1
|
Akerele CA, Koralnik LR, Lafont E, Gilman C, Walsh-Messinger J, Malaspina D. Nutrition and brain health: Implications of Mediterranean diet elements for psychiatric disorders. Schizophr Res 2025; 281:30-44. [PMID: 40315757 DOI: 10.1016/j.schres.2025.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 04/03/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
The Mediterranean diet is an anti-inflammatory diet now recognized for prevention and intervention against cardiometabolic disorders, although emerging literature also shows its benefits for mental health. This paper surveys literature pertinent to the Mediterranean diet with respect to schizophrenia, bipolar and unipolar depression and cognition. The National Library of Medicine database of literature was searched for publications relating to our topic through June 2024. Our results highlight the Mediterranean diet's potential role in mediating inflammation, potentially through the gut-brain axis, and its neuroprotective role against cognitive decline. Moreover, individual components of the Mediterranean diet are potentially therapeutic as well as protective, particularly fruits, vegetables, fatty fish, and whole grains. These dietary components reduce systemic inflammation, modulate gut microbiota and influence critical pathways such as moderating oxidative stress. Overall, this survey of recent literature highlights the potential of the Mediterranean diet to promote mental health and reduce the risk or severity of psychiatric and neurodegenerative disorders. The analysis underscores broader implications for the Mediterranean diet in advancing mental health outcomes and emphasizes the need for further studies and increased public education to encourage healthier eating habits and dietary interventions.
Collapse
Affiliation(s)
| | | | - Ezequiel Lafont
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY 10029, USA
| | - Caitlin Gilman
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Dolores Malaspina
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY 10029, USA.
| |
Collapse
|
2
|
Ścibior A, Llopis J, Dobrakowski PP, Męcik-Kronenberg T. Magnesium (Mg) and Neurodegeneration: A Comprehensive Overview of Studies on Mg Levels in Biological Specimens in Humans Affected Some Neurodegenerative Disorders with an Update on Therapy and Clinical Trials Supplemented with Selected Animal Studies. Int J Mol Sci 2024; 25:12595. [PMID: 39684308 DOI: 10.3390/ijms252312595] [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: 09/25/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegenerative diseases, characterized by neuron loss, are a group of neurological disorders that adversely affect the lives of millions of people worldwide. Although several medicines have been approved for managing neurodegenerative diseases, new therapies allowing for a significant slowdown in the progression of neurodegenerative syndromes are constantly being sought. Magnesium (Mg), a crucial mineral necessary for the functioning of organisms, is important to normal central nervous system (CNS) activity. Although the effects of this bioelement on the CNS are relatively well recognized, its role in the pathophysiology of neurological disorders in humans is not yet well characterized. Therefore, the main goal of this review is to collect data about a possible association between Mg and neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's Disease (PD), and Amyotrophic lateral sclerosis (ALS) in humans. Hence, the levels of Mg in blood, cerebrospinal fluid (CSF), urine, and hair from subjects with AD, PD, and ALS are compiled to detect possible variations in the levels of this mineral in the biological specimens of people with neurodegenerative illnesses. Additionally, the findings from an animal model are summarized to offer the reader a deeper insight into studies on Mg in the context of neuroprotection and neurodegeneration. Data provided in the present review indicate that Mg, due to its neuroprotective, antioxidant, anti-inflammatory, and mitochondrial-supportive properties, could be a potential therapeutic agent for AD, PD, and ALS. However, more epidemiological studies with standardized methods of dietary assessment and Mg measurement are necessary to recognize its exact role in neurodegenerative disorders. Moreover, extensive well-designed clinical trials are also needed to establish definitive therapeutic protocols and optimal dosages, and to ensure long-term safety of this mineral supplementation in AD, PD, and ALS patients.
Collapse
Affiliation(s)
- Agnieszka Ścibior
- Laboratory of Oxidative Stress, Department of Biomedicine and Environmental Research, Institute of Biological Sciences, Faculty of Medicine, The John Paul II Catholic University of Lublin, Konstantynów St. 1J, 20-708 Lublin, Poland
| | - Juan Llopis
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Centre, University of Granada, 18100 Armilla, Spain
- Sport and Health Research Centre, University of Granada, 18016 Granada, Spain
| | - Paweł P Dobrakowski
- Psychology Institute, Humanitas University in Sosnowiec, Jana Kilińskiego St. 43, 41-200 Sosnowiec, Poland
| | - Tomasz Męcik-Kronenberg
- Department of Pathomorphology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 3 Maja St. 13, 41-800 Zabrze, Poland
- Collegium Medicum im. Dr Władysław Biegański, Jan Długosz University, Washington St. 4/8, 42-200 Częstochowa, Poland
| |
Collapse
|
3
|
Talebi S, Khodagholi F, Bahaeddin Z, Ansari Dezfouli M, Zeinaddini-Meymand A, Berchi Kankam S, Foolad F, Alijaniha F, Fayazi Piranghar F. Does hazelnut consumption affect brain health and function against neurodegenerative diseases? Nutr Neurosci 2024; 27:1008-1024. [PMID: 38151890 DOI: 10.1080/1028415x.2023.2296164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
INTRODUCTION A healthy daily diet and consuming certain nutrients, such as polyphenols, vitamins, and unsaturated fatty acids, may help neuronal health maintenance. Polyphenolic chemicals, which have antioxidant and anti-inflammatory properties, are involved in the neuroprotective pathway. Because of their nutritional value, nuts have been shown in recent research to be helpful in neuroprotection. OBJECTIVE Hazelnut is often consumed worldwide in various items, including processed foods, particularly in bakery, chocolate, and confectionery products. This nut is an excellent source of vitamins, amino acids, tocopherols, phytosterols, polyphenols, minerals, and unsaturated fatty acids. Consuming hazelnut may attenuate the risk of neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, and Huntington's disease due to its anti-inflammatory and anti-oxidant qualities. RESULTS Many documents introduce hazelnut as an excellent choice to provide neuroprotection against neurodegenerative disorders and there is some direct proof of its neuroprotective effects. DISCUSSION So hazelnut consumption in daily diet may reduce neurodegenerative disease risk and be advantageous in reducing the imposed costs of dealing with neurodegenerative diseases.
Collapse
Affiliation(s)
- Shadi Talebi
- Traditional Medicine Clinical Trial Research Center, Shahed University, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Bahaeddin
- Traditional Medicine Clinical Trial Research Center, Shahed University, Tehran, Iran
| | - Mitra Ansari Dezfouli
- Faculty of Medicine, Department of Neurology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | | | - Forough Foolad
- Faculty of Medical Sciences, Department of Physiology, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Alijaniha
- Traditional Medicine Clinical Trial Research Center, Shahed University, Tehran, Iran
- School of Persian Medicine, Department of Traditional Persian Medicine, Shahed University, Tehran, Iran
| | | |
Collapse
|
4
|
Huang L, Lin R, Zhang C, Zheng S, Wang Y, Wu Z, Chen S, Shen Y, Zhang G, Qi Y, Lin L. The Neuroprotective and Anxiolytic Effects of Magnesium Sulfate on Retinal Dopaminergic Neurons in 6-OHDA-Induced Parkinsonian Rats: A Pilot Study. Brain Sci 2024; 14:861. [PMID: 39335357 PMCID: PMC11430011 DOI: 10.3390/brainsci14090861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
This study investigates the protective effects of magnesium sulfate on dopamine neurons in the retinas of rats with 6-hydroxydopamine (6-OHDA)-induced Parkinson's disease (PD). Rapidly progressing cognitive decline often precedes or coincides with the motor symptoms associated with PD. PD patients also frequently exhibit visual function abnormalities. However, the specific mechanisms underlying visual dysfunction in PD patients are not yet fully understood. Therefore, this study aims to investigate whether magnesium homeostasis affects dopaminergic neurons in the retina of PD rats. Thirty-six rats were divided into four groups: (1) control, (2) control with magnesium sulfate (control/MgSO4), (3) Parkinson's disease (PD), and (4) Parkinson's disease with magnesium sulfate (PD/MgSO4). The apomorphine-induced (APO) rotation test assessed the success of the PD models. The open-field experiment measured the rats' anxiety levels. Tyrosine hydroxylase (TH) and glutamate levels, indicators of dopamine neuron survival, were detected using immunofluorescence staining. Protein levels of solute carrier family 41 A1 (SCL41A1), magnesium transporter 1 (MagT1), and cyclin M2 (CNNM2) in the retina were analyzed using Western blot. Results showed that, compared to the PD group, rats in the PD/MgSO4 group had improved psychological states and motor performance at two and four weeks post-surgery. The PD/MgSO4 group also exhibited significantly higher TH fluorescence intensity in the left retinas and lower glutamate fluorescence intensity than the PD group. Additional experiments indicated that the protein levels of SLC41A1, MagT1, and CNNM2 were generally higher in the retinas of the PD/MgSO4 group, along with an increase in retinal magnesium ion content. This suggests that magnesium sulfate may reduce glutamate levels and protect dopamine neurons in the retina. Thus, magnesium sulfate might have therapeutic potential for visual functional impairments in PD patients.
Collapse
Affiliation(s)
- Leyi Huang
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
- Key Laboratory of Brain Aging and Neurodegenerative Disease, Fujian Medical University, Fuzhou 350122, China
| | - Renxi Lin
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
- Key Laboratory of Brain Aging and Neurodegenerative Disease, Fujian Medical University, Fuzhou 350122, China
- Experimental Teaching Center of Basic Medicine, Fujian Medical University, Fuzhou 350122, China
| | - Chunying Zhang
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
| | - Shaoqing Zheng
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China; (S.Z.); (Y.W.); (Z.W.); (S.C.); (Y.S.)
| | - Yiyang Wang
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China; (S.Z.); (Y.W.); (Z.W.); (S.C.); (Y.S.)
| | - Zeyu Wu
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China; (S.Z.); (Y.W.); (Z.W.); (S.C.); (Y.S.)
| | - Sihao Chen
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China; (S.Z.); (Y.W.); (Z.W.); (S.C.); (Y.S.)
| | - Yihan Shen
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China; (S.Z.); (Y.W.); (Z.W.); (S.C.); (Y.S.)
| | - Guoheng Zhang
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
| | - Yuanlin Qi
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
| | - Ling Lin
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
- Key Laboratory of Brain Aging and Neurodegenerative Disease, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
5
|
Cibulka M, Brodnanova M, Halasova E, Kurca E, Kolisek M, Grofik M. The Role of Magnesium in Parkinson's Disease: Status Quo and Implications for Future Research. Int J Mol Sci 2024; 25:8425. [PMID: 39125993 PMCID: PMC11312984 DOI: 10.3390/ijms25158425] [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: 06/14/2024] [Revised: 07/15/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Neurodegenerative diseases represent an increasing economic, social, and, above all, medical burden worldwide. The second most prevalent disease in this category is Parkinson's disease, surpassed only by Alzheimer's. It is a treatable but still incurable systemic disease with a pathogenesis that has not yet been elucidated. Several theories are currently being developed to explain the causes and progression of Parkinson's disease. Magnesium is one of the essential macronutrients and is absolutely necessary for life as we know it. The magnesium cation performs several important functions in the cell in the context of energetic metabolism, substrate metabolism, cell signalling, and the regulation of the homeostasis of other ions. Several of these cellular processes have been simultaneously described as being disrupted in the development and progression of Parkinson's disease. The relationship between magnesium homeostasis and the pathogenesis of Parkinson's disease has received little scientific attention to date. The aim of this review is to summarise and critically evaluate the current state of knowledge on the possible role of magnesium in the pathogenesis of Parkinson's disease and to outline possible future directions for research in this area.
