1
|
Leuci R, Brunetti L, Tufarelli V, Cerini M, Paparella M, Puvača N, Piemontese L. Role of copper chelating agents: between old applications and new perspectives in neuroscience. Neural Regen Res 2025; 20:751-762. [PMID: 38886940 PMCID: PMC11433910 DOI: 10.4103/nrr.nrr-d-24-00140] [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: 02/02/2024] [Revised: 03/12/2024] [Accepted: 04/03/2024] [Indexed: 06/20/2024] Open
Abstract
The role of copper element has been an increasingly relevant topic in recent years in the fields of human and animal health, for both the study of new drugs and innovative food and feed supplements. This metal plays an important role in the central nervous system, where it is associated with glutamatergic signaling, and it is widely involved in inflammatory processes. Thus, diseases involving copper (II) dyshomeostasis often have neurological symptoms, as exemplified by Alzheimer's and other diseases (such as Parkinson's and Wilson's diseases). Moreover, imbalanced copper ion concentrations have also been associated with diabetes and certain types of cancer, including glioma. In this paper, we propose a comprehensive overview of recent results that show the importance of these metal ions in several pathologies, mainly Alzheimer's disease, through the lens of the development and use of copper chelators as research compounds and potential therapeutics if included in multi-target hybrid drugs. Seeing how copper homeostasis is important for the well-being of animals as well as humans, we shortly describe the state of the art regarding the effects of copper and its chelators in agriculture, livestock rearing, and aquaculture, as ingredients for the formulation of feed supplements as well as to prevent the effects of pollution on animal productions.
Collapse
Affiliation(s)
- Rosalba Leuci
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, Bari, Italy
| | - Leonardo Brunetti
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Tufarelli
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePRe-J), Section of Veterinary Science and Animal Production, University of Bari Aldo Moro, Bari, Italy
| | - Marco Cerini
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, Bari, Italy
| | - Marco Paparella
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, Bari, Italy
| | - Nikola Puvača
- Department of Engineering Management in Biotechnology, Faculty of Economics and Engineering Management in Novi Sad, University Business Academy in Novi Sad, Novi Sad, Serbia
| | - Luca Piemontese
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
2
|
Mizuno D, Kawahara M, Konoha-Mizuno K, Hama R, Ogawara T. The Role of Zinc in the Development of Vascular Dementia and Parkinson's Disease and the Potential of Carnosine as Their Therapeutic Agent. Biomedicines 2024; 12:1296. [PMID: 38927502 PMCID: PMC11201809 DOI: 10.3390/biomedicines12061296] [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: 04/25/2024] [Revised: 05/10/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Synaptic zinc ions (Zn2+) play an important role in the development of vascular dementia (VD) and Parkinson's disease (PD). In this article, we reviewed the current comprehension of the Zn2+-induced neurotoxicity that leads to the pathogenesis of these neuronal diseases. Zn2+-induced neurotoxicity was investigated by using immortalised hypothalamic neurons (GT1-7 cells). This cell line is useful for the development of a rapid and convenient screening system for investigating Zn2+-induced neurotoxicity. GT1-7 cells were also used to search for substances that prevent Zn2+-induced neurotoxicity. Among the tested substances was a protective substance in the extract of Japanese eel (Anguilla japonica), and we determined its structure to be like carnosine (β-alanylhistidine). Carnosine may be a therapeutic drug for VD and PD. Furthermore, we reviewed the molecular mechanisms that involve the role of carnosine as an endogenous protector and its protective effect against Zn2+-induced cytotoxicity and discussed the prospects for the future therapeutic applications of this dipeptide for neurodegenerative diseases and dementia.
Collapse
Affiliation(s)
- Dai Mizuno
- Department of Forensic Medicine, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata-shi 990-9585, Yamagata, Japan; (K.K.-M.); (R.H.); (T.O.)
| | - Masahiro Kawahara
- Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shin-machi, Nishitokyo-shi 202-8585, Tokyo, Japan;
| | - Keiko Konoha-Mizuno
- Department of Forensic Medicine, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata-shi 990-9585, Yamagata, Japan; (K.K.-M.); (R.H.); (T.O.)
| | - Ryoji Hama
- Department of Forensic Medicine, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata-shi 990-9585, Yamagata, Japan; (K.K.-M.); (R.H.); (T.O.)
| | - Terumasa Ogawara
- Department of Forensic Medicine, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata-shi 990-9585, Yamagata, Japan; (K.K.-M.); (R.H.); (T.O.)
| |
Collapse
|
3
|
Fukazawa R, Ishii R, Higashimoto Y, Hanya M, Shimizu Y, Shinomoto M, Fujii A, Mizuno T. Zinc Administration Favorably Affects Prophylactic Therapy-refractory Migraine Attacks: A Case Series. Intern Med 2024; 63:1631-1635. [PMID: 37926551 PMCID: PMC11189699 DOI: 10.2169/internalmedicine.2111-23] [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: 04/11/2023] [Accepted: 09/18/2023] [Indexed: 11/07/2023] Open
Abstract
Notably, certain nutrients are effective in preventing migraine. Nonetheless, zinc replacement therapy for migraine treatment has yet to be explored. We herein report four patients with migraine who were refractory to prophylactic therapy and whose headache frequency and severity improved with zinc supplementation. Zinc administration may be an option for treating patients with prophylaxis-refractory migraine. Further investigation is required to determine the efficacy of zinc replacement therapy as a treatment option for migraine.
Collapse
Affiliation(s)
- Ryosuke Fukazawa
- Department of Neurology, Kyoto Prefectural University of Medicine, Japan
- Department of Neurology, Saiseikai Shiga Hospital, Japan
| | - Ryotaro Ishii
- Department of Neurology, Kyoto Prefectural University of Medicine, Japan
| | - Yuki Higashimoto
- Department of Neurology, Kyoto Prefectural University of Medicine, Japan
- Department of Neurology, Japanese Red Cross Kyoto Daini Hospital, Japan
| | - Misaki Hanya
- Department of Neurology, Kyoto Prefectural University of Medicine, Japan
- Department of Neurology, North Medical Center Kyoto Prefectural University of Medicine, Japan
| | - Yuki Shimizu
- Department of Neurology, Kyoto Prefectural University of Medicine, Japan
- Department of Neurology, Japanese Red Cross Kyoto Daini Hospital, Japan
| | - Makiko Shinomoto
- Department of Neurology, Kyoto Prefectural University of Medicine, Japan
| | - Akihiro Fujii
- Department of Neurology, Saiseikai Shiga Hospital, Japan
| | - Toshiki Mizuno
- Department of Neurology, Kyoto Prefectural University of Medicine, Japan
- Department of Neurology, Japan Community Health Care Organization Kyoto Kuramaguchi Medical Center, Japan
| |
Collapse
|
4
|
Blacher C, Abramov-Harpaz K, Miller Y. Primary Nucleation of Polymorphic α-Synuclein Dimers Depends on Copper Concentrations and Definite Copper-Binding Site. Biomolecules 2024; 14:627. [PMID: 38927031 PMCID: PMC11201572 DOI: 10.3390/biom14060627] [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: 05/01/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
The primary nucleation process of α-synuclein (AS) that forms toxic oligomeric species is the early stage of the pathological cause of Parkinson's disease. It is well-known that copper influences this primary nucleation process. While significant efforts have been made to solve the structures of polymorphic AS fibrils, the structures of AS oligomers and the copper-bound AS oligomers at the molecular level and the effect of copper concentrations on the primary nucleation are elusive. Here, we propose and demonstrate new molecular mechanism pathways of primary nucleation of AS that are tuned by distinct copper concentrations and by a specific copper-binding site. We present the polymorphic AS dimers bound to different copper-binding sites at the atomic resolution in high- and low-copper concentrations, using extensive molecular dynamics simulations. Our results show the complexity of the primary nucleation pathways that rely on the copper concentrations and the copper binding site. From a broader perspective, our study proposes a new strategy to control the primary nucleation of other toxic amyloid oligomers in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Carmia Blacher
- Department of Chemistry, Ben-Gurion University of the Negev, Beér-Sheva 8410501, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beér-Sheva 8410501, Israel
- The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beér-Sheva 8410501, Israel
| | - Karina Abramov-Harpaz
- Department of Chemistry, Ben-Gurion University of the Negev, Beér-Sheva 8410501, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beér-Sheva 8410501, Israel
- The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beér-Sheva 8410501, Israel
| | - Yifat Miller
- Department of Chemistry, Ben-Gurion University of the Negev, Beér-Sheva 8410501, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beér-Sheva 8410501, Israel
- The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beér-Sheva 8410501, Israel
| |
Collapse
|
5
|
An D, Xu Y. Environmental risk factors provoke new thinking for prevention and treatment of dementia with Lewy bodies. Heliyon 2024; 10:e30175. [PMID: 38707435 PMCID: PMC11068646 DOI: 10.1016/j.heliyon.2024.e30175] [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: 01/05/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024] Open
Abstract
In recent years, environmental factors have received attention in the pathogenesis of neurodegenerative diseases. Other than genetic factors, the identification of environmental factors and modifiable risk factors may create opportunities to delay the onset or slow the progression of Lewy body disease. Researchers have made significant progress in understanding environmental and modifiable risk factors over the past 30 years. To date, despite the increasing number of articles assessing risk factors for Lewy body disease, few reviews have focused on their role in its onset. In this review, we reviewed the literature investigating the relationship between Lewy body disease and several environmental and other modifiable factors. We found that some air pollutants, exposure to some metals, and infection with some microorganisms may increase the risk of Lewy body disease. Coffee intake and the Mediterranean diet are protective factors. However, it is puzzling that low educational levels and smoking may have some protective effects. In addition, we proposed specific protocols for subsequent research directions on risk factors for neurodegenerative diseases and improved methods. By conducting additional case-control studies, we could explore the role of these factors in the etiopathogenesis of Lewy body disease, establishing a foundation for strategies aimed at preventing and reducing the onset and burden of the disease.
Collapse
Affiliation(s)
- Dinghao An
- Department of Neurology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Nanjing Neurology Clinical Medical Center, Nanjing, China
| |
Collapse
|
6
|
Zhou QY, Ren C, Li JY, Wang L, Duan Y, Yao RQ, Tian YP, Yao YM. The crosstalk between mitochondrial quality control and metal-dependent cell death. Cell Death Dis 2024; 15:299. [PMID: 38678018 PMCID: PMC11055915 DOI: 10.1038/s41419-024-06691-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Mitochondria are the centers of energy and material metabolism, and they also serve as the storage and dispatch hubs of metal ions. Damage to mitochondrial structure and function can cause abnormal levels and distribution of metal ions, leading to cell dysfunction and even death. For a long time, mitochondrial quality control pathways such as mitochondrial dynamics and mitophagy have been considered to inhibit metal-induced cell death. However, with the discovery of new metal-dependent cell death including ferroptosis and cuproptosis, increasing evidence shows that there is a complex relationship between mitochondrial quality control and metal-dependent cell death. This article reviews the latest research results and mechanisms of crosstalk between mitochondrial quality control and metal-dependent cell death in recent years, as well as their involvement in neurodegenerative diseases, tumors and other diseases, in order to provide new ideas for the research and treatment of related diseases.
