of Pediatric Allergy, Koc University School of Medicine, Istanbul, Turkey
| | - Ömer Kalayci
- Department of Pediatric Allergy and Asthma, Hacettepe University School of Medicine, Ankara, Turkey
| |
Collapse
|
259
|
Resveratrol and Alzheimer's disease. From molecular pathophysiology to clinical trials. Exp Gerontol 2018; 113:36-47. [DOI: 10.1016/j.exger.2018.09.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/04/2018] [Accepted: 09/21/2018] [Indexed: 12/18/2022]
|
260
|
Zeinsteger PA, Barberón JL, Leaden PJ, Palacios A. Antioxidant properties of Calendula officinalis L. (Asteraceae) on Fe2+-initiated peroxidation of rat brain mitochondria. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2254-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
261
|
Kim HJ, Jung SW, Kim SY, Cho IH, Kim HC, Rhim H, Kim M, Nah SY. Panax ginseng as an adjuvant treatment for Alzheimer's disease. J Ginseng Res 2018; 42:401-411. [PMID: 30337800 PMCID: PMC6190533 DOI: 10.1016/j.jgr.2017.12.008] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 11/29/2017] [Accepted: 12/12/2017] [Indexed: 01/22/2023] Open
Abstract
Longevity in medicine can be defined as a long life without mental or physical deficits. This can be prevented by Alzheimer's disease (AD). Current conventional AD treatments only alleviate the symptoms without reversing AD progression. Recent studies demonstrated that Panax ginseng extract improves AD symptoms in patients with AD, and the two main components of ginseng might contribute to AD amelioration. Ginsenosides show various AD-related neuroprotective effects. Gintonin is a newly identified ginseng constituent that contains lysophosphatidic acids and attenuates AD-related brain neuropathies. Ginsenosides decrease amyloid β-protein (Aβ) formation by inhibiting β- and γ-secretase activity or by activating the nonamyloidogenic pathway, inhibit acetylcholinesterase activity and Aβ-induced neurotoxicity, and decrease Aβ-induced production of reactive oxygen species and neuroinflammatory reactions. Oral administration of ginsenosides increases the expression levels of enzymes involved in acetylcholine synthesis in the brain and alleviates Aβ-induced cholinergic deficits in AD models. Similarly, gintonin inhibits Aβ-induced neurotoxicity and activates the nonamyloidogenic pathway to reduce Aβ formation and to increase acetylcholine and choline acetyltransferase expression in the brain through lysophosphatidic acid receptors. Oral administration of gintonin attenuates brain amyloid plaque deposits, boosting hippocampal cholinergic systems and neurogenesis, thereby ameliorating learning and memory impairments. It also improves cognitive functions in patients with AD. Ginsenosides and gintonin attenuate AD-related neuropathology through multiple routes. This review focuses research demonstrating that ginseng constituents could be a candidate as an adjuvant for AD treatment. However, clinical investigations including efficacy and tolerability analyses may be necessary for the clinical acceptance of ginseng components in combination with conventional AD drugs.
Collapse
Key Words
- AChE, acetylcholinesterase
- AD, Alzheimer's disease
- APP, amyloid precursor protein
- Adjuvant
- Alzheimer's disease
- Aβ, amyloid β-protein
- BDNF, brain-derived neurotrophic factor
- EGF, Epidermal growth factor
- GLP151, ginseng major latex-like protein 151
- Ginsenoside
- Gintonin
- LPA, Lysophosphatidic acid
- NGF, nerve growth factor
- NMDA, n-methyl-d-aspartic acid
- PI3K, phosphoinositide-3 kinase
- PPARγ, peroxisome proliferator-activated receptor-γ
- Panax ginseng
- ROS, reactive oxygen species
- sAPPα, soluble amyloid precursor protein α
Collapse
Affiliation(s)
- Hyeon-Joong Kim
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Seok-Won Jung
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Seog-Young Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine and Institute of Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and toxicology program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Manho Kim
- Department of Neurology, Neuroscience Research Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
262
|
Ferreira CA, Ni D, Rosenkrans ZT, Cai W. Scavenging of reactive oxygen and nitrogen species with nanomaterials. NANO RESEARCH 2018; 11:4955-4984. [PMID: 30450165 PMCID: PMC6233906 DOI: 10.1007/s12274-018-2092-y] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/01/2018] [Accepted: 05/04/2018] [Indexed: 05/03/2023]
Abstract
Reactive oxygen and nitrogen species (RONS) are essential for normal physiological processes and play important roles in cell signaling, immunity, and tissue homeostasis. However, excess radical species are implicated in the development and augmented pathogenesis of various diseases. Several antioxidants may restore the chemical balance, but their use is limited by disappointing results of clinical trials. Nanoparticles are an attractive therapeutic alternative because they can change the biodistribution profile of antioxidants, and possess intrinsic ability to scavenge RONS. Herein, we review the types of RONS, how they are implicated in several diseases, and the types of nanoparticles with inherent antioxidant capability, their mechanisms of action, and their biological applications.
Collapse
Affiliation(s)
- Carolina A. Ferreira
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dalong Ni
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
263
|
Li X, Jiang LH. A critical role of the transient receptor potential melastatin 2 channel in a positive feedback mechanism for reactive oxygen species-induced delayed cell death. J Cell Physiol 2018; 234:3647-3660. [PMID: 30229906 DOI: 10.1002/jcp.27134] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 07/05/2018] [Indexed: 12/19/2022]
Abstract
Transient receptor potential melastatin 2 (TRPM2) channel activation by reactive oxygen species (ROS) plays a critical role in delayed neuronal cell death, responsible for postischemia brain damage via altering intracellular Zn2+ homeostasis, but a mechanistic understanding is still lacking. Here, we showed that H2 O2 induced neuroblastoma SH-SY5Y cell death with a significant delay, dependently of the TRPM2 channel and increased [Zn2+ ]i , and therefore used this cell model to investigate the mechanisms underlying ROS-induced TRPM2-mediated delayed cell death. H2 O2 increased concentration-dependently the [Zn2+ ]i and caused lysosomal dysfunction and Zn2+ loss and, furthermore, mitochondrial Zn2+ accumulation, fragmentation, and ROS generation. Such effects were suppressed by preventing poly(adenosine diphosphate ribose, ADPR) polymerase-1-dependent TRPM2 channel activation with PJ34 and 3,3',5,5'-tetra-tert-butyldiphenoquinone, inhibiting the TRPM2 channel with 2-aminoethoxydiphenyl borate (2-APB) and N-(p-amylcinnamoyl)anthranilic acid, or chelating Zn2+ with N,N,N,N-tetrakis(2-pyridylmethyl)-ethylenediamine (TPEN). Bafilomycin-induced lysosomal dysfunction also resulted in mitochondrial Zn2+ accumulation, fragmentation, and ROS generation that were inhibited by PJ34 or 2-APB, suggesting that these mitochondrial events are TRPM2 dependent and sequela of lysosomal dysfunction. Mitochondrial TRPM2 expression was detected and exposure to ADPR-induced Zn2+ uptake in isolated mitochondria, which was prevented by TPEN. H2 O2 -induced delayed cell death was inhibited by apocynin and diphenyleneiodonium, nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) oxidase (NOX) inhibitors, GKT137831, an NOX1/4-specific inhibitor, or Gö6983, a protein kinase C (PKC) inhibitor. Moreover, inhibition of PKC/NOX prevented H2 O2 -induced ROS generation, lysosomal dysfunction and Zn2+ release, and mitochondrial Zn2+ accumulation, fragmentation and ROS generation. Collectively, these results support a critical role for the TRPM2 channel in coupling PKC/NOX-mediated ROS generation, lysosomal Zn2+ release, and mitochondrial Zn2+ accumulation, and ROS generation to form a vicious positive feedback signaling mechanism for ROS-induced delayed cell death.