Collapse
Affiliation(s)
- Michal Cibulka
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (M.C.); (M.B.); (E.H.)
| | - Maria Brodnanova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (M.C.); (M.B.); (E.H.)
| | - Erika Halasova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (M.C.); (M.B.); (E.H.)
| | - Egon Kurca
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Martin Kolisek
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (M.C.); (M.B.); (E.H.)
| | - Milan Grofik
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| |
Collapse
|
6
|
Al-Kuraishy HM, Fahad EH, Al-Windy S, El-Sherbeni SA, Negm WA, Batiha GES. The effects of cholesterol and statins on Parkinson's neuropathology: a narrative review. Inflammopharmacology 2024; 32:917-925. [PMID: 38499742 DOI: 10.1007/s10787-023-01400-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 11/14/2023] [Indexed: 03/20/2024]
Abstract
Parkinson disease (PD) is chronic and progressive neurodegenerative disease of the brain characterized by motor symptoms including tremors, rigidity, postural instability, and bradykinesia. PD neuropathology is due to the progressive degeneration of dopaminergic neurons in the substantia nigra and accumulation of Lewy bodies in the survival neurons. The brain contains a largest amount of cholesterol which is mainly synthesized from astrocytes and glial cells. Cholesterol is intricate in the pathogenesis of PD and may be beneficial or deleterious. Therefore, there are controversial points concerning the role of cholesterol in PD neuropathology. In addition, cholesterol-lowering agents' statins can affect brain cholesterol. Different studies highlighted that statins, via inhibition of brain HMG-CoA, can affect neuronal integrity through suppression of neuronal cholesterol, which regulates synaptic plasticity and neurotransmitter release. Furthermore, statins affect the development and progression of different neurodegenerative diseases in bidirectional ways that could be beneficial or detrimental. Therefore, the objective of the present review was to clarify the double-sward effects of cholesterol and statins on PD neuropathology.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, 14132, Iraq
| | - Esraa H Fahad
- Department of Pharmacology and Toxicology, College of Pharmacy, Mustansiriyah University, Baghdad, 14132, Iraq
| | - Salah Al-Windy
- Department of Biology, College of Science, Baghdad University, Baghdad, Iraq
| | - Suzy A El-Sherbeni
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
7
|
Zeng Z, Cen Y, Xiong L, Hong G, Luo Y, Luo X. Dietary Copper Intake and Risk of Parkinson's Disease: a Cross-sectional Study. Biol Trace Elem Res 2024; 202:955-964. [PMID: 37462848 PMCID: PMC10803382 DOI: 10.1007/s12011-023-03750-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/24/2023] [Indexed: 01/23/2024]
Abstract
Copper is an essential trace element for the human body. The epidemiological evidence for the association of dietary intake of copper with the risk of Parkinson's disease (PD) is limited. We conducted an evaluation of the cross-sectional data gathered from the National Health and Nutrition Examination Surveys spanning from 2007 to 2018, which comprised a total of 17,948 participants. To discern the distinct characteristics of the participants, we performed a univariate analysis and utilized a 1:2 ratio propensity score matching method to minimize the effects of selection bias. We employed weighted univariate as well as three multivariate logistic regression models both prior to and following matching, with the aim of examining the association between dietary copper intake and PD risk. Finally, we used the restricted cubic spline (RCS) methodology in order to investigate possible non-linear relationships. Furthermore, subgroup analysis was undertaken to elicit further understanding concerning the association between copper intake and PD. A negative correlation resulted between dietary copper intake and PD risk in both univariate and multivariate logistic regression models, prior to and following matching. Our findings demonstrate that there is a nonlinear, dose-dependent relationship between copper intake and PD, according to our RCS analysis. In subgroup analysis, copper intake was identified as an important protective factor for individuals who were non-Hispanic White, unmarried, and had completed higher education. Dietary copper intake was associated with the risk of PD. Supplementation of dietary copper may have potentially beneficial effects.
Collapse
Affiliation(s)
- Zhaohao Zeng
- Department of Neurology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518001, Guangdong, China
- The First Clinical Medical College of Jinan University, Guangzhou, 510632, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yanmei Cen
- The First Clinical Medical College of Jinan University, Guangzhou, 510632, Guangdong, China
| | - Lijiao Xiong
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518001, Guangdong, China
| | - Guo Hong
- Department of Neurology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518001, Guangdong, China
- The First Clinical Medical College of Jinan University, Guangzhou, 510632, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yu Luo
- Department of Neurology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518001, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Xiaoguang Luo
- Department of Neurology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518001, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| |
Collapse
|
8
|
Fan YG, Wu TY, Zhao LX, Jia RJ, Ren H, Hou WJ, Wang ZY. From zinc homeostasis to disease progression: Unveiling the neurodegenerative puzzle. Pharmacol Res 2024; 199:107039. [PMID: 38123108 DOI: 10.1016/j.phrs.2023.107039] [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: 10/07/2023] [Revised: 11/16/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
Zinc is a crucial trace element in the human body, playing a role in various physiological processes such as oxidative stress, neurotransmission, protein synthesis, and DNA repair. The zinc transporters (ZnTs) family members are responsible for exporting intracellular zinc, while Zrt- and Irt-like proteins (ZIPs) are involved in importing extracellular zinc. These processes are essential for maintaining cellular zinc homeostasis. Imbalances in zinc metabolism have been linked to the development of neurodegenerative diseases. Disruptions in zinc levels can impact the survival and activity of neurons, thereby contributing to the progression of neurodegenerative diseases through mechanisms like cell apoptosis regulation, protein phase separation, ferroptosis, oxidative stress, and neuroinflammation. Therefore, conducting a systematic review of the regulatory network of zinc and investigating the relationship between zinc dysmetabolism and neurodegenerative diseases can enhance our understanding of the pathogenesis of these diseases. Additionally, it may offer new insights and approaches for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yong-Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| | - Ting-Yao Wu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Ling-Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Rong-Jun Jia
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Hang Ren
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Wen-Jia Hou
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhan-You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| |
Collapse
|
9
|
Liu L, Shen Q, Bao Y, Xu F, Zhang D, Huang H, Tu L, Xu Y. Association between dietary intake and risk of Parkinson's disease: cross-sectional analysis of survey data from NHANES 2007-2016. Front Nutr 2023; 10:1278128. [PMID: 38192644 PMCID: PMC10773772 DOI: 10.3389/fnut.2023.1278128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/23/2023] [Indexed: 01/10/2024] Open
Abstract
Background While dietary factors have shown an association with Parkinson's disease (PD), the available data remains a subject of ongoing debate and controversy. Aim We sought to evaluate potential relationships between dietary consumption of nutrients and micronutrients and risk of PD in a large sample. Methods Cross-sectional data were retrospectively analyzed for 10,651 adults aged 40-80 years that had been collected in the US between 2007 and 2016 as a component of the nationwide National Health and Nutrition Examination Survey. Aspects of dietary intake were compared between those who reported having specific PD medication regimens or not when they completed the survey, and potential associations between diet and risk of PD were explored using binomial logistic regression. We employed Propensity Score Matching (PSM) to minimize the impact of potential confounding factors, thus enhancing the reliability of the results. Additionally, subgroup analysis based on gender and age was conducted to investigate these relationships. Results Higher dietary intake of iron was linked to greater PD risk [odds ratio (OR) 1.065, 95% confidence interval (CI) 1.019-1.114, p = 0.006], whereas risk decreased with higher intake of vitamin K (OR 0.999, 95% CI 0.998-1.000, p = 0.024) or vitamin C (OR 0.998, 95% CI 0.996-0.999, p = 0.039). Even after applying PSM, the connection between dietary iron intake and dietary vitamin C intake with PD risk remained substantial. Subgroup analysis results revealed a significant positive association between dietary intake of iron from food and the PD risk, which was evident among individuals under 60 years of age and among males. Conclusion The intake of micronutrients can influence risk of PD, which should be verified and explored further in prospective samples with other dietary habits and ethnic backgrounds.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yanming Xu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Turck D, Bohn T, Castenmiller J, de Henauw S, Hirsch‐Ernst K, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Bornhorst J, Cubadda F, Dopter A, FitzGerald R, de Sesmaisons Lecarré A, das Neves Ferreira P, Fabiani L, Horvath Z, Matijević L, Naska A. Scientific opinion on the tolerable upper intake level for manganese. EFSA J 2023; 21:e8413. [PMID: 38075631 PMCID: PMC10704406 DOI: 10.2903/j.efsa.2023.8413] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024] Open
Abstract
Following a request from the European Commission (EC), the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver a scientific opinion on the tolerable upper intake level (UL) for manganese. Systematic reviews of the literature of human and animal data were conducted to assess evidence regarding excess manganese intake (including authorised manganese salts) and the priority adverse health effect, i.e. manganese-induced neurotoxicity. Available human and animal studies support neurotoxicity as a critical effect, however, data are not sufficient and suitable to characterise a dose-response relationship and identify a reference point for manganese-induced neurotoxicity. In the absence of adequate data to establish an UL, estimated background dietary intakes (i.e. manganese intakes from natural dietary sources only) observed among high consumers (95th percentile) were used to provide an indication of the highest level of intake where there is reasonable confidence on the absence of adverse effects. A safe level of intake of 8 mg/day was established for adults ≥ 18 years (including pregnant and lactating women) and ranged between 2 and 7 mg/day for other population groups. The application of the safe level of intake is more limited than an UL because the intake level at which the risk of adverse effects starts to increase is not defined.
Collapse
|
11
|
Karpenko MN, Muruzheva ZM, Ilyechova EY, Babich PS, Puchkova LV. Abnormalities in Copper Status Associated with an Elevated Risk of Parkinson's Phenotype Development. Antioxidants (Basel) 2023; 12:1654. [PMID: 37759957 PMCID: PMC10525645 DOI: 10.3390/antiox12091654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023] Open
Abstract
In the last 15 years, among the many reasons given for the development of idiopathic forms of Parkinson's disease (PD), copper imbalance has been identified as a factor, and PD is often referred to as a copper-mediated disorder. More than 640 papers have been devoted to the relationship between PD and copper status in the blood, which include the following markers: total copper concentration, enzymatic ceruloplasmin (Cp) concentration, Cp protein level, and non-ceruloplasmin copper level. Most studies measure only one of these markers. Therefore, the existence of a correlation between copper status and the development of PD is still debated. Based on data from the published literature, meta-analysis, and our own research, it is clear that there is a connection between the development of PD symptoms and the number of copper atoms, which are weakly associated with the ceruloplasmin molecule. In this work, the link between the risk of developing PD and various inborn errors related to copper metabolism, leading to decreased levels of oxidase ceruloplasmin in the circulation and cerebrospinal fluid, is discussed.
Collapse
Affiliation(s)
- Marina N. Karpenko
- I.P. Pavlov Department of Physiology, Research Institute of Experimental Medicine, 197376 St. Petersburg, Russia; (M.N.K.); (Z.M.M.)
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia;
| | - Zamira M. Muruzheva
- I.P. Pavlov Department of Physiology, Research Institute of Experimental Medicine, 197376 St. Petersburg, Russia; (M.N.K.); (Z.M.M.)