Collapse
Affiliation(s)
- Qi-Yuan Zhou
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Chao Ren
- Department of Pulmonary and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing-Yan Li
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lu Wang
- Department of Critical Care Medicine, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yu Duan
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou, 423000, China
| | - Ren-Qi Yao
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
- Medical Innovation Research Division, Translational Medicine Research Center and the Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| | - Ying-Ping Tian
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Yong-Ming Yao
- Medical Innovation Research Division, Translational Medicine Research Center and the Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
7
|
Jäntti H, Jonk S, Gómez Budia M, Ohtonen S, Fagerlund I, Fazaludeen MF, Aakko-Saksa P, Pebay A, Lehtonen Š, Koistinaho J, Kanninen KM, Jalava PI, Malm T, Korhonen P. Particulate matter from car exhaust alters function of human iPSC-derived microglia. Part Fibre Toxicol 2024; 21:6. [PMID: 38360668 PMCID: PMC10870637 DOI: 10.1186/s12989-024-00564-y] [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: 04/20/2023] [Accepted: 01/25/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Air pollution is recognized as an emerging environmental risk factor for neurological diseases. Large-scale epidemiological studies associate traffic-related particulate matter (PM) with impaired cognitive functions and increased incidence of neurodegenerative diseases such as Alzheimer's disease. Inhaled components of PM may directly invade the brain via the olfactory route, or act through peripheral system responses resulting in inflammation and oxidative stress in the brain. Microglia are the immune cells of the brain implicated in the progression of neurodegenerative diseases. However, it remains unknown how PM affects live human microglia. RESULTS Here we show that two different PMs derived from exhausts of cars running on EN590 diesel or compressed natural gas (CNG) alter the function of human microglia-like cells in vitro. We exposed human induced pluripotent stem cell (iPSC)-derived microglia-like cells (iMGLs) to traffic related PMs and explored their functional responses. Lower concentrations of PMs ranging between 10 and 100 µg ml-1 increased microglial survival whereas higher concentrations became toxic over time. Both tested pollutants impaired microglial phagocytosis and increased secretion of a few proinflammatory cytokines with distinct patterns, compared to lipopolysaccharide induced responses. iMGLs showed pollutant dependent responses to production of reactive oxygen species (ROS) with CNG inducing and EN590 reducing ROS production. CONCLUSIONS Our study indicates that traffic-related air pollutants alter the function of human microglia and warrant further studies to determine whether these changes contribute to adverse effects in the brain and on cognition over time. This study demonstrates human iPSC-microglia as a valuable tool to study functional microglial responses to environmental agents.
Collapse
Affiliation(s)
- Henna Jäntti
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steffi Jonk
- Division of Eye and Vision, Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Mireia Gómez Budia
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sohvi Ohtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ilkka Fagerlund
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | | | - Alice Pebay
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Šárka Lehtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Koistinaho
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Katja M Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pasi I Jalava
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Paula Korhonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
8
|
Abd El-Karim SS, Anwar MM, Ahmed NS, Syam YM, Elseginy SA, Aly HF, Younis EA, Khalil WKB, Ahmed KA, Mohammed FF, Rizk M. Discovery of novel benzofuran-based derivatives as acetylcholinesterase inhibitors for the treatment of Alzheimer's disease: Design, synthesis, biological evaluation, molecular docking and 3D-QSAR investigation. Eur J Med Chem 2023; 260:115766. [PMID: 37678141 DOI: 10.1016/j.ejmech.2023.115766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023]
Abstract
A series of novel benzofuran-based compounds 7a-s were designed, synthesized, and investigated in vitro as acetylcholinesterase inhibitors (AChEIs). Compounds 7c and 7e displayed promising inhibitory activity with IC50 values of 0.058 and 0.086 μM in comparison to donepezil with an IC50 value of 0.049 μM. The new molecules' antioxidant evaluation revealed that 7c, 7e, 7j, 7n, and 7q produced the strongest DPPH scavenging activity when compared to vitamin C. As it was the most promising AChEI, compound 7c was selected for further biological evaluation. Acute and chronic toxicity studies exhibited that 7c showed no signs of toxicity or adverse events, no significant differences in the blood profile, and an insignificant difference in hepatic enzymes, glucose, urea, creatinine, and albumin levels in the experimental rat group. Furthermore, 7c did not produce histopathological damage to normal liver, kidney, heart, and brain tissues, ameliorated tissue malonaldehyde (MDA) and glutathione (GSH) levels and reduced the expression levels of the APP and Tau genes in AD rats. Molecular docking results of compounds 7c and 7e showed good binding modes in the active site of the acetylcholinesterase enzyme, which are similar to the native ligand donepezil. 3D-QSAR analysis revealed the importance of the alkyl group in positions 2 and 3 of the phenyl moiety for the activity. Overall, these findings suggested that compound 7c could be deemed a promising candidate for the management of Alzheimer's disease.
Collapse
Affiliation(s)
- Somaia S Abd El-Karim
- Department of Therapeutic Chemistry, National Research Centre, P.O. Box 12262 El-Bohouth St, Cairo, Egypt.
| | - Manal M Anwar
- Department of Therapeutic Chemistry, National Research Centre, P.O. Box 12262 El-Bohouth St, Cairo, Egypt.
| | - Nesreen S Ahmed
- Department of Therapeutic Chemistry, National Research Centre, P.O. Box 12262 El-Bohouth St, Cairo, Egypt
| | - Yasmin M Syam
- Department of Therapeutic Chemistry, National Research Centre, P.O. Box 12262 El-Bohouth St, Cairo, Egypt
| | - Samia A Elseginy
- Green Chemistry Department, Chemical Industries Research Division, National Research Centre, P. O. Box 12622, El-Bohouth St, Dokki, Cairo, Egypt
| | - Hanan F Aly
- Department of Therapeutic Chemistry, National Research Centre, P.O. Box 12262 El-Bohouth St, Cairo, Egypt
| | - Eman A Younis
- Department of Therapeutic Chemistry, National Research Centre, P.O. Box 12262 El-Bohouth St, Cairo, Egypt
| | - Wagdy K B Khalil
- Department of Cell Biology, National Research Centre, P.O. Box 12262 El-Bohouth St, Dokki, Cairo, Egypt
| | - Kawkab A Ahmed
- Pathology Departments, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Faten F Mohammed
- Pathology Departments, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Maha Rizk
- Department of Therapeutic Chemistry, National Research Centre, P.O. Box 12262 El-Bohouth St, Cairo, Egypt
| |
Collapse
|
9
|
Kell DB, Pretorius E. Are fibrinaloid microclots a cause of autoimmunity in Long Covid and other post-infection diseases? Biochem J 2023; 480:1217-1240. [PMID: 37584410 DOI: 10.1042/bcj20230241] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023]
Abstract
It is now well established that the blood-clotting protein fibrinogen can polymerise into an anomalous form of fibrin that is amyloid in character; the resultant clots and microclots entrap many other molecules, stain with fluorogenic amyloid stains, are rather resistant to fibrinolysis, can block up microcapillaries, are implicated in a variety of diseases including Long COVID, and have been referred to as fibrinaloids. A necessary corollary of this anomalous polymerisation is the generation of novel epitopes in proteins that would normally be seen as 'self', and otherwise immunologically silent. The precise conformation of the resulting fibrinaloid clots (that, as with prions and classical amyloid proteins, can adopt multiple, stable conformations) must depend on the existing small molecules and metal ions that the fibrinogen may (and is some cases is known to) have bound before polymerisation. Any such novel epitopes, however, are likely to lead to the generation of autoantibodies. A convergent phenomenology, including distinct conformations and seeding of the anomalous form for initiation and propagation, is emerging to link knowledge in prions, prionoids, amyloids and now fibrinaloids. We here summarise the evidence for the above reasoning, which has substantial implications for our understanding of the genesis of autoimmunity (and the possible prevention thereof) based on the primary process of fibrinaloid formation.
Collapse
Affiliation(s)
- Douglas B Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| |
Collapse
|
10
|
Kamel NN, Aly HF, Fouad GI, Abd El-Karim SS, Anwar MM, Syam YM, Elseginy SA, Ahmed KA, Booles HF, Shalaby MB, Khalil WKB, Sandhir R, Deshwal S, Rizk MZ. Anti-Alzheimer activity of new coumarin-based derivatives targeting acetylcholinesterase inhibition. RSC Adv 2023; 13:18496-18510. [PMID: 37346948 PMCID: PMC10280131 DOI: 10.1039/d3ra02344c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/29/2023] [Indexed: 06/23/2023] Open
Abstract
New 2-oxo-chromene-7-oxymethylene acetohydrazide derivatives 4a-d were designed and synthesized with a variety of bioactive chemical fragments. The newly synthesized compounds were evaluated as acetylcholinesterase (AChE) inhibitors and antioxidant agents in comparison to donepezil and ascorbic acid, respectively. Compound 4c exhibited a promising inhibitory impact with an IC50 value of 0.802 μM and DPPH scavenging activity of 57.14 ± 2.77%. Furthermore, biochemical and haematological studies revealed that compound 4c had no effect on the blood profile, hepatic enzyme levels (AST, ALT, and ALP), or total urea in 4c-treated rats compared to the controls. Moreover, the histopathological studies of 4c-treated rats revealed the normal architecture of the hepatic lobules and renal parenchyma, as well as no histopathological damage in the examined hepatic, kidney, heart, and brain tissues. In addition, an in vivo study investigated the amelioration in the cognitive function of AD-rats treated with 4c through the T-maze and beam balance behavioural tests. Also, 4c detectably ameliorated MDA and GSH, reaching 90.64 and 27.17%, respectively, in comparison to the standard drug (90.64% and 35.03% for MDA and GSH, respectively). The molecular docking study exhibited a good fitting of compound 4c in the active site of the AChE enzyme and a promising safety profile. Compound 4c exhibited a promising anti-Alzheimer's disease efficiency compared to the standard drug donepezil.
Collapse
Affiliation(s)
- Nahla N Kamel
- Department of Therapeutic Chemistry, National Research Centre 12262 El-Bohouth St Cairo Egypt
| | - Hanan F Aly
- Department of Therapeutic Chemistry, National Research Centre 12262 El-Bohouth St Cairo Egypt
| | - Ghadha I Fouad
- Department of Therapeutic Chemistry, National Research Centre 12262 El-Bohouth St Cairo Egypt
| | - Somaia S Abd El-Karim
- Department of Therapeutic Chemistry, National Research Centre 12262 El-Bohouth St Cairo Egypt
| | - Manal M Anwar
- Department of Therapeutic Chemistry, National Research Centre 12262 El-Bohouth St Cairo Egypt
| | - Yasmin M Syam
- Department of Therapeutic Chemistry, National Research Centre 12262 El-Bohouth St Cairo Egypt
| | - Samia A Elseginy
- Green Chemistry Department, Chemical Industries Research Division, National Research Centre P. O. Box 12622 Egypt
| | - Kawkab A Ahmed
- Pathology Departments, Faculty of Veterinary Medicine, Cairo University Giza 12211 Egypt
| | - Hoda F Booles
- Department of Cell Biology, National Research Centre 12262 El-Bohouth St Cairo Egypt
| | - Mohamed B Shalaby
- Toxicology Research Department, Research Institute of Medical Entomology (RIME), General Organization of Teaching Hospitals and Institutes (GOTHI), Ministry of Health and Population (MoHP) Dokki, P. O. Box 12311 Cairo Egypt
| | - Wagdy K B Khalil
- Department of Cell Biology, National Research Centre 12262 El-Bohouth St Cairo Egypt
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University Chandigarh India
| | - Sonam Deshwal
- Department of Biochemistry, Panjab University Chandigarh India
| | - Maha Z Rizk
- Department of Therapeutic Chemistry, National Research Centre 12262 El-Bohouth St Cairo Egypt
| |
Collapse
|
11
|
Kawahara M, Kato-Negishi M, Tanaka KI. Dietary Trace Elements and the Pathogenesis of Neurodegenerative Diseases. Nutrients 2023; 15:2067. [PMID: 37432185 PMCID: PMC10180548 DOI: 10.3390/nu15092067] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 07/12/2023] Open
Abstract
Trace elements such as iron (Fe), zinc (Zn), copper (Cu), and manganese (Mn) are absorbed from food via the gastrointestinal tract, transported into the brain, and play central roles in normal brain functions. An excess of these trace elements often produces reactive oxygen species and damages the brain. Moreover, increasing evidence suggests that the dyshomeostasis of these metals is involved in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease, prion diseases, and Lewy body diseases. The disease-related amyloidogenic proteins can regulate metal homeostasis at the synapses, and thus loss of the protective functions of these amyloidogenic proteins causes neurodegeneration. Meanwhile, metal-induced conformational changes of the amyloidogenic proteins contribute to enhancing their neurotoxicity. Moreover, excess Zn and Cu play central roles in the pathogenesis of vascular-type senile dementia. Here, we present an overview of the intake, absorption, and transport of four essential elements (Fe, Zn, Cu, Mn) and one non-essential element (aluminum: Al) in food and their connections with the pathogenesis of neurodegenerative diseases based on metal-protein, and metal-metal cross-talk.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Midori Kato-Negishi
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Ken-Ichiro Tanaka
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| |
Collapse
|
12
|
Zhang X, Liu J, Wang H. The cGAS-STING-autophagy pathway: Novel perspectives in neurotoxicity induced by manganese exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 315:120412. [PMID: 36240967 DOI: 10.1016/j.envpol.2022.120412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/28/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Chronic high-level heavy metal exposure increases the risk of developing different neurodegenerative diseases. Chronic excessive manganese (Mn) exposure is known to lead to neurodegenerative diseases. In addition, some evidence suggests that autophagy dysfunction plays an important role in the pathogenesis of various neurodegenerative diseases. Over the past decade, the DNA-sensing receptor cyclic GMP-AMP synthase (cGAS) and its downstream signal-efficient interferon gene stimulator (STING), as well as the molecular composition and regulatory mechanisms of this pathway have been well understood. The cGAS-STING pathway has emerged as a crucial mechanism to induce effective innate immune responses by inducing type I interferons in mammalian cells. Moreover, recent studies have found that Mn2+ is the second activator of the cGAS-STING pathway besides dsDNA, and inducing autophagy is a primitive function for the activation of the cGAS-STING pathway. However, overactivation of the immune response can lead to tissue damage. This review discusses the mechanism of neurotoxicity induced by Mn exposure from the cGAS-STING-autophagy pathway. Future work exploiting the cGAS-STING-autophagy pathway may provide a novel perspective for manganese neurotoxicity.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
13
|
Bacchella C, Dell'Acqua S, Nicolis S, Monzani E, Casella L. The reactivity of copper complexes with neuronal peptides promoted by catecholamines and its impact on neurodegeneration. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
14
|
Kawahara M, Tanaka KI, Kato-Negishi M. Crosstalk of copper and zinc in the pathogenesis of vascular dementia. J Clin Biochem Nutr 2022; 71:7-15. [PMID: 35903609 PMCID: PMC9309079 DOI: 10.3164/jcbn.22-40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/12/2022] [Indexed: 11/23/2022] Open
Abstract
Copper and zinc are essential for normal brain functions. Both are localized in presynaptic vesicles and are secreted into synaptic clefts during neuronal excitation. Despite their significance, excesses of copper and zinc are neurotoxic. In particular, excess zinc after transient global ischemia plays a central role in the ischemia-induced neurodegeneration and pathogenesis of vascular type senile dementia. We previously found that sub-lethal concentrations of copper remarkably exacerbated zinc-induced neurotoxicity, and we investigated the molecular pathways of copper-enhanced zinc-induced neurotoxicity. The endoplasmic reticulum stress pathway, the stress-activated protein kinases/c-Jun amino-terminal kinases pathway, and mitochondrial energy production failure were revealed to be involved in the neurodegenerative processes. Regarding the upstream factors of these pathways, we focused on copper-derived reactive oxygen species and the disruption of calcium homeostasis. Because excess copper and zinc may be present in the synaptic clefts during ischemia, it is possible that secreted copper and copper-induced reactive oxygen species may enhance zinc neurotoxicity and eventually contribute to the pathogenesis of vascular type senile dementia.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Ken-Ichiro Tanaka
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Midori Kato-Negishi
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| |
Collapse
|
15
|
Tang Y, Liu L, Nong Q, Guo H, Zhou Q, Wang D, Yin Y, Shi J, He B, Hu L, Jiang G. Sensitive determination of metalloprotein in salt-rich matrices by size exclusion chromatography coupled with inductively coupled plasma-mass spectrometry. J Chromatogr A 2022; 1677:463303. [DOI: 10.1016/j.chroma.2022.463303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/27/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022]
|
16
|
Oblak AL, Kotredes KP, Pandey RS, Reagan AM, Ingraham C, Perkins B, Lloyd C, Baker D, Lin PB, Soni DM, Tsai AP, Persohn SA, Bedwell AA, Eldridge K, Speedy R, Meyer JA, Peters JS, Figueiredo LL, Sasner M, Territo PR, Sukoff Rizzo SJ, Carter GW, Lamb BT, Howell GR. Plcg2M28L Interacts With High Fat/High Sugar Diet to Accelerate Alzheimer's Disease-Relevant Phenotypes in Mice. Front Aging Neurosci 2022; 14:886575. [PMID: 35813947 PMCID: PMC9263289 DOI: 10.3389/fnagi.2022.886575] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
Obesity is recognized as a significant risk factor for Alzheimer's disease (AD). Studies have supported the notion that obesity accelerates AD-related pathophysiology in mouse models of AD. The majority of studies, to date, have focused on the use of early-onset AD models. Here, we evaluate the impact of genetic risk factors on late-onset AD (LOAD) in mice fed with a high fat/high sugar diet (HFD). We focused on three mouse models created through the IU/JAX/PITT MODEL-AD Center. These included a combined risk model with APOE4 and a variant in triggering receptor expressed on myeloid cells 2 (Trem2R47H ). We have termed this model, LOAD1. Additional variants including the M28L variant in phospholipase C Gamma 2 (Plcg2M28L ) and the 677C > T variant in methylenetetrahydrofolate reductase (Mthfr 677C > T ) were engineered by CRISPR onto LOAD1 to generate LOAD1.Plcg2M28L and LOAD1.Mthfr 677C > T . At 2 months of age, animals were placed on an HFD that induces obesity or a control diet (CD), until 12 months of age. Throughout the study, blood was collected to assess the levels of cholesterol and glucose. Positron emission tomography/computed tomography (PET/CT) was completed prior to sacrifice to image for glucose utilization and brain perfusion. After the completion of the study, blood and brains were collected for analysis. As expected, animals fed a HFD, showed a significant increase in body weight compared to those fed a CD. Glucose increased as a function of HFD in females only with cholesterol increasing in both sexes. Interestingly, LOAD1.Plcg2M28L demonstrated an increase in microglia density and alterations in regional brain glucose and perfusion on HFD. These changes were not observed in LOAD1 or LOAD1.Mthfr 677C > T animals fed with HFD. Furthermore, LOAD1.Plcg2M28L but not LOAD1.Mthfr 677C > T or LOAD1 animals showed transcriptomics correlations with human AD modules. Our results show that HFD affects the brain in a genotype-specific manner. Further insight into this process may have significant implications for the development of lifestyle interventions for the treatment of AD.
Collapse
Affiliation(s)
- Adrian L. Oblak
- Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | | | - Ravi S. Pandey
- The Jackson Laboratory, Bar Harbor, ME, United States
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | | | - Cynthia Ingraham
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Bridget Perkins
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Christopher Lloyd
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Deborah Baker
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Peter B. Lin
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Disha M. Soni
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Andy P. Tsai
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Scott A. Persohn
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Amanda A. Bedwell
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Kierra Eldridge
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Rachael Speedy
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Jill A. Meyer
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Johnathan S. Peters
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Lucas L. Figueiredo
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | | | - Paul R. Territo
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Stacey J. Sukoff Rizzo
- Department of Medicine, Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Bruce T. Lamb
- Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | | |
Collapse
|
17
|
de Jesus JR, Galazzi RM, Lopes Júnior CA, Arruda MAZ. Trace element homeostasis in the neurological system after SARS-CoV-2 infection: Insight into potential biochemical mechanisms. J Trace Elem Med Biol 2022; 71:126964. [PMID: 35240553 PMCID: PMC8881805 DOI: 10.1016/j.jtemb.2022.126964] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/10/2022] [Accepted: 02/23/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Several studies have suggested that COVID-19 is a systemic disease that can affect several organs, including the brain. In the brain, specifically, viral infection can cause dyshomeostasis of some trace elements that promote complex biochemical reactions in specialized neurological functions. OBJECTIVE Understand the neurovirulence of SARS-CoV-2 and the relationship between trace elements and neurological disorders after infection, and provide new insights on the drug development for the treatment of SARS-CoV-2 infections. METHODS The main databases were used to search studies published up September 2021, focusing on the role of trace elements during viral infection and on the correct functioning of the brain. RESULTS The imbalance of important trace elements can accelerate SARS-CoV-2 neurovirulence and increase the neurotoxicity since many neurological processes can be associated with the homeostasis of metal and metalloproteins. Some studies involving animals and humans have suggested the synapse as a vulnerable region of the brain to neurological disorders after viral infection. Considering the combined evidence, some mechanisms have been suggested to understand the relationship between neurological disorders and imbalance of trace elements in the brain after viral infection. CONCLUSION Trace elements play important roles in viral infections, such as helping to activate immune cells, produce antibodies, and inhibit virus replication. However, the relationship between trace elements and virus infections is complex since the specific functions of several elements remain largely undefined. Therefore, there is still a lot to be explored to understand the biochemical mechanisms involved between trace elements and viral infections, especially in the brain.
Collapse
Affiliation(s)
- Jemmyson Romário de Jesus
- Research Laboratory in Bionanomaterials, LPbio, Brazil; Chemistry Department, Federal University of Viçosa, UFV, Viçosa, Minas Gerais, Brazil.
| | - Rodrigo Moretto Galazzi
- Analytical Instrumentation Division, Analytik Jena GmbH, an Endress & Hauser Company, São Paulo, SP 04029-901, Brazil.
| | - Cícero Alves Lopes Júnior
- Grupo de Estudos em Bioanalítica - GEBIO, Department of Chemistry, Federal University of Piauí, 64049-550 Teresina, PI, Brazil.
| | - Marco Aurélio Zezzi Arruda
- Spectrometry, Sample Preparation and Mechanization Group, GEPAM, Institute of Chemistry, University of Campinas, UNICAMP, Campinas, Brazil; National Institute of Science and Technology for Bioanalytics, Brazil.
| |
Collapse
|
18
|
Elmaidomy AH, Abdelmohsen UR, Alsenani F, Aly HF, Eldin Shams SG, Younis EA, Ahmed KA, Sayed AM, Owis AI, Afifi N, El Amir D. The anti-Alzheimer potential of Tamarindus indica: an in vivo investigation supported by in vitro and in silico approaches. RSC Adv 2022; 12:11769-11785. [PMID: 35481086 PMCID: PMC9015909 DOI: 10.1039/d2ra01340a] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/30/2022] [Indexed: 01/06/2023] Open
Abstract
Tamarindus indica Linn. (Tamarind, F. Fabaceae) is one of the most widely consumed fruits in the world. A crude extract and different fractions of T. indica (using n-hexane, dichloromethane, ethyl acetate, and n-butanol) were evaluated in vitro with respect to their DPPH scavenging and AchE inhibition activities. The results showed that the dichloromethane and ethyl acetate fractions showed the highest antioxidant activities, with 84.78 and 86.96% DPPH scavenging at 0.10 μg mL-1. The n-hexane, dichloromethane, and ethyl acetate fractions inhibited AchE activity in a dose-dependent manner, and the n-hexane fraction showed the highest inhibition at 20 μg mL-1. The results were confirmed by using n-hexane, dichloromethane, and ethyl acetate fractions in vivo to regress the neurodegenerative features of Alzheimer's dementia in an aluminum-intoxicated rat model. Phytochemical investigations of those three fractions afforded two new diphenyl ether derivative compounds 1-2, along with five known ones (3-7). The structures of the isolated compounds were confirmed via 1D and 2D NMR and HRESIMS analyses. The isolated compounds were subjected to extensive in silico-based investigations to putatively highlight the most probable compounds responsible for the anti-Alzheimer activity of T. indica. Inverse docking studies followed by molecular dynamics simulation (MDS) and binding free energy (ΔG) investigations suggested that both compounds 1 and 2 could be promising AchE inhibitors. The results presented in this study may provide potential dietary supplements for the management of Alzheimer's disease.
Collapse
Affiliation(s)
- Abeer H Elmaidomy
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University Beni-Suef 62514 Egypt
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University Minia 61519 Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University 7 Universities Zone New Minia 61111 Egypt
| | - Faisal Alsenani
- Department of Pharmacognosy, Faculty of Pharmacy, Umm Al-Qura University Makkah 21955 Saudi Arabia
| | - Hanan F Aly
- Therapeutic Chemistry Department, National Research Centre (NRC) El-Bouth St. P.O. 12622 Cairo Egypt
| | - Shams Gamal Eldin Shams
- Therapeutic Chemistry Department, National Research Centre (NRC) El-Bouth St. P.O. 12622 Cairo Egypt
| | - Eman A Younis
- Therapeutic Chemistry Department, National Research Centre (NRC) El-Bouth St. P.O. 12622 Cairo Egypt
| | - Kawkab A Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University Giza 12211 Egypt
| | - Ahmed M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University Beni-Suef 62513 Egypt
| | - Asmaa I Owis
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University Beni-Suef 62514 Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Heliopolis University for Sustainable Development Cairo Egypt
| | - Naglaa Afifi
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University Beni-Suef 62514 Egypt
| | - Dalia El Amir
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University Beni-Suef 62514 Egypt
| |
Collapse
|
19
|
Maung MT, Carlson A, Olea-Flores M, Elkhadragy L, Schachtschneider KM, Navarro-Tito N, Padilla-Benavides T. The molecular and cellular basis of copper dysregulation and its relationship with human pathologies. FASEB J 2021; 35:e21810. [PMID: 34390520 DOI: 10.1096/fj.202100273rr] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/23/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022]
Abstract
Copper (Cu) is an essential micronutrient required for the activity of redox-active enzymes involved in critical metabolic reactions, signaling pathways, and biological functions. Transporters and chaperones control Cu ion levels and bioavailability to ensure proper subcellular and systemic Cu distribution. Intensive research has focused on understanding how mammalian cells maintain Cu homeostasis, and how molecular signals coordinate Cu acquisition and storage within organs. In humans, mutations of genes that regulate Cu homeostasis or facilitate interactions with Cu ions lead to numerous pathologic conditions. Malfunctions of the Cu+ -transporting ATPases ATP7A and ATP7B cause Menkes disease and Wilson disease, respectively. Additionally, defects in the mitochondrial and cellular distributions and homeostasis of Cu lead to severe neurodegenerative conditions, mitochondrial myopathies, and metabolic diseases. Cu has a dual nature in carcinogenesis as a promotor of tumor growth and an inducer of redox stress in cancer cells. Cu also plays role in cancer treatment as a component of drugs and a regulator of drug sensitivity and uptake. In this review, we provide an overview of the current knowledge of Cu metabolism and transport and its relation to various human pathologies.
Collapse
Affiliation(s)
- May T Maung
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Alyssa Carlson
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Monserrat Olea-Flores
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | - Lobna Elkhadragy
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Napoleon Navarro-Tito
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | | |
Collapse
|
20
|
Genetic and environmental factors in Alzheimer's and Parkinson's diseases and promising therapeutic intervention via fecal microbiota transplantation. NPJ Parkinsons Dis 2021; 7:70. [PMID: 34381040 PMCID: PMC8357954 DOI: 10.1038/s41531-021-00213-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 07/23/2021] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases are characterized by neuronal impairment and loss of function, and with the major shared histopathological hallmarks of misfolding and aggregation of specific proteins inside or outside cells. Some genetic and environmental factors contribute to the promotion of the development and progression of neurodegenerative diseases. Currently, there are no effective treatments for neurodegenerative diseases. It has been revealed that bidirectional communication exists between the brain and the gut. The gut microbiota is a changeable and experience-dependent ecosystem and can be modified by genetic and environmental factors. The gut microbiota provides potential therapeutic targets that can be regulated as new interventions for neurodegenerative diseases. In this review, we discuss genetic and environmental risk factors for neurodegenerative diseases, summarize the communication among the components of the microbiota-gut-brain axis, and discuss the treatment strategy of fecal microbiota transplantation (FMT). FMT is a promising treatment for neurodegenerative diseases, and restoration of the gut microbiota to a premorbid state is a novel goal for prevention and treatment strategies.
Collapse
|
21
|
Moreda-Piñeiro J, Cocho JA, Couce ML, Moreda-Piñeiro A, Bermejo-Barrera P. Trace elements in dried blood spots as potential discriminating features for metabolic disorder diagnosis in newborns. Metallomics 2021; 13:6261194. [PMID: 33930140 PMCID: PMC8755940 DOI: 10.1093/mtomcs/mfab018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/26/2021] [Accepted: 04/08/2021] [Indexed: 11/13/2022]
Abstract
Trace elements in dried blood spots (DBSs) from newborns were determined by laser ablation coupled with inductively coupled plasma mass spectrometry, and data were subjected to chemometric evaluation in an attempt to classify healthy newborns and newborns suffering from metabolic disorders. Unsupervised [principal component analysis (PCA) and cluster analysis (CA)] and supervised [linear discriminant analysis (LDA) and soft independent modeling by class analogy (SIMCA)] pattern recognition techniques were used as classification techniques. PCA and CA have shown a clear tendency to form two groups (healthy newborns and newborns suffering from metabolic disorders). LDA and SIMCA have predicted that 90.5% and 83.9% of originally grouped healthy newborn cases were correctly classified by LDA and SIMCA, respectively. In addition, these percentages were 97.6% (LDA) and 80.6% (SIMCA) for DBSs from newborns suffering from metabolic disorders. However, SIMCA has only detected one misclassified DBS from the healthy group, and the lower percentage is attributed to four DBSs from the healthy newborn group and five DBSs from newborns with disorders that were found as belonging to both categories (healthy newborns and newborns with disorders) in the training set. LDA also gave a percentage of grouped maple syrup urine disease (MSUD) cases correctly classified of 100%, although the percentage fells to 66.7% when classifying phenylketonuria (PKU) cases. Finally, essential elements such as Fe, K, Rb, and Zn were found to be matched (correlated) with the concentration of amino acids such as phenylalanine, valine, and leucine, biomarkers linked with MSUD and PKU diseases.
Collapse
Affiliation(s)
- Jorge Moreda-Piñeiro
- Department of Chemistry, Faculty of Sciences, Universidade da Coruña, Grupo Química Analítica Aplicada (QANAP), University Institute of Research in Environmental Studies (IUMA), Centro de Investigaciones Científicas Avanzadas (CICA), Campus de A Coruña, s/n, 15071 A Coruña, Spain
| | - José A Cocho
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases, Department of Pediatrics, University Hospital of Santiago de Compostela, IDIS, CIBERER, A Choupana, s/n, 15706 Santiago de Compostela, Spain
| | - María Luz Couce
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases, Department of Pediatrics, University Hospital of Santiago de Compostela, IDIS, CIBERER, A Choupana, s/n, 15706 Santiago de Compostela, Spain
| | - Antonio Moreda-Piñeiro
- Group of Trace Elements, Speciation and Spectroscopy (GETEE), Strategic Grouping in Materials (AEMAT), Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Chemistry, Universidade de Santiago de Compostela, Avenida das Ciencias, s/n, 15782 Santiago de Compostela, Spain
| | - Pilar Bermejo-Barrera
- Group of Trace Elements, Speciation and Spectroscopy (GETEE), Strategic Grouping in Materials (AEMAT), Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Chemistry, Universidade de Santiago de Compostela, Avenida das Ciencias, s/n, 15782 Santiago de Compostela, Spain
| |
Collapse
|
22
|
Kawahara M, Kato-Negishi M, Tanaka KI. Neurometals in the Pathogenesis of Prion Diseases. Int J Mol Sci 2021; 22:ijms22031267. [PMID: 33525334 PMCID: PMC7866166 DOI: 10.3390/ijms22031267] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 12/29/2022] Open
Abstract
Prion diseases are progressive and transmissive neurodegenerative diseases. The conformational conversion of normal cellular prion protein (PrPC) into abnormal pathogenic prion protein (PrPSc) is critical for its infection and pathogenesis. PrPC possesses the ability to bind to various neurometals, including copper, zinc, iron, and manganese. Moreover, increasing evidence suggests that PrPC plays essential roles in the maintenance of homeostasis of these neurometals in the synapse. In addition, trace metals are critical determinants of the conformational change and toxicity of PrPC. Here, we review our studies and other new findings that inform the current understanding of the links between trace elements and physiological functions of PrPC and the neurotoxicity of PrPSc.
Collapse
|
23
|
Lakey-Beitia J, Burillo AM, Penna GL, Hegde ML, Rao K. Polyphenols as Potential Metal Chelation Compounds Against Alzheimer's Disease. J Alzheimers Dis 2021; 82:S335-S357. [PMID: 32568200 PMCID: PMC7809605 DOI: 10.3233/jad-200185] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease affecting more than 50 million people worldwide. The pathology of this multifactorial disease is primarily characterized by the formation of amyloid-β (Aβ) aggregates; however, other etiological factors including metal dyshomeostasis, specifically copper (Cu), zinc (Zn), and iron (Fe), play critical role in disease progression. Because these transition metal ions are important for cellular function, their imbalance can cause oxidative stress that leads to cellular death and eventual cognitive decay. Importantly, these transition metal ions can interact with the amyloid-β protein precursor (AβPP) and Aβ42 peptide, affecting Aβ aggregation and increasing its neurotoxicity. Considering how metal dyshomeostasis may substantially contribute to AD, this review discusses polyphenols and the underlying chemical principles that may enable them to act as natural chelators. Furthermore, polyphenols have various therapeutic effects, including antioxidant activity, metal chelation, mitochondrial function, and anti-amyloidogenic activity. These combined therapeutic effects of polyphenols make them strong candidates for a moderate chelation-based therapy for AD.
Collapse
Affiliation(s)
- Johant Lakey-Beitia
- Centre for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Clayton, City of Knowledge, Panama
| | - Andrea M. Burillo
- Centre for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Clayton, City of Knowledge, Panama
| | - Giovanni La Penna
- National Research Council, Institute of Chemistry of Organometallic Compounds, Sesto Fiorentino (FI), Italy
| | - Muralidhar L. Hegde
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX, USA
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Weill Medical College of Cornell University, New York, NY, USA
| | - K.S. Rao
- Centre for Neuroscience, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Clayton, City of Knowledge, Panama
- Zhongke Jianlan Medical Institute, Hangzhou, Republic of China
| |
Collapse
|
24
|
de Jesus JR, Arruda MAZ. Unravelling neurological disorders through metallomics-based approaches. Metallomics 2020; 12:1878-1896. [PMID: 33237082 DOI: 10.1039/d0mt00234h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Understanding the biological process involving metals and biomolecules in the brain is essential for establishing the origin of neurological disorders, such as neurodegenerative and psychiatric diseases. From this perspective, this critical review presents recent advances in this topic, showing possible mechanisms involving the disruption of metal homeostasis and the pathogenesis of neurological disorders. We also discuss the main challenges observed in metallomics studies associated with neurological disorders, including those related to sample preparation and analyte quantification.
Collapse
|
25
|
Calabrò M, Rinaldi C, Santoro G, Crisafulli C. The biological pathways of Alzheimer disease: a review. AIMS Neurosci 2020; 8:86-132. [PMID: 33490374 PMCID: PMC7815481 DOI: 10.3934/neuroscience.2021005] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer disease is a progressive neurodegenerative disorder, mainly affecting older people, which severely impairs patients' quality of life. In the recent years, the number of affected individuals has seen a rapid increase. It is estimated that up to 107 million subjects will be affected by 2050 worldwide. Research in this area has revealed a lot about the biological and environmental underpinnings of Alzheimer, especially its correlation with β-Amyloid and Tau related mechanics; however, the precise molecular events and biological pathways behind the disease are yet to be discovered. In this review, we focus our attention on the biological mechanics that may lie behind Alzheimer development. In particular, we briefly describe the genetic elements and discuss about specific biological processes potentially associated with the disease.
Collapse
Affiliation(s)
| | | | | | - Concetta Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| |
Collapse
|
26
|
Disease Ionomics: Understanding the Role of Ions in Complex Disease. Int J Mol Sci 2020; 21:ijms21228646. [PMID: 33212764 PMCID: PMC7697569 DOI: 10.3390/ijms21228646] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
Ionomics is a novel multidisciplinary field that uses advanced techniques to investigate the composition and distribution of all minerals and trace elements in a living organism and their variations under diverse physiological and pathological conditions. It involves both high-throughput elemental profiling technologies and bioinformatic methods, providing opportunities to study the molecular mechanism underlying the metabolism, homeostasis, and cross-talk of these elements. While much effort has been made in exploring the ionomic traits relating to plant physiology and nutrition, the use of ionomics in the research of serious diseases is still in progress. In recent years, a number of ionomic studies have been carried out for a variety of complex diseases, which offer theoretical and practical insights into the etiology, early diagnosis, prognosis, and therapy of them. This review aims to give an overview of recent applications of ionomics in the study of complex diseases and discuss the latest advances and future trends in this area. Overall, disease ionomics may provide substantial information for systematic understanding of the properties of the elements and the dynamic network of elements involved in the onset and development of diseases.
Collapse
|
27
|
Martins AC, Gubert P, Villas Boas GR, Paes MM, Santamaría A, Lee E, Tinkov AA, Bowman AB, Aschner M. Manganese-induced neurodegenerative diseases and possible therapeutic approaches. Expert Rev Neurother 2020; 20:1109-1121. [PMID: 32799578 PMCID: PMC7657997 DOI: 10.1080/14737175.2020.1807330] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and prion disease represent important public health concerns. Exposure to high levels of heavy metals such as manganese (Mn) may contribute to their development. AREAS COVERED In this critical review, we address the role of Mn in the etiology of neurodegenerative diseases and discuss emerging treatments of Mn overload, such as chelation therapy. In addition, we discuss natural and synthetic compounds under development as prospective therapeutics. Moreover, bioinformatic approaches to identify new potential targets and therapeutic substances to reverse the neurodegenerative diseases are discussed. EXPERT OPINION Here, the authors highlight the importance of better understanding the molecular mechanisms of toxicity associated with neurodegenerative diseases, and the role of Mn in these diseases. Additional emphasis should be directed to the discovery of new agents to treat Mn-induced diseases, since present day chelator therapies have limited bioavailability. Furthermore, the authors encourage the scientific community to develop research using libraries of compounds to screen those compounds that show efficacy in regulating brain Mn levels. In addition, bioinformatics may provide novel insight for pathways and clinical treatments associated with Mn-induced neurodegeneration, leading to a new direction in Mn toxicological research.
Collapse
Affiliation(s)
- Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Priscila Gubert
- Department of Biochemistry, Laboratory of Immunopathology Keizo Asami, LIKA, Federal, University of Pernambuco, Recife, Brazil
- Postgraduate Program in Pure and Applied Chemistry, Federal University of Western of Bahia, Bahia, Brazil
| | - Gustavo R Villas Boas
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Marina Meirelles Paes
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32301, USA
| | - Alexey A. Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Yaroslavl State University, Yaroslavl, Russia
- Federal Research Centre of Biological Systems and Agro-Technologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
28
|
Development of Conformational Antibodies to Detect Bcl-xL's Amyloid Aggregates in Metal-Induced Apoptotic Neuroblastoma Cells. Int J Mol Sci 2020; 21:ijms21207625. [PMID: 33076337 PMCID: PMC7589975 DOI: 10.3390/ijms21207625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/06/2020] [Accepted: 10/10/2020] [Indexed: 12/31/2022] Open
Abstract
Bcl-xL, a member of the Bcl-2 family, is a pro-survival protein involved in apoptosis regulation. We have previously reported the ability of Bcl-xL to form various types of fibers, from native to amyloid conformations. Here, we have mimicked the effect of apoptosis-induced caspase activity on Bcl-xL by limited proteolysis using trypsin. We show that cleaved Bcl-xL (ΔN-Bcl-xL) forms fibers that exhibit the features of amyloid structures (BclxLcf37). Moreover, three monoclonal antibodies (mAbs), produced by mouse immunization and directed against ΔN-Bcl-xL or Bcl-xL fibers, were selected and characterized. Our results show that these mAbs specifically target ΔN-Bcl-xL in amyloid fibers in vitro. Upon metal-stress-induced apoptosis, these mAbs are able to detect the presence of Bcl-xL in amyloid aggregates in neuroblastoma SH-SY5Y cell lines. In conclusion, these specific mAbs directed against amyloidogenic conformations of Bcl-xL constitute promising tools for studying, in vitro and in cellulo, the contribution of Bcl-xL in apoptosis. These mAbs may further help in developing new diagnostics and therapies, considering Bcl-xL as a strategic target for treating brain lesions relevant to stroke and neurodegenerative diseases.
Collapse
|
29
|
Andreev A, Erdinc B, Shivaraj K, Schmutz J, Levochkina O, Bhowmik D, Farag F, Money KM, Primavera LH, Gotlieb V, Sahni S. The Association Between Anemia of Chronic Inflammation and Alzheimer's Disease and Related Dementias. J Alzheimers Dis Rep 2020; 4:379-391. [PMID: 33163899 PMCID: PMC7592836 DOI: 10.3233/adr-200178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Dementia is a spectrum of neurological diseases characterized by memory impairment and cognitive decline with the pathogenesis and effective management remaining elusive. Several studies have identified a correlation between anemia and Alzheimer's disease and related dementias (ADRD); however, anemia subtypes and association with ADRD have yet to be studied conclusively. Objective To study an association between ADRD and anemia of chronic inflammation. Methods We conducted a retrospective case-control study of the patients, diagnosed with ADRD at Brookdale Hospital. Pair-wise comparisons between means of controls and cases in terms of iron studies and laboratory results were performed using a Mann-Whitney U test. Pair-wise comparisons between anemia subgroups (moderate and severe) were performed using a Two Sample proportion Z-Test, where for each couple of normally distributed population. Results There was a total of 4,517 (1,274 ADRD group; 3,243 Control group) patients. There was significant difference in hemoglobin 10.15 versus 11.04 [p-value <0.001]. Iron studies showed a significant difference in ferritin 395±488.18 versus 263±1023.4 [p < 0.001], total iron binding capacity 225±84.08 versus 266±82.30 [p < 0.001] and serum iron level 64±39.34 versus 53±41.83 [p < 0.001]. Folic acid and vitamin B12 levels were normal in both groups. Severe and moderate anemia in the ADRD group were respectively 6.2% [95% CI: 4.2-8.4] and 13% [95% CI: 9.8-16.2] higher. Overall, incidence of moderate-to-severe anemia was found to be 19% higher in ADRD group [95% CI: 15.8-22.1]. Conclusion We demonstrated an association between ADRD and anemia of chronic inflammation independent of age, renal function, and HgbA1C levels.
Collapse
Affiliation(s)
- Alexander Andreev
- Brookdale University Hospital Medical Center, Department of Internal Medicine, Brooklyn, NY, USA.,Beth Israel Deaconess Medical Center, Department of Neurology, Boston, MA, USA
| | - Burak Erdinc
- Brookdale University Hospital Medical Center, Department of Internal Medicine, Brooklyn, NY, USA
| | - Kiran Shivaraj
- Brookdale University Hospital Medical Center, Department of Internal Medicine, Brooklyn, NY, USA
| | - Julia Schmutz
- Ross University School of Medicine, Miramar, FL, USA
| | - Olga Levochkina
- Richmond University Medical Center, Department of Psychiatry, Staten Island, NY, USA
| | - Dhrity Bhowmik
- Brookdale University Hospital Medical Center, Department of Internal Medicine, Brooklyn, NY, USA
| | - Fady Farag
- Brookdale University Hospital Medical Center, Department of Internal Medicine, Brooklyn, NY, USA
| | - Kelli M Money
- Beth Israel Deaconess Medical Center, Department of Neurology, Boston, MA, USA
| | | | - Vladimir Gotlieb
- Brookdale University Hospital Medical Center, Department of Internal Medicine, Division of Hematology & Oncology, Brooklyn, NY, USA
| | - Sonu Sahni
- Brookdale University Hospital Medical Center, Department of Internal Medicine, Brooklyn, NY, USA.,Department of Primary Care, Touro College of Osteopathic Medicine, New York, NY, USA.,Department of Research Medicine, NYIT College of Osteopathic Medicine, Glen Head, NY, USA
| |
Collapse
|
30
|
Kawahara M, Sadakane Y, Mizuno K, Kato-Negishi M, Tanaka KI. Carnosine as a Possible Drug for Zinc-Induced Neurotoxicity and Vascular Dementia. Int J Mol Sci 2020; 21:ijms21072570. [PMID: 32272780 PMCID: PMC7177235 DOI: 10.3390/ijms21072570] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence suggests that the metal homeostasis is involved in the pathogenesis of various neurodegenerative diseases including senile type of dementia such as Alzheimer’s disease, dementia with Lewy bodies, and vascular dementia. In particular, synaptic Zn2+ is known to play critical roles in the pathogenesis of vascular dementia. In this article, we review the molecular pathways of Zn2+-induced neurotoxicity based on our and numerous other findings, and demonstrated the implications of the energy production pathway, the disruption of calcium homeostasis, the production of reactive oxygen species (ROS), the endoplasmic reticulum (ER)-stress pathway, and the stress-activated protein kinases/c-Jun amino-terminal kinases (SAPK/JNK) pathway. Furthermore, we have searched for substances that protect neurons from Zn2+-induced neurotoxicity among various agricultural products and determined carnosine (β-alanyl histidine) as a possible therapeutic agent for vascular dementia.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan; (M.K.-N.); (K.T.)
- Correspondence: ; Tel.: +81–42–468–8299
| | - Yutaka Sadakane
- Graduate School of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka 513-8670, Japan;
| | - Keiko Mizuno
- Department of Forensic Medicine, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan;
| | - Midori Kato-Negishi
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan; (M.K.-N.); (K.T.)
| | - Ken-ichiro Tanaka
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan; (M.K.-N.); (K.T.)
| |
Collapse
|
31
|
Kawahara M, Kato-Negishi M, Tanaka KI. Amyloids: Regulators of Metal Homeostasis in the Synapse. Molecules 2020; 25:molecules25061441. [PMID: 32210005 PMCID: PMC7145306 DOI: 10.3390/molecules25061441] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022] Open
Abstract
Conformational changes in amyloidogenic proteins, such as β-amyloid protein, prion proteins, and α-synuclein, play a critical role in the pathogenesis of numerous neurodegenerative diseases, including Alzheimer’s disease, prion disease, and Lewy body disease. The disease-associated proteins possess several common characteristics, including the ability to form amyloid oligomers with β-pleated sheet structure, as well as cytotoxicity, although they differ in amino acid sequence. Interestingly, these amyloidogenic proteins all possess the ability to bind trace metals, can regulate metal homeostasis, and are co-localized at the synapse, where metals are abundantly present. In this review, we discuss the physiological roles of these amyloidogenic proteins in metal homeostasis, and we propose hypothetical models of their pathogenetic role in the neurodegenerative process as the loss of normal metal regulatory functions of amyloidogenic proteins. Notably, these amyloidogenic proteins have the capacity to form Ca2+-permeable pores in membranes, suggestive of a toxic gain of function. Therefore, we focus on their potential role in the disruption of Ca2+ homeostasis in amyloid-associated neurodegenerative diseases.
Collapse
|
32
|
Leng Y, Yu D, Hu Y, Yu SS, Ye Z. Dynamic behaviors of hyperbolic-type memristor-based Hopfield neural network considering synaptic crosstalk. CHAOS (WOODBURY, N.Y.) 2020; 30:033108. [PMID: 32237757 DOI: 10.1063/5.0002076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 06/11/2023]
Abstract
Crosstalk phenomena taking place between synapses can influence signal transmission and, in some cases, brain functions. It is thus important to discover the dynamic behaviors of the neural network infected by synaptic crosstalk. To achieve this, in this paper, a new circuit is structured to emulate the Coupled Hyperbolic Memristors, which is then utilized to simulate the synaptic crosstalk of a Hopfield Neural Network (HNN). Thereafter, the HNN's multi-stability, asymmetry attractors, and anti-monotonicity are observed with various crosstalk strengths. The dynamic behaviors of the HNN are presented using bifurcation diagrams, dynamic maps, and Lyapunov exponent spectrums, considering different levels of crosstalk strengths. Simulation results also reveal that different crosstalk strengths can lead to wide-ranging nonlinear behaviors in the HNN systems.
Collapse
Affiliation(s)
- Yang Leng
- School of Electrical and Power Engineering, China University of Mining and Technology, No. 1 Daxue Road, Xuzhou, Jiangsu 221116, China
| | - Dongsheng Yu
- School of Electrical and Power Engineering, China University of Mining and Technology, No. 1 Daxue Road, Xuzhou, Jiangsu 221116, China
| | - Yihua Hu
- School of Electrical and Power Engineering, China University of Mining and Technology, No. 1 Daxue Road, Xuzhou, Jiangsu 221116, China
| | - Samson Shenglong Yu
- School of Engineering, Deakin University, 75 Pigdons Road, Waurn Ponds, Victoria 3216, Australia
| | - Zongbin Ye
- School of Electrical and Power Engineering, China University of Mining and Technology, No. 1 Daxue Road, Xuzhou, Jiangsu 221116, China
| |
Collapse
|
33
|
Bisaglia M, Bubacco L. Copper Ions and Parkinson's Disease: Why Is Homeostasis So Relevant? Biomolecules 2020; 10:biom10020195. [PMID: 32013126 PMCID: PMC7072482 DOI: 10.3390/biom10020195] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/20/2020] [Accepted: 01/28/2020] [Indexed: 12/18/2022] Open
Abstract
The involvement of copper in numerous physiological processes makes this metal ion essential for human life. Alterations in copper homeostasis might have deleterious consequences, and several neurodegenerative disorders, including Parkinson’s disease (PD), have been associated with impaired copper levels. In the present review, we describe the molecular mechanisms through which copper can exert its toxicity, by considering how it can interfere with other cellular processes known to play a role in PD, such as dopamine metabolism, oxidative stress, and α-synuclein aggregation. The recent experimental evidence that associates copper deficiency and the formation of superoxide dismutase 1 (SOD1) aggregates with the progression of PD is also discussed together with its therapeutic implication. Overall, the recent discoveries described in this review show how either copper deficiency or excessive levels can promote detrimental effects, highlighting the importance of preserving copper homeostasis and opening unexplored therapeutic avenues in the definition of novel disease-modifying drugs.
Collapse
|
34
|
Jing MJ, Liu K, Liu C, Yan DY, Ma Z, Wang C, Deng Y, Liu W, Xu B. Protective effects of trehalose against Mn-induced α-synuclein oligomerization in mice: Involvement of oxidative stress and autophagy. ENVIRONMENTAL TOXICOLOGY 2020; 35:55-65. [PMID: 31497924 DOI: 10.1002/tox.22842] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 06/10/2023]
Abstract
Overexposure to manganese (Mn) is widely known to induce alpha-synuclein (α-Syn) oligomerization, which has been attributed to the oxidative damage of α-Syn protein. Trehalose has been shown to induce autophagy and serve as a chemical chaperone, but little information has been reported about its effect on Mn-induced α-Syn oligomerization. In this study, we investigate whether trehalose can effectively interfere with Mn-induced α-Syn oligomerization, using different concentrations of trehalose (2% and 4% (g/vol [mL])) in a mouse model of manganism. After 6 weeks of exposure to Mn, both oxidative stress and autophagy were activated and resulted in α-Syn oligomerization and neuronal cell damage in the mouse brain tissue. Our results also revealed that pretreatment with trehalose significantly reduced the oxidative damage to α-Syn protein and increased autophagy activation. These findings clearly demonstrated that trehalose can relieve Mn-induced α-Syn oligomerization and neuronal cell damage through its anti-oxidative and autophagy-inducing effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
35
|
Bacchella C, Nicolis S, Dell'Acqua S, Rizzarelli E, Monzani E, Casella L. Membrane Binding Strongly Affecting the Dopamine Reactivity Induced by Copper Prion and Copper/Amyloid-β (Aβ) Peptides. A Ternary Copper/Aβ/Prion Peptide Complex Stabilized and Solubilized in Sodium Dodecyl Sulfate Micelles. Inorg Chem 2019; 59:900-912. [PMID: 31869218 DOI: 10.1021/acs.inorgchem.9b03153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The combination between dyshomeostatic levels of catecholamine neurotransmitters and redox-active metals such as copper and iron exacerbates the oxidative stress condition that typically affects neurodegenerative diseases. We report a comparative study of the oxidative reactivity of copper complexes with amyloid-β (Aβ40) and the prion peptide fragment 76-114 (PrP76-114), containing the high-affinity binding site, toward dopamine and 4-methylcatechol, in aqueous buffer and in sodium dodecyl sulfate micelles, as a model membrane environment. The competitive oxidative and covalent modifications undergone by the peptides were also evaluated. The high binding affinity of Cu/peptide to micelles and lipid membranes leads to a strong reduction (Aβ40) and quenching (PrP76-114) of the oxidative efficiency of the binary complexes and to a stabilization and redox silencing of the ternary complex CuII/Aβ40/PrP76-114, which is highly reactive in solution. The results improve our understanding of the pathological and protective effects associated with these complexes, depending on the physiological environment.
Collapse
Affiliation(s)
- Chiara Bacchella
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Stefania Nicolis
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Simone Dell'Acqua
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Enrico Rizzarelli
- Istituto di Biostrutture e Bioimmagini , Consiglio Nazionale delle Ricerche , Via P. Gaifami 18 , 95125 Catania , Italy
| | - Enrico Monzani
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Luigi Casella
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| |
Collapse
|
36
|
Sheng J, Olrichs NK, Geerts WJ, Kaloyanova DV, Helms JB. Metal ions and redox balance regulate distinct amyloid-like aggregation pathways of GAPR-1. Sci Rep 2019; 9:15048. [PMID: 31636315 PMCID: PMC6803662 DOI: 10.1038/s41598-019-51232-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/26/2019] [Indexed: 01/10/2023] Open
Abstract
Members of the CAP superfamily (Cysteine-rich secretory proteins, Antigen 5, and Pathogenesis-Related 1 proteins) are characterized by the presence of a structurally conserved CAP domain. The common structure-function relationship of this domain is still poorly understood. In this study, we unravel specific molecular mechanisms modulating the quaternary structure of the mammalian CAP protein GAPR-1 (Golgi-Associated plant Pathogenesis-Related protein 1). Copper ions are shown to induce a distinct amyloid-like aggregation pathway of GAPR-1 in the presence of heparin. This involves an immediate shift from native multimers to monomers which are prone to form amyloid-like fibrils. The Cu2+-induced aggregation pathway is independent of a conserved metal-binding site and involves the formation of disulfide bonds during the nucleation process. The elongation process occurs independently of the presence of Cu2+ ions, and amyloid-like aggregation can proceed under oxidative conditions. In contrast, the Zn2+-dependent aggregation pathway was found to be independent of cysteines and was reversible upon removal of Zn2+ ions. Together, our results provide insight into the regulation of the quaternary structure of GAPR-1 by metal ions and redox homeostasis with potential implications for regulatory mechanisms of other CAP proteins.
Collapse
Affiliation(s)
- Jie Sheng
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Nick K Olrichs
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Willie J Geerts
- Biomolecular Imaging, Bijvoet Center, Utrecht University, Utrecht, The Netherlands
| | - Dora V Kaloyanova
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - J Bernd Helms
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
37
|
Capucciati A, Galliano M, Bubacco L, Zecca L, Casella L, Monzani E, Nicolis S. Neuronal Proteins as Targets of 3-Hydroxykynurenine: Implications in Neurodegenerative Diseases. ACS Chem Neurosci 2019; 10:3731-3739. [PMID: 31298828 DOI: 10.1021/acschemneuro.9b00265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The neurotoxic activity of the tryptophan metabolite 3-hydroxykynurenine (3OHKyn) in neurodegenerative disorders, such as Parkinson's and Alzheimer's diseases, is related to oxidative stress and 3OHKyn interaction with cellular proteins. The pattern of protein modification induced by 3OHKyn involves the nucleophilic side chains of Cys, His, and Lys residues, similarly to the one promoted by dopamine and other catecholamines. In the present work, we have analyzed the reactivity of 3OHKyn toward the neuronal targets α-synuclein (and its N-terminal fragments 1-6 and 1-15) and amyloid-β peptides (1-16 and 1-28) and characterized the resulting conjugates through spectrometric (LC-MS/MS) and spectroscopic (UV-vis, fluorescence, NMR) techniques. The amino acid residues of α-synuclein and amyloid-β peptides involved in derivatizations by 3OHKyn and its autoxidation products (belonging to the xanthommatin family) are Lys and His, respectively. The pattern of protein modification is expanded in the conjugates obtained in the presence of the metal ions copper(II) or iron(III), reflecting a more oxidizing environment that in addition to adducts with protein/peptide residues also favors the fragmentation of the protein. These results open the perspective to using the 3OHKyn-protein/peptide synthetic conjugates to explore their competence to activate microglia cell cultures as well as to unravel their role in neuroinflammatory conditions.
Collapse
Affiliation(s)
| | - Monica Galliano
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Luigi Bubacco
- Department of Biology, University of Padova, 35121 Padova, Italy
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, 20090 Segrate, Milano, Italy
| | - Luigi Casella
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy
| | - Enrico Monzani
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy
| | - Stefania Nicolis
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
38
|
Mezzaroba L, Alfieri DF, Colado Simão AN, Vissoci Reiche EM. The role of zinc, copper, manganese and iron in neurodegenerative diseases. Neurotoxicology 2019; 74:230-241. [PMID: 31377220 DOI: 10.1016/j.neuro.2019.07.007] [Citation(s) in RCA: 300] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 01/16/2023]
Abstract
Metals are involved in different pathophysiological mechanisms associated with neurodegenerative diseases (NDDs), including Alzheimer's disease (AD), Parkinson's disease (PD) and multiple sclerosis (MS). The aim of this study was to review the effects of the essential metals zinc (Zn), copper (Cu), manganese (Mn) and iron (Fe) on the central nervous system (CNS), as well as the mechanisms involved in their neurotoxicity. Low levels of Zn as well as high levels of Cu, Mn, and Fe participate in the activation of signaling pathways of the inflammatory, oxidative and nitrosative stress (IO&NS) response, including nuclear factor kappa B and activator protein-1. The imbalance of these metals impairs the structural, regulatory, and catalytic functions of different enzymes, proteins, receptors, and transporters. Neurodegeneration occurs via association of metals with proteins and subsequent induction of aggregate formation creating a vicious cycle by disrupting mitochondrial function, which depletes adenosine triphosphate and induces IO&NS, cell death by apoptotic and/or necrotic mechanisms. In AD, at low levels, Zn suppresses β-amyloid-induced neurotoxicity by selectively precipitating aggregation intermediates; however, at high levels, the binding of Zn to β-amyloid may enhance formation of fibrillar β-amyloid aggregation, leading to neurodegeneration. High levels of Cu, Mn and Fe participate in the formation α-synuclein aggregates in intracellular inclusions, called Lewy Body, that result in synaptic dysfunction and interruption of axonal transport. In PD, there is focal accumulation of Fe in the substantia nigra, while in AD a diffuse accumulation of Fe occurs in various regions, such as cortex and hippocampus, with Fe marginally increased in the senile plaques. Zn deficiency induces an imbalance between T helper (Th)1 and Th2 cell functions and a failure of Th17 down-regulation, contributing to the pathogenesis of MS. In MS, elevated levels of Fe occur in certain brain regions, such as thalamus and striatum, which may be due to inflammatory processes disrupting the blood-brain barrier and attracting Fe-rich macrophages. Delineating the specific mechanisms by which metals alter redox homeostasis is essential to understand the pathophysiology of AD, PD, and MS and may provide possible new targets for their prevention and treatment of the patients affected by these NDDs.
Collapse
Affiliation(s)
- Leda Mezzaroba
- Laboratory of Applied Immunology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil; Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil
| | - Daniela Frizon Alfieri
- Laboratory of Applied Immunology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil
| | - Andrea Name Colado Simão
- Laboratory of Applied Immunology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil; Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil
| | - Edna Maria Vissoci Reiche
- Laboratory of Applied Immunology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil; Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil.
| |
Collapse
|
39
|
Two-Dimensional Polyacrylamide Gel Electrophoresis for Metalloprotein Analysis Based on Differential Chemical Structure Recognition by CBB Dye. Sci Rep 2019; 9:10566. [PMID: 31332224 PMCID: PMC6646366 DOI: 10.1038/s41598-019-46955-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/08/2019] [Indexed: 02/05/2023] Open
Abstract
In an effort to develop an analytical method capable of finding new metalloproteins, this is the first report of a new diagonal gel electrophoresis method to isolate and identify metalloproteins, based on the molecular recognition of holo- and apo-metalloproteins (metalbound and -free forms, respectively) by CBB G-250 dye and employing metal ion contaminant sweeping-blue native-polyacrylamide gel electrophoresis (MICS-BN-PAGE). The difference in electrophoretic mobilities between holo- and apo-forms was exaggerated as a result of interactions between the metalloproteins and the dye with no metal ion dissociation. The different binding modes of proteins with CBB G-250 dye, primarily related to hydrogen bonding, were confirmed by capillary zone electrophoresis (CZE) and molecular docking simulations. Due to in-gel holo/apo conversion between the first and second dimensions of PAGE, holo-metalloproteins in the original sample were completely isolated as spots off the diagonal line in the second dimension of PAGE. To prove the high efficiency of this method for metalloprotein analysis, we successfully identified a copper-binding protein from a total bacterial soluble extract for the first time.
Collapse
|
40
|
Monzani E, Nicolis S, Dell'Acqua S, Capucciati A, Bacchella C, Zucca FA, Mosharov EV, Sulzer D, Zecca L, Casella L. Dopamin, oxidativer Stress und Protein‐Chinonmodifikationen bei Parkinson und anderen neurodegenerativen Erkrankungen. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201811122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Enrico Monzani
- Department of ChemistryUniversity of Pavia 27100 Pavia Italien
| | | | | | | | | | - Fabio A. Zucca
- Institute of Biomedical TechnologiesNational Research Council of Italy Segrate (Mailand) Italien
| | - Eugene V. Mosharov
- Department of PsychiatryColumbia University Medical CenterNew York State Psychiatric Institute New York NY USA
- Departments Neurology, PharmacologyColumbia University Medical Center New York NY USA
| | - David Sulzer
- Department of PsychiatryColumbia University Medical CenterNew York State Psychiatric Institute New York NY USA
- Departments Neurology, PharmacologyColumbia University Medical Center New York NY USA
| | - Luigi Zecca
- Institute of Biomedical TechnologiesNational Research Council of Italy Segrate (Mailand) Italien
- Department of PsychiatryColumbia University Medical CenterNew York State Psychiatric Institute New York NY USA
| | - Luigi Casella
- Department of ChemistryUniversity of Pavia 27100 Pavia Italien
| |
Collapse
|
41
|
Monzani E, Nicolis S, Dell'Acqua S, Capucciati A, Bacchella C, Zucca FA, Mosharov EV, Sulzer D, Zecca L, Casella L. Dopamine, Oxidative Stress and Protein-Quinone Modifications in Parkinson's and Other Neurodegenerative Diseases. Angew Chem Int Ed Engl 2019; 58:6512-6527. [PMID: 30536578 DOI: 10.1002/anie.201811122] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/10/2018] [Indexed: 12/19/2022]
Abstract
Dopamine (DA) is the most important catecholamine in the brain, as it is the most abundant and the precursor of other neurotransmitters. Degeneration of nigrostriatal neurons of substantia nigra pars compacta in Parkinson's disease represents the best-studied link between DA neurotransmission and neuropathology. Catecholamines are reactive molecules that are handled through complex control and transport systems. Under normal conditions, small amounts of cytosolic DA are converted to neuromelanin in a stepwise process involving melanization of peptides and proteins. However, excessive cytosolic or extraneuronal DA can give rise to nonselective protein modifications. These reactions involve DA oxidation to quinone species and depend on the presence of redox-active transition metal ions such as iron and copper. Other oxidized DA metabolites likely participate in post-translational protein modification. Thus, protein-quinone modification is a heterogeneous process involving multiple DA-derived residues that produce structural and conformational changes of proteins and can lead to aggregation and inactivation of the modified proteins.
Collapse
Affiliation(s)
- Enrico Monzani
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| | - Stefania Nicolis
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| | | | | | - Chiara Bacchella
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| | - Fabio A Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate (Milano), Italy
| | - Eugene V Mosharov
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA.,Departments of Neurology and Pharmacology, Columbia University Medical Center, New York, NY, USA
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate (Milano), Italy.,Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Luigi Casella
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| |
Collapse
|
42
|
Young TR, Wedd AG, Xiao Z. Evaluation of Cu(i) binding to the E2 domain of the amyloid precursor protein - a lesson in quantification of metal binding to proteins via ligand competition. Metallomics 2019; 10:108-119. [PMID: 29215101 DOI: 10.1039/c7mt00291b] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The extracellular domain E2 of the amyloid precursor protein (APP) features a His-rich metal-binding site (denoted as the M1 site). In conjunction with surrounding basic residues, the site participates in interactions with components of the extracellular matrix including heparins, a class of negatively charged polysaccharide molecules of varying length. This work studied the chemistry of Cu(i) binding to APP E2 with the probe ligands Bcs, Bca, Fz and Fs. APP E2 forms a stable Cu(i)-mediated ternary complex with each of these anionic ligands. The complex with Bca was selected for isolation and characterization and was demonstrated, by native ESI-MS analysis, to have the stoichiometry E2 : Cu(i) : Bca = 1 : 1 : 1. Formation of these ternary complexes is specific for the APP E2 domain and requires Cu(i) coordination to the M1 site. Mutation of the M1 site was consistent with the His ligands being part of the E2 ligand set. It is likely that interactions between the negatively charged probe ligands and a positively charged patch on the surface of APP E2 are one aspect of the generation of the stable ternary complexes. Their formation prevented meaningful quantification of the affinity of Cu(i) binding to the M1 site with these probe ligands. However, the ternary complexes are disrupted by heparin, allowing reliable determination of a picomolar Cu(i) affinity for the E2/heparin complex with the Fz or Bca probe ligands. This is the first documented example of the formation of stable ternary complexes between a Cu(i) binding protein and a probe ligand. The ready disruption of the complexes by heparin identified clear 'tell-tale' signs for diagnosis of ternary complex formation and allowed a systematic review of conditions and criteria for reliable determination of affinities for metal binding via ligand competition. This study also provides new insights into a potential correlation of APP functions regulated by copper binding and heparin interaction.
Collapse
Affiliation(s)
- Tessa R Young
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | |
Collapse
|
43
|
Yamada Y, Prosser RA. Copper in the suprachiasmatic circadian clock: A possible link between multiple circadian oscillators. Eur J Neurosci 2018; 51:47-70. [PMID: 30269387 DOI: 10.1111/ejn.14181] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 09/05/2018] [Accepted: 09/17/2018] [Indexed: 01/07/2023]
Abstract
The mammalian circadian clock in the suprachiasmatic nucleus (SCN) is very robust, able to coordinate our daily physiological and behavioral rhythms with exquisite accuracy. Simultaneously, the SCN clock is highly sensitive to environmental timing cues such as the solar cycle. This duality of resiliency and sensitivity may be sustained in part by a complex intertwining of three cellular oscillators: transcription/translation, metabolic/redox, and membrane excitability. We suggest here that one of the links connecting these oscillators may be forged from copper (Cu). Cellular Cu levels are highly regulated in the brain and peripherally, and Cu affects cellular metabolism, redox state, cell signaling, and transcription. We have shown that both Cu chelation and application induce nighttime phase shifts of the SCN clock in vitro and that these treatments affect glutamate, N-methyl-D-aspartate receptor, and associated signaling processes differently. More recently we found that Cu induces mitogen-activated protein kinase-dependent phase shifts, while the mechanisms by which Cu removal induces phase shifts remain unclear. Lastly, we have found that two Cu transporters are expressed in the SCN, and that one of these transporters (ATP7A) exhibits a day/night rhythm. Our results suggest that Cu homeostasis is tightly regulated in the SCN, and that changes in Cu levels may serve as a time cue for the circadian clock. We discuss these findings in light of the existing literature and current models of multiple coupled circadian oscillators in the SCN.
Collapse
Affiliation(s)
- Yukihiro Yamada
- Department of Biochemistry & Cellular and Molecular Biology, NeuroNET Research Center, University of Tennessee, Knoxville, Tennessee
| | - Rebecca A Prosser
- Department of Biochemistry & Cellular and Molecular Biology, NeuroNET Research Center, University of Tennessee, Knoxville, Tennessee
| |
Collapse
|
44
|
Abeyawardhane DL, Fernández RD, Heitger DR, Crozier MK, Wolver JC, Lucas HR. Copper Induced Radical Dimerization of α-Synuclein Requires Histidine. J Am Chem Soc 2018; 140:17086-17094. [DOI: 10.1021/jacs.8b08947] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | - Ricardo D. Fernández
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Denver R. Heitger
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Madeleine K. Crozier
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Julia C. Wolver
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Heather R. Lucas
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| |
Collapse
|
45
|
Aliès B, Borghesani V, Noël S, Sayen S, Guillon E, Testemale D, Faller P, Hureau C. Mutations of Histidine 13 to Arginine and Arginine 5 to Glycine Are Responsible for Different Coordination Sites of Zinc(II) to Human and Murine Peptides. Chemistry 2018; 24:14233-14241. [PMID: 29978925 DOI: 10.1002/chem.201802759] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Indexed: 01/21/2023]
Abstract
Because mice and rats do not naturally develop Alzheimer's disease, genetically modified animals are required to study this pathology. This striking difference in terms of disease onset could be due to three alterations in the murine sequence (R5G, Y10F and H13R) of the amyloid-β peptide with respect to the human counterpart. Whether the metal-ion binding properties of the murine peptide are at the origin of such different amyloidogenicity of the two peptides is still an open question. Herein, the main zinc binding site to the murine amyloid-β at physiological pH has been determined through the combination of several spectroscopic and analytical methods applied to a series of six peptides with one or two of the key mutations. These results have been compared with the zinc binding site encountered in the human peptide. A coordination mechanism that demonstrates the importance of the H13R and R5G mutations in the different zinc environments present in the murine and human peptides is proposed. The nature of the minor zinc species present at physiological pH is also suggested for both peptides. Finally, the biological relevance and fallouts of the differences determined in zinc binding to human versus murine amyloid-β are also discussed.
Collapse
Affiliation(s)
- Bruno Aliès
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France.,Current address: Université de Bordeaux, ChemBioPharm INSERM U1212 CNRS UMR 5320, 33076, Bordeaux, France
| | | | - Sabrina Noël
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Stephanie Sayen
- Université Reims Champagne Ardenne, Institut de Chimie Moléculaire de Reims (ICMR), UMR 7312 CNRS-URCA, Moulin de la Housse, BP 1039, 51687, Reims Cedex 2, France
| | - Emmanuel Guillon
- Université Reims Champagne Ardenne, Institut de Chimie Moléculaire de Reims (ICMR), UMR 7312 CNRS-URCA, Moulin de la Housse, BP 1039, 51687, Reims Cedex 2, France
| | - Denis Testemale
- Univ. Grenoble Alpes, CNRS, Grenoble INP, Institut Néel, 38000, Grenoble, France.,BM30B/FAME, ESRF, The European Synchrotron, 71 avenue des Martyrs, 38000, Grenoble, France
| | - Peter Faller
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France.,Current address: Institut de Chimie, UMR 7177 CNRS-Université de Strasbourg, 4 rue Blaise Pascal, Institut Le Bel, 67008, Strasbourg, France
| | | |
Collapse
|
46
|
Liu JL, Fan YG, Yang ZS, Wang ZY, Guo C. Iron and Alzheimer's Disease: From Pathogenesis to Therapeutic Implications. Front Neurosci 2018; 12:632. [PMID: 30250423 PMCID: PMC6139360 DOI: 10.3389/fnins.2018.00632] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/22/2018] [Indexed: 12/28/2022] Open
Abstract
As people age, iron deposits in different areas of the brain may impair normal cognitive function and behavior. Abnormal iron metabolism generates hydroxyl radicals through the Fenton reaction, triggers oxidative stress reactions, damages cell lipids, protein and DNA structure and function, and ultimately leads to cell death. There is an imbalance in iron homeostasis in Alzheimer's disease (AD). Excessive iron contributes to the deposition of β-amyloid and the formation of neurofibrillary tangles, which in turn, promotes the