Collapse
Affiliation(s)
- Xin Li
- Sino-UK Joint Laboratory of Brain Function and Injury, Xinxiang Medical University, Xinxiang, China.,Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury, Xinxiang Medical University, Xinxiang, China.,Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
264
|
Yang L, Shen Q, Xia Y, Lei X, Peng J. Sevoflurane‑induced neurotoxicity is driven by OXR1 post‑transcriptional downregulation involving hsa‑miR‑302e. Mol Med Rep 2018; 18:4657-4665. [PMID: 30221705 DOI: 10.3892/mmr.2018.9442] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 04/13/2018] [Indexed: 11/05/2022] Open
Abstract
Sevoflurane is a common anesthetic agent used in surgical settings and previous studies have indicated that it exerts a neurotoxic effect. However, the molecular mechanism underlying this side effect is unknown. In addition, the human microRNA‑302 (hsa‑miR‑302) family members have been reported to be involved in neuronal cell development and biology. Thus, the present study aimed to investigate the potential implication of hsa‑miR‑302e in the sevoflurane‑induced cytotoxicity on human hippocampal cells (HN‑h). HN‑h cells were transfected with hsa‑miR‑302e mimic, hsa‑miR‑302e inhibitor or negative controls and subsequently exposed to different concentrations of sevoflurane. An MTT assay was used to assess the cytotoxicity of sevoflurane on HN‑h cells. Cell apoptosis was determined by flow cytometry. The levels of lactate dehydrogenase release, reactive oxygen species, lipid peroxidation and intracellular calcium (Ca2+) were additionally detected. Reverse transcription‑quantitative polymerase chain reaction and western blotting were conducted to determine mRNA and protein expression, respectively. A luciferase assay was performed for validating the targeting of OXR1 by hsa‑miR‑302e. The results indicated that sevoflurane induced a decrease in cell viability, malondialdehyde and reactive oxygen species production, lactate dehydrogenase release, intracellular Ca2+ production, calcium/calmodulin‑dependent protein kinase II phosphorylation and apoptosis. In addition, treatment with sevoflurane induced the expression of hsa‑miR‑302e while the expression of its target, oxidation resistance gene 1 (OXR1), was significantly downregulated. Inhibition of hsa‑miR‑302e expression protected neuronal cells from sevoflurane cytotoxicity. Mechanistic studies demonstrated that OXR1 was a direct target of hsa‑miR‑302e. Furthermore, the overexpression of OXR1 abolished the effect of sevoflurane on neuronal cells. The results of the present study indicated that sevoflurane exerts its neurotoxic effect by regulating the hsa‑miR‑302e/OXR1 axis. Therefore, the manipulation of the hsa‑miR‑302e/OXR1 pathway will be useful for preventing sevoflurane‑induced neurotoxicity.
Collapse
Affiliation(s)
- Leilei Yang
- Department of Anesthesiology, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei 430060, P.R. China
| | - Qian Shen
- Department of Anesthesiology, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei 430060, P.R. China
| | - Yanqiong Xia
- Department of Anesthesiology, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei 430060, P.R. China
| | - Xueheng Lei
- Department of Anesthesiology, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei 430060, P.R. China
| | - Jian Peng
- Department of Anesthesiology, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
265
|
Ahmadian N, Hejazi S, Mahmoudi J, Talebi M. Tau Pathology of Alzheimer Disease: Possible Role of Sleep Deprivation. Basic Clin Neurosci 2018; 9:307-316. [PMID: 30719245 PMCID: PMC6360494 DOI: 10.32598/bcn.9.5.307] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/17/2017] [Accepted: 02/18/2018] [Indexed: 12/24/2022] Open
Abstract
Sleep deprivation is a common complaint in modern societies. Insufficient sleep has increased the risk of catching neurodegenerative diseases such as Alzheimer’s. Several studies have indicated that restricted sleep increases the level of deposition of β-amyloid and formation of neurofibrillary tangles, the major brain microstructural hallmarks for Alzheimer disease. The mechanisms by which sleep deprivation affects the pathology of Alzheimer disease has not yet been fully and definitively identified. However, risk factors like apolipoprotein E risk alleles, kinases and phosphatases dysregulation, reactive oxygen species, endoplasmic reticulum damages, glymphatic system dysfunctions and orexinergic system inefficacy have been identified as the most important factors which mediates between the two conditions. In this review, these factors are briefly discussed.
Collapse
Affiliation(s)
- Nahid Ahmadian
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajjad Hejazi
- Department of Anatomy, Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahnaz Talebi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
266
|
O'Hara DM, Kalia SK, Kalia LV. Emerging disease-modifying strategies targeting α-synuclein for the treatment of Parkinson's disease. Br J Pharmacol 2018; 175:3080-3089. [PMID: 29722028 PMCID: PMC6031880 DOI: 10.1111/bph.14345] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/30/2018] [Accepted: 04/06/2018] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease is the most common neurodegenerative movement disorder. It arises as a result of neuronal cell death in specific brain regions, notably the substantia nigra pars compacta, and is characterized by the accumulation of α-synuclein in these brain regions. Current pharmacological therapies alleviate the motor symptoms of the disease and are particularly effective in the early stages of the disease. Ongoing drug development efforts focus on disease-modifying strategies that aim to halt or slow disease progression. In this review, we explore a number of emerging disease-modifying strategies with a focus on direct and indirect targeting of α-synuclein dysfunction. We summarize newer classes of small molecules and biological agents intended to attenuate protein aggregation or to target enzymes that may increase the degradation of the pathogenic forms of α-synuclein. Finally, we discuss emerging strategies that are demonstrating the potential for disease modification at the preclinical stage.
Collapse
Affiliation(s)
- Darren M O'Hara
- Krembil Research Institute, Toronto Western HospitalUniversity Health NetworkTorontoCanada
| | - Suneil K Kalia
- Krembil Research Institute, Toronto Western HospitalUniversity Health NetworkTorontoCanada
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoCanada
| | - Lorraine V Kalia
- Krembil Research Institute, Toronto Western HospitalUniversity Health NetworkTorontoCanada
- Division of Neurology, Department of MedicineUniversity of TorontoTorontoCanada
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Division of Neurology, Department of Medicine, Toronto Western HospitalUniversity Health NetworkTorontoCanada
- Tanz Centre for Research in Neurodegenerative DiseasesUniversity of TorontoTorontoCanada
| |
Collapse
|
267
|
Nieto CI, Cornago MP, Cabildo MP, Sanz D, Claramunt RM, Torralba MC, Torres MR, Martínez Casanova D, Sánchez-Alegre YR, Escudero E, Lavandera JL. Evaluation of the Antioxidant and Neuroprotectant Activities of New Asymmetrical 1,3-Diketones. Molecules 2018; 23:E1837. [PMID: 30042315 PMCID: PMC6222706 DOI: 10.3390/molecules23081837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 01/08/2023] Open
Abstract
A series of fourteen new asymmetrical 1,3-diketone derivatives have been synthesized and evaluated in the ABTS, FRAP and DPPH assays as a new chemotype with antioxidant and drug-like properties. All the compounds displayed low cytotoxicity in comparison to curcumin against the human neuroblastoma SH-SY5Y cell line. Among them, (3Z,5E)-6-(2,5-difluoro-4-hydroxy-phenyl)-1,1,1-trifluoro-4-hydroxyhexa-3,5-dien-2-one (6b) and (3Z,5E)-6-(2,3-difluoro-4-hydroxy-phenyl)-1,1,1-trifluoro-4-hydroxyhexa-3,5-dien-2-one (7b) with excellent solubility and chemical stability in biorelevant media, have also shown a similar Fe+2 chelation behavior to that of curcumin. Additionally, both derivatives 6b and 7b have afforded good neuroprotection activity against H₂O₂ induced oxidative stress in the same neuronal cell line, with a significant reduction of intracellular ROS levels, in parallel with a good recovery of the Mitochondrial Membrane Potential (ΔΨm). Compounds 6b and 7b with a promising antioxidant and drug-like profile, with low cytotoxic and good neuroprotectant activity, constitute a new interesting chemical class with high potential as new therapeutic agents against neurodegenerative diseases.
Collapse
Affiliation(s)
- Carla I Nieto
- Departamento de Química Orgánica y Bio-Orgánica, Facultad de Ciencias, Universidad Nacional de Educación a distancia (UNED), Paseo Senda del Rey 9, E-28040 Madrid, Spain.
| | - María Pilar Cornago
- Departamento de Química Orgánica y Bio-Orgánica, Facultad de Ciencias, Universidad Nacional de Educación a distancia (UNED), Paseo Senda del Rey 9, E-28040 Madrid, Spain.
| | - María Pilar Cabildo
- Departamento de Química Orgánica y Bio-Orgánica, Facultad de Ciencias, Universidad Nacional de Educación a distancia (UNED), Paseo Senda del Rey 9, E-28040 Madrid, Spain.
| | - Dionisia Sanz
- Departamento de Química Orgánica y Bio-Orgánica, Facultad de Ciencias, Universidad Nacional de Educación a distancia (UNED), Paseo Senda del Rey 9, E-28040 Madrid, Spain.
| | - Rosa M Claramunt
- Departamento de Química Orgánica y Bio-Orgánica, Facultad de Ciencias, Universidad Nacional de Educación a distancia (UNED), Paseo Senda del Rey 9, E-28040 Madrid, Spain.
| | - María Carmen Torralba
- Departamento de Química Inorgánica I and CAI de Difracción de Rayos-X, Facultad de Ciencias Químicas, Universidad Complutense de Madrid (UCM), E-28040 Madrid, Spain.
| | - María Rosario Torres
- Departamento de Química Inorgánica I and CAI de Difracción de Rayos-X, Facultad de Ciencias Químicas, Universidad Complutense de Madrid (UCM), E-28040 Madrid, Spain.
| | - Diana Martínez Casanova
- Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad CEU San Pablo, Campus de Montepríncipe, Boadilla, E-28668 Madrid, Spain.
| | - Yaiza Rebeca Sánchez-Alegre
- Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad CEU San Pablo, Campus de Montepríncipe, Boadilla, E-28668 Madrid, Spain.
| | - Esther Escudero
- Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad CEU San Pablo, Campus de Montepríncipe, Boadilla, E-28668 Madrid, Spain.
| | - José Luis Lavandera
- Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad CEU San Pablo, Campus de Montepríncipe, Boadilla, E-28668 Madrid, Spain.
| |
Collapse
|
268
|
Branca C, Ferreira E, Nguyen TV, Doyle K, Caccamo A, Oddo S. Genetic reduction of Nrf2 exacerbates cognitive deficits in a mouse model of Alzheimer's disease. Hum Mol Genet 2018; 26:4823-4835. [PMID: 29036636 DOI: 10.1093/hmg/ddx361] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 09/07/2017] [Indexed: 12/20/2022] Open
Abstract
Aging is the major risk factor for several neurodegenerative diseases, including Alzheimer's disease (AD). However, the mechanisms by which aging contributes to neurodegeneration remain elusive. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a transcription factor that regulates expression of a vast number of genes by binding to the antioxidant response element. Nrf2 levels decrease as a function of age, and reduced Nrf2 levels have been reported in postmortem human brains and animal models of AD. Nevertheless, it is still unknown whether Nrf2 plays a role in the cognitive deficits associated with AD. To address this question, we used a genetic approach to remove the Nrf2 gene from APP/PS1 mice, a widely used animal model of AD. We found that the lack of Nrf2 significantly exacerbates cognitive deficits in APP/PS1, without altering gross motor function. Specifically, we found an exacerbation of deficits in spatial learning and memory, as well as in working and associative memory. Different brain regions control these behavioral tests, indicating that the lack of Nrf2 has a global effect on brain function. The changes in cognition were linked to an increase in Aβ and interferon-gamma (IFNγ) levels, and microgliosis. The changes in IFNγ levels are noteworthy as previously published evidence indicates that IFNγ can increase microglia activation and induce Aβ production. Our data suggest a clear link between Nrf2 and AD-mediated cognitive decline and further strengthen the connection between Nrf2 and AD.
Collapse
Affiliation(s)
- Caterina Branca
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Eric Ferreira
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Thuy-Vi Nguyen
- Department of Immunobiology and Department of Neurology, University of Arizona, Tucson, AZ 85719, USA
| | - Kristian Doyle
- Department of Immunobiology and Department of Neurology, University of Arizona, Tucson, AZ 85719, USA.,The Arizona Center on Aging, University of Arizona, Tucson, AZ 85719, USA
| | - Antonella Caccamo
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Salvatore Oddo
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA.,School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
269
|
Yang L, Han W, Luo Y, Hu X, Xu Y, Li H, Hu C, Huang D, Ma J, Yang Y, Chen Q, Li Y, Zhang J, Xia H, Chen Z, Wang H, Ran D, Yang J. Adapentpronitrile, a New Dipeptidyl Peptidase-IV Inhibitor, Ameliorates Diabetic Neuronal Injury Through Inhibiting Mitochondria-Related Oxidative Stress and Apoptosis. Front Cell Neurosci 2018; 12:214. [PMID: 30072873 PMCID: PMC6058014 DOI: 10.3389/fncel.2018.00214] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 06/28/2018] [Indexed: 12/19/2022] Open
Abstract
Our previous studies indicated that adapentpronitrile, a new adamantane-based dipeptidyl peptidase-IV (DPP-IV) inhibitor, has a hypoglycemic effect and ameliorates rat pancreatic β cell dysfunction in type 2 diabetes mellitus through inhibiting DPP-IV activity. However, the effect of adapentpronitrile on the neurodegenerative diseases has not been studied. In the present study, we first found that adapentpronitrile significantly ameliorated neuronal injury and decreased amyloid precursor protein (APP) and amyloid beta (Aβ) expression in the hippocampus and cortex in the high fat diet/STZ rat model of diabetes. Furthermore, adapentpronitrile significantly attenuated oxidative stress, downregulated expression of the pro-apoptotic proteins BAX, cytochrome c, caspase-9, and caspase-3, and upregulated expression of the anti-apoptotic protein Bcl-2, although there was no effect on GLP-1R expression. At 30 min post-injection of adapentpronitrile (50 mg/kg) via the tail vein, its concentration in normal rat brain was 0.2034 ± 0.0094 μg/g. Subsequently, we further confirmed the neuroprotective effects and mechanism of adapentpronitrile in HT22 cells treated with high glucose (HG) and aluminum maltolate [Al(mal)3] overload, respectively. Our results showed significant decreases in mitochondrial membrane potential (MTP) and Bcl-2 expression, accompanied by a significant increase in apoptosis, reactive oxygen species (ROS) generation, and the expression of pro-apoptotic proteins in HT22 cells exposed to these stimuli. Adapentpronitrile treatment protected against neuronal injury, suppressed ROS generation, and reduced MTP and mitochondrial apoptosis in HT22 cells; however, DPP-IV activity was not detected. Our results suggest that adapentpronitrile protects against diabetic neuronal injury, at least partially, by inhibiting mitochondrial oxidative stress and the apoptotic pathway in a DPP-IV-independent manner.
Collapse
Affiliation(s)
- Lu Yang
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Wenli Han
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Ying Luo
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xiangnan Hu
- Department of Pharmacology, The Laboratory of Pharmaceutical Chemistry, Chongqing Medical University, Chongqing, China
| | - Ying Xu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York (SUNY), Buffalo, NY, United States
| | - Huan Li
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Congli Hu
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Dan Huang
- Department of Pharmacology, The Laboratory of Pharmaceutical Analysis, Chongqing Medical University, Chongqing, China
| | - Jie Ma
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yang Yang
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Qi Chen
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yuke Li
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Jiahua Zhang
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Hui Xia
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Zhihao Chen
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Hong Wang
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Dongzhi Ran
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Junqing Yang
- Department of Pharmacology, The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
270
|
Abstract
Exfoliation syndrome (XFS) produces deleterious ocular aging and has protean systemic manifestations. Local ocular production of TGFβ1 is of central importance in XFS. TGFβ1 appears to induce the expression of LOXL1 and the production of other extracellular matrix components which are known to be present in exfoliation material. Furthermore, results from several studies find that the aqueous humor of exfoliation glaucoma patients exhibits a decreased antioxidant defense and increased oxidative stress systems. Finally, studies show that the levels of interleukin-6 and interleukin-8 in the aqueous humor of XFS patients were 3-fold higher than in controls. Overall TGFβ1, as well as a prooxidative and proinflammatory environment seems to play an important role in XFS.
Collapse
Affiliation(s)
- Teresa Borrás
- Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, NC
| |
Collapse
|
271
|
Rutin as a Potent Antioxidant: Implications for Neurodegenerative Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6241017. [PMID: 30050657 PMCID: PMC6040293 DOI: 10.1155/2018/6241017] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 04/29/2018] [Indexed: 12/16/2022]
Abstract
A wide range of neurodegenerative diseases (NDs), including Alzheimer's disease, Parkinson's disease, Huntington's disease, and prion diseases, share common mechanisms such as neuronal loss, apoptosis, mitochondrial dysfunction, oxidative stress, and inflammation. Intervention strategies using plant-derived bioactive compounds have been offered as a form of treatment for these debilitating conditions, as there are currently no remedies to prevent, reverse, or halt the progression of neuronal loss. Rutin, a glycoside of the flavonoid quercetin, is found in many plants and fruits, especially buckwheat, apricots, cherries, grapes, grapefruit, plums, and oranges. Pharmacological studies have reported the beneficial effects of rutin in many disease conditions, and its therapeutic potential in several models of NDs has created considerable excitement. Here, we have summarized the current knowledge on the neuroprotective mechanisms of rutin in various experimental models of NDs. The mechanisms of action reviewed in this article include reduction of proinflammatory cytokines, improved antioxidant enzyme activities, activation of the mitogen-activated protein kinase cascade, downregulation of mRNA expression of PD-linked and proapoptotic genes, upregulation of the ion transport and antiapoptotic genes, and restoration of the activities of mitochondrial complex enzymes. Taken together, these findings suggest that rutin may be a promising neuroprotective compound for the treatment of NDs.
Collapse
|
272
|
Insights into Structure-Activity Relationships of 3-Arylhydrazonoindolin-2-One Derivatives for Their Multitarget Activity on β-Amyloid Aggregation and Neurotoxicity. Molecules 2018; 23:molecules23071544. [PMID: 29949947 PMCID: PMC6100259 DOI: 10.3390/molecules23071544] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/18/2018] [Accepted: 06/24/2018] [Indexed: 12/31/2022] Open
Abstract
Despite the controversial outcomes of clinical trials executed so far, the prevention of β-amyloid (Aβ) deposition and neurotoxicity by small molecule inhibitors of Aβ aggregation remains a target intensively pursued in the search of effective drugs for treating Alzheimer’s disease (AD) and related neurodegeneration syndromes. As a continuation of previous studies, a series of new 3-(2-arylhydrazono)indolin-2-one derivatives was synthesized and assayed, investigating the effects of substitutions on both the indole core and arylhydrazone moiety. Compared with the reference compound 1, we disclosed equipotent derivatives bearing alkyl substituents at the indole nitrogen, and fairly tolerated bioisosteric replacements at the arylhydrazone moiety. For most of the investigated compounds, the inhibition of Aβ40 aggregation (expressed as pIC50) was found to be correlated with lipophilicity, as assessed by a reversed-phase HPLC method, through a bilinear relationship. The N1-cyclopropyl derivative 28 was tested in cell-based assays of Aβ42 oligomer toxicity and oxidative stress induced by hydrogen peroxide, showing significant cytoprotective effects. This study confirmed the versatility of isatin in preparing multitarget small molecules affecting different biochemical pathways involved in AD.
Collapse
|
273
|
Organoruthenium(II) Complexes Ameliorates Oxidative Stress and Impedes the Age Associated Deterioration in Caenorhabditis elegans through JNK-1/DAF-16 Signalling. Sci Rep 2018; 8:7688. [PMID: 29769649 PMCID: PMC5955923 DOI: 10.1038/s41598-018-25984-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 05/02/2018] [Indexed: 12/21/2022] Open
Abstract
New ruthenium(II) complexes were synthesised and characterized by various spectro analytical techniques. The structure of the complexes 3 and 4 has been confirmed by X-ray crystallography. The complexes were subjected to study their anti-oxidant profile and were exhibited significantly greater in vitro DPPH radical scavenging activity than vitamin C. We found that complexes 1–4 confered tolerance to oxidative stress and extend the mean lifespan of mev-1 mutant worms and wild-type Caenorhabditis elegans. Further, mechanistic study and reporter gene expression analysis revealed that Ru(ƞ6-p-cymene) complexes maintained the intracellular redox status and offers stress resistance through activating JNK-1/DAF-16 signaling axis and possibly by other antioxidant response pathway. Notably, complex 3 and 4 ameliorates the polyQ (a Huntington’s disease associated protein) mediated proteotoxicity and related behavioural deficits in Huntington’s disease models of C. elegans. From these observations, we hope that new Ru(ƞ6-p-cymene) complexes could be further considered as a potential drug to retard aging and age-related neurodegenerative diseases.
Collapse
|
274
|
Kabel AM, Omar MS, Alhadhrami A, Alharthi SS, Alrobaian MM. Linagliptin potentiates the effect of l-dopa on the behavioural, biochemical and immunohistochemical changes in experimentally-induced Parkinsonism: Role of toll-like receptor 4, TGF-β1, NF-κB and glucagon-like peptide 1. Physiol Behav 2018; 188:108-118. [DOI: 10.1016/j.physbeh.2018.01.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/12/2018] [Accepted: 01/29/2018] [Indexed: 12/25/2022]
|
275
|
Xu SF, Ji LL, Wu Q, Li J, Liu J. Ontogeny and aging of Nrf2 pathway genes in livers of rats. Life Sci 2018; 203:99-104. [PMID: 29689272 DOI: 10.1016/j.lfs.2018.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/22/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
Abstract
The Nrf2/Keap1 antioxidant system plays important roles in protecting against oxidative stress and toxic stimuli, which may vary in infants, elderly, and females. AIM The constitutive expression of the Nrf2 genes during development and aging in both sexes would help our understanding of the Nrf2/Keap1 pathway in toxicological studies. MAIN METHODS Sprague Dawley rat livers were collected at 11 age points from prenatal (-2 d), neonatal (1, 7, 14 and 21 d), at puberty (28 and 35 d), at adulthood (60 and 180 d), to aging (540 and 800 d) from both sexes. Total RNA and proteins were extracted for real-time RT-PCR and Western-blot analysis. KEY FINDINGS The abundant mRNA expression was in the order of Nrf2, Gclm, Nqo1, Gclc, Ho-1, and Keap1. The expression of these genes except Gclc was high in fetal livers, decreased at birth, reached the first peak at 7 days of age, and gradually decreased to adult levels till 180 days of age. All these genes remained high at 540 days of age, but declined at 800 days of age, with more increases with Nqo1 and Ho-1. Females had lower fetal, neonatal, and aged levels than males. Protein expressions of Nrf2, Nqo1, Ho-1, GCLC and GCLM agree with mRNA analysis. SIGNIFICANCE This study characterized the age- and sex-related changes of Nrf2-related gene/proteins in livers of rats, and higher expressions in newborns and aged rats could cope with increased oxidative stress in infants and elderly.
Collapse
Affiliation(s)
- Shang-Fu Xu
- Key Lab for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China; The MOE Key Lab for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Li Ji
- The MOE Key Lab for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qin Wu
- Key Lab for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China
| | - Jin Li
- Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, China
| | - Jie Liu
- Key Lab for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
276
|
Cui H, Tao F, Hou Y, Lu Y, Zheng T, Sang S, Lv L. Dual effects of propyl gallate and its methylglyoxal adduct on carbonyl stress and oxidative stress. Food Chem 2018; 265:227-232. [PMID: 29884377 DOI: 10.1016/j.foodchem.2018.04.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 11/26/2022]
Abstract
In the present study, we investigated the trapping of methylglyoxal (MGO) by propyl gallate (PG), a known food grade antioxidant, and the anti-carbonyl and anti-oxidative properties of the mono-MGO adduct of PG (MM-PG). Our result indicated that more than 77.5% MGO was suppressed by PG after a 30 min incubation of PG with MGO, which was much more effective than gallic acid (15.2%). For the first time, MM-PG was purified, and its structure was elucidated based on the analysis of its 1H, 13C, and 2D-NMR data. We also demonstrated that MM-PG had strong anti-oxidative and anti-carbonyl activities. Furthermore, PG could trap the MGO generated during the preparation of roasted pork, and both mono- and di- MGO adducts of PG were detected in the roasted pork system using LC/MS technique. Thus, PG could be widely applied in the food system for inhibiting the formation of both carbonyl species and oxidative species.
Collapse
Affiliation(s)
- Hengqing Cui
- Department of Food Science and Technology, Nanjing Normal University, 122# Ninghai Road, Nanjing 210097, PR China
| | - Fei Tao
- College of Standardization, China Jiliang University, 258# Xueyuan Street, Hangzhou 310018, PR China
| | - Yu Hou
- Department of Food Science and Technology, Nanjing Normal University, 122# Ninghai Road, Nanjing 210097, PR China
| | - Yongling Lu
- Department of Food Science and Technology, Nanjing Normal University, 122# Ninghai Road, Nanjing 210097, PR China
| | - Tiesong Zheng
- Department of Food Science and Technology, Nanjing Normal University, 122# Ninghai Road, Nanjing 210097, PR China
| | - Shengmin Sang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, 500 Laureate Way, Kannapolis, NC 28081, United States.
| | - Lishuang Lv
- Department of Food Science and Technology, Nanjing Normal University, 122# Ninghai Road, Nanjing 210097, PR China.
| |
Collapse
|
277
|
Harris SA, Harris EA. Molecular Mechanisms for Herpes Simplex Virus Type 1 Pathogenesis in Alzheimer's Disease. Front Aging Neurosci 2018; 10:48. [PMID: 29559905 PMCID: PMC5845560 DOI: 10.3389/fnagi.2018.00048] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022] Open
Abstract
This review focuses on research in the areas of epidemiology, neuropathology, molecular biology and genetics that implicates herpes simplex virus type 1 (HSV-1) as a causative agent in the pathogenesis of sporadic Alzheimer’s disease (AD). Molecular mechanisms whereby HSV-1 induces AD-related pathophysiology and pathology, including neuronal production and accumulation of amyloid beta (Aβ), hyperphosphorylation of tau proteins, dysregulation of calcium homeostasis, and impaired autophagy, are discussed. HSV-1 causes additional AD pathologies through mechanisms that promote neuroinflammation, oxidative stress, mitochondrial damage, synaptic dysfunction, and neuronal apoptosis. The AD susceptibility genes apolipoprotein E (APOE), phosphatidylinositol binding clathrin assembly protein (PICALM), complement receptor 1 (CR1) and clusterin (CLU) are involved in the HSV lifecycle. Polymorphisms in these genes may affect brain susceptibility to HSV-1 infection. APOE, for example, influences susceptibility to certain viral infections, HSV-1 viral load in the brain, and the innate immune response. The AD susceptibility gene cholesterol 25-hydroxylase (CH25H) is upregulated in the AD brain and is involved in the antiviral immune response. HSV-1 interacts with additional genes to affect cognition-related pathways and key enzymes involved in Aβ production, Aβ clearance, and hyperphosphorylation of tau proteins. Aβ itself functions as an antimicrobial peptide (AMP) against various pathogens including HSV-1. Evidence is presented supporting the hypothesis that Aβ is produced as an AMP in response to HSV-1 and other brain infections, leading to Aβ deposition and plaque formation in AD. Epidemiologic studies associating HSV-1 infection with AD and cognitive impairment are discussed. Studies are reviewed supporting subclinical chronic reactivation of latent HSV-1 in the brain as significant in the pathogenesis of AD. Finally, the rationale for and importance of clinical trials treating HSV-1-infected MCI and AD patients with antiviral medication is discussed.
Collapse
Affiliation(s)
- Steven A Harris
- St. Vincent Medical Group, Northside Internal Medicine, Indianapolis, IN, United States
| | - Elizabeth A Harris
- Department of Neurology, University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
278
|
Selvasundaram R, Manoharan S, Buddhan R, Neelakandan M, Murali Naidu R. Chemopreventive potential of esculetin in 7,12-dimethylbenz(a)anthracene-induced hamster buccal pouch carcinogenesis. Mol Cell Biochem 2018; 448:145-153. [PMID: 29435870 DOI: 10.1007/s11010-018-3321-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 02/07/2018] [Indexed: 12/28/2022]
Abstract
7,12-Dimethylbenz(a)anthracene (DMBA)-induced hamster buccal pouch carcinogenesis is widely preferred to assess the tumor-inhibiting efficacy of the medicinal plants or their constituents. The present study explores the tumor-inhibiting potential of esculetin by utilizing the status of lipid peroxidation by products (thiobarbituric acid reactive substances), antioxidants (vitamin E, reduced glutathione, superoxide dismutase, catalase, and glutathione peroxidase), and phase I and phase II detoxification agents as biochemical end points and by using histopathological studies as well in DMBA-induced hamster buccal pouch carcinogenesis. Oral tumors developed in the buccal pouch were subjected to histopathological studies, and were confirmed as oral squamous cell carcinoma. Hamsters treated with DMBA alone showed an abnormal pattern of lipid peroxidation, antioxidants, and detoxification agents as compared to control hamsters. The status of the above-mentioned biochemical markers and histopathological abnormalities were found to be reversed in DMBA + esculetin-treated hamsters. The result of the present study thus indicates the tumor preventive potential of esculetin in DMBA-induced oral carcinogenesis.
Collapse
Affiliation(s)
- Renganathan Selvasundaram
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, 608002, India
| | - Shanmugam Manoharan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, 608002, India.
| | - Rajamanickam Buddhan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, 608002, India
| | - Mani Neelakandan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, 608002, India
| | - Radhakrishnan Murali Naidu
- Department of Oral & Maxillofacial Pathology, Rajah Muthiah Dental College & Hospital, Annamalai University, Annamalainagar, Tamil Nadu, India
| |
Collapse
|
279
|
Sørvik IB, Solum EJ, Labba NA, Hansen TV, Paulsen RE. Differential effects of some novel synthetic oestrogen analogs on oxidative PC12 cell death caused by serum deprivation. Free Radic Res 2018; 52:273-287. [DOI: 10.1080/10715762.2018.1430363] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Irene B. Sørvik
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Eirik Johansson Solum
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Nils A. Labba
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Trond Vidar Hansen
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Ragnhild E. Paulsen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
280
|
Martinez JH, Alaimo A, Gorojod RM, Porte Alcon S, Fuentes F, Coluccio Leskow F, Kotler ML. Drp-1 dependent mitochondrial fragmentation and protective autophagy in dopaminergic SH-SY5Y cells overexpressing alpha-synuclein. Mol Cell Neurosci 2018; 88:107-117. [PMID: 29414102 DOI: 10.1016/j.mcn.2018.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/10/2018] [Accepted: 01/15/2018] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease is a neurodegenerative movement disorder caused by the loss of dopaminergic neurons from substantia nigra. It is characterized by the accumulation of aggregated α-synuclein as the major component of the Lewy bodies. Additional common features of this disease are the mitochondrial dysfunction and the activation/inhibition of autophagy both events associated to the intracellular accumulation of α-synuclein. The mechanism by which these events contribute to neural degeneration remains unknown. In the present work we investigated the effect of α-synuclein on mitochondrial dynamics and autophagy/mitophagy in SH-SY5Y cells, an in vitro model of Parkinson disease. We demonstrated that overexpression of wild type α-synuclein causes moderated toxicity, ROS generation and mitochondrial dysfunction. In addition, α-synuclein induces the mitochondrial fragmentation on a Drp-1-dependent fashion. Overexpression of the fusion protein Opa-1 prevented both mitochondrial fragmentation and cytotoxicity. On the other hand, cells expressing α-synuclein showed activated autophagy and particularly mitophagy. Employing a genetic strategy we demonstrated that autophagy is triggered in order to protect cells from α-synuclein-induced cell death. Our results clarify the role of Opa-1 and Drp-1 in mitochondrial dynamics and cell survival, a controversial α-synuclein research issue. The findings presented point to the relevance of mitochondrial homeostasis and autophagy in the pathogenesis of PD. Better understanding of the molecular interaction between these processes could give rise to novel therapeutic methods for PD prevention and amelioration.
Collapse
Affiliation(s)
- Jimena Hebe Martinez
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio Interdisciplinario de Dinámica Celular y Nanoherramientas, Argentina.
| | - Agustina Alaimo
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| | - Roxana Mayra Gorojod
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| | - Soledad Porte Alcon
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| | - Federico Fuentes
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Federico Coluccio Leskow
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio Interdisciplinario de Dinámica Celular y Nanoherramientas, Argentina.
| | - Mónica Lidia Kotler
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| |
Collapse
|
281
|
Kuciel-Lewandowska J, Gnus J, Pawlik-Sobecka L, Płaczkowska S, Kokot I, Kasperczak M, Paprocka-Borowicz M. The Assessment of the Integrated Antioxidant System of the Body in the Course of Radon Therapy: A Pilot Study. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6038106. [PMID: 29487870 PMCID: PMC5816895 DOI: 10.1155/2018/6038106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/08/2017] [Accepted: 03/14/2017] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The sources of Reactive Oxidative Species (ROS) in the organism are the respiratory processes occurring in cells catalyzed by different enzymes. Operation of ROS is balanced by antioxidants, the compounds; although present in low concentrations, they significantly inhibit the degree of oxidation of particular molecules. THE AIM OF THE STUDY The aim of this study was to assess the changes in the integrated antioxidant system under the influence of radon therapy in osteoarthritis patients. MATERIAL AND METHODS Observation included 35 patients suffering from degenerative joints and disc disease (mean age 56.5 years) undergoing radon water therapy and control group that consisted of 15 osteoarthritis patients (mean age 54.2) without contact with radon water. Before therapy and after 18 days of treatment, serum total antioxidant status (TAS) was assessed with the use of standard colorimetric assay. RESULTS In the study group, we observed trends to increase TAS concentration, whereas, in the control group, TAS concentration was decreasing. CONCLUSIONS (1) Radon waters treatment influenced the level of TAS of osteoarthritis patients treated with the radon water. (2) The change in TAS concentrations in the study group may be the result of low doses of ionizing radiation, but further studies on larger patient's groups are demanded. This study is registered with number NCT03274128.
Collapse
Affiliation(s)
| | - Jan Gnus
- Department of Physiotherapy, Medical University of Wroclaw, Wroclaw, Poland
| | - Lilla Pawlik-Sobecka
- Department of Professional Training in Clinical Chemistry, Medical University of Wroclaw, Wroclaw, Poland
| | - Sylwia Płaczkowska
- Department of Diagnostics Laboratory for Teaching and Research, Medical University of Wroclaw, Wroclaw, Poland
| | - Izabela Kokot
- Department of Professional Training in Clinical Chemistry, Medical University of Wroclaw, Wroclaw, Poland
| | - Michał Kasperczak
- Department of Physiotherapy, Medical University of Wroclaw, Wroclaw, Poland
| | | |
Collapse
|
282
|
Curry DW, Stutz B, Andrews ZB, Elsworth JD. Targeting AMPK Signaling as a Neuroprotective Strategy in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2018; 8:161-181. [PMID: 29614701 PMCID: PMC6004921 DOI: 10.3233/jpd-171296] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. It is characterized by the accumulation of intracellular α-synuclein aggregates and the degeneration of nigrostriatal dopaminergic neurons. While no treatment strategy has been proven to slow or halt the progression of the disease, there is mounting evidence from preclinical PD models that activation of 5'-AMP-activated protein kinase (AMPK) may have broad neuroprotective effects. Numerous dietary supplements and pharmaceuticals (e.g., metformin) that increase AMPK activity are available for use in humans, but clinical studies of their effects in PD patients are limited. AMPK is an evolutionarily conserved serine/threonine kinase that is activated by falling energy levels and functions to restore cellular energy balance. However, in response to certain cellular stressors, AMPK activation may exacerbate neuronal atrophy and cell death. This review describes the regulation and functions of AMPK, evaluates the controversies in the field, and assesses the potential of targeting AMPK signaling as a neuroprotective treatment for PD.
Collapse
Affiliation(s)
- Daniel W Curry
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Bernardo Stutz
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zane B Andrews
- Department of Physiology, Monash University, Melbourne, VIC, Australia
- Monash Biomedicine Discovery Institute, Monash University, VIC, Australia
| | - John D Elsworth
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
283
|
Liu R, Li X. Radix Ophiopogonis polysaccharide extracts alleviate MPP +-induced PC-12 cell injury through inhibition of Notch signaling pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:99-109. [PMID: 31938091 PMCID: PMC6957974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/28/2017] [Indexed: 06/10/2023]
Abstract
Background: Parkinson's disease (PD) is a degenerative disease of central nervous system. 1-Methyl-4-phenylpyridine (MPP+) is a non-selective dopaminergic neurotoxin that induces cell injury similar to PD. This study aimed to explore the protective effects of Radix Ophiopogonis polysaccharide extracts (ROP) on the MPP+-induced PC-12 cell injury. Methods: PC-12 cells were exposed to MPP+ with or without ROP treatment. Then the cell viability, apoptosis, reactive oxygen species (ROS) level, calcium (Ca2+) concentration, mitochondrial membrane potential (MMP), Cyctochrome C release, mitochondrial ATP synthesis, and the expression level of Notch signaling pathway were detected by CellTiter 96 AQueous One Solution Cell Proliferation assay, fluorescent staining, flow cytometer analysis, and western blotting, respectively. Results: MPP+ treatment obviously induced PC-12 cell injury as evidenced by the cell viability loss and cell apoptosis enhancement. MPP+ markedly increased the concentrations of ROS and Ca2+ and the mitochondrial dysfunction in PC-12 cells. Moreover, the activation of Notch signaling pathway was found after MPP+ treatment. ROP significantly reversed the MPP+-induced PC-12 cell viability loss, apoptosis increase, intracellular oxidative stress and endoplasmic reticulum (ER) stress rise, mitochondrial dysfunction and the activation of Notch signaling pathways in PC-12 cells. Conclusion: ROP protected PC-12 cells from the MPP+-induced injury through suppressing the increase of the intracellular oxidative stress and ER stress and activation of Notch signaling pathway. These findings will be helpful for understanding the protective roles of ROP in nerve cell injury and provide potential therapeutic drug for PD.
Collapse
|
284
|
Wu N, Wan Y, Song L, Qi C, Liu Z, Gan J. The abnormal activation of D1R/Shp-2 complex involved in levodopa-induced dyskinesia in 6-hydroxydopamine-lesioned Parkinson's rats. Neuropsychiatr Dis Treat 2018; 14:1779-1786. [PMID: 30013350 PMCID: PMC6038854 DOI: 10.2147/ndt.s162562] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Levodopa-induced dyskinesia (LID) is a troublesome problem in the treatment of Parkinson's disease (PD). The mechanisms of LID are still mysterious. Recently, the interaction between Shp-2 and D1 dopamine receptor (D1R) has been identified to be indispensable in the D1R-mediated extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation and the occurrence of LID. However, the role of Shp-2 in the D1R-mediated signaling pathway of dyskinetic rat models is not fully clear. We designed this study with the purpose of exploring the role of D1R/Shp-2 complex in the D1R-mediated signaling pathway in the occurrence of LID. MATERIALS AND METHODS The 6-hydroxydopamine (6-OHDA) was injected unilaterally to produce the rat models of PD. Successful PD rat models were randomly divided into three groups to receive the treatment with L-3,4-dihydroxyphenylalanine (l-DOPA) + benserazide, l-DOPA + benserazide + D1R antagonist (SCH23390) or D1R agonist (SKF38393). Abnormal involuntary movements were assessed in different groups during the treatment. The interaction between D1R and Shp-2 was confirmed in the sham and LID rats through the methods of coimmunoprecipitation. In addition, the levels of p-Shp-2, p-ERK1/2 and p-mTOR were determined by Western blot in different groups. RESULTS After the treatment with l-DOPA + benserazide for 22 days, PD rats presented with dyskinesia. D1R agonist, SKF38393, induced similar involuntary movements in PD rats. In contrast, the dyskinetic movements were not induced by coadministration of l-DOPA + D1R antagonist (SCH23390). The interaction between D1R and Shp-2 in the normal rats was kept stable after the long-term use of l-DOPA. Moreover, we found that the pulsatile levodopa administration induced hyperphosphorylation of Shp-2, ERK1/2 and mTOR, while the coadministration of l-DOPA and D1R antagonist, SCH23390, did not induce the hyperphosphorylation of these proteins. CONCLUSION These data verified the existence of D1R/Shp-2 complex and its crucial role in the D1R-mediated signaling pathway in dyskinetic rats. Focus on the D1R/Shp-2 complex might be a potential treatment of LID in the future.
Collapse
Affiliation(s)
- Na Wu
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China, ;
| | - Ying Wan
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China, ;
| | - Lu Song
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China, ;
| | - Chen Qi
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China, ;
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China, ;
| | - Jing Gan
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China, ;
| |
Collapse
|
285
|
Li B, Evivie SE, Lu J, Jiao Y, Wang C, Li Z, Liu F, Huo G. Lactobacillus helveticus KLDS1.8701 alleviates d-galactose-induced aging by regulating Nrf-2 and gut microbiota in mice. Food Funct 2018; 9:6586-6598. [DOI: 10.1039/c8fo01768a] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We first revealed a close association between probiotic-manipulated gut microbiota and hepatic Nrf-2 dependent mechanisms to suppress d-galactose-induced aging.
Collapse
Affiliation(s)
- Bailiang Li
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Smith Etareri Evivie
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Jingjing Lu
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Yuehua Jiao
- Center of Drug Safety Evaluation
- Heilongjiang University of Chinese Medicine
- Harbin 150040
- China
| | - Chengfeng Wang
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Ziye Li
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Fei Liu
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Guicheng Huo
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| |
Collapse
|
286
|
Shi L, Huang C, Luo Q, Xia Y, Liu H, Li L, Liu W, Ma W, Fang J, Tang L, Zeng W, Chen Z. Pilot study: molecular risk factors for diagnosing sporadic Parkinson's disease based on gene expression in blood in MPTP-induced rhesus monkeys. Oncotarget 2017; 8:105606-105614. [PMID: 29285276 PMCID: PMC5739663 DOI: 10.18632/oncotarget.22348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/17/2017] [Indexed: 11/25/2022] Open
Abstract
Clinical diagnosis of Parkinson's disease (PD) is characterized by the classical features of tremor, bradykinesia and rigidity, which are present only when more than 70%-80% degeneration of dopaminergic (DA) neurons in the substantia nigra. The lack of means for early diagnosis of PD has elicited interest in searching for its risk factors, which, by now, are almost obtained at a single time point in PD process, and little developing risk factors, obtained from completely normal situation to the onset or even advanced stage of PD in individual person which could monitor the progress of PD, are present. Here we have detected some potential factors in the blood of MPTP induced PD monkeys along with the progress of the disease. All the PD monkeys showed mild PD symptoms since the 9th week and gradually reached a classic and stable parkinsonism stage at the 18th week. Our results have found that the expression of Parkin, USP30, MUL1, PINK1, and LRRK2 significantly increased at 1st, 3th, 3th, 5th, and 8th week respectively and remained high till the end; The expression of UCHL1 and TRIM24 significantly increased at the 1st and 18th week, respectively, then gradually decreased and significantly lower than normal value; DJ-1 showed significantly decreased since the 12th week, while SNCA showed no significantly changed excepted at the 5th week. And, the terminal results of whole blood were highly consistent with those of in SN. These results support that these genes change may as biomarkers to monitor the progress of PD, and may facilitate the development of biomarkers for early diagnosis.
Collapse
Affiliation(s)
- Liangqin Shi
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Chao Huang
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Qihui Luo
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Ya'an, Sichuan 625014, China
| | - Yu Xia
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Heng Liu
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Like Li
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Wentao Liu
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Wenjing Ma
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jing Fang
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Li Tang
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Wen Zeng
- Sichuan Primed Biological Technology Co., Ltd, National Experimental Macaque Reproduce Laboratory, Ya'an, Sichuan 625014, China
| | - Zhengli Chen
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Ya'an, Sichuan 625014, China
| |
Collapse
|
287
|
Emborg ME. Nonhuman Primate Models of Neurodegenerative Disorders. ILAR J 2017; 58:190-201. [PMID: 28985333 PMCID: PMC5886328 DOI: 10.1093/ilar/ilx021] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 05/18/2017] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's (AD), Huntington's (HD), and Parkinson's (PD) disease are age-related neurodegenerative disorders characterized by progressive neuronal cell death. Although each disease has particular pathologies and symptoms, accumulated evidence points to similar mechanisms of neurodegeneration, including inflammation, oxidative stress, and protein aggregation. A significant body of research is ongoing to understand how these pathways affect each other and what ultimately triggers the onset of the disease. Experiments in nonhuman primates (NHPs) account for only 5% of all research in animals. Yet the impact of NHP studies for clinical translation is much greater, especially for neurodegenerative disorders, as NHPs have a complex cognitive and motor functions and highly developed neuroanatomy. New NHP models are emerging to better understand pathology and improve the platform in which to test novel therapies. The goal of this report is to review NHP models of AD, HD, and PD in the context of the current understanding of these diseases and their contribution to the development of novel therapies.
Collapse
Affiliation(s)
- Marina E Emborg
- Marina E. Emborg, MD, PhD, is the director of the Preclinical Parkinson’s Research Program at the Wisconsin National Primate Research Center and an associate professor in the department of Medical Physics at the University of Wisconsin in Madison, Wisconsin.
| |
Collapse
|
288
|
Manca A, Alladio E, Massarenti P, Puccinelli MP, De Francesco A, Del Grosso E, Mengozzi G, Pazzi M, Vincenti M. “One-pot” ethyl chloroformate derivatization and liquid-liquid extraction of reduced glutathione in erythrocyte and its quantitative GC–MS analysis. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1070:15-22. [DOI: 10.1016/j.jchromb.2017.10.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 10/13/2017] [Accepted: 10/14/2017] [Indexed: 11/17/2022]
|
289
|
Dietary supplementation with cysteine prevents adverse metabolic outcomes of repeated cures with paracetamol in old rats. Br J Nutr 2017; 118:889-896. [PMID: 29173208 DOI: 10.1017/s0007114517002847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cysteine (Cys), a conditionally indispensable amino acid, is required for the detoxification of paracetamol (acetaminophen, N-acetyl-para-aminophenol, 4-hydroxy-acetanilide, APAP), a drug of widespread use in older persons. We recently reported that repeated APAP cures could worsen sarcopenia in old rats, likely to be due to the impairment of Cys/GSH homoeostasis. The aim of the study was to evaluate whether a dietary Cys supplementation during APAP cures could improve Cys/GSH homoeostasis and thus preserve skeletal muscle. Male 21·5-month-old Wistar rats received three 2-week-long cures of APAP (1 % of diet) alone or with extra Cys (0·5 % of diet), intercalated with washout periods of 2 weeks (APAP and APAP-Cys groups, respectively). They were compared with untreated control rats (CT group). CT and APAP-Cys groups were pair-fed to the APAP group. Dietary Cys supplementation was efficient to prevent increase in liver mass (P<0·0001), decrease in liver GSH (P<0·0001), increase in blood GSH concentration (P<0·0001), and to some extent, decrease in plasma free Cys concentration (P<0·05), all induced by repeated APAP cures. The addition of Cys to APAP cures decreased plasma alanine transaminase (P<0·05), the fractional synthesis rate of liver proteins (P<0·01), and increased masses of extensor digitorum longus (P<0·01), and soleus (P<0·05), compared with the APAP group. Cys supplementation prevented alteration in Cys/GSH homoeostasis and increased some muscle masses in old rats under repeated cures with a non-toxic dose of APAP.
Collapse
|
290
|
Ragassi Fiorini AM, Barbalho SM, Guiguer ÉL, Oshiiwa M, Mendes CG, Vieites RL, Chies AB, de Oliveira PB, de Souza MDSS, Nicolau CCT. Dipteryx alata Vogel May Improve Lipid Profile and Atherogenic Indices in Wistar Rats Dipteryx alata and Atherogenic Indices. J Med Food 2017; 20:1121-1126. [DOI: 10.1089/jmf.2017.0052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
| | - Sandra Maria Barbalho
- Department of Biochemistry and Nutrition, Faculty of Food Technology of Marília, Marília, SP, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, SP, Brazil
| | - Élen Landgraf Guiguer
- Department of Biochemistry and Nutrition, Faculty of Food Technology of Marília, Marília, SP, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, SP, Brazil
| | - Marie Oshiiwa
- Department of Biochemistry and Nutrition, Faculty of Food Technology of Marília, Marília, SP, Brazil
| | - Claudemir Gregório Mendes
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, SP, Brazil
| | - Rogério Lopes Vieites
- Faculty of Agronomic Sciences, University Estadual Paulista–UNESP, Botucatu, SP, Brazil
| | - Agnaldo Bruno Chies
- Department of Pharmacology, Marilia Faculty of Medicine, FAMEMA, Marília, SP, Brazil
| | | | | | | |
Collapse
|
291
|
Angeloni C, Malaguti M, Barbalace MC, Hrelia S. Bioactivity of Olive Oil Phenols in Neuroprotection. Int J Mol Sci 2017; 18:ijms18112230. [PMID: 29068387 PMCID: PMC5713200 DOI: 10.3390/ijms18112230] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 12/12/2022] Open
Abstract
Neurological disorders such as stroke, Alzheimer’s and Parkinson’s diseases are associated with high morbidity and mortality, and few or no effective options are available for their treatment. These disorders share common pathological characteristics like the induction of oxidative stress, abnormal protein aggregation, perturbed Ca2+ homeostasis, excitotoxicity, inflammation and apoptosis. A large body of evidence supports the beneficial effects of the Mediterranean diet in preventing neurodegeneration. As the Mediterranean diet is characterized by a high consumption of extra-virgin olive oil it has been hypothesized that olive oil, and in particular its phenols, could be responsible for the beneficial effect of the Mediterranean diet. This review provides an updated vision of the beneficial properties of olive oil and olive oil phenols in preventing/counteracting both acute and chronic neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy.
| | - Maria Cristina Barbalace
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy.
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
292
|
Consales C, Cirotti C, Filomeni G, Panatta M, Butera A, Merla C, Lopresto V, Pinto R, Marino C, Benassi B. Fifty-Hertz Magnetic Field Affects the Epigenetic Modulation of the miR-34b/c in Neuronal Cells. Mol Neurobiol 2017; 55:5698-5714. [PMID: 29039021 DOI: 10.1007/s12035-017-0791-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/26/2017] [Indexed: 12/24/2022]
Abstract
The exposure to extremely low-frequency magnetic fields (ELF-MFs) has been associated to increased risk of neurodegenerative diseases, although the underlying molecular mechanisms are still undefined. Since epigenetic modulation has been recently encountered among the key events leading to neuronal degeneration, we here aimed at assessing if the control of gene expression mediated by miRNAs, namely miRs-34, has any roles in driving neuronal cell response to 50-Hz (1 mT) magnetic field in vitro. We demonstrate that ELF-MFs drive an early reduction of the expression level of miR-34b and miR-34c in SH-SY5Y human neuroblastoma cells, as well as in mouse primary cortical neurons, by affecting the transcription of the common pri-miR-34. This modulation is not p53 dependent, but attributable to the hyper-methylation of the CpG island mapping within the miR-34b/c promoter. Incubation with N-acetyl-l-cysteine or glutathione ethyl-ester fails to restore miR-34b/c expression, suggesting that miRs-34 are not responsive to ELF-MF-induced oxidative stress. By contrast, we show that miRs-34 control reactive oxygen species production and affect mitochondrial oxidative stress triggered by ELF-MFs, likely by modulating mitochondria-related miR-34 targets identified by in silico analysis. We finally demonstrate that ELF-MFs alter the expression of the α-synuclein, which is specifically stimulated upon ELF-MFs exposure via both direct miR-34 targeting and oxidative stress. Altogether, our data highlight the potential of the ELF-MFs to tune redox homeostasis and epigenetic control of gene expression in vitro and shed light on the possible mechanism(s) producing detrimental effects and predisposing neurons to degeneration.
Collapse
Affiliation(s)
- Claudia Consales
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.
| | - Claudia Cirotti
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy.,Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Martina Panatta
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Alessio Butera
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Caterina Merla
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.,Vectorology and Anticancer Therapies, UMR 8203, CNRS, Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Vanni Lopresto
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Rosanna Pinto
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Carmela Marino
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Barbara Benassi
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.
| |
Collapse
|
293
|
Cavallaro RA, Nicolia V, Fiorenza MT, Scarpa S, Fuso A. S-Adenosylmethionine and Superoxide Dismutase 1 Synergistically Counteract Alzheimer's Disease Features Progression in TgCRND8 Mice. Antioxidants (Basel) 2017; 6:antiox6040076. [PMID: 28973985 PMCID: PMC5745486 DOI: 10.3390/antiox6040076] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 01/30/2023] Open
Abstract
Recent evidence emphasizes the role of dysregulated one-carbon metabolism in Alzheimer’s Disease (AD). Exploiting a nutritional B-vitamin deficiency paradigm, we have previously shown that PSEN1 and BACE1 activity is modulated by one-carbon metabolism, leading to increased amyloid production. We have also demonstrated that S-adenosylmethionine (SAM) supplementation contrasted the AD-like features, induced by B-vitamin deficiency. In the present study, we expanded these observations by investigating the effects of SAM and SOD (Superoxide dismutase) association. TgCRND8 AD mice were fed either with a control or B-vitamin deficient diet, with or without oral supplementation of SAM + SOD. We measured oxidative stress by lipid peroxidation assay, PSEN1 and BACE1 expression by Real-Time Polymerase Chain Reaction (PCR), amyloid deposition by ELISA assays and immunohistochemistry. We found that SAM + SOD supplementation prevents the exacerbation of AD-like features induced by B vitamin deficiency, showing synergistic effects compared to either SAM or SOD alone. SAM + SOD supplementation also contrasts the amyloid deposition typically observed in TgCRND8 mice. Although the mechanisms underlying the beneficial effect of exogenous SOD remain to be elucidated, our findings identify that the combination of SAM + SOD could be carefully considered as co-adjuvant of current AD therapies.
Collapse
Affiliation(s)
- Rosaria A Cavallaro
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Vincenzina Nicolia
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Maria Teresa Fiorenza
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00183 Rome, Italy.
- IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64-65, 00143 Rome, Italy.
| | - Sigfrido Scarpa
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Andrea Fuso
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00183 Rome, Italy.
| |
Collapse
|
294
|
Martínez-Huélamo M, Rodríguez-Morató J, Boronat A, de la Torre R. Modulation of Nrf2 by Olive Oil and Wine Polyphenols and Neuroprotection. Antioxidants (Basel) 2017; 6:E73. [PMID: 28954417 PMCID: PMC5745483 DOI: 10.3390/antiox6040073] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 12/11/2022] Open
Abstract
Strong adherence to a Mediterranean diet is associated with improved cognitive function and a lower prevalence of mild cognitive impairment. Olive oil and red wine are rich sources of polyphenols which are responsible in part for the beneficial effects on cognitive functioning. Polyphenols induce endogenous antioxidant defense mechanisms by modulating transcription factors such as the nuclear factor (erythroid-derived 2)-like 2 (Nrf2). This review discusses the scientific data supporting the modulating effect of olive oil and red wine polyphenols on Nrf2 expression, and the potential health benefits associated with cognitive functioning.
Collapse
Affiliation(s)
- Miriam Martínez-Huélamo
- Integrated Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, IMIM-Institut Hospital del Mar d'Investigacions Mèdiques, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Jose Rodríguez-Morató
- Integrated Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, IMIM-Institut Hospital del Mar d'Investigacions Mèdiques, Dr. Aiguader 88, 08003 Barcelona, Spain.
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra (CEXS-UPF), Dr. Aiguader 80, 08003 Barcelona, Spain.
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN, CB06/03/028), 15706 Santiago de Compostela, Spain.
| | - Anna Boronat
- Integrated Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, IMIM-Institut Hospital del Mar d'Investigacions Mèdiques, Dr. Aiguader 88, 08003 Barcelona, Spain.
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra (CEXS-UPF), Dr. Aiguader 80, 08003 Barcelona, Spain.
| | - Rafael de la Torre
- Integrated Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, IMIM-Institut Hospital del Mar d'Investigacions Mèdiques, Dr. Aiguader 88, 08003 Barcelona, Spain.
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra (CEXS-UPF), Dr. Aiguader 80, 08003 Barcelona, Spain.
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN, CB06/03/028), 15706 Santiago de Compostela, Spain.
| |
Collapse
|
295
|
Metallothionein in Brain Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:5828056. [PMID: 29085556 PMCID: PMC5632493 DOI: 10.1155/2017/5828056] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/17/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022]
Abstract
Metallothioneins are a family of proteins which are able to bind metals intracellularly, so their main function is to regulate the cellular metabolism of essential metals. There are 4 major isoforms of MTs (I-IV), three of which have been localized in the central nervous system. MT-I and MT-II have been localized in the spinal cord and brain, mainly in astrocytes, whereas MT-III has been found mainly in neurons. MT-I and MT-II have been considered polyvalent proteins whose main function is to maintain cellular homeostasis of essential metals such as zinc and copper, but other functions have also been considered: detoxification of heavy metals, regulation of gene expression, processes of inflammation, and protection against free radicals generated by oxidative stress. On the other hand, the MT-III has been related in events of pathogenesis of neurodegenerative diseases such as Parkinson and Alzheimer. Likewise, the participation of MTs in other neurological disorders has also been reported. This review shows recent evidence about the role of MT in the central nervous system and its possible role in neurodegenerative diseases as well as in brain disorders.
Collapse
|
296
|
Bhore N, Wang BJ, Chen YW, Liao YF. Critical Roles of Dual-Specificity Phosphatases in Neuronal Proteostasis and Neurological Diseases. Int J Mol Sci 2017; 18:ijms18091963. [PMID: 28902166 PMCID: PMC5618612 DOI: 10.3390/ijms18091963] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/01/2017] [Accepted: 09/07/2017] [Indexed: 12/31/2022] Open
Abstract
Protein homeostasis or proteostasis is a fundamental cellular property that encompasses the dynamic balancing of processes in the proteostasis network (PN). Such processes include protein synthesis, folding, and degradation in both non-stressed and stressful conditions. The role of the PN in neurodegenerative disease is well-documented, where it is known to respond to changes in protein folding states or toxic gain-of-function protein aggregation. Dual-specificity phosphatases have recently emerged as important participants in maintaining balance within the PN, acting through modulation of cellular signaling pathways that are involved in neurodegeneration. In this review, we will summarize recent findings describing the roles of dual-specificity phosphatases in neurodegeneration and offer perspectives on future therapeutic directions.
Collapse
Affiliation(s)
- Noopur Bhore
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 11529, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| | - Bo-Jeng Wang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| | - Yun-Wen Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| | - Yung-Feng Liao
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 11529, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
297
|
Liu Z, Wang Y, Wang Y, Dong W, Xia X, Song E, Song Y. Effect of Subcellular Translocation of Protein Disulfide Isomerase on Tetrachlorobenzoquinone-Induced Signaling Shift from Endoplasmic Reticulum Stress to Apoptosis. Chem Res Toxicol 2017; 30:1804-1814. [DOI: 10.1021/acs.chemrestox.7b00118] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Zixuan Liu
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Yawen Wang
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Yuxin Wang
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Wenjing Dong
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Xiaomin Xia
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Erqun Song
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Yang Song
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| |
Collapse
|
298
|
Guo X, Namekata K, Kimura A, Harada C, Harada T. ASK1 in neurodegeneration. Adv Biol Regul 2017; 66:63-71. [PMID: 28882588 DOI: 10.1016/j.jbior.2017.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (NDDs) such as glaucoma, multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD) are characterized by the progressive loss of neurons, causing irreversible damage to patients. Longer lifespans may be leading to an increase in the number of people affected by NDDs worldwide. Among the pathways strongly impacting the pathogenesis of NDDs, oxidative stress, a condition that occurs because of an imbalance in oxidant and antioxidant levels, has been known to play a vital role in the pathophysiology of NDDs. One of the molecules activated by oxidative stress is apoptosis signal-regulating kinase 1 (ASK1), which has been shown to play a role in NDDs. ASK1 activation is regulated by multiple steps, including oligomerization, phosphorylation, and protein-protein interactions. In the oxidative stress state, reactive oxygen species (ROS) induce the dissociation of thioredoxin, a protein regulating cellular reduction and oxidation (redox), from the N-terminal region of ASK1, and ASK1 is subsequently activated by the oligomerization and phosphorylation of a critical threonine residue, leading to cell death. Here, we review experimental evidence that links ASK1 signaling with the pathogenesis of several NDDs. We propose that ASK1 may be a new point of therapeutic intervention to prevent or treat NDDs.
Collapse
Affiliation(s)
- Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
299
|
Testosterone Upregulates the Expression of Mitochondrial ND1 and ND4 and Alleviates the Oxidative Damage to the Nigrostriatal Dopaminergic System in Orchiectomized Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1202459. [PMID: 29138672 PMCID: PMC5613679 DOI: 10.1155/2017/1202459] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 08/01/2017] [Accepted: 08/07/2017] [Indexed: 11/17/2022]
Abstract
Testosterone deficiency, as a potential risk factor for aging and aging-related neurodegenerative disorders, might induce mitochondrial dysfunction and facilitate the declines of the nigrostriatal dopaminergic system by exacerbating the mitochondrial defects and increasing the oxidative damage. Thus, how testosterone levels influence the mitochondrial function in the substantia nigra was investigated in the study. The present studies showed that testosterone deficiency impaired the mitochondrial function in the substantia nigra and induced the oxidative damage to the substantia nigra as well as the deficits in the nigrostriatal dopaminergic system. Of four mitochondrial respiratory chain complexes, castration of male rats reduced the activity of mitochondrial complex I and downregulated the expression of ND1 and ND4 of 7 mitochondrial DNA- (mtDNA-) encoded subunits of complex I in the substantia nigra. Supplements of testosterone propionate to castrated male rats ameliorated the activity of mitochondrial complex I and upregulated the expression of mitochondrial ND1 and ND4. These results suggest an important role of testosterone in maintaining the mitochondrial function in the substantia nigra and the vulnerability of mitochondrial complex I to testosterone deficiency. Mitochondrial ND1 and ND4, as potential testosterone targets, were implicated in the oxidative damage to the nigrostriatal dopaminergic system.
Collapse
|
300
|
Abstract
Purpose of Review In this review, we summarise the evidence on the association between cardiovascular disease (CVD) and cognitive impairment and explore the role of the nitric oxide (NO) pathway as a causal mechanism. Recent Findings Evidence from epidemiological studies suggests that the presence of CVD and its risk factors in midlife is associated with an increased risk of later life cognitive impairment and dementia. It is unclear what is driving this association but risk may be conveyed via an increase in neurodegeneration (e.g. amyloid deposition), vascular changes (e.g. small vessel disease) and mechanistically due to increased levels of oxidative stress and inflammation as well as changes in NO bioavailability. Summary CVDs and dementia are major challenges to global health worldwide. The NO pathway may be a promising biological candidate for future studies focused on reducing not only CVD but also risk of cognitive decline and dementia.
Collapse
|