- State Budgetary Institution of Health Care “Leningrad Regional Clinical Hospital”, 194291 St. Petersburg, Russia
| | - Ekaterina Yu. Ilyechova
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia;
- Research Center of Advanced Functional Materials and Laser Communication Systems, ADTS Institute, ITMO University, 197101 St. Petersburg, Russia
- Department of Molecular Genetics, Research Institute of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Polina S. Babich
- Department of Zoology and Genetics, Faculty of Biology, Herzen State Pedagogical University of Russia, 191186 St. Petersburg, Russia;
| | - Ludmila V. Puchkova
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia;
- Research Center of Advanced Functional Materials and Laser Communication Systems, ADTS Institute, ITMO University, 197101 St. Petersburg, Russia
- Department of Molecular Genetics, Research Institute of Experimental Medicine, 197376 St. Petersburg, Russia
| |
Collapse
|
12
|
Zhao Y, Ray A, Portengen L, Vermeulen R, Peters S. Metal Exposure and Risk of Parkinson Disease: A Systematic Review and Meta-Analysis. Am J Epidemiol 2023; 192:1207-1223. [PMID: 37022311 PMCID: PMC10326611 DOI: 10.1093/aje/kwad082] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 09/29/2022] [Accepted: 04/04/2023] [Indexed: 04/07/2023] Open
Abstract
Metal exposure has been suggested as a possible environmental risk factor for Parkinson disease (PD). We searched the PubMed, EMBASE, and Cochrane databases to systematically review the literature on the relationship between metal exposure and PD risk and to examine the overall quality of each study and the exposure assessment method. A total of 83 case-control studies and 5 cohort studies published during the period 1963-July 2021 were included, of which 73 were graded as being of low or moderate overall quality. Investigators in 69 studies adopted self-reported exposure and biomonitoring after disease diagnosis for exposure assessment approaches. The meta-analyses showed that concentrations of copper and iron in serum and concentrations of zinc in either serum or plasma were lower, while concentrations of magnesium in CSF and zinc in hair were higher, among PD cases as compared with controls. Cumulative lead levels in bone were found to be associated with increased risk of PD. We did not find associations between other metals and PD. The current level of evidence for associations between metals and PD risk is limited, as biases from methodological limitations cannot be ruled out. High-quality studies assessing metal levels before disease onset are needed to improve our understanding of the role of metals in the etiology of PD.
Collapse
Affiliation(s)
| | | | | | | | - Susan Peters
- Correspondence to Dr. Susan Peters, Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 2, 3584 CM Utrecht, the Netherlands (e-mail: )
| |
Collapse
|
13
|
Hnath B, Chen J, Reynolds J, Choi E, Wang J, Zhang D, Sha CM, Dokholyan NV. Big versus small: The impact of aggregate size in disease. Protein Sci 2023; 32:e4686. [PMID: 37243896 PMCID: PMC10273386 DOI: 10.1002/pro.4686] [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: 02/10/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
Protein aggregation results in an array of different size soluble oligomers and larger insoluble fibrils. Insoluble fibrils were originally thought to cause neuronal cell deaths in neurodegenerative diseases due to their prevalence in tissue samples and disease models. Despite recent studies demonstrating the toxicity associated with soluble oligomers, many therapeutic strategies still focus on fibrils or consider all types of aggregates as one group. Oligomers and fibrils require different modeling and therapeutic strategies, targeting the toxic species is crucial for successful study and therapeutic development. Here, we review the role of different-size aggregates in disease, and how factors contributing to aggregation (mutations, metals, post-translational modifications, and lipid interactions) may promote oligomers opposed to fibrils. We review two different computational modeling strategies (molecular dynamics and kinetic modeling) and how they are used to model both oligomers and fibrils. Finally, we outline the current therapeutic strategies targeting aggregating proteins and their strengths and weaknesses for targeting oligomers versus fibrils. Altogether, we aim to highlight the importance of distinguishing the difference between oligomers and fibrils and determining which species is toxic when modeling and creating therapeutics for protein aggregation in disease.
Collapse
Affiliation(s)
- Brianna Hnath
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Jiaxing Chen
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Joshua Reynolds
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Esther Choi
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Medical Scientist Training ProgramPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Jian Wang
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Dongyan Zhang
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Congzhou M. Sha
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Medical Scientist Training ProgramPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of Engineering Science and MechanicsPenn State UniversityUniversity ParkPennsylvaniaUSA
| | - Nikolay V. Dokholyan
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of Engineering Science and MechanicsPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of Biochemistry & Molecular BiologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of ChemistryPenn State UniversityUniversity ParkPennsylvaniaUSA
| |
Collapse
|
14
|
Cerasuolo M, Di Meo I, Auriemma MC, Trojsi F, Maiorino MI, Cirillo M, Esposito F, Polito R, Colangelo AM, Paolisso G, Papa M, Rizzo MR. Iron and Ferroptosis More than a Suspect: Beyond the Most Common Mechanisms of Neurodegeneration for New Therapeutic Approaches to Cognitive Decline and Dementia. Int J Mol Sci 2023; 24:9637. [PMID: 37298586 PMCID: PMC10253771 DOI: 10.3390/ijms24119637] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/27/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Neurodegeneration is a multifactorial process that involves multiple mechanisms. Examples of neurodegenerative diseases are Parkinson's disease, multiple sclerosis, Alzheimer's disease, prion diseases such as Creutzfeldt-Jakob's disease, and amyotrophic lateral sclerosis. These are progressive and irreversible pathologies, characterized by neuron vulnerability, loss of structure or function of neurons, and even neuron demise in the brain, leading to clinical, functional, and cognitive dysfunction and movement disorders. However, iron overload can cause neurodegeneration. Dysregulation of iron metabolism associated with cellular damage and oxidative stress is reported as a common event in several neurodegenerative diseases. Uncontrolled oxidation of membrane fatty acids triggers a programmed cell death involving iron, ROS, and ferroptosis, promoting cell death. In Alzheimer's disease, the iron content in the brain is significantly increased in vulnerable regions, resulting in a lack of antioxidant defenses and mitochondrial alterations. Iron interacts with glucose metabolism reciprocally. Overall, iron metabolism and accumulation and ferroptosis play a significant role, particularly in the context of diabetes-induced cognitive decline. Iron chelators improve cognitive performance, meaning that brain iron metabolism control reduces neuronal ferroptosis, promising a novel therapeutic approach to cognitive impairment.
Collapse
Affiliation(s)
- Michele Cerasuolo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Irene Di Meo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Maria Chiara Auriemma
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Maria Ida Maiorino
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Mario Cirillo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Fabrizio Esposito
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Rita Polito
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Anna Maria Colangelo
- Laboratory of Neuroscience “R. Levi-Montalcini”, Department of Biotechnology and Biosciences, NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126 Milano, Italy;
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| |
Collapse
|
15
|
Chen HL, Lei WY, Wang JH, Bair MJ, Chen CL. Proton pump inhibitor use and the risk for Parkinson's disease: A nationwide population-based study in Taiwan. Medicine (Baltimore) 2023; 102:e33711. [PMID: 37171333 PMCID: PMC10174385 DOI: 10.1097/md.0000000000033711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 05/13/2023] Open
Abstract
Previous studies have shown that proton pump inhibitors (PPIs) are associated with an increased risk of dementia. However, little is known about the relationship between PPIs use and Parkinson's disease (PD). This study aimed to examine whether PPI use was associated with an increased risk of developing clinically verified PD. This used data from the Taiwan National Health Insurance Research Database for the period between 1999 and 2011, and patients with PPI use were compared with 1 to 1 propensity score-matched controls by age, sex, cohort entry year, and comorbidity. A multivariate analysis was performed using Cox proportional hazards models to estimate the association between PPI use and PD risk. Subgroup analyses according to sex, age, and comorbidities were also conducted. In total, 56,785 PPI users and 56,785 matched controls were enrolled in this study. In the PPI cohort, 366 patients developed PD during a median follow-up of 5.0 years. The incidence rate of PD was 1.48-fold higher in PPI users than in non-PPI users (90.0 vs 133.2 per 100,000 person-years), with an adjusted hazard ratio of 1.76 (95% confidence interval, 1.48-2.08). In the subgroup analysis, the adjusted risk of PD in the PPI and non-PPI cohorts increased in the subgroups regardless of age, sex, and comorbidities. The results of this retrospective, nationwide, population-based cohort study in Taiwan indicate that PPI use is associated with the risk of PD development. Further mechanistic studies on the effect of PPI on PD are needed.
Collapse
Affiliation(s)
- Huan-Lin Chen
- Division of Gastroenterology, Department of Internal Medicine, Taitung MacKay Memorial Hospital, Taitung, Taiwan
- Mackay Medical College, New Taipei, Taiwan
| | - Wei-Yi Lei
- Division of Gastroenterology, Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Jen-Hung Wang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ming-Jong Bair
- Division of Gastroenterology, Department of Internal Medicine, Taitung MacKay Memorial Hospital, Taitung, Taiwan
- Mackay Medical College, New Taipei, Taiwan
| | - Chien-Lin Chen
- Division of Gastroenterology, Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
16
|
Xue J, Li F, Dai P. The Potential of ANK1 to Predict Parkinson's Disease. Genes (Basel) 2023; 14:genes14010226. [PMID: 36672967 PMCID: PMC9859451 DOI: 10.3390/genes14010226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
The main cause of Parkinson's disease (PD) remains unknown and the pathologic changes in the brain limit rapid diagnosis. Herein, differentially expressed genes (DEGs) in the Gene Expression Omnibus (GEO) database (GSE8397 and GSE22491) were assessed using linear models for microarray analysis (limma). Ankyrin 1 (ANK1) was the only common gene differentially down-regulated in lateral substantia nigra (LSN), medial substantia nigra (MSN) and blood. Additionally, DEGs between high ANK1 and low ANK1 in GSE99039 were picked out and then uploaded to the Database for Annotation, Visualization and Integrated Discovery (DAVID) for gene ontology (GO) functional annotation analysis. GO analysis displayed that these DEGs were mainly enriched in oxygen transport, myeloid cell development and gas transport (biological process (BP)); hemoglobin complex, haptoglobin-hemoglobin complex and cortical cytoskeleton (cellular component (CC)); and oxygen transporter activity, haptoglobin binding and oxygen binding (molecular function (MF)). Receiver operating characteristic (ROC) curve analysis showed ANK1 had good diagnostic accuracy and increased the area under the curve (AUC) value when combined with other biomarkers. Consistently, intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropy-ridi-ne (MPTP) in C57BL/6J mice reduced ANK1 mRNA expression in both substantia nigra and blood compared to the control group. Thus, ANK1 may serve as a candidate biomarker for PD diagnosis.
Collapse
|
17
|
Liu J, Chen Y, Lu X, Xu X, Bulloch G, Zhu S, Zhu Z, Ge Z, Wang W, Shang X, He M. The Association between Dietary Iron Intake and Incidence of Dementia in Adults Aged 60 Years or over in the UK Biobank. Nutrients 2023; 15:nu15020260. [PMID: 36678132 PMCID: PMC9865143 DOI: 10.3390/nu15020260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/06/2023] Open
Abstract
Background Several studies have investigated the association between dietary iron intake and cognitive impairment, but little is known about the relationship between iron intake and dementia incidence. Objectives This study explored the association between dietary iron intake and incident dementia in males and females. Whether this association was modified by factors such as age and medical diseases was also examined. Methods We included 41,213 males and 48,892 females aged 60 years or over, from the UK-Biobank cohort. Dietary iron intake was measured using a web-based 24-h dietary recall questionnaire from between 2009 and 2012. Incident dementia was ascertained using hospital inpatient records and death registers until April 2021. Cox proportional regression models examined the association between iron intake and incident dementia, and hazard ratio curves were constructed with knots from the analysis indicating insufficient or excessive iron intake. Results During a mean follow-up of 11.8 years, 560 males and 492 females developed dementia. A non-linear relationship between iron intake and incident dementia was observed in both males and females. The lowest incidence rates were observed in the higher iron intake quintile (Q4: ≥15.73, <17.57 mg/day) for males, and the intermediate iron intake quintile (Q3: ≥12.4, <13.71 mg/day) for females. Among those aged 60 and above, all-cause dementia in males was associated with deficient iron intake (Q1 versus Q4: Hazard ratio [HR]: 1.37, 95% Confidence interval [95%CI]: 1.01−1.86, p = 0.042) and excessive iron intake (Q5 versus Q4: HR: 1.49, 95%CI: 1.14−1.96, p = 0.003), whilst significant associations between all-cause dementia and deficient iron intake were only observed in females without hypertension. Smoking status was a significant moderator (p-value for trend = 0.017) for dementia in males only. Conclusions Excessive iron intake (≥17.57 mg/day) is associated with a higher incidence of all-cause dementia in males and smoking status modified this association amongst males. Deficient iron intake (<10.93 mg/day) was associated with a higher incidence of all-cause dementia in females without a history of hypertension.
Collapse
Affiliation(s)
- Jiahao Liu
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC 3002, Australia
| | - Yutong Chen
- Faculty of Medicine, Nursing and Health Science, Monash University, Clayton, VIC 3800, Australia
| | - Xi Lu
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Xiaojing Xu
- Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Gabriella Bulloch
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC 3002, Australia
| | - Susan Zhu
- Austin Hospital, University of Melbourne, Melbourne, VIC 3084, Australia
| | - Zhuoting Zhu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC 3002, Australia
| | - Zongyuan Ge
- Monash e-Research Center, Faculty of Engineering, Airdoc Research, Nvidia AI Technology Research Center, Monash University, Melbourne, VIC 3800, Australia
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Xianwen Shang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC 3002, Australia
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Mingguang He
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC 3002, Australia
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Correspondence:
| |
Collapse
|
18
|
Zhang X, Wang K, Ren XL, Zhang MD, Wu KN, Wu H, Chu ZW, Liu SS, Jiang XX, Zhu JH, Wu HM. Zinc Deficiency Exacerbates Behavioral Impediments and Dopaminergic Neuron Degeneration in a Mouse Model of Parkinson Disease. J Nutr 2023; 153:167-175. [PMID: 36913450 DOI: 10.1016/j.tjnut.2022.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/12/2022] [Accepted: 11/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Circulating zinc (Zn) concentrations are lower than normal in patients with Parkinson disease (PD). It is unknown whether Zn deficiency increases the susceptibility to PD. OBJECTIVES The study aimed to investigate the effect of dietary Zn deficiency on behaviors and dopaminergic neurons in a mouse model of PD and to explore potential mechanisms. METHODS Male C57BL/6J mice aged 8-10 wk were fed Zn adequate (ZnA; 30 μg/g) or Zn deficient (ZnD; <5 μg/g) diet throughout the experiments. Six weeks later 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was injected to generate the PD model. Controls were injected with saline. Thus, 4 groups (Saline-ZnA, Saline-ZnD, MPTP-ZnA, and MPTP-ZnD) were formed. The experiment lasted 13 wk. Open field test, rotarod test, immunohistochemistry, and RNA sequencing were performed. Data were analyzed with t-test, 2-factor ANOVA, or Kruskal-Wallis test. RESULTS Both MPTP and ZnD diet treatments led to a significant reduction in blood Zn concentrations (PMPTP = 0.012, PZn = 0.014), reduced total distance traveled (PMPTP < 0.001, PZn = 0.031), and affected the degeneration of dopaminergic neurons in the substantia nigra (PMPTP < 0.001, PZn = 0.020). In the MPTP-treated mice, the ZnD diet significantly reduced total distance traveled by 22.4% (P = 0.026), decreased latency to fall by 49.9% (P = 0.026), and reduced dopaminergic neurons by 59.3% (P = 0.002) compared with the ZnA diet. RNA sequencing analysis revealed a total of 301 differentially expressed genes (156 upregulated; 145 downregulated) in the substantia nigra of ZnD mice compared with ZnA mice. The genes were involved in a number of processes, including protein degradation, mitochondria integrity, and α-synuclein aggregation. CONCLUSIONS Zn deficiency aggravates movement disorders in PD mice. Our results support previous clinical observations and suggest that appropriate Zn supplementation may be beneficial for PD.
Collapse
Affiliation(s)
- Xiong Zhang
- Institute of Geriatric Neurology, Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ke Wang
- Institute of Geriatric Neurology, Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Neurology, Lishui Hospital of Zhejiang University (the Central Hospital of Lishui), Lishui, Zhejiang, China
| | - Xiao-Li Ren
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Meng-Di Zhang
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kai-Nian Wu
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Han Wu
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhong-Wei Chu
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shu-Shu Liu
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao-Xia Jiang
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian-Hong Zhu
- Institute of Geriatric Neurology, Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Hong-Mei Wu
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
19
|
Magnesium and the Brain: A Focus on Neuroinflammation and Neurodegeneration. Int J Mol Sci 2022; 24:ijms24010223. [PMID: 36613667 PMCID: PMC9820677 DOI: 10.3390/ijms24010223] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Magnesium (Mg) is involved in the regulation of metabolism and in the maintenance of the homeostasis of all the tissues, including the brain, where it harmonizes nerve signal transmission and preserves the integrity of the blood-brain barrier. Mg deficiency contributes to systemic low-grade inflammation, the common denominator of most diseases. In particular, neuroinflammation is the hallmark of neurodegenerative disorders. Starting from a rapid overview on the role of magnesium in the brain, this narrative review provides evidences linking the derangement of magnesium balance with multiple sclerosis, Alzheimer's, and Parkinson's diseases.
Collapse
|
20
|
Association between egg consumption and cognitive function among Chinese adults: long-term effect and interaction effect of iron intake. Br J Nutr 2022; 128:1180-1189. [PMID: 34736543 DOI: 10.1017/s0007114521004402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The association between egg consumption and cognitive function is inclusive. We aimed to assess the association between egg consumption and cognitive function in Chinese adults and tested the interaction between egg consumption and Fe intake. The data used were from a nationwide sample (n 4852, age ≥ 55 years) from the China Health and Nutrition Survey between 1991 and 2006. Assessment of cognitive function was conducted in 1997, 2000, 2004 and 2006. Dietary egg intake was obtained by 24-h dietary recalls of 3 consecutive days during home visits between 1991 and 2006. Multivariable mixed linear regression and logistic regression were used. Egg intake was positively associated with global cognitive function. In fully adjusted models, across the quartiles of egg intake the regression coefficients were 0, 0·11 (95 % CI -0·28, 0·51), 0·79 (95 % CI 0·36, 1·22) and 0·92 (95 % CI 0·43, 1·41), respectively. There was a significant interaction between egg intake and Fe intake. The association between high egg intake and cognitive function was stronger among those with low Fe intake than those with high Fe intake. In addition, there was a significant interaction between egg consumption and sex, with the association mainly observed in women but not men. Furthermore, compared with non-consumers, those with higher egg consumption (Q4) had the OR of 0·93 (95 % CI 0·74, 1·19), 0·84 (95 % CI 0·69, 1·02) for self-reported poor memory and self-reported memory decline, respectively. Higher egg intake is associated with better cognition in Chinese adults among those with low Fe intake.
Collapse
|
21
|
Pal A, Cerchiaro G, Rani I, Ventriglia M, Rongioletti M, Longobardi A, Squitti R. Iron in Alzheimer's Disease: From Physiology to Disease Disabilities. Biomolecules 2022; 12:1248. [PMID: 36139084 PMCID: PMC9496246 DOI: 10.3390/biom12091248] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/19/2022] Open
Abstract
Reactive oxygen species (ROS) play a key role in the neurodegeneration processes. Increased oxidative stress damages lipids, proteins, and nucleic acids in brain tissue, and it is tied to the loss of biometal homeostasis. For this reason, attention has been focused on transition metals involved in several biochemical reactions producing ROS. Even though a bulk of evidence has uncovered the role of metals in the generation of the toxic pathways at the base of Alzheimer's disease (AD), this matter has been sidelined by the advent of the Amyloid Cascade Hypothesis. However, the link between metals and AD has been investigated in the last two decades, focusing on their local accumulation in brain areas known to be critical for AD. Recent evidence revealed a relation between iron and AD, particularly in relation to its capacity to increase the risk of the disease through ferroptosis. In this review, we briefly summarize the major points characterizing the function of iron in our body and highlight why, even though it is essential for our life, we have to monitor its dysfunction, particularly if we want to control our risk of AD.
Collapse
Affiliation(s)
- Amit Pal
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Kalyani 741245, West Bengal, India
| | - Giselle Cerchiaro
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André 09210-580, SP, Brazil
| | - Isha Rani
- Department of Biochemistry, Maharishi Markandeshwar University (MMU), Mullana, Ambala 133203, Haryana, India
| | - Mariacarla Ventriglia
- Fatebenefratelli Foundation for Health Research and Education, AFaR Division, 00186 Rome, Italy
| | - Mauro Rongioletti
- Department of Laboratory Medicine, Research and Development Division, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy
| | - Antonio Longobardi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Rosanna Squitti
- Department of Laboratory Medicine, Research and Development Division, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy
| |
Collapse
|
22
|
Talebi S, Ghoreishy SM, Jayedi A, Travica N, Mohammadi H. Dietary Antioxidants and Risk of Parkinson's Disease: A Systematic Review and Dose-Response Meta-analysis of Observational Studies. Adv Nutr 2022; 13:1493-1504. [PMID: 35030236 PMCID: PMC9526846 DOI: 10.1093/advances/nmac001] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/09/2021] [Accepted: 01/10/2022] [Indexed: 01/28/2023] Open
Abstract
The aim of the current review was to explore the association between various dietary antioxidants and the risk of developing Parkinson's disease (PD). PubMed, Scopus, Web of Science, and Google Scholar were searched up to March 2021. Prospective, observational cohort studies, nested case-control, and case-control designs that investigated the association between antioxidants and PD risk were included. A random-effects model was used to pool the RRs. The certainty of the evidence was rated using the GRADE (Grading of Recommendations Assessment, Development, and Evaluations) scoring system. In addition, a dose-response relation was examined between antioxidant intake and PD risk. Six prospective cohort studies and 2 nested case-control (total n = 448,737 with 4654 cases), as well as 6 case-control (1948 controls, 1273 cases) studies were eligible. The pooled RR was significantly lower for the highest compared with the lowest intake categories of vitamin E (n = 7; 0.84; 95% CI: 0.71, 0.99) and anthocyanins (n = 2; 0.76; 95% CI: 0.61, 0.96) in cohort studies. Conversely, a significantly higher risk of PD was observed for higher lutein intake (n = 3; 1.86; 95% CI: 1.20, 2.88) among case-control studies. Dose-response meta-analyses indicated a significant association between a 50-mg/d increase in vitamin C (n = 6; RR: 0.94; 95% CI: 0.88, 0.99), a 5-mg/d increment in vitamin E (n = 7; RR: 0.84; 95% CI: 0.70, 0.99), a 2-mg/d increment in β-carotene (n = 6; RR: 0.94, 95% CI: 0.89, 0.99), and a 1-mg/d increment in zinc (n = 1; OR: 0.65; 95% CI: 0.49, 0.86) and a reduced risk of PD. Overall, higher intake of antioxidant-rich foods may be associated with a lower risk of PD. Future well-designed prospective studies are needed to validate the present findings. The protocol was registered in the International Prospective Register of Systematic Reviews (PROSPERO) database (https://www.crd.york.ac.uk/PROSPERO, CRD42021242511).
Collapse
Affiliation(s)
- Sepide Talebi
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran,Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mojtaba Ghoreishy
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Jayedi
- Social Determinant of Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Nikolaj Travica
- Deakin University, IMPACT–the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | | |
Collapse
|
23
|
Litvinenko IV, Lobzin VY. On a New Paradigm of the Development of Neurodegenerative Diseases by the Example of Alzheimer’s Disease and Parkinson’s Disease. ADVANCES IN GERONTOLOGY 2022; 12. [PMCID: PMC9774074 DOI: 10.1134/s2079057022040117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The role of neuronal inflammation developing during the formation of amyloid plaques and Lewy bodies is investigated. The influence of various exogenous and endogenous factors on the development of neuroinflammation is established, but the role of various infectious agents in the development of this process is much less studied. Today, the existence of a universal trigger mechanism of the neurodegenerative process is obvious: a specific pathogen of a bacterial or viral nature (including long-term persistent in nervous tissue in a latent state), reactivating, penetrates into certain cerebral structures, where it is influenced by either Aβ or resident macrophages of the central nervous system, which, in turn, are activated and induce the release of proinflammatory cytokines, leading to the development of neuronal inflammation, autophagy and neurodegeneration. The reactivation of latent infection, such as herpes, in APOE4 carriers significantly increases the risk of development of Alzheimer’s disease. Class-II genes of the HLA locus (HLA II) may be related to the progression of neurodegenerative diseases. An increase in iron levels in the glia is induced by inflammation, which leads to neurodegeneration. Disruption of the homeostasis of redox-active metals, iron and copper, is an integral part of the pathogenesis of Alzheimer’s disease and Parkinson’s disease. The developing neuroinflammation leads to intensification of the processes of peroxidation, oxidation of metals and the development of ferroptosis.
Collapse
Affiliation(s)
| | - V. Yu. Lobzin
- Kirov Military Medical Academy, 194044 St. Petersburg, Russia ,Mechnikov North-Western State Medical University, 191015 St. Petersburg, Russia ,Children’s Research and Clinical Center of Infectious Diseases, 197022 St. Petersburg, Russia
| |
Collapse
|
24
|
SOZEN E, DEMIREL-YALCINER T, ECE A, ISMICOGLU A, KARTAL ÖZER N. Effect of High Cholesterol Diet and α-Tocopherol Supplementation on Endoplasmic Retüculum Stress and Apoptosis in Hippocampus Tissue. CLINICAL AND EXPERIMENTAL HEALTH SCIENCES 2021. [DOI: 10.33808/clinexphealthsci.972222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
25
|
Dos Santos AB, Bezerra MA, Rocha ME, Barreto GE, Kohlmeier KA. Lower calcium levels in hair of Parkinson's disease patients are associated with presence of sleeping disturbances. Nutr Neurosci 2021; 25:2577-2587. [PMID: 34693879 DOI: 10.1080/1028415x.2021.1990464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Objectives: To investigate the correlation between sleep disorders and the concentrations of three metals analyzed from hair samples of PD patients.The hypothesis of an involvement of an imbalance of metals in the development of Parkinson's Disease (PD) has been strengthened by several clinical chemistry studies. Interestingly, while sparse, some studies have correlated the imbalance of metals in PD patients with comorbidities present in this disease. Although not all PD sufferers present sleep disturbances, significant disorders of sleep are common in this population. Methods: Sleep evaluation was divided into three parameters: sleep quality, excessive daytime sleepiness and clinically probable REM Sleep Behavior Disorder. Flame atomic absorption spectrometry (F AAS) was used to assess the concentrations of calcium, iron and zinc in hair samples collected from a population of PD patients registered in a Brazilian city and from controls (a total of 53 subjects). All subjects lived within a restricted geographical region and were exposed to similar environmental conditions. Results: PD patients with poor sleep quality and excessive daytime sleepiness exhibited significant differences in concentrations of calcium, but not iron or zinc when compared to levels found in controls and PD patients who do not report these sleeping problems. Discussion: Our data suggest that different subgroups of PD patients exist, and clinical chemistry could be useful as a biomarker for these subgroups, which needs to be confirmed in a larger patient population. Further, our data raise the question regarding whether normalization of calcium levels could improve the sleep quality and somnolence in PD patients.
Collapse
Affiliation(s)
- Altair Brito Dos Santos
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Bahia, Brazil
| | - Marcos A Bezerra
- Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Bahia, Brazil
| | - Marcelo E Rocha
- Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Bahia, Brazil
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Wang YC, Huang APH, Yuan SP, Huang CY, Wu CC, Poly TN, Atique S, Kung WM. Association between Anemia and Risk of Parkinson Disease. Behav Neurol 2021; 2021:8360627. [PMID: 34306250 PMCID: PMC8279865 DOI: 10.1155/2021/8360627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/19/2021] [Indexed: 02/08/2023] Open
Abstract
METHODS We systematically searched articles on electronic databases such as PubMed, Embase, Scopus, and Google Scholar between January 1, 2000 and July 30, 2020. Articles were independently evaluated by two authors. We included observational studies (case-control and cohort) and calculated the risk ratios (RRs) for associated with anemia and PD. Heterogeneity among the studies was assessed using the Q and I 2 statistic. We utilized the random-effect model to calculate the overall RR with 95% CI. RESULTS A total of 342 articles were identified in the initial searches, and 7 full-text articles were evaluated for eligibility. Three articles were further excluded for prespecified reasons including insufficient data and duplications, and 4 articles were included in our systematic review and meta-analysis. A random effect model meta-analysis of all 4 studies showed no increased risk of PD in patients with anemia (N = 4, RRadjusted = 1.17 (95% CI: 0.94-1.45, p = 0.15). However, heterogeneity among the studies was significant (I 2 = 92.60, p = <0.0001). The pooled relative risk of PD in female patients with anemia was higher (N = 3, RRadjusted = 1.14 (95% CI: 0.83-1.57, p = 0.40) as compared to male patients with anemia (N = 3, RRadjusted = 1.09 (95% CI: 0.83-1.42, p = 0.51). CONCLUSION This is the first meta-analysis that shows that anemia is associated with higher risk of PD when compared with patients without anemia. However, more studies are warranted to evaluate the risk of PD among patients with anemia.
Collapse
Affiliation(s)
- Yao-Chin Wang
- Department of Emergency, Min-Sheng General Hospital, Taoyuan, Taiwan
- Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Abel Po-Hao Huang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Sheng-Po Yuan
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Department of Otorhinolaryngology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Otorhinolaryngology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chu-Ya Huang
- Taiwan College of Healthcare Executives, Taipei, Taiwan
| | - Chieh-Chen Wu
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei, Taiwan
| | - Tahmina Nasrin Poly
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Suleman Atique
- Department of Health Informatics, College of Public Health and Health Informatics, University of Ha'il, Ha'il, Saudi Arabia
| | - Woon-Man Kung
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei, Taiwan
| |
Collapse
|
27
|
Kim JH, Oh JK, Wee JH, Min CY, Yoo DM, Choi HG. The Association between Anemia and Parkinson's Disease: A Nested Case-Control Study Using a National Health Screening Cohort. Brain Sci 2021; 11:623. [PMID: 34068124 PMCID: PMC8152756 DOI: 10.3390/brainsci11050623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 01/02/2023] Open
Abstract
(1) Background: Controversy exists regarding the relationship between anemia and Parkinson's disease (PD). This study aimed to evaluate the risk of PD related to anemia in the Korean population. (2) Methods: The Korean National Health Insurance Service-National Sample Cohort, which includes adults over 40 years of age, was assessed from 2002 to 2015. A total of 5844 PD patients were matched by age, sex, income, and region of residence with 23,376 control participants at a ratio of 1:4. The analyzed covariates included age, sex, blood pressure, fasting blood glucose, obesity, smoking status, and alcohol consumption. A multiple logistic regression analysis was conducted for case-control analyses. (3) Results: The adjusted odds ratio (OR) for the risk of PD associated with anemia was 1.09 after adjusting for potential confounders (95% confidence interval (CI) 1.01-1.18, p = 0.030). Among men younger than 70 years, the adjusted OR of PD was 1.34 (95% CI 1.13-1.60, p = 0.001). (4) Conclusions: Our findings suggest that anemia may increase the risk of PD, particularly in men younger than 70 years. Further research is required to elucidate the causal relationship between these two diseases.
Collapse
Affiliation(s)
- Ji Hee Kim
- Department of Neurosurgery, Hallym University College of Medicine, Anyang 14068, Korea; (J.H.K.); (J.K.O.)
| | - Jae Keun Oh
- Department of Neurosurgery, Hallym University College of Medicine, Anyang 14068, Korea; (J.H.K.); (J.K.O.)
| | - Jee Hye Wee
- Department of Otorhinolaryngology, Hallym University College of Medicine, Anyang 14068, Korea;
| | - Chan Yang Min
- Hallym Data Science Laboratory, Hallym University College of Medicine, Anyang 14068, Korea; (C.Y.M.); (D.M.Y.)
| | - Dae Myoung Yoo
- Hallym Data Science Laboratory, Hallym University College of Medicine, Anyang 14068, Korea; (C.Y.M.); (D.M.Y.)
| | - Hyo Geun Choi
- Hallym Data Science Laboratory, Department of Otorhinolaryngology-Head & Neck Surgery, Hallym University College of Medicine, Anyang 14068, Korea
| |
Collapse
|
28
|
Synaptic Zinc: An Emerging Player in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22094724. [PMID: 33946908 PMCID: PMC8125092 DOI: 10.3390/ijms22094724] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 02/03/2023] Open
Abstract
Alterations of zinc homeostasis have long been implicated in Parkinson's disease (PD). Zinc plays a complex role as both deficiency and excess of intracellular zinc levels have been incriminated in the pathophysiology of the disease. Besides its role in multiple cellular functions, Zn2+ also acts as a synaptic transmitter in the brain. In the forebrain, subset of glutamatergic neurons, namely cortical neurons projecting to the striatum, use Zn2+ as a messenger alongside glutamate. Overactivation of the cortico-striatal glutamatergic system is a key feature contributing to the development of PD symptoms and dopaminergic neurotoxicity. Here, we will cover recent evidence implicating synaptic Zn2+ in the pathophysiology of PD and discuss its potential mechanisms of actions. Emphasis will be placed on the functional interaction between Zn2+ and glutamatergic NMDA receptors, the most extensively studied synaptic target of Zn2+.
Collapse
|
29
|
Fiorentini D, Cappadone C, Farruggia G, Prata C. Magnesium: Biochemistry, Nutrition, Detection, and Social Impact of Diseases Linked to Its Deficiency. Nutrients 2021; 13:1136. [PMID: 33808247 PMCID: PMC8065437 DOI: 10.3390/nu13041136] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
Magnesium plays an important role in many physiological functions. Habitually low intakes of magnesium and in general the deficiency of this micronutrient induce changes in biochemical pathways that can increase the risk of illness and, in particular, chronic degenerative diseases. The assessment of magnesium status is consequently of great importance, however, its evaluation is difficult. The measurement of serum magnesium concentration is the most commonly used and readily available method for assessing magnesium status, even if serum levels have no reliable correlation with total body magnesium levels or concentrations in specific tissues. Therefore, this review offers an overview of recent insights into magnesium from multiple perspectives. Starting from a biochemical point of view, it aims at highlighting the risk due to insufficient uptake (frequently due to the low content of magnesium in the modern western diet), at suggesting strategies to reach the recommended dietary reference values, and at focusing on the importance of detecting physiological or pathological levels of magnesium in various body districts, in order to counteract the social impact of diseases linked to magnesium deficiency.
Collapse
Affiliation(s)
| | | | - Giovanna Farruggia
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy; (D.F.); (C.C.); (C.P.)
| | | |
Collapse
|
30
|
Grubić Kezele T, Ćurko-Cofek B. Age-Related Changes and Sex-Related Differences in Brain Iron Metabolism. Nutrients 2020; 12:E2601. [PMID: 32867052 PMCID: PMC7551829 DOI: 10.3390/nu12092601] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/21/2022] Open
Abstract
Iron is an essential element that participates in numerous cellular processes. Any disruption of iron homeostasis leads to either iron deficiency or iron overload, which can be detrimental for humans' health, especially in elderly. Each of these changes contributes to the faster development of many neurological disorders or stimulates progression of already present diseases. Age-related cellular and molecular alterations in iron metabolism can also lead to iron dyshomeostasis and deposition. Iron deposits can contribute to the development of inflammation, abnormal protein aggregation, and degeneration in the central nervous system (CNS), leading to the progressive decline in cognitive processes, contributing to pathophysiology of stroke and dysfunctions of body metabolism. Besides, since iron plays an important role in both neuroprotection and neurodegeneration, dietary iron homeostasis should be considered with caution. Recently, there has been increased interest in sex-related differences in iron metabolism and iron homeostasis. These differences have not yet been fully elucidated. In this review we will discuss the latest discoveries in iron metabolism, age-related changes, along with the sex differences in iron content in serum and brain, within the healthy aging population and in neurological disorders such as multiple sclerosis, Parkinson's disease, Alzheimer's disease, and stroke.
Collapse
Affiliation(s)
- Tanja Grubić Kezele
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia;
- Clinical Department for Clinical Microbiology, Clinical Hospital Center Rijeka, Krešimirova 42, 51000 Rijeka, Croatia
| | - Božena Ćurko-Cofek
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia;
| |
Collapse
|
31
|
McCarty MF, Lerner A. Nutraceuticals Targeting Generation and Oxidant Activity of Peroxynitrite May Aid Prevention and Control of Parkinson's Disease. Int J Mol Sci 2020; 21:3624. [PMID: 32455532 PMCID: PMC7279222 DOI: 10.3390/ijms21103624] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/29/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a chronic low-grade inflammatory process in which activated microglia generate cytotoxic factors-most prominently peroxynitrite-which induce the death and dysfunction of neighboring dopaminergic neurons. Dying neurons then release damage-associated molecular pattern proteins such as high mobility group box 1 which act on microglia via a range of receptors to amplify microglial activation. Since peroxynitrite is a key mediator in this process, it is proposed that nutraceutical measures which either suppress microglial production of peroxynitrite, or which promote the scavenging of peroxynitrite-derived oxidants, should have value for the prevention and control of PD. Peroxynitrite production can be quelled by suppressing activation of microglial NADPH oxidase-the source of its precursor superoxide-or by down-regulating the signaling pathways that promote microglial expression of inducible nitric oxide synthase (iNOS). Phycocyanobilin of spirulina, ferulic acid, long-chain omega-3 fatty acids, good vitamin D status, promotion of hydrogen sulfide production with taurine and N-acetylcysteine, caffeine, epigallocatechin-gallate, butyrogenic dietary fiber, and probiotics may have potential for blunting microglial iNOS induction. Scavenging of peroxynitrite-derived radicals may be amplified with supplemental zinc or inosine. Astaxanthin has potential for protecting the mitochondrial respiratory chain from peroxynitrite and environmental mitochondrial toxins. Healthful programs of nutraceutical supplementation may prove to be useful and feasible in the primary prevention or slow progression of pre-existing PD. Since damage to the mitochondria in dopaminergic neurons by environmental toxins is suspected to play a role in triggering the self-sustaining inflammation that drives PD pathogenesis, there is also reason to suspect that plant-based diets of modest protein content, and possibly a corn-rich diet high in spermidine, might provide protection from PD by boosting protective mitophagy and thereby aiding efficient mitochondrial function. Low-protein diets can also promote a more even response to levodopa therapy.
Collapse
Affiliation(s)
| | - Aaron Lerner
- B. Rappaport School of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
| |
Collapse
|
32
|
The Role of Xenobiotics and Trace Metals in Parkinson’s Disease. Mol Neurobiol 2019; 57:1405-1417. [DOI: 10.1007/s12035-019-01832-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/01/2019] [Indexed: 12/21/2022]
|
33
|
Shen X, Yang H, Zhang D, Jiang H. Iron Concentration Does Not Differ in Blood but Tends to Decrease in Cerebrospinal Fluid in Parkinson's Disease. Front Neurosci 2019; 13:939. [PMID: 31616238 PMCID: PMC6775209 DOI: 10.3389/fnins.2019.00939] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022] Open
Abstract
Background Iron accumulation in the substantia nigra in PD patients was acknowledged, but the studies on alteration of iron levels in blood and cerebrospinal fluids (CSF) reported inconsistent results. Objective To determinate the alterations of blood and CSF levels of iron in PD patients, a case-control study and a meta-analysis both in blood and CSF were conducted. Methods In the case-control study, 43 PD patients and 33 controls were recruited to test iron metabolism, 15 normal and 12 PD patients donated CSF. Levels in iron were quantified by inductively coupled atomic emission spectrometry. Iron metabolism was analyzed by routine blood tests. In the meta-analysis, a comprehensive literature search was performed on relevant studies published from Jan 1980 to Dec 2018 in PubMed, Web of Science and EMBASE databases. The pooled standard mean difference (SMD) with random effects model was selected to estimate the association between iron levels and PD. Results In the case-control study, the iron level in serum in the controls and PD patients were 110.00 ± 48.75 μg/dl and 107.21 ± 34.25 μg/dl, respectively, no significant difference was found between them (p = 0.850), with a small effect size (Cohen’s d: 0.12; 95% CI: 0.08–0.17). Ferritin level in PD patients was lower than controls (p = 0.014). The CSF levels of iron in control and the PD patients were 20.14 ± 3.35 ng/dl and 16.26 ± 4.82 ng/dl, respectively. CSF levels of iron were lower in PD compared with that of controls (p = 0.021), with a moderate effect size (Cohen’s d: 0.51; 95% CI: 0.43–0.65). In the meta-analysis, 22 eligible studies and a total of 3607 participants were identified. Blood levels of iron did not differ significant between PD patients and the controls [SMD (95% CI): −0.03 (−0.30, 0.24)], but CSF iron levels tended to be lower in PD patients compared with that in the controls [SMD (95% CI): −0.33 (−0.65, −0.00)]. Conclusion Iron homeostasis may be disturbed in CSF, but not in the peripheral blood in PD.
Collapse
Affiliation(s)
- Xiaoli Shen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Qingdao University, Qingdao, China.,Department of Epidemiology and Health Statistics, Qingdao University, Qingdao, China
| | - Huazhen Yang
- Department of Epidemiology and Health Statistics, West China School of Public Health, Sichuan University, Chengdu, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Qingdao University, Qingdao, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Qingdao University, Qingdao, China
| |
Collapse
|
34
|
Shi Z, El-Obeid T, Li M, Xu X, Liu J. Iron-related dietary pattern increases the risk of poor cognition. Nutr J 2019; 18:48. [PMID: 31464628 PMCID: PMC6716885 DOI: 10.1186/s12937-019-0476-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION High iron intake has been shown to be associated with poor cognition. We aimed to examine the association between iron-related dietary pattern (IDP) and cognitive function in Chinese adults. METHOD Longitudinal study data from the China Health and Nutrition Survey (CHNS) during 1991-2006 were used (N = 4852, ≥55 years old). Dietary intake was obtained from a 3-day food record during home visits. Reduced rank regression was used to construct IDP with iron intake as a response variable. Cognitive function was assessed in 1997, 2000, 2004 and 2006. Multivariable mixed linear regression and logistic regression were used in the analyses. RESULTS IDP was characterised by high intake of fresh vegetable, wheat, legume, beverage, offal, rice and whole grain. High IDP intake was associated with poor cognition. In fully adjusted models, across the quartiles of IDP, the odds ratio (95% CI) for poor cognitive function were: 1.00, 1.06 (0.86-1.30), 1.24 (0.99-1.54), and 1.50 (1.17-1.93), respectively. There was a borderline significant interaction between IDP and meat intake (p interaction 0.085). The association between high IDP and poor cognition was only observed among those with no or low intake of meat. With the adjustment of carbohydrate or iron intake, the IDP and cognition association became non-significant. IDP was positively associated with lead intake. The association between IDP and poor cognition was partly mediated by lead intake. CONCLUSIONS Iron-related dietary pattern is associated with poor cognition in Chinese adults, partly due to high intake of carbohydrate, iron and lead.
Collapse
Affiliation(s)
- Zumin Shi
- Human Nutrition Department, College of Health Science, QU Health, Qatar University, Doha, Qatar
| | - Tahra El-Obeid
- Human Nutrition Department, College of Health Science, QU Health, Qatar University, Doha, Qatar
| | - Ming Li
- Centre for Population Health Research, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Xiaoyue Xu
- Faculty of Health, University of Technology Sydney, Sydney, Australia
| | - Jianghong Liu
- University of Pennsylvania School of Nursing, Philadelphia, USA
| |
Collapse
|
35
|
Magnesium Is a Key Player in Neuronal Maturation and Neuropathology. Int J Mol Sci 2019; 20:ijms20143439. [PMID: 31336935 PMCID: PMC6678825 DOI: 10.3390/ijms20143439] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/06/2019] [Accepted: 07/09/2019] [Indexed: 01/05/2023] Open
Abstract
Magnesium (Mg) is the second most abundant cation in mammalian cells, and it is essential for numerous cellular processes including enzymatic reactions, ion channel functions, metabolic cycles, cellular signaling, and DNA/RNA stabilities. Because of the versatile and universal nature of Mg2+, the homeostasis of intracellular Mg2+ is physiologically linked to growth, proliferation, differentiation, energy metabolism, and death of cells. On the cellular and tissue levels, maintaining Mg2+ within optimal levels according to the biological context, such as cell types, developmental stages, extracellular environments, and pathophysiological conditions, is crucial for development, normal functions, and diseases. Hence, Mg2+ is pathologically involved in cancers, diabetes, and neurodegenerative diseases, such as Parkinson's disease, Alzheimer's disease, and demyelination. In the research field regarding the roles and mechanisms of Mg2+ regulation, numerous controversies caused by its versatility and complexity still exist. As Mg2+, at least, plays critical roles in neuronal development, healthy normal functions, and diseases, appropriate Mg2+ supplementation exhibits neurotrophic effects in a majority of cases. Hence, the control of Mg2+ homeostasis can be a candidate for therapeutic targets in neuronal diseases. In this review, recent results regarding the roles of intracellular Mg2+ and its regulatory system in determining the cell phenotype, fate, and diseases in the nervous system are summarized, and an overview of the comprehensive roles of Mg2+ is provided.
Collapse
|
36
|
Dos Santos AB, Bezerra MA, Rocha ME, Barreto GE, Kohlmeier KA. Higher zinc concentrations in hair of Parkinson’s disease are associated with psychotic complications and depression. J Neural Transm (Vienna) 2019; 126:1291-1301. [DOI: 10.1007/s00702-019-02041-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/29/2019] [Indexed: 12/12/2022]
|
37
|
The effects of retinol oral supplementation in 6-hydroxydopamine dopaminergic denervation model in Wistar rats. Neurochem Int 2019; 125:25-34. [DOI: 10.1016/j.neuint.2019.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/02/2019] [Accepted: 02/04/2019] [Indexed: 12/23/2022]
|
38
|
Xie F, Gao X, Yang W, Chang Z, Yang X, Wei X, Huang Z, Xie H, Yue Z, Zhou F, Wang Q. Advances in the Research of Risk Factors and Prodromal Biomarkers of Parkinson's Disease. ACS Chem Neurosci 2019; 10:973-990. [PMID: 30590011 DOI: 10.1021/acschemneuro.8b00520] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in the world. With the advent of an aging population and improving life expectancy worldwide, the number of PD patients is expected to increase, which may lead to an urgent need for effective preventive and diagnostic strategies for PD. Although there is increasing research regarding the pathogenesis of PD, there is limited knowledge regarding the prevention of PD. Moreover, the diagnosis of PD depends on clinical criteria, which require the occurrence of bradykinesia and at least one symptom of rest tremor or rigidity. However, converging evidence from clinical, genetic, neuropathological, and imaging studies suggests the initiation of PD-specific pathology prior to the initial presentation of these classical motor clinical features by years or decades. This latent stage of neurodegeneration in PD is a particularly important stage for effective neuroprotective therapies, which might retard the progression or prevent the onset of PD. Therefore, the exploration of risk factors and premotor biomarkers is not only crucial to the early diagnosis of PD but is also helpful in the development of effective neuroprotection and health care strategies for appropriate populations at risk for PD. In this review, we searched and summarized ∼249 researches and 31 reviews focusing on the risk factors and prodromal biomarkers of PD and published in MEDLINE.
Collapse
Affiliation(s)
- Fen Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Xiaoya Gao
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Wanlin Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Zihan Chang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Xiaohua Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Xiaobo Wei
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Zifeng Huang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Huifang Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Hess Research Center Ninth Floor, New York, New York 10029, United States
| | - Fengli Zhou
- Department of Respiratory Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| |
Collapse
|
39
|
Shi Z, Li M, Wang Y, Liu J, El-Obeid T. High iron intake is associated with poor cognition among Chinese old adults and varied by weight status-a 15-y longitudinal study in 4852 adults. Am J Clin Nutr 2019; 109:109-116. [PMID: 30649164 PMCID: PMC6900563 DOI: 10.1093/ajcn/nqy254] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/24/2018] [Indexed: 02/07/2023] Open
Abstract
Background High body iron status has been shown to be associated with adverse health outcomes. However, the relation between high body iron status, body mass index (BMI), and cognition is still understudied. Objective This study aimed to examine the association between iron intake and cognitive function in Chinese adults and tested the interaction effect of iron intake and BMI on cognition. Design Longitudinal study data from a nationwide sample (n = 4852; age ≥55 y) from the China Health and Nutrition Survey during 1991-2006 were used. Of the participants, 3302 had completed cognitive screening tests in ≥2 surveys. Cognitive function was assessed in 1997, 2000, 2004, and 2006. Dietary iron intake was obtained from a 3-d food record during home visits in 1991, 1993, 1997, 2000, 2004, and 2006. Multivariable mixed linear regression and logistic regression were used. Results The cumulative mean ± SD iron intake in 1997 of tested subjects was 23.7 ± 11.3 mg/d (25.4 mg/d in men and 22.2 mg/d in women). High iron intake was associated with poor cognition. In fully adjusted models, across the quartiles of iron intake the regression coefficients (95% CIs) were 0, -0.39 (-0.77, -0.01), -0.55 (-0.95, -0.15), and -0.90 (-1.33, -0.47), respectively. Comparing extreme quartiles of iron intake (high), the OR (95% CI) for poor cognitive function was 1.30 (1.04, 1.64). There was a significant interaction between iron intake and BMI. The association between high iron intake and poor cognition was stronger among those with a high BMI than those with a low BMI. Among those with a BMI (kg/m2) >24, across quartiles of iron intake the ORs (95% CIs) for poor cognitive function were 1.00, 1.27 (0.91, 1.78), 1.41 (0.97, 2.04), and 2.04 (1.38, 3.01), respectively. Conclusion Higher iron intake is associated with poor cognition in Chinese adults, especially among those with a high BMI.
Collapse
Affiliation(s)
- Zumin Shi
- Human Nutrition Department, Qatar University, Doha, Qatar
| | - Ming Li
- Center for Population Health Research, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Youfa Wang
- Global Health Institute, Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jianghong Liu
- University of Pennsylvania School of Nursing, Philadelphia, PA
| | - Tahra El-Obeid
- Human Nutrition Department, Qatar University, Doha, Qatar
| |
Collapse
|
40
|
Lin L, Yan M, Wu B, Lin R, Zheng Z. Expression of magnesium transporter SLC41A1 in the striatum of 6-hydroxydopamine-induced parkinsonian rats. Brain Res Bull 2018; 142:338-343. [DOI: 10.1016/j.brainresbull.2018.08.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/13/2018] [Accepted: 08/28/2018] [Indexed: 11/30/2022]
|
41
|
Kirkland AE, Sarlo GL, Holton KF. The Role of Magnesium in Neurological Disorders. Nutrients 2018; 10:E730. [PMID: 29882776 PMCID: PMC6024559 DOI: 10.3390/nu10060730] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/30/2018] [Accepted: 06/04/2018] [Indexed: 12/14/2022] Open
Abstract
Magnesium is well known for its diverse actions within the human body. From a neurological standpoint, magnesium plays an essential role in nerve transmission and neuromuscular conduction. It also functions in a protective role against excessive excitation that can lead to neuronal cell death (excitotoxicity), and has been implicated in multiple neurological disorders. Due to these important functions within the nervous system, magnesium is a mineral of intense interest for the potential prevention and treatment of neurological disorders. Current literature is reviewed for migraine, chronic pain, epilepsy, Alzheimer’s, Parkinson’s, and stroke, as well as the commonly comorbid conditions of anxiety and depression. Previous reviews and meta-analyses are used to set the scene for magnesium research across neurological conditions, while current research is reviewed in greater detail to update the literature and demonstrate the progress (or lack thereof) in the field. There is strong data to suggest a role for magnesium in migraine and depression, and emerging data to suggest a protective effect of magnesium for chronic pain, anxiety, and stroke. More research is needed on magnesium as an adjunct treatment in epilepsy, and to further clarify its role in Alzheimer’s and Parkinson’s. Overall, the mechanistic attributes of magnesium in neurological diseases connote the macromineral as a potential target for neurological disease prevention and treatment.
Collapse
Affiliation(s)
- Anna E Kirkland
- Department of Psychology, Behavior, Cognition and Neuroscience Program, American University, Washington, DC 20016, USA.
| | - Gabrielle L Sarlo
- Department of Psychology, Behavior, Cognition and Neuroscience Program, American University, Washington, DC 20016, USA.
| | - Kathleen F Holton
- Department of Health Studies, American University, Washington, DC 20016, USA.
- Center for Behavioral Neuroscience, American University, Washington, DC 20016, USA.
| |
Collapse
|
42
|
Dos Santos AB, Kohlmeier KA, Rocha ME, Barreto GE, Barreto JA, de Souza ACA, Bezerra MA. Hair in Parkinson's disease patients exhibits differences in Calcium, Iron and Zinc concentrations measured by flame atomic absorption spectrometry - FAAS. J Trace Elem Med Biol 2018; 47:134-139. [PMID: 29544800 DOI: 10.1016/j.jtemb.2018.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/29/2018] [Accepted: 02/03/2018] [Indexed: 10/18/2022]
Abstract
Imbalances in metals have emerged as playing a role in the pathophysiology of Parkinson's Disease (PD). Monitoring of metal levels could serve as a biomarker of presence, or future development, of this disease. To this end, we evaluated the ability of flame atomic absorption spectrometry (FAAS) to assess the concentrations of Ca, Fe and Zn in hair of PD patients and to investigate if there was an association with age and disease duration. Hair samples were collected from 26 clinically-diagnosed PD patients, and 33 healthy individuals. Concentrations of Ca and Fe were lower in PD patients when compared to control, whereas, a higher concentration of Zn was detected in PD patients. Levels of Ca and Fe did not vary with age nor with the duration of PD. While Zn did not present variation with duration of the disease, there was a correlation with age as PD patients older than 65 years exhibited a higher concentration of Zn than controls. We conclude that FAAS is useful for detecting differences in Fe, Ca and Zn in hair samples of patients with PD. Hair samples required for this method are easy to collect, and the technique relies on a simple method of digestion of the organic matrix. The ease of use of FAAS should allow for more frequent monitoring of metallic levels in patients in a variety of small clinical situations, thereby offering the hope of allowing systematic tracking of metal levels as the disease progresses, or prior to the defining motor symptoms.
Collapse
Affiliation(s)
- Altair B Dos Santos
- Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Brazil; Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Marcelo E Rocha
- Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Brazil
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Jeferson A Barreto
- Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Brazil
| | | | - Marcos A Bezerra
- Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Brazil.
| |
Collapse
|
43
|
Two mTOR inhibitors, rapamycin and Torin 1, differentially regulate iron-induced generation of mitochondrial ROS. Biometals 2017; 30:975-980. [DOI: 10.1007/s10534-017-0059-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 10/13/2017] [Indexed: 12/21/2022]
|
44
|
The Central Role of Biometals Maintains Oxidative Balance in the Context of Metabolic and Neurodegenerative Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8210734. [PMID: 28751933 PMCID: PMC5511683 DOI: 10.1155/2017/8210734] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/19/2017] [Accepted: 05/28/2017] [Indexed: 12/13/2022]
Abstract
Traditionally, oxidative stress as a biological aspect is defined as an imbalance between the free radical generation and antioxidant capacity of living systems. The intracellular imbalance of ions, disturbance in membrane dynamics, hypoxic conditions, and dysregulation of gene expression are all molecular pathogenic mechanisms closely associated with oxidative stress and underpin systemic changes in the body. These also include aspects such as chronic immune system activation, the impairment of cellular structure renewal, and alterations in the character of the endocrine secretion of diverse tissues. All of these mentioned features are crucial for the correct function of the various tissue types in the body. In the present review, we summarize current knowledge about the common roots of metabolic and neurodegenerative disorders induced by oxidative stress. We discuss these common roots with regard to the way that (1) the respective metal ions are involved in the maintenance of oxidative balance and (2) the metabolic and signaling disturbances of the most important biometals, such as Mg2+, Zn2+, Se2+, Fe2+, or Cu2+, can be considered as the central connection point between the pathogenesis of both types of disorders and oxidative stress.
Collapse
|
45
|
Sturgeon M, Wu P, Cornell R. SLC41A1 and TRPM7 in magnesium homeostasis and genetic risk for Parkinson's disease. JOURNAL OF NEUROLOGY & NEUROMEDICINE 2016; 1:23-28. [PMID: 31187092 PMCID: PMC6557452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder of the central nervous system with a clinically heterogeneous presentation that includes progressive loss of dopaminergic (DA) neurons in the substantia nigra. A minority of PD cases are familial and are caused by mutations in single genes. Most cases, however, are idiopathic PD, a complex multifactorial disorder with environmental and genetic contributors to etiology. Here, we first briefly summarize published evidence that among environmental contributors is dietary deficiency of magnesium. We then review genetic data suggesting that mutations in genes encoding two proteins contributing to cellular magnesium homeostasis confer risk for PD or other Parkinsonian conditions. First, the gene encoding magnesium transporter SLC41A1 is, among others, a candidate for the causative gene in the PARK16 locus where variation is associated with risk for idiopathic Parkinsonian disease. Studies of the function of SLC41A1 in animal models are needed to test whether this protein has a role in maintenance of dopaminergic neurons. Second, in a small study, a hypomorphic variant of TRPM7, a magnesium-permeable channel, was over-represented in cases of amyotrophic lateral sclerosis/ Parkinson dementia complex versus controls from the same ethnic group. Although this association was not detected in a second study, in zebrafish Trpm7 is necessary for terminal differentiation and reduction of toxin-sensitivity in dopaminergic neurons. Overall, epidemiological results support the possibility that mutations in genes relevant to magnesium homeostasis would alter PD risk, but deeper genetic analyses of PD patients are necessary to confirm whether SLC41A1 and TRPM7 are among such genes.
Collapse
Affiliation(s)
- Morgan Sturgeon
- Molecular and Cell Biology Graduate Program, University of Iowa, Iowa City, Iowa
| | - Perry Wu
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa
| | - Robert Cornell
- Molecular and Cell Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa
| |
Collapse
|
46
|
Lin L, Ke Z, Lv M, Lin R, Wu B, Zheng Z. Effects of MgSO 4 and magnesium transporters on 6-hydroxydopamine-induced SH-SY5Y cells. Life Sci 2016; 172:48-54. [PMID: 28011227 DOI: 10.1016/j.lfs.2016.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/04/2016] [Accepted: 12/19/2016] [Indexed: 11/16/2022]
Abstract
AIMS The magnesium ion (Mg2+) fulfils several important functions for living organisms. We investigated whether there is a protective effect of MgSO4 on 6-OHDA-induced neurotoxicity in SH-SY5Y cells, and gained insight into the effects of cellular mRNA and protein expression of the magnesium transporters SLC41A1, NIPA1, MagT1 and CNNM2 on 6-OHDA-induced neurotoxicity. MAIN METHODS The effect of MgSO4 on cell viability in 6-OHDA-treated SH-SY5Y cells was measured using a CCK-8 kit. The mRNA and protein expression of SLC41A1, NIPA1, MagT1, and CNNM2 were detected using reverse transcription-qPCR and Western blot. KEY FINDINGS The results showed that SH-SY5Y cells treated with 25-50μM 6-OHDA for 24h significantly decreased cell viability, while if pre-incubated with 0.125-1mM MgSO4 for 1h before adding 6-OHDA it partially prevented the cell damage. There was a significant decrease in cellular mRNA and protein expression of SLC41A1, NIPA1, MagT1 and CNNM2 in 6-OHDA treated SH-SY5Y cells, and MgSO4 can reverse its decline. SIGNIFICANCE Our results suggest that MgSO4 may protect SH-SY5Y cells against 6-OHDA-induced cell injury and that gene expression of SLC41A1, NIPA1, MagT1, and CNNM2 might be involved in dopaminergic neurons.
Collapse
Affiliation(s)
- Ling Lin
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou 350122, China; Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China
| | - Zili Ke
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China
| | - Meiqi Lv
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China
| | - Renxi Lin
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China
| | - Bin Wu
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China
| | - Zhihong Zheng
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou 350122, China; Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
47
|
Preciados M, Yoo C, Roy D. Estrogenic Endocrine Disrupting Chemicals Influencing NRF1 Regulated Gene Networks in the Development of Complex Human Brain Diseases. Int J Mol Sci 2016; 17:E2086. [PMID: 27983596 PMCID: PMC5187886 DOI: 10.3390/ijms17122086] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/21/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
During the development of an individual from a single cell to prenatal stages to adolescence to adulthood and through the complete life span, humans are exposed to countless environmental and stochastic factors, including estrogenic endocrine disrupting chemicals. Brain cells and neural circuits are likely to be influenced by estrogenic endocrine disruptors (EEDs) because they strongly dependent on estrogens. In this review, we discuss both environmental, epidemiological, and experimental evidence on brain health with exposure to oral contraceptives, hormonal therapy, and EEDs such as bisphenol-A (BPA), polychlorinated biphenyls (PCBs), phthalates, and metalloestrogens, such as, arsenic, cadmium, and manganese. Also we discuss the brain health effects associated from exposure to EEDs including the promotion of neurodegeneration, protection against neurodegeneration, and involvement in various neurological deficits; changes in rearing behavior, locomotion, anxiety, learning difficulties, memory issues, and neuronal abnormalities. The effects of EEDs on the brain are varied during the entire life span and far-reaching with many different mechanisms. To understand endocrine disrupting chemicals mechanisms, we use bioinformatics, molecular, and epidemiologic approaches. Through those approaches, we learn how the effects of EEDs on the brain go beyond known mechanism to disrupt the circulatory and neural estrogen function and estrogen-mediated signaling. Effects on EEDs-modified estrogen and nuclear respiratory factor 1 (NRF1) signaling genes with exposure to natural estrogen, pharmacological estrogen-ethinyl estradiol, PCBs, phthalates, BPA, and metalloestrogens are presented here. Bioinformatics analysis of gene-EEDs interactions and brain disease associations identified hundreds of genes that were altered by exposure to estrogen, phthalate, PCBs, BPA or metalloestrogens. Many genes modified by EEDs are common targets of both 17 β-estradiol (E2) and NRF1. Some of these genes are involved with brain diseases, such as Alzheimer's Disease (AD), Parkinson's Disease, Huntington's Disease, Amyotrophic Lateral Sclerosis, Autism Spectrum Disorder, and Brain Neoplasms. For example, the search of enriched pathways showed that top ten E2 interacting genes in AD-APOE, APP, ATP5A1, CALM1, CASP3, GSK3B, IL1B, MAPT, PSEN2 and TNF-underlie the enrichment of the Kyoto Encyclopedia of Genes and Genomes (KEGG) AD pathway. With AD, the six E2-responsive genes are NRF1 target genes: APBB2, DPYSL2, EIF2S1, ENO1, MAPT, and PAXIP1. These genes are also responsive to the following EEDs: ethinyl estradiol (APBB2, DPYSL2, EIF2S1, ENO1, MAPT, and PAXIP1), BPA (APBB2, EIF2S1, ENO1, MAPT, and PAXIP1), dibutyl phthalate (DPYSL2, EIF2S1, and ENO1), diethylhexyl phthalate (DPYSL2 and MAPT). To validate findings from Comparative Toxicogenomics Database (CTD) curated data, we used Bayesian network (BN) analysis on microarray data of AD patients. We observed that both gender and NRF1 were associated with AD. The female NRF1 gene network is completely different from male human AD patients. AD-associated NRF1 target genes-APLP1, APP, GRIN1, GRIN2B, MAPT, PSEN2, PEN2, and IDE-are also regulated by E2. NRF1 regulates targets genes with diverse functions, including cell growth, apoptosis/autophagy, mitochondrial biogenesis, genomic instability, neurogenesis, neuroplasticity, synaptogenesis, and senescence. By activating or repressing the genes involved in cell proliferation, growth suppression, DNA damage/repair, apoptosis/autophagy, angiogenesis, estrogen signaling, neurogenesis, synaptogenesis, and senescence, and inducing a wide range of DNA damage, genomic instability and DNA methylation and transcriptional repression, NRF1 may act as a major regulator of EEDs-induced brain health deficits. In summary, estrogenic endocrine disrupting chemicals-modified genes in brain health deficits are part of both estrogen and NRF1 signaling pathways. Our findings suggest that in addition to estrogen signaling, EEDs influencing NRF1 regulated communities of genes across genomic and epigenomic multiple networks may contribute in the development of complex chronic human brain health disorders.
Collapse
Affiliation(s)
- Mark Preciados
- Department of Environmental & Occupational Health, Florida International University, Miami, FL 33199, USA.
| | - Changwon Yoo
- Department of Biostatistics, Florida International University, Miami, FL 33199, USA.
| | - Deodutta Roy
- Department of Environmental & Occupational Health, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
48
|
Komiya Y, Runnels LW. TRPM channels and magnesium in early embryonic development. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2016; 59:281-8. [PMID: 26679946 DOI: 10.1387/ijdb.150196lr] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Magnesium (Mg(2+)) is the second most abundant cellular cation and is essential for all stages of life, from the early embryo to adult. Mg(2+) deficiency causes or contributes to many human diseases, including migraine headaches, Parkinson's disease, Alzheimer's disease, hypotension, type 2 diabetes mellitus and cardiac arrhythmias. Although the concentration of Mg(2+) in the extracellular environment can vary significantly, the total intracellular Mg(2+) concentration is actively maintained within a relatively narrow range (14 - 20 mM) via tight, yet poorly understood, regulation of intracellular Mg(2+)by Mg(2+) transporters and Mg(2+)-permeant ion channels. Recent studies have continued to add to the growing number of Mg(2+) transporters and ion channels involved in Mg(2+) homeostasis, including TRPM6 and TRPM7, members of the transient receptor potential (TRP) ion channel family. Mutations in TRPM6, including amino acid substitutions that prevent its heterooligomerization with TRPM7, occur in the rare autosomal-recessive disease hypomagnesemia with secondary hypocalcemia (HSH). Genetic ablation of either gene in mice results in early embryonic lethality, raising the question of whether these channels' capacity to mediate Mg(2+) influx plays an important role in embryonic development. Here we review what is known of the function of Mg(2+) in early development and summarize recent findings regarding the function of the TRPM6 and TRPM7 ion channels during embryogenesis.
Collapse
Affiliation(s)
- Yuko Komiya
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | | |
Collapse
|
49
|
Nandipati S, Litvan I. Environmental Exposures and Parkinson's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13090881. [PMID: 27598189 PMCID: PMC5036714 DOI: 10.3390/ijerph13090881] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/29/2016] [Accepted: 08/30/2016] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD) affects millions around the world. The Braak hypothesis proposes that in PD a pathologic agent may penetrate the nervous system via the olfactory bulb, gut, or both and spreads throughout the nervous system. The agent is unknown, but several environmental exposures have been associated with PD. Here, we summarize and examine the evidence for such environmental exposures. We completed a comprehensive review of human epidemiologic studies of pesticides, selected industrial compounds, and metals and their association with PD in PubMed and Google Scholar until April 2016. Most studies show that rotenone and paraquat are linked to increased PD risk and PD-like neuropathology. Organochlorines have also been linked to PD in human and laboratory studies. Organophosphates and pyrethroids have limited but suggestive human and animal data linked to PD. Iron has been found to be elevated in PD brain tissue but the pathophysiological link is unclear. PD due to manganese has not been demonstrated, though a parkinsonian syndrome associated with manganese is well-documented. Overall, the evidence linking paraquat, rotenone, and organochlorines with PD appears strong; however, organophosphates, pyrethroids, and polychlorinated biphenyls require further study. The studies related to metals do not support an association with PD.
Collapse
Affiliation(s)
- Sirisha Nandipati
- Department of Neurosciences Movement Disorders Center, University of California, San Diego, CA 92093, USA.
| | - Irene Litvan
- Department of Neurosciences Movement Disorders Center, University of California, San Diego, CA 92093, USA.
| |
Collapse
|
50
|
Coherent and Contradictory Facts, Feats and Fictions Associated with Metal Accumulation in Parkinson's Disease: Epicenter or Outcome, Yet a Demigod Question. Mol Neurobiol 2016; 54:4738-4755. [PMID: 27480264 DOI: 10.1007/s12035-016-0016-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/12/2016] [Indexed: 01/30/2023]
Abstract
Unwarranted exposure due to liberal use of metals for maintaining the lavish life and to achieve the food demand for escalating population along with an incredible boost in the average human life span owing to orchestrated progress in rejuvenation therapy have gradually increased the occurrence of Parkinson's disease (PD). Etiology is albeit elusive; association of PD with metal accumulation has never been overlooked due to noteworthy similitude between metal-exposure symptoms and a few cardinal features of disease. Even though metals are entailed in the vital functions, a hysterical shift, primarily augmentation, escorts the stern nigrostriatal dopaminergic neurodegeneration. An increase in the passage of metals through the blood brain barrier and impaired metabolic activity and elimination system could lead to metal accumulation in the brain, which eventually makes dopaminergic neurons quite susceptible. In the present article, an update on implication of metal accumulation in PD/Parkinsonism has been provided. Moreover, encouraging and paradoxical facts and fictions associated with metal accumulation in PD/Parkinsonism have also been compiled. Systematic literature survey of PD is performed to describe updated information if metal accumulation is an epicenter or merely an outcome. Finally, a perspective on the association of metal accumulation with pesticide-induced Parkinsonism has been explained to unveil the likely impact of the former in the latter.
Collapse
|