1
|
Bjørklund G, Oliinyk P, Khavrona O, Lozynska I, Lysiuk R, Darmohray R, Antonyak H, Dub N, Zayachuk V, Antoniv O, Rybak O, Peana M. The Effects of Fisetin and Curcumin on Oxidative Damage Caused by Transition Metals in Neurodegenerative Diseases. Mol Neurobiol 2025; 62:1225-1246. [PMID: 38970766 DOI: 10.1007/s12035-024-04321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/19/2024] [Indexed: 07/08/2024]
Abstract
Neurodegenerative diseases pose a significant health challenge for the elderly. The escalating presence of toxic metals and chemicals in the environment is a potential contributor to central nervous system dysfunction and the onset of neurodegenerative conditions. Transition metals play a crucial role in various pathophysiological mechanisms associated with prevalent neurodegenerative diseases such as Alzheimer's and Parkinson's. Given the ubiquitous exposure to metals from diverse sources in everyday life, the workplace, and the environment, most of the population faces regular contact with different forms of these metals. Disturbances in the levels and homeostasis of certain transition metals are closely linked to the manifestation of neurodegenerative disorders. Oxidative damage further exacerbates the progression of neurological consequences. Presently, there exists no curative therapy for individuals afflicted by neurodegenerative diseases, with treatment approaches primarily focusing on alleviating pathological symptoms. Within the realm of biologically active compounds derived from plants, flavonoids and curcuminoids stand out for their extensively documented antioxidant, antiplatelet, and neuroprotective properties. The utilization of these compounds holds the potential to formulate highly effective therapeutic strategies for managing neurodegenerative diseases. This review provides a comprehensive overview of the impact of abnormal metal levels, particularly copper, iron, and zinc, on the initiation and progression of neurodegenerative diseases. Additionally, it aims to elucidate the potential of fisetin and curcumin to inhibit or decelerate the neurodegenerative process.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, 8610, Mo I Rana, Norway.
| | - Petro Oliinyk
- Department of Disaster Medicine and Military Medicine, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Oksana Khavrona
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Biological Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Iryna Lozynska
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Biological Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Roman Lysiuk
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Pharmacognosy and Botany, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Roman Darmohray
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Pharmacognosy and Botany, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Halyna Antonyak
- Department of Ecology, Ivan Franko National University of Lviv, Lviv, 79005, Ukraine
| | - Natalia Dub
- Andrei Krupynskyi Lviv Medical Academy, Lviv, 79000, Ukraine
| | - Vasyl Zayachuk
- Department of Botany, Ukrainian National Forestry University, Wood Science and Non-Wood Forest Products, Lviv, 79057, Ukraine
| | - Olha Antoniv
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Pharmacology, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Oksana Rybak
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Pharmacognosy and Botany, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, 07100, Sassari, Italy.
| |
Collapse
|
2
|
Wei R, Wei P, Yuan H, Yi X, Aschner M, Jiang YM, Li SJ. Inflammation in Metal-Induced Neurological Disorders and Neurodegenerative Diseases. Biol Trace Elem Res 2024; 202:4459-4481. [PMID: 38206494 DOI: 10.1007/s12011-023-04041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024]
Abstract
Essential metals play critical roles in maintaining human health as they participate in various physiological activities. Nonetheless, both excessive accumulation and deficiency of these metals may result in neurotoxicity secondary to neuroinflammation and the activation of microglia and astrocytes. Activation of these cells can promote the release of pro-inflammatory cytokines. It is well known that neuroinflammation plays a critical role in metal-induced neurotoxicity as well as the development of neurological disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Initially seen as a defense mechanism, persistent inflammatory responses are now considered harmful. Astrocytes and microglia are key regulators of neuroinflammation in the central nervous system, and their excessive activation may induce sustained neuroinflammation. Therefore, in this review, we aim to emphasize the important role and molecular mechanisms underlying metal-induced neurotoxicity. Our objective is to raise the awareness on metal-induced neuroinflammation in neurological disorders. However, it is not only just neuroinflammation that different metals could induce; they can also cause harm to the nervous system through oxidative stress, apoptosis, and autophagy, to name a few. The primary pathophysiological mechanism by which these metals induce neurological disorders remains to be determined. In addition, given the various pathways through which individuals are exposed to metals, it is necessary to also consider the effects of co-exposure to multiple metals on neurological disorders.
Collapse
Affiliation(s)
- Ruokun Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Peiqi Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Haiyan Yuan
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Xiang Yi
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Michael Aschner
- The Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yue-Ming Jiang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| | - Shao-Jun Li
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| |
Collapse
|
3
|
Mohammed-Geba K, ElShaarawy RS, Alian A, Ibrahim HM, Galal-Khallaf A. Unraveling the Red Sea soft coral Sarcophyton convolutum potentials against oxidative and inflammatory stresses in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2024; 147:109442. [PMID: 38354966 DOI: 10.1016/j.fsi.2024.109442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/01/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
The Red Sea is one of the world's hotspots for biodiversity, and for marine natural products (MNPs) as well. These MNPs attract special interest for their capabilities to combat inflammatory and oxidative stress-related diseases, being some of the most serious health problems worldwide nowadays. The current study aimed to identify the bioactive ingredients of the Red Sea soft coral Sarcophyton convolutum, and to assess its protective potentials against oxidative and inflammatory stresses. Coral extract (CE) was analyzed using GC-MS and HPLC. In a protection trial, adult zebrafish were intraperitoneally injected with two doses of crab extract, i.e. 50 and 500 μg/fish in 1 % DMSO as a vehicle, then challenged with 30 μg L-1 of CuSO4 for 48 h. All groups, but the negative control one, were challenged with 30 μg L-1 of CuSO4. Total antioxidant activity, as well as mRNA levels of proinflammatory markers and antioxidant enzyme genes were measured. The results showed richness of S. convolutum extract with various bioactive ingredients, including phenolic compounds, flavonoids, alkanes, fatty acids, sesquiterpenes, and pheromone-like substances. CuSO4 significantly induced the expected signals of inflammatory and oxidative stress, reducing both the antioxidant activity and increasing proinflammatory marker genes. However, CE, especially the low dose, showed significant capability to reduce proinflammatory markers and elevating the total antioxidant activity. Therefore, we concluded that S. convolutum can be a promising source for future efforts of drug discovery and a wide spectrum of pharmaceutical products.
Collapse
Affiliation(s)
- Khaled Mohammed-Geba
- Zoology Department, Faculty of Science, Menoufia University, 32511, Shebin El-Kom, Menoufia, Egypt.
| | - Reham Salah ElShaarawy
- Zoology Department, Faculty of Science, Menoufia University, 32511, Shebin El-Kom, Menoufia, Egypt
| | - AbdAllah Alian
- Zoology Department, Faculty of Science, Al-Azhar University, Assiut, 71524, Egypt
| | - Hany Mohammed Ibrahim
- Zoology Department, Faculty of Science, Menoufia University, 32511, Shebin El-Kom, Menoufia, Egypt
| | - Asmaa Galal-Khallaf
- Zoology Department, Faculty of Science, Menoufia University, 32511, Shebin El-Kom, Menoufia, Egypt.
| |
Collapse
|
4
|
Takemura Mariano MV, Paganotto Leandro L, Gomes KK, Dos Santos AB, de Rosso VO, Dafre AL, Farina M, Posser T, Franco JL. Assessing the disparity: comparative toxicity of Copper in zebrafish larvae exposes alarming consequences of permissible concentrations in Brazil. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:166-184. [PMID: 38073470 DOI: 10.1080/15287394.2023.2290630] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Copper (Cu) is a naturally occurring metal with essential micronutrient properties. However, this metal might also pose increased adverse environmental and health risks due to industrial and agricultural activities. In Brazil, the maximum allowable concentration of Cu in drinking water is 2 mg/L. Despite this standard, the impact of such concentrations on aquatic organisms remains unexplored. This study aimed to evaluate the toxicity of CuSO4 using larval zebrafish at environmentally relevant concentrations. Zebrafish (Danio rerio) larvae at 72 hr post-fertilization (hpf) were exposed to nominal CuSO4 concentrations ranging from 0.16 to 48 mg/L to determine the median lethal concentration (LC50), established at 8.4 mg/L. Subsequently, non-lethal concentrations of 0.16, 0.32, or 1.6 mg/L were selected for assessing CuSO4 -induced toxicity. Morphological parameters, including body length, yolk sac area, and swim bladder area, were adversely affected by CuSO4 exposure, particularly at 1.6 mg/L (3.31 mm ±0.1, 0.192 mm2 ±0.01, and 0.01 mm2 ±0.05, respectively). In contrast, the control group exhibited values of 3.62 mm ±0.09, 0.136 mm2 ±0.013, and 0.3 mm2 ±0.06, respectively. Behavioral assays demonstrated impairments in escape response and swimming capacity, accompanied by increased levels of reactive oxygen species (ROS) and lipid peroxidation. In addition, decreased levels of non-protein thiols and reduced cellular viability were noted. Data demonstrated that exposure to CuSO4 at similar concentrations as those permitted in Brazil for Cu adversely altered morphological, biochemical, and behavioral endpoints in zebrafish larvae. This study suggests that the permissible Cu concentrations in Brazil need to be reevaluated, given the potential enhanced adverse health risks of exposure to environmental metal contamination.
Collapse
Affiliation(s)
- Maria Vitória Takemura Mariano
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| | - Luana Paganotto Leandro
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
- Department of Molecular Biology and Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Karen Kich Gomes
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| | - Ana Beatriz Dos Santos
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| | - Vitor Oliveira de Rosso
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| | - Alcir Luiz Dafre
- Department of Biochemistry, Center for Biological Sciences, Federal University of Santa Catarina, Santa Catarina, Brazil
| | - Marcelo Farina
- Department of Biochemistry, Center for Biological Sciences, Federal University of Santa Catarina, Santa Catarina, Brazil
| | - Thaís Posser
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| | - Jeferson Luis Franco
- Oxidative Stress and Cell Signaling Research Group. Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Federal University of Pampa, São Gabriel, Brazil
| |
Collapse
|
5
|
Zhong G, Wang X, Li J, Xie Z, Wu Q, Chen J, Wang Y, Chen Z, Cao X, Li T, Liu J, Wang Q. Insights Into the Role of Copper in Neurodegenerative Diseases and the Therapeutic Potential of Natural Compounds. Curr Neuropharmacol 2024; 22:1650-1671. [PMID: 38037913 PMCID: PMC11284712 DOI: 10.2174/1570159x22666231103085859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/23/2023] [Accepted: 06/05/2023] [Indexed: 12/02/2023] Open
Abstract
Neurodegenerative diseases encompass a collection of neurological disorders originating from the progressive degeneration of neurons, resulting in the dysfunction of neurons. Unfortunately, effective therapeutic interventions for these diseases are presently lacking. Copper (Cu), a crucial trace element within the human body, assumes a pivotal role in various biological metabolic processes, including energy metabolism, antioxidant defense, and neurotransmission. These processes are vital for the sustenance, growth, and development of organisms. Mounting evidence suggests that disrupted copper homeostasis contributes to numerous age-related neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), Wilson's disease (WD), Menkes disease (MD), prion diseases, and multiple sclerosis (MS). This comprehensive review investigates the connection between the imbalance of copper homeostasis and neurodegenerative diseases, summarizing pertinent drugs and therapies that ameliorate neuropathological changes, motor deficits, and cognitive impairments in these conditions through the modulation of copper metabolism. These interventions include Metal-Protein Attenuating Compounds (MPACs), copper chelators, copper supplements, and zinc salts. Moreover, this review highlights the potential of active compounds derived from natural plant medicines to enhance neurodegenerative disease outcomes by regulating copper homeostasis. Among these compounds, polyphenols are particularly abundant. Consequently, this review holds significant implications for the future development of innovative drugs targeting the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Wang
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiaqi Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhouyuan Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiqing Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaxin Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiyun Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziying Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianyao Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinman Liu
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
6
|
Li X, Ling J, Hu Q, Fang C, Mei K, Wu Y, Huang J, Ling Q, Chen Y, Yu P, Liu X, Li J. Association of serum copper (Cu) with cardiovascular mortality and all-cause mortality in a general population: a prospective cohort study. BMC Public Health 2023; 23:2138. [PMID: 37915007 PMCID: PMC10621106 DOI: 10.1186/s12889-023-17018-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 10/19/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Copper (Cu) homeostasis and Cu-induced cell death are gaining recognition as crucial processes in the pathogenesis of cardiovascular disease (CVD). Circulating Cu associated with CVD and mortality is yet to be fully elucidated. OBJECTIVE This national prospective cohort study is to estimate relationship between serum Cu and the risk of CVD and all-cause mortality. METHODS This study included participants from the National Health and Nutrition Examination Survey 2011-2016. Weighted Cox proportional hazards regression analysis and exposure-response curves were applied. RESULTS This included 5,412 adults, representing 76,479,702 individuals. During a mean of 5.85 years of follow-up (31,653 person-years), 96 CVD and 356 all-cause mortality events occurred. Age and sex-adjusted survival curves showed that individuals with higher levels of serum Cu experienced increased CVD and all-cause death rates (tertiles, p < 0.05). Compared with the participant in tertile 1 of serum Cu (< 16.31 mol/L), those in tertile 3 (≥ 19.84 mol/L) were significantly associated with CVD mortality (HR: 7.06, 95%CI: 1.85,26.96), and all-cause mortality (HR: 2.84, 95% CI: 1.66,4.87). The dose-response curve indicated a linear relationship between serum Cu and CVD mortality (p -nonlinear = 0.48) and all-cause (p -nonlinear = 0.62). A meta-analysis included additional three prospective cohorts with 13,189 patients confirmed the association between higher serum Cu and CVD (HR: 2.08, 95% CI: 1.63,2.65) and all-cause mortality (HR: 1.89, 95%CI: 1.58,2.25). CONCLUSION The present study suggests excessive serum Cu concentrations are associated with the risk of CVD and all-cause mortality in American adults. Our findings and the causal relationships require further investigation.
Collapse
Affiliation(s)
- Xiaozhong Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jitao Ling
- Department of Endocrinology Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qingwen Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Changchang Fang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Kaibo Mei
- Department of Anesthesiology, The People's Hospital of Shanggrao, Shanggrao, Jiangxi, China
| | - Yifan Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jingyi Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Qin Ling
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yixuan Chen
- Department of Endocrinology Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- Department of Endocrinology Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong, China.
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Guangzhou, Guangdong, China.
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
7
|
Oza MJ, Gaikwad AB, Kulkarni YA. Effect of diet and nutrition on neuroinflammation: An overview. DIET AND NUTRITION IN NEUROLOGICAL DISORDERS 2023:597-611. [DOI: 10.1016/b978-0-323-89834-8.00030-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
8
|
Aaseth JO, Nurchi VM. Chelation Combination-A Strategy to Mitigate the Neurotoxicity of Manganese, Iron, and Copper? Biomolecules 2022; 12:1713. [PMID: 36421727 PMCID: PMC9687779 DOI: 10.3390/biom12111713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/13/2022] [Accepted: 11/16/2022] [Indexed: 01/19/2024] Open
Abstract
The chelating thiol dimercaptosuccinate (DMSA) and the traditional agent D-penicillamine (PSH) are effective in enhancing the urinary excretion of copper (Cu) and lead (Pb) in poisoned individuals. However, DMSA, PSH, EDTA (ethylenediamine tetraacetate), and deferoxamine (DFOA) are water-soluble agents with limited access to the central nervous system (CNS). Strategies for mobilization of metals such as manganese (Mn), iron (Fe), and Cu from brain deposits may require the combined use of two agents: one water-soluble agent to remove circulating metal into urine, in addition to an adjuvant shuttler to facilitate the brain-to-blood mobilization. The present review discusses the chemical basis of metal chelation and the ligand exchange of metal ions. To obtain increased excretion of Mn, Cu, and Fe, early experiences showed promising results for CaEDTA, PSH, and DFOA, respectively. Recent experiments have indicated that p-amino salicylate (PAS) plus CaEDTA may be a useful combination to remove Mn from binding sites in CNS, while the deferasirox-DFOA and the tetrathiomolybdate-DMSA combinations may be preferable to promote mobilization of Fe and Cu, respectively, from the CNS. Further research is requested to explore benefits of chelator combinations.
Collapse
Affiliation(s)
- Jan O. Aaseth
- Department of Research, Innlandet Hospital Trust, P.O. Box 104, N-2381 Brumunddal, Norway
- Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, P.O. Box 104, N-2418 Elverum, Norway
| | - Valeria M. Nurchi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy
| |
Collapse
|
9
|
Mo X, Hu D, Yang P, Li Y, Bashir S, Nai A, Ma F, Jia G, Xu M. A novel cuproptosis-related prognostic lncRNA signature and lncRNA MIR31HG/miR-193a-3p/TNFRSF21 regulatory axis in lung adenocarcinoma. Front Oncol 2022; 12:927706. [PMID: 35936736 PMCID: PMC9353736 DOI: 10.3389/fonc.2022.927706] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/27/2022] [Indexed: 01/10/2023] Open
Abstract
Lung adenocarcinoma (LUAD) remains the most common subtype of lung malignancy. Cuproptosis is a newly identified cell death which could regulate tumor cell proliferation and progression. Long non-coding RNAs (lncRNAs) are key molecules and potential biomarkers for diagnosing and treating various diseases. However, the effects of cuproptosis-related lncRNAs on LUAD are still unclear. In our study, 7 cuproptosis-related lncRNAs were selected to establish a prognostic model using univariate Cox regression analysis, LASSO algorithm, and multivariate analysis. Furthermore, we evaluated AC008764.2, AL022323.1, ELN-AS1, and LINC00578, which were identified as protective lncRNAs, while AL031667.3, AL606489.1, and MIR31HG were identified as risk lncRNAs. The risk score calculated by the prognostic model proved to be an effective independent factor compared with other clinical features by Cox regression analyses [univariate analysis: hazard ratio (HR) = 1.065, 95% confidence interval (CI) = 1.043–1.087, P < 0.001; multivariate analysis: HR = 1.067, 95% CI = 1.044–1.091, P < 0.001]. In addition, both analyses (ROC and nomogram) were used to corroborate the accuracy and reliability of this signature. The correlation between cuproptosis-related lncRNAs and immune microenvironment was elucidated, where 7 immune cells and 8 immune-correlated pathways were found to be differentially expressed between two risk groups. Furthermore, our results also identified and verified the ceRNA of cuproptosis-related lncRNA MIR31HG/miR-193a-3p/TNFRSF21 regulatory axis using bioinformatics tools. MIR31HG was highly expressed in LUAD specimens and some LUAD cell lines. Inhibition of MIR31HG clearly reduced the proliferation, migration, and invasion of the LUAD cells. MIR31HG showed oncogenic features via sponging miR-193a-3p and tended to positively regulate TNFRSF21 expression. In a word, lncRNA MIR31HG acts as an oncogene in LUAD by targeting miR-193a-3p to modulate TNFRSF21, which may be beneficial to the gene therapy of LUAD.
Collapse
Affiliation(s)
- Xiaocong Mo
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Di Hu
- Department of Neurology and Stroke Centre, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Pingshan Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yin Li
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Shoaib Bashir
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Aitao Nai
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Feng Ma
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Guoxia Jia
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Meng Xu
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- *Correspondence: Meng Xu,
| |
Collapse
|
10
|
He Z, Zheng L, Zhao X, Li X, Xue H, Zhao Q, Ren B, Li N, Ni J, Zhang Y, Liu Q. An Adequate Supply of Bis(ethylmaltolato)oxidovanadium(IV) Remarkably Reversed the Pathological Hallmarks of Alzheimer's Disease in Triple-Transgenic Middle-Aged Mice. Biol Trace Elem Res 2022; 200:3248-3264. [PMID: 35031965 DOI: 10.1007/s12011-021-02938-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/22/2021] [Indexed: 11/02/2022]
Abstract
Alzheimer's disease (AD) is a complex and progressive neurodegenerative disease with impaired synapse, imbalanced mineral metabolism, protein mis-folding and aggregation. Bis(ethylmaltolato)oxidovanadium(IV) (BEOV), an organic bioactive vanadium compound with low toxicity and high bioavailability, has been studied as therapeutic agent against tuberculosis and diabetes. However, its neuroprotective effects have rarely been reported. Therefore, in this study, the potential application of BEOV in intervening AD cognitive dysfunction and neuropathology was evaluated. Both low- and high-dose of BEOV (0.2 mmol/L and 1.0 mmol/L) supplementation for 2 months improved the spatial learning and memory deficits of the triple-transgenic AD (3 × Tg AD) mice and mitigated the loss of synaptic proteins and synaptic dysfunction. By inhibiting the expression of amyloid-β precursor protein and β-secretase, and the phosphorylation of tau protein at Ser262, Ser396, Ser404, and Ser202/Thr205 residues, BEOV reduced the amyloid-β deposition and neurofibrillary tangle formation in AD mouse brains and primarily cultured neurons. Further analysis of the brain ionome revealed that BEOV supplementation could significantly affect the concentrations of a variety of metals, most of which, including several AD risk metals, showed reduced levels, particularly with a high-dose intake. Additionally, the elemental correlation network identified both conserved and specific elemental correlations, implying a highly complex and dynamic crosstalk between vanadium and other elements during long-term BEOV supplementation. Overall, our results suggest that BEOV is effective in AD intervention via both ameliorating the disease related pathology and regulating metal homeostasis.
Collapse
Affiliation(s)
- Zhijun He
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Lin Zheng
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Xu Zhao
- Food Inspection & Quarantine Center, Shenzhen Entry-Exit Inspection and Quarantine Bureau, Shenzhen, 518045, China
| | - Xiaoqian Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Hua Xue
- National Quality Supervision and Inspection Center for Selenium-Enriched Products, Enshi, 445000, China
| | - Qionghui Zhao
- Food Inspection & Quarantine Center, Shenzhen Entry-Exit Inspection and Quarantine Bureau, Shenzhen, 518045, China
| | - Bingyu Ren
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Nan Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Jiazuan Ni
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Yan Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
11
|
Arbo BD, Schimith LE, Goulart dos Santos M, Hort MA. Repositioning and development of new treatments for neurodegenerative diseases: Focus on neuroinflammation. Eur J Pharmacol 2022; 919:174800. [DOI: 10.1016/j.ejphar.2022.174800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/18/2022] [Accepted: 02/02/2022] [Indexed: 11/03/2022]
|
12
|
Liu L, Zhou Q, Lin C, He L, Wei L. Histological alterations, oxidative stress, and inflammatory response in the liver of swamp eel (Monopterus albus) acutely exposed to copper. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:1865-1878. [PMID: 34564773 DOI: 10.1007/s10695-021-01014-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 08/30/2021] [Indexed: 06/13/2023]
Abstract
Copper (Cu) is widely used as an essential trace element in diets as well as a therapeutic chemical. However, excessive Cu has deleterious effects on organisms, including teleosts. Although numerous toxic effects of Cu have been reported, the effects of Cu exposure on the swamp eel (Monopterus albus) as well as the underlying mechanisms have not yet been elucidated. In this study, swamp eels were acutely exposed to 100, 200, and 400 μg/L of Cu for 96 h to evaluate liver histopathology, oxidative stress, and inflammation. Dissolution of hepatocyte membrane, vacuolar degeneration, and inflammatory cell infiltration were detected in the livers of the Cu-treated swamp eels, especially in the 400 μg Cu/L group. Cu-induced hepatic dysfunction was further verified by the elevated activities of glutamate oxaloacetate transaminase (GOT) and glutamate pyruvate transaminase (GPT) and transcript levels of GOT and GPT genes. In addition, Cu exposure decreased the activities of total superoxide dismutase T-SOD and catalase (CAT) and the contents of glutathione (GSH) and total antioxidant capacity (T-AOC) and increased the levels of malondialdehyde (MDA). Cu exposure also significantly decreased the transcript levels of glutathione synthetase (GSS) and increased the transcript levels of SOD1, SOD2, CAT, and heme oxygenase-1 (HO-1) genes. Furthermore, pro-inflammatory genes such as interleukin (IL)-1β, tumor necrosis factor-α (TNF-α), and IL-8 were significantly upregulated. These results indicate that Cu induces oxidative stress and inflammatory response and causes pathological changes in the liver of the swamp eel.
Collapse
Affiliation(s)
- Lin Liu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, People's Republic of China
| | - Qiubai Zhou
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, People's Republic of China
| | - Changgao Lin
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, People's Republic of China
| | - Li He
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, People's Republic of China
| | - Lili Wei
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, People's Republic of China.
| |
Collapse
|
13
|
Pal A, Rani I, Pawar A, Picozza M, Rongioletti M, Squitti R. Microglia and Astrocytes in Alzheimer's Disease in the Context of the Aberrant Copper Homeostasis Hypothesis. Biomolecules 2021; 11:1598. [PMID: 34827595 PMCID: PMC8615684 DOI: 10.3390/biom11111598] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 10/09/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022] Open
Abstract
Evidence of copper's (Cu) involvement in Alzheimer's disease (AD) is available, but information on Cu involvement in microglia and astrocytes during the course of AD has yet to be structurally discussed. This review deals with this matter in an attempt to provide an updated discussion on the role of reactive glia challenged by excess labile Cu in a wide picture that embraces all the major processes identified as playing a role in toxicity induced by an imbalance of Cu in AD.
Collapse
Affiliation(s)
- Amit Pal
- Department of Biochemistry, AIIMS, Kalyani 741245, West Bengal, India
| | - Isha Rani
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research (MMIMSR), Maharishi Markandeshwar University (MMU), Mullana, Ambala 133207, Haryana, India;
| | - Anil Pawar
- Department of Zoology, DAV University, Jalandhar 144012, Punjab, India;
| | - Mario Picozza
- Neuroimmunology Unit, IRCSS Fondazione Santa Lucia, 00143 Rome, Italy;
| | - Mauro Rongioletti
- Department of Laboratory Medicine, Research and Development Division, San Giovanni Calibita Fatebenefratelli Hospital, Isola Tiberina, 00186 Rome, Italy;
| | - Rosanna Squitti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| |
Collapse
|
14
|
Gromadzka G, Tarnacka B, Flaga A, Adamczyk A. Copper Dyshomeostasis in Neurodegenerative Diseases-Therapeutic Implications. Int J Mol Sci 2020; 21:E9259. [PMID: 33291628 PMCID: PMC7730516 DOI: 10.3390/ijms21239259] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/12/2022] Open
Abstract
Copper is one of the most abundant basic transition metals in the human body. It takes part in oxygen metabolism, collagen synthesis, and skin pigmentation, maintaining the integrity of blood vessels, as well as in iron homeostasis, antioxidant defense, and neurotransmitter synthesis. It may also be involved in cell signaling and may participate in modulation of membrane receptor-ligand interactions, control of kinase and related phosphatase functions, as well as many cellular pathways. Its role is also important in controlling gene expression in the nucleus. In the nervous system in particular, copper is involved in myelination, and by modulating synaptic activity as well as excitotoxic cell death and signaling cascades induced by neurotrophic factors, copper is important for various neuronal functions. Current data suggest that both excess copper levels and copper deficiency can be harmful, and careful homeostatic control is important. This knowledge opens up an important new area for potential therapeutic interventions based on copper supplementation or removal in neurodegenerative diseases including Wilson's disease (WD), Menkes disease (MD), Alzheimer's disease (AD), Parkinson's disease (PD), and others. However, much remains to be discovered, in particular, how to regulate copper homeostasis to prevent neurodegeneration, when to chelate copper, and when to supplement it.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Collegium Medicum, Faculty of Medicine, Cardinal Stefan Wyszynski University, Wóycickiego 1/3 Street, 01-938 Warsaw, Poland;
| | - Beata Tarnacka
- Department of Rehabilitation, Eleonora Reicher National Institute of Geriatrics, Rheumatology and Rehabilitation, Rehabilitation Clinic, Medical University of Warsaw, Spartańska 1 Street, 02-637 Warsaw, Poland;
| | - Anna Flaga
- Collegium Medicum, Faculty of Medicine, Cardinal Stefan Wyszynski University, Wóycickiego 1/3 Street, 01-938 Warsaw, Poland;
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Street, 02-106 Warsaw, Poland;
| |
Collapse
|
15
|
Lee S, Chung CYS, Liu P, Craciun L, Nishikawa Y, Bruemmer KJ, Hamachi I, Saijo K, Miller EW, Chang CJ. Activity-Based Sensing with a Metal-Directed Acyl Imidazole Strategy Reveals Cell Type-Dependent Pools of Labile Brain Copper. J Am Chem Soc 2020; 142:14993-15003. [PMID: 32815370 PMCID: PMC7877313 DOI: 10.1021/jacs.0c05727] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Copper is a required nutrient for life and particularly important to the brain and central nervous system. Indeed, copper redox activity is essential to maintaining normal physiological responses spanning neural signaling to metabolism, but at the same time copper misregulation is associated with inflammation and neurodegeneration. As such, chemical probes that can track dynamic changes in copper with spatial resolution, especially in loosely bound, labile forms, are valuable tools to identify and characterize its contributions to healthy and disease states. In this report, we present an activity-based sensing (ABS) strategy for copper detection in live cells that preserves spatial information by a copper-dependent bioconjugation reaction. Specifically, we designed copper-directed acyl imidazole dyes that operate through copper-mediated activation of acyl imidazole electrophiles for subsequent labeling of proximal proteins at sites of elevated labile copper to provide a permanent stain that resists washing and fixation. To showcase the utility of this new ABS platform, we sought to characterize labile copper pools in the three main cell types in the brain: neurons, astrocytes, and microglia. Exposure of each of these cell types to physiologically relevant stimuli shows distinct changes in labile copper pools. Neurons display translocation of labile copper from somatic cell bodies to peripheral processes upon activation, whereas astrocytes and microglia exhibit global decreases and increases in intracellular labile copper pools, respectively, after exposure to inflammatory stimuli. This work provides foundational information on cell type-dependent homeostasis of copper, an essential metal in the brain, as well as a starting point for the design of new activity-based probes for metals and other dynamic signaling and stress analytes in biology.
Collapse
Affiliation(s)
| | | | | | | | - Yuki Nishikawa
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- ERATO Innovative Molecular Technology for Neuroscience Project, Japan Science and Technology Agency (JST), Kyoto 615-8530, Japan
| | | | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- ERATO Innovative Molecular Technology for Neuroscience Project, Japan Science and Technology Agency (JST), Kyoto 615-8530, Japan
| | | | | | | |
Collapse
|
16
|
Zhao X, Li S, Gaur U, Zheng W. Artemisinin Improved Neuronal Functions in Alzheimer's Disease Animal Model 3xtg Mice and Neuronal Cells via Stimulating the ERK/CREB Signaling Pathway. Aging Dis 2020; 11:801-819. [PMID: 32765947 PMCID: PMC7390534 DOI: 10.14336/ad.2019.0813] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 08/13/2019] [Indexed: 01/03/2023] Open
Abstract
The most common form of dementia is Alzheimer’s disease which is characterized by memory loss and cognitive disorders. The pathogenesis of Alzheimer’s disease is not known at present but toxicity of amyloid-beta is one of the central hypotheses. Amyloid-beta can stimulate the production of reactive oxygen species (ROS), cause oxidative stress, damage mitochondrial, cause inflammatory reactions and activate apoptosis related factors which lead to the neuronal death. In this study, we found that artemisinin, a first line antimalarial drug used in clinic for decades, improved the cognitive functions in Alzheimer’s disease animal model 3xTg mice. Further study showed that artemisinin reduced the deposition of amyloid-beta and tau protein, reduced the release of inflammation factors and apoptosis factors, and thereby reduced the neuronal cell death. Western blot assay showed that artemisinin stimulated the activation of ERK/CREB signaling pathway. Consistent with these results, artemisinin concentration-dependently promoted the survival of SH-SY5Y cell against toxicity of amyloid-beta protein 1-42 induced ROS accumulation, caspase activation and apoptosis. Artemisinin also stimulated the phosphorylation of ERK1/2 and CREB in SH-SY5Y cells in time and concentration-dependent manner. Inhibition of ERK/CREB pathway attenuated the protective effect of artemisinin. These data put together suggested that artemisinin has the potential to protect neuronal cells in vitro as well as in vivo animal model 3xTg mice via, at least in part, the activation of the ERK/CREB pathway. Our findings also strongly support the potential of artemisinin as a new multi-target drug that can be used for preventing and treating the Alzheimer’s disease.
Collapse
Affiliation(s)
- Xia Zhao
- Center of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Shuai Li
- Center of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Uma Gaur
- Center of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wenhua Zheng
- Center of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
17
|
Wang L, Yin YL, Liu XZ, Shen P, Zheng YG, Lan XR, Lu CB, Wang JZ. Current understanding of metal ions in the pathogenesis of Alzheimer's disease. Transl Neurodegener 2020; 9:10. [PMID: 32266063 PMCID: PMC7119290 DOI: 10.1186/s40035-020-00189-z] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
Background The homeostasis of metal ions, such as iron, copper, zinc and calcium, in the brain is crucial for maintaining normal physiological functions. Studies have shown that imbalance of these metal ions in the brain is closely related to the onset and progression of Alzheimer's disease (AD), the most common neurodegenerative disorder in the elderly. Main body Erroneous deposition/distribution of the metal ions in different brain regions induces oxidative stress. The metal ions imbalance and oxidative stress together or independently promote amyloid-β (Aβ) overproduction by activating β- or γ-secretases and inhibiting α-secretase, it also causes tau hyperphosphorylation by activating protein kinases, such as glycogen synthase kinase-3β (GSK-3β), cyclin-dependent protein kinase-5 (CDK5), mitogen-activated protein kinases (MAPKs), etc., and inhibiting protein phosphatase 2A (PP2A). The metal ions imbalances can also directly or indirectly disrupt organelles, causing endoplasmic reticulum (ER) stress; mitochondrial and autophagic dysfunctions, which can cause or aggravate Aβ and tau aggregation/accumulation, and impair synaptic functions. Even worse, the metal ions imbalance-induced alterations can reversely exacerbate metal ions misdistribution and deposition. The vicious cycles between metal ions imbalances and Aβ/tau abnormalities will eventually lead to a chronic neurodegeneration and cognitive deficits, such as seen in AD patients. Conclusion The metal ions imbalance induces Aβ and tau pathologies by directly or indirectly affecting multiple cellular/subcellular pathways, and the disrupted homeostasis can reversely aggravate the abnormalities of metal ions transportation/deposition. Therefore, adjusting metal balance by supplementing or chelating the metal ions may be potential in ameliorating AD pathologies, which provides new research directions for AD treatment.
Collapse
Affiliation(s)
- Lu Wang
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Ya-Ling Yin
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Xin-Zi Liu
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Peng Shen
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Yan-Ge Zheng
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Xin-Rui Lan
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Cheng-Biao Lu
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Jian-Zhi Wang
- 2Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
18
|
Exposure to CuO Nanoparticles Mediates NFκB Activation and Enhances Amyloid Precursor Protein Expression. Biomedicines 2020; 8:biomedicines8030045. [PMID: 32120908 PMCID: PMC7175332 DOI: 10.3390/biomedicines8030045] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/23/2020] [Accepted: 02/25/2020] [Indexed: 12/14/2022] Open
Abstract
Amyloid precursor protein (APP) is directly related to Aβ amyloidosis—a hallmark of Alzheimer’s disease (AD). However, the impact of environmental factors upon APP biology and Aβ amyloid pathology have not been well studied. The increased use of nanoparticles (NPs) or engineered nanomaterials (ENMs) has led to a growing body of evidence suggesting that exposure to metal/metal oxide NPs, such as Fe2O3, CuO, and ZnO, may contribute to the pathophysiology of neurodegenerative diseases such as AD through neuroinflammation. Our previous studies indicated that exposure to CuO nanoparticles (CuONPs) induce potent in vitro neurotoxicity. Herein, we investigated the effects on APP expression in neuronal cells exposed to different metal oxide NPs. We found a low dose of CuONPs effectively activated the NFκB signaling pathway and increased APP expression. Moreover, the inhibition of p65 expression using siRNA abolished CuONP-mediated APP expression, suggesting that NFκB-regulated APP expression in response to CuONP exposure may be associated with AD pathology.
Collapse
|
19
|
Anti-Inflammatory and Antioxidant Properties of the Ethanol Extract of Clerodendrum Cyrtophyllum Turcz in Copper Sulfate-Induced Inflammation in Zebrafish. Antioxidants (Basel) 2020; 9:antiox9030192. [PMID: 32106612 PMCID: PMC7139517 DOI: 10.3390/antiox9030192] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/09/2023] Open
Abstract
Oxidative stress and inflammation are commonly present in many chronic diseases. These responses are closely related to pathophysiological processes. The inflammatory process can induce oxidative stress and vice versa through the activation of multiple pathways. Therefore, agents with antioxidant and/or anti-inflammatory activities are very useful in the treatment of many pathologies. Clerodendrum cyrthophyllum Turcz, a plant belonging to the Verbenaceae family, is used in Vietnamese traditional medicine for treating migraine, hypertension, inflammation of the throat, and rheumatic arthritis. Despite its usefulness, studies on its biological properties are still scarce. In this study, ethanol extract (EE) of leaves of C. cyrtophyllum showed protective activity against CuSO4 toxicity. The protective activity was proven to relate to antioxidant and anti-inflammatory properties. EE exhibited relatively high antioxidant activity (IC50 of 16.45 µg/mL) as measured by DPPH assay. In an in vivo anti-antioxidant test, three days post fertilization (dpf) zebrafish larvae were treated with different concentrations of EE for 1 h and then exposed to 10 µM CuSO4 for 20 min to induce oxidative stress. Fluorescent probes were used to detect and quantify oxidative stress by measuring the fluorescent intensity (FI) in larvae. FI significantly decreased in the presence of EE at 5 and 20 µg/mL, demonstrating EE’s profound antioxidant effects, reducing or preventing oxidative stress from CuSO4. Moreover, the co-administration of EE also protected zebrafish larvae against oxidative damage from CuSO4 through down-regulation of hsp70 and gadd45bb expression and upregulation of sod. Due to copper accumulation in zebrafish tissues, the damage and oxidative stress were exacerbated overtime, resulting in the upregulation of genes related to inflammatory processes such as cox-2, pla2, c3a, mpo, and pro- and anti-inflammatory cytokines (il-1ß, il-8, tnf-α, and il-10, respectively). However, the association of CuSO4 with EE significantly decreased the expression of cox-2, pla2, c3a, mpo, il-8, and il-1ß. Taken together, the results suggest that EE has potent antioxidant and anti-inflammatory activities and may be useful in the treatment of various inflammatory diseases.
Collapse
|
20
|
Overcoming Resistance to Therapies Targeting the MAPK Pathway in BRAF-Mutated Tumours. JOURNAL OF ONCOLOGY 2020; 2020:1079827. [PMID: 32411231 PMCID: PMC7199609 DOI: 10.1155/2020/1079827] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/21/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
Overactivation of the mitogen-activated protein kinase (MAPK) pathway is an important driver of many human cancers. First line, FDA-approved therapies targeting MAPK signalling, which include BRAF and MEK inhibitors, have variable success across cancers, and a significant number of patients quickly develop resistance. In recent years, a number of preclinical studies have reported alternative methods of overcoming resistance, which include promoting apoptosis, modulating autophagy, and targeting mitochondrial metabolism. This review summarizes mechanisms of resistance to approved MAPK-targeted therapies in BRAF-mutated cancers and discusses novel preclinical approaches to overcoming resistance.
Collapse
|
21
|
Effects of Huang-Lian-Jie-Du Decoction on Oxidative Stress and AMPK-SIRT1 Pathway in Alzheimer's Disease Rat. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:6212907. [PMID: 31976005 PMCID: PMC6959142 DOI: 10.1155/2020/6212907] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/08/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022]
Abstract
Huang-Lian-Jie-Du Decoction (HLJDD), traditional Chinese medicine (TCM), is proven to have ameliorative effects on learning and memory deficits of Alzheimer's disease (AD). The current study aims to reveal the underlying mechanism of HLJDD in the treatment of AD by simultaneous determination on the regulation of HLJDD on oxidative stress, neurotransmitters, and AMPK-SIRT1 pathway in AD. AD model rat was successfully established by injection of D-galactose and Aβ25-35-ibotenic acid. Morris Water Maze (MWM) test was used to evaluate the success of AD modelling. On this basis, an advanced technique with UPLC-QqQ MS/MS was built up and applied to determine the levels of 8 neurotransmitters in rat plasma. Significant alternation in methionine, glutamine, and tryptophan was observed in AD rats' plasma after the administration of HLJDD, relative to the model group. Meanwhile, HLJDD could upregulate the levels of SOD, GSH-Px, AMPK, and SIRT1 and downregulate the content of MDA in the peripheral system of the AD rats. The underlying therapeutic mechanism of HLJDD for the treatment of AD was associated with alleviating oxidation stress, inflammation, neurotransmitters, and energy metabolism. These data provide solid foundation for the potential use of HLJDD to treat AD.
Collapse
|
22
|
Wang Y, Xia J, Shen M, Zhou Y, Wu Z, Shi Y, Xu J, Hou L, Zhang R, Qiu Z, Xie Q, Chen H, Zhang Y, Wang H. Effects of BIS-MEP on Reversing Amyloid Plaque Deposition and Spatial Learning and Memory Impairments in a Mouse Model of β-Amyloid Peptide- and Ibotenic Acid-Induced Alzheimer's Disease. Front Aging Neurosci 2019; 11:3. [PMID: 30723404 PMCID: PMC6349730 DOI: 10.3389/fnagi.2019.00003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/08/2019] [Indexed: 12/29/2022] Open
Abstract
Alzheimer’s disease (AD) is the main type of dementia and is characterized by progressive memory loss and a notable decrease in cholinergic neuron activity. As classic drugs currently used in the clinic, acetylcholinesterase inhibitors (AChEIs) restore acetylcholine levels and relieve the symptoms of AD, but are insufficient at delaying the onset of AD. Based on the multi-target-directed ligand (MTDL) strategy, bis-(-)-nor-meptazinol (BIS-MEP) was developed as a multi-target AChEI that mainly targets AChE catalysis and the β-amyloid (Aβ) aggregation process. In this study, we bilaterally injected Aβ oligomers and ibotenic acid (IBO) into the hippocampus of ICR mice and then subcutaneously injected mice with BIS-MEP to investigate its therapeutic effects and underlying mechanisms. According to the results from the Morris water maze test, BIS-MEP significantly improved the spatial learning and memory impairments in AD model mice. Compared with the vehicle control, the BIS-MEP treatment obviously inhibited the AChE activity in the mouse brain, consistent with the findings from the behavioral tests. The BIS-MEP treatment also significantly reduced the Aβ plaque area in both the hippocampus and cortex, suggesting that BIS-MEP represents a direct intervention for AD pathology. Additionally, the immunohistochemistry and ELISA results revealed that microglia (ionized calcium-binding adapter molecule 1, IBA1) and astrocyte (Glial fibrillary acidic protein, GFAP) activation and the secretion of relevant inflammatory factors (TNFα and IL-6) induced by Aβ were decreased by the BIS-MEP treatment. Furthermore, BIS-MEP showed more advantages than donepezil (an approved AChEI) as an Aβ intervention. Based on our findings, BIS-MEP improved spatial learning and memory deficits in AD mice by regulating acetylcholinesterase activity, Aβ deposition and the inflammatory response in the brain.
Collapse
Affiliation(s)
- Yu Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Xia
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengjun Shen
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yifan Zhou
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhe Wu
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuhuan Shi
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianrong Xu
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lina Hou
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Zhang
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuibai Qiu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongfang Zhang
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
23
|
Inflammatory cytokines expression in Wilson's disease. Neurol Sci 2019; 40:1059-1066. [PMID: 30644005 DOI: 10.1007/s10072-018-3680-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 12/06/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND Wilson's disease (WD) is an autosomal recessive inherited disorder of copper (Cu) metabolism. Inflammation is a self-defensive reaction aimed at eliminating or neutralizing injurious stimuli, and restoring tissue integrity. Copper deposition may lead to inflammation in the organs and tissues of WD patients. OBJECTIVE The aim of this study was to compare the plasma levels of inflammatory cytokines in patients with WD and healthy group, and also to assess whether inflammatory cytokines affects the clinical manifestation of WD. METHODS Ninety-nine patients with WD and 32 controls were recruited for this study. Ray Biotech antibody microarray was used to detect the levels of plasma inflammatory cytokines. RESULTS AND CONCLUSION Our results showed significant increase in T helper (Th) 1 cells (IL-2, TNF-α, and TNF-β), Th2 cells (IL-5, IL-10, and IL-13), and Th17 (IL-23) (p < 0.05). Higher plasma Th 1 cells (IL-2, TNF-α, and TNF-β), Th 2 cells (IL-13), and Th 17 (TGF-β1, IL-23) levels were found in neurological patients compared with control groups (p < 0.01). Besides, we found Th 1 cells (TNF-α and TNF-β), Th 3 (TGF-β1), and Th 17 (IL-23) levels were significantly higher in hepatic and neurological patients (p < 0.05). In addition, the higher Th1 cells (IL-2, TNF-α, and TNF-β), Th2 cells (IL-13), and Th17 (TGF-β1, IL-23) and the course of WD were associated with the severity of the neurological symptoms for WD patients. Altogether, our results indicated that dysregulation of cytokines, mainly increased expression of cytokines and chemokines, occurred in WD patients.
Collapse
|
24
|
Lee JY, Nam JH, Nam Y, Nam HY, Yoon G, Ko E, Kim SB, Bautista MR, Capule CC, Koyanagi T, Leriche G, Choi HG, Yang J, Kim J, Hoe HS. The small molecule CA140 inhibits the neuroinflammatory response in wild-type mice and a mouse model of AD. J Neuroinflammation 2018; 15:286. [PMID: 30309372 PMCID: PMC6182807 DOI: 10.1186/s12974-018-1321-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/19/2018] [Indexed: 12/23/2022] Open
Abstract
Background Neuroinflammation is associated with neurodegenerative diseases, including Alzheimer’s disease (AD). Thus, modulating the neuroinflammatory response represents a potential therapeutic strategy for treating neurodegenerative diseases. Several recent studies have shown that dopamine (DA) and its receptors are expressed in immune cells and are involved in the neuroinflammatory response. Thus, we recently developed and synthesized a non-self-polymerizing analog of DA (CA140) and examined the effect of CA140 on neuroinflammation. Methods To determine the effects of CA140 on the neuroinflammatory response, BV2 microglial cells were pretreated with lipopolysaccharide (LPS, 1 μg/mL), followed by treatment with CA140 (10 μM) and analysis by reverse transcription-polymerase chain reaction (RT-PCR). To examine whether CA140 alters the neuroinflammatory response in vivo, wild-type mice were injected with both LPS (10 mg/kg, intraperitoneally (i.p.)) and CA140 (30 mg/kg, i.p.), and immunohistochemistry was performed. In addition, familial AD (5xFAD) mice were injected with CA140 or vehicle daily for 2 weeks and examined for microglial and astrocyte activation. Results Pre- or post-treatment with CA140 differentially regulated proinflammatory responses in LPS-stimulated microglia and astrocytes. Interestingly, CA140 regulated D1R levels to alter LPS-induced proinflammatory responses. CA140 significantly downregulated LPS-induced phosphorylation of ERK and STAT3 in BV2 microglia cells. In addition, CA140-injected wild-type mice exhibited significantly decreased LPS-induced microglial and astrocyte activation. Moreover, CA140-injected 5xFAD mice exhibited significantly reduced microglial and astrocyte activation. Conclusions CA140 may be beneficial for preventing and treating neuroinflammatory-related diseases, including AD. Electronic supplementary material The online version of this article (10.1186/s12974-018-1321-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ju-Young Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| | - Jin Han Nam
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| | - Youngpyo Nam
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| | - Hye Yeon Nam
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| | - Gwangho Yoon
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| | - Eunhwa Ko
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu, 41061, South Korea
| | - Sang-Bum Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu, 41061, South Korea
| | - Mahealani R Bautista
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Christina C Capule
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Takaoki Koyanagi
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Geoffray Leriche
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Hwan Geun Choi
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu, 41061, South Korea
| | - Jerry Yang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0358, USA
| | - Jeongyeon Kim
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea.
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea.
| |
Collapse
|
25
|
Choo XY, Liddell JR, Huuskonen MT, Grubman A, Moujalled D, Roberts J, Kysenius K, Patten L, Quek H, Oikari LE, Duncan C, James SA, McInnes LE, Hayne DJ, Donnelly PS, Pollari E, Vähätalo S, Lejavová K, Kettunen MI, Malm T, Koistinaho J, White AR, Kanninen KM. Cu II(atsm) Attenuates Neuroinflammation. Front Neurosci 2018; 12:668. [PMID: 30319344 PMCID: PMC6165894 DOI: 10.3389/fnins.2018.00668] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 09/05/2018] [Indexed: 12/31/2022] Open
Abstract
Background: Neuroinflammation and biometal dyshomeostasis are key pathological features of several neurodegenerative diseases, including Alzheimer’s disease (AD). Inflammation and biometals are linked at the molecular level through regulation of metal buffering proteins such as the metallothioneins. Even though the molecular connections between metals and inflammation have been demonstrated, little information exists on the effect of copper modulation on brain inflammation. Methods: We demonstrate the immunomodulatory potential of the copper bis(thiosemicarbazone) complex CuII(atsm) in an neuroinflammatory model in vivo and describe its anti-inflammatory effects on microglia and astrocytes in vitro. Results: By using a sophisticated in vivo magnetic resonance imaging (MRI) approach, we report the efficacy of CuII(atsm) in reducing acute cerebrovascular inflammation caused by peripheral administration of bacterial lipopolysaccharide (LPS). CuII(atsm) also induced anti-inflammatory outcomes in primary microglia [significant reductions in nitric oxide (NO), monocyte chemoattractant protein 1 (MCP-1), and tumor necrosis factor (TNF)] and astrocytes [significantly reduced NO, MCP-1, and interleukin 6 (IL-6)] in vitro. These anti-inflammatory actions were associated with increased cellular copper levels and increased the neuroprotective protein metallothionein-1 (MT1) in microglia and astrocytes. Conclusion: The beneficial effects of CuII(atsm) on the neuroimmune system suggest copper complexes are potential therapeutics for the treatment of neuroinflammatory conditions.
Collapse
Affiliation(s)
- Xin Yi Choo
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia.,Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, Australia
| | - Jeffrey R Liddell
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, Australia
| | - Mikko T Huuskonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Alexandra Grubman
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia.,Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Diane Moujalled
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Jessica Roberts
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Kai Kysenius
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Lauren Patten
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Hazel Quek
- Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Lotta E Oikari
- Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Clare Duncan
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Simon A James
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Australian Synchrotron, Clayton, VIC, Australia
| | - Lachlan E McInnes
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| | - David J Hayne
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul S Donnelly
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| | - Eveliina Pollari
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Suvi Vähätalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Katarína Lejavová
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mikko I Kettunen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Anthony R White
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia.,Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Katja M Kanninen
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia.,A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
26
|
Wang Z, Zhang YH, Guo C, Gao HL, Zhong ML, Huang TT, Liu NN, Guo RF, Lan T, Zhang W, Wang ZY, Zhao P. Tetrathiomolybdate Treatment Leads to the Suppression of Inflammatory Responses through the TRAF6/NFκB Pathway in LPS-Stimulated BV-2 Microglia. Front Aging Neurosci 2018. [PMID: 29535623 PMCID: PMC5835334 DOI: 10.3389/fnagi.2018.00009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although the positive relationship between copper and Alzheimer's disease (AD) was reported by a lot of epidemiological data, the mechanism is not completely known. Copper is a redox metal and serves as a mediator of inflammation. Because the homeostasis of copper is altered in Aβ precursor protein (APP) and presenilin 1 (PS1) transgenic (Tg) mice, the using of copper chelators is a potential therapeutic strategy for AD. Here we report that a copper chelator, tetrathiomolybdate (TM), is a potential therapeutic drug of AD. We investigated whether TM treatment led to a decrease of pro-inflammatory cytokines in vivo and in vitro, and found that TM treatment reduced the expression of iNOS and TNF-α in APP/PS1 Tg mice through up-regulating superoxide dismutase 1 (SOD1) activity. In vitro, once stimulated, microglia secretes a variety of proinflammatory cytokines, so we utilized LPS-stimulated BV-2 cells as the inflammatory cell model to detect the anti-inflammatory effects of TM. Our results indicated that TM-pretreatment suppressed the ubiquitination of TRAF6 and the activation of NFκB without affecting the expression of TLR4 and Myd88 in vitro. By detecting the activity of SOD1 and the production of reactive oxygen species (ROS), we found that the anti-inflammatory effects of TM could be attributed to its ability to reduce the amount of intracellular bioavailable copper, and the production of ROS which is an activator of the TRAF6 auto-ubiquitination. Hence, our results revealed that TM-treatment could reduce the production of inflammatory cytokines by the suppression of ROS/TRAF6/AKT/NFκB signaling pathway.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Ya-Hong Zhang
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Chuang Guo
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Hui-Ling Gao
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Man-Li Zhong
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Ting-Ting Huang
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Na-Na Liu
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Rui-Fang Guo
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Tian Lan
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Wei Zhang
- Department of Hepatobiliary Surgery, General Hospital of Shenyang Military Area Command, Shenyang, China
| | - Zhan-You Wang
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Pu Zhao
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
27
|
Wang Y, Zhao H, Shao Y, Liu J, Li J, Xing M. Copper or/and arsenic induce oxidative stress-cascaded, nuclear factor kappa B-dependent inflammation and immune imbalance, trigging heat shock response in the kidney of chicken. Oncotarget 2017; 8:98103-98116. [PMID: 29228677 PMCID: PMC5716717 DOI: 10.18632/oncotarget.21463] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/22/2017] [Indexed: 12/26/2022] Open
Abstract
Excessive amount of copper (Cu) and inorganic arsenic (iAs) coexists in drinking water in many regions, this is associated with high risk of nephropathy, defined as chronic structural and functional disorders of the kidney. However, the underlying mechanisms are not well understood. In this study, a total of 72 day-old Hy-line chickens were exposed to 300 mg/kg copper sulphate or/and 30 mg/kg arsenic trioxide for 12 weeks. Indicators of oxidative stress, inflammation and heat shock proteins (HSPs) production were analyzed in kidney. The results showed that, when the toxicant was administrated alone, there is an antagonism between redox homeostasis during the first 4 weeks, which follows a collapse of antioxidant system manifested by damaged biomembrane structure. What's worse, oxidative damage-cascaded histopathological lesions were accompanied by increases of proinflammatory mediators and an imbalance of "Th1/Th2 drift" (Th, helper T cell) regulated by nuclear factor kappa B (NF-κB). Simultaneously, intense heat shock response went with the organism. The above-mentioned renal lesions and indicators changes were time-dependent, more complex and deteriorated effects were observed in Cu/iAs combined groups compared with the others. This study supports Cu and iAs have a synergistic type on the nephro-toxicological process additively. In conclusion, oxidative stress and inflammatory induced by Cu or/and iAs are potential mechanisms in their nephrotoxicity, increased heat shock response may play a renoprotection function in tissues damage.
Collapse
Affiliation(s)
- Yu Wang
- Department of Physiology, College of Wildlife Resources, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Hongjing Zhao
- Department of Physiology, College of Wildlife Resources, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Yizhi Shao
- Department of Physiology, College of Wildlife Resources, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Juanjuan Liu
- Department of Physiology, College of Wildlife Resources, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Jinglun Li
- Department of Physiology, College of Wildlife Resources, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Mingwei Xing
- Department of Physiology, College of Wildlife Resources, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| |
Collapse
|
28
|
Wang H, Cheng N, Dong J, Wang X, Han Y, Yang R, Han Y. Serum pentraxin 3 is elevated in patients with neurological Wilson's disease. Clin Chim Acta 2016; 462:178-182. [PMID: 27553858 DOI: 10.1016/j.cca.2016.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 08/14/2016] [Accepted: 08/15/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Wilson's disease (WD) is an autosomal recessive inherited disorder of copper (Cu) metabolism, resulting in pathological accumulation of Cu in many organs and tissues, predominantly in the liver and brain. Cu deposition may lead to neuroinflammation in the brain of WD patients. Pentraxin 3 (PTX3) may play an important role in innate immunity and in WD. We compared plasma PTX3 concentrations in WD patients and healthy controls, and to determine whether PTX3 concentration was associated with neurological disease severity. METHODS This study included 86 WD patients and 28 controls. Plasma PTX3 and C-reactive protein (CRP) concentration levels were measured using specific enzyme-linked immunosorbent assays. Disease severity was determined using the neurological Global Assessment Scale (GAS) for WD. RESULTS Plasma PTX3 levels were significantly higher in patients with neurological WD than in controls. PTX3 levels in WD patients were associated with neurological disease severity. However, there was no correlation between CRP and neurological GAS scores. CONCLUSIONS PTX3 represents a potential biochemical marker of disease severity in patients with neurological WD.
Collapse
Affiliation(s)
- Honghao Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Nan Cheng
- Hospital affiliated to Institute of Neurology, Anhui College of TCM, Hefei, China; Hefei Institute of Physical Science, CAS, China
| | - Jianjian Dong
- Hospital affiliated to Institute of Neurology, Anhui College of TCM, Hefei, China
| | - Xun Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongsheng Han
- Hospital affiliated to Institute of Neurology, Anhui College of TCM, Hefei, China
| | - Renmin Yang
- Hospital affiliated to Institute of Neurology, Anhui College of TCM, Hefei, China
| | - Yongzhu Han
- Hospital affiliated to Institute of Neurology, Anhui College of TCM, Hefei, China.
| |
Collapse
|
29
|
Pereira TCB, Campos MM, Bogo MR. Copper toxicology, oxidative stress and inflammation using zebrafish as experimental model. J Appl Toxicol 2016; 36:876-85. [PMID: 26888422 DOI: 10.1002/jat.3303] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/17/2015] [Accepted: 01/12/2016] [Indexed: 12/26/2022]
Abstract
Copper is an essential micronutrient and a key catalytic cofactor in a wide range of enzymes. As a trace element, copper levels are tightly regulated and both its deficit and excess are deleterious to the organism. Under inflammatory conditions, serum copper levels are increased and trigger oxidative stress responses that activate inflammatory responses. Interestingly, copper dyshomeostasis, oxidative stress and inflammation are commonly present in several chronic diseases. Copper exposure can be easily modeled in zebrafish; a consolidated model in toxicology with increasing interest in immunity-related research. As a result of developmental, economical and genetic advantages, this freshwater teleost is uniquely suitable for chemical and genetic large-scale screenings, representing a powerful experimental tool for a whole-organism approach, mechanistic studies, disease modeling and beyond. Copper toxicological and more recently pro-inflammatory effects have been investigated in both larval and adult zebrafish with breakthrough findings. Here, we provide an overview of copper metabolism in health and disease and its effects on oxidative stress and inflammation responses in zebrafish models. Copper-induced inflammation is highlighted owing to its potential to easily mimic pro-oxidative and pro-inflammatory features that combined with zebrafish genetic tractability could help further in the understanding of copper metabolism, inflammatory responses and related diseases. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Talita Carneiro Brandão Pereira
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Porto Alegre, Brasil.,Laboratório de Biologia Genômica e Molecular, PUCRS, Porto Alegre, Brasil
| | - Maria Martha Campos
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Porto Alegre, Brasil.,Instituto de Toxicologia e Farmacologia, PUCRS, Porto Alegre, Brasil.,Programa de Pós-Graduação em Odontologia, PUCRS, Porto Alegre, Brasil
| | - Maurício Reis Bogo
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Porto Alegre, Brasil.,Laboratório de Biologia Genômica e Molecular, PUCRS, Porto Alegre, Brasil.,Instituto de Toxicologia e Farmacologia, PUCRS, Porto Alegre, Brasil.,Programa de Pós-Graduação em Biologia Celular e Molecular, PUCRS, Porto Alegre, Brasil
| |
Collapse
|
30
|
Zhang Q, Wu HH, Wang Y, Gu GJ, Zhang W, Xia R. Neural stem cell transplantation decreases neuroinflammation in a transgenic mouse model of Alzheimer's disease. J Neurochem 2015; 136:815-825. [PMID: 26525612 DOI: 10.1111/jnc.13413] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/17/2015] [Accepted: 10/20/2015] [Indexed: 12/13/2022]
Abstract
Inflammatory processes are considered to play an important role in the progression of neurodegenerative changes in Alzheimer's disease (AD). A number of studies have reported that inflammatory processes are highly correlated with cognitive deficits in AD-like mice. Transplantation of neural stem cells (NSCs) has been considered as a potential new therapy for the treatment of AD because of its effects in improving cognitive ability. However, NSCs have not been evaluated for their protective effects against inflammatory changes in AD. Here, we injected NSCs into amyloid precursor protein (APP)/PS1 transgenic mice to analyse cognitive function and to measure glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor molecule-1 (Iba-1) and toll-like receptors 4(TLR4) activation. We also quantified TLR-4 pathway-related agents, Aβ concentration and the levels of proinflammatory mediators. Our results showed that in NSC-injected APP/PS1 mice, activation of GFAP, Iba-1, TLR4 and TLR4 pathway-related agents (MyD88, TRIF, P38 MAPK and NF-κB P65) were significantly decreased with decreased expression of proinflammatory mediators (IL-1, IL-6, TNF-α and PGE2). These changes were associated with the amelioration of cognitive deficits, but no difference was found in Aβ concentration. Our results provide novel evidence that NSC transplantation in APP/PS1 mice significantly improved cognitive deficits and was accompanied by the attenuation of inflammatory injury via suppression of glial and TLR4-mediated inflammatory pathway activation. Our data indicate that these pathways may potentially be important therapeutic targets to prevent or delay AD. This study investigated the neuroprotective effect of neural stem cell (NSC) transplantation against Alzheimer's disease (AD) inflammation. We found that NSC treatment in APP/PS1 mice significantly improved cognitive deficits and was accompanied by the attenuation of inflammatory injury via suppression of glial and toll-like receptor 4 (TLR4) activation and its downstream signalling pathways. Our findings indicate that these pathways may be potentially important therapeutic targets to prevent or delay AD.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Hua-Hui Wu
- Harbin Hospital of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Yuan Wang
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Guo-Jun Gu
- Department of Medical Imaging, Tongji Hospital, Medical School of Tongji University, Shanghai, China
| | - Wei Zhang
- Department of Medical Imaging, Renji Hospital, Medical School of Jiaotong University, Shanghai, China
| | - Rong Xia
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Yu F, Gong P, Hu Z, Qiu Y, Cui Y, Gao X, Chen H, Li J. Cu(II) enhances the effect of Alzheimer's amyloid-β peptide on microglial activation. J Neuroinflammation 2015; 12:122. [PMID: 26104799 PMCID: PMC4490619 DOI: 10.1186/s12974-015-0343-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 06/12/2015] [Indexed: 12/14/2022] Open
Abstract
Background Aggregated forms of amyloid-β (Aβ) peptides are important triggers for microglial activation, which is an important pathological component in the brains of Alzheimer’s patients. Cu(II) ions are reported to be coordinated to monomeric Aβ, drive Aβ aggregation, and potentiate Aβ neurotoxicity. Here we investigated whether Cu(II) binding modulates the effect of Aβ on microglial activation and the subsequent neurotoxicity. Methods Aβ peptides were incubated with Cu(II) at an equimolar ratio to obtain the Cu(II)-Aβ complex. Primary and BV-2 microglial cells were treated with Cu(II)-Aβ, Aβ, or Cu(II). The tumor necrosis factor-α (TNF-α) and nitric oxide levels in the media were determined. Extracellular hydrogen peroxide was quantified by a fluorometric assay with Amplex Red. Mitochondrial superoxide was detected by MitoSOX oxidation. Results Incubation of Cu(II) with Aβ confers different chemical properties on the resulting complex. At the subneurotoxic concentrations, Cu(II)-Aβ (but not Aβ or Cu(II) alone) treatment induced an activating morphological phenotype of microglia and induced the microglial release of TNF-α and nitric oxide as well as microglia-mediated neuronal damage. Cu(II)-Aβ-triggered microglial activation was blocked by nuclear factor (NF)-κB inhibitors and was accompanied with NF-κB activation. Moreover, Cu(II)-Aβ induced hydrogen peroxide release, which was not affected by NADPH oxidase inhibitors. Mitochondrial superoxide production was increased after Cu(II)-Aβ stimulation. N-acetyl-cysteine, a scavenger of reactive oxygen species (ROS), inhibited Cu(II)-Aβ-elicited microglial release of TNF-α and nitric oxide as well as the microglia-mediated neurotoxic effect. Conclusion Our observations suggest that Cu(II) enhances the effect of Aβ on microglial activation and the subsequent neurotoxicity. The Cu(II)-Aβ-triggered microglial activation involves NF-κB activation and mitochondrial ROS production. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0343-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fengxiang Yu
- Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Ping Gong
- Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Zhuqin Hu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yu Qiu
- Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yongyao Cui
- Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Xiaoling Gao
- Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Hongzhuan Chen
- Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Juan Li
- Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
32
|
Esparham AE. Nutritional and Metabolic Biomarkers in Autism Spectrum Disorders: An Exploratory Study. Integr Med (Encinitas) 2015; 14:40-53. [PMID: 26770138 PMCID: PMC4566479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
CONTEXT Autism spectrum disorder (ASD) is currently on the rise, now affecting approximately 1 in 68 children in the United States according to a 2010 surveillance summary from the Centers for Disease Control and Prevention (CDC). This figure is an estimated increase of 78% from the figure in 2002. The CDC suggests that more investigation is needed to understand this astounding increase in autism in such a short period. OBJECTIVE The aim of this pilot study was to determine whether a group of children with ASD exhibited similar variations in a broad array of potential correlates, including medical histories, symptoms, genetics, and multiple nutritional and metabolic biomarkers. DESIGN This study was a retrospective, descriptive chart review. SETTING The study took place at the University of Kansas Medical Center (KUMC). PARTICIPANTS Participants were 7 children with ASD who had sought treatment at the Integrative Medicine Clinic at the medical center. RESULTS A majority of the children exhibited an elevated copper:zinc ratio and abnormal vitamin D levels. Children also demonstrated abnormal levels of the essential fatty acids: (1) α-linolenic acid (ALA)- C13:3W3, and (2) linoleic acid (LA)-C18:2W6; high levels of docosahexaenoic acid (DHA); and an elevated ω-6:ω-3 ratio. Three of 7 children demonstrated abnormal manganese levels. Children did not demonstrate elevated urine pyruvate or lactate but did have abnormal detoxification markers. Three of 7 patients demonstrated abnormalities in citric acid metabolites, bacterial metabolism, and fatty acid oxidation markers. A majority demonstrated elevated serum immunoglobulin G (IgG) antibodies to casein, egg whites, egg yolks, and peanuts. A majority had absent glutathione S-transferase (GSTM) at the 1p13.3 location, and 3 of 7 children were heterozygous for the glutathione S-transferase I105V (GSTP1). A majority also exhibited genetic polymorphism of the mitochondrial gene superoxide dismutase A16V (SOD2). CONCLUSIONS The findings from this small group of children with ASD points to the existence of nutritional, metabolic, and genetic correlates of ASD. These factors appear to be important potential abnormalities that warrant a case control study to evaluate their reliability and validity as markers of ASD.
Collapse
Affiliation(s)
- Anna E. Esparham
- Integrative medicine department at the University of Kansas Medical Center (KUMC) in Kansas City, Kansas
| |
Collapse
|
33
|
Cui Z, Zhou L, Liu C, Zhu G, Wu X, Yan Y, Xia X, Ben Z, Song Y, Zhou Y, Zhang H, Zhang D. The role of Homer1b/c in neuronal apoptosis following LPS-induced neuroinflammation. Neurochem Res 2014; 40:204-15. [PMID: 25503822 DOI: 10.1007/s11064-014-1460-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 10/16/2014] [Accepted: 10/18/2014] [Indexed: 02/07/2023]
Abstract
Homer, also designated Vesl, is one member of the newly found postsynaptic density scaffold proteins, playing a vital role in maintaining synaptic integrity, regulating intracellular calcium mobilization, and being critical for the regulation of cellular apoptosis. However, its function in the inflamed central nervous system (CNS) is not fully elucidated. Here, we investigated the role of Homer1b/c, a long form of Homer1, in lipopolysaccharide (LPS) induced neuroinflammation in CNS. Western blot analysis indicated that LPS administration significantly increased the expression of Homer1b/c in rat brain. Moreover, double immunofluorescent staining suggested Homer1b/c was mainly distributed in the cytoplasm of neurons and had a close association with cleaved caspase-3 level in neurons in rat brain after LPS injection. In vitro studies indicated that up-regulation of Homer1b/c might be related to the subsequent apoptosis in neurons treated by conditioned media (CM), collected from LPS-stimulated mixed glial cultures (MGC). We also found down-regulation of Homer1b/c partly blocked the increase of cleaved caspase-3 and the proportion of Bax/Bcl-2 in neurons induced by MGC-CM. Taken together, these findings suggested that Homer1b/c might promote neuronal apoptosis via the Bax/Bcl-2 dependent pathway during neuroinflammation in CNS, and inhibiting Homer1b/c expression might provide a novel neuroprotective strategy against the inflammation-related neuronal apoptosis.
Collapse
Affiliation(s)
- Zhiming Cui
- Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226002, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Copper is an essential element in many biological processes. The critical functions associated with copper have resulted from evolutionary harnessing of its potent redox activity. This same property also places copper in a unique role as a key modulator of cell signal transduction pathways. These pathways are the complex sequence of molecular interactions that drive all cellular mechanisms and are often associated with the interplay of key enzymes including kinases and phosphatases but also including intracellular changes in pools of smaller molecules. A growing body of evidence is beginning to delineate the how, when and where of copper-mediated control over cell signal transduction. This has been driven by research demonstrating critical changes to copper homeostasis in many disorders including cancer and neurodegeneration and therapeutic potential through control of disease-associated cell signalling changes by modulation of copper-protein interactions. This timely review brings together for the first time the diverse actions of copper as a key regulator of cell signalling pathways and discusses the potential strategies for controlling disease-associated signalling processes using copper modulators. It is hoped that this review will provide a valuable insight into copper as a key signal regulator and stimulate further research to promote our understanding of copper in disease and therapy.
Collapse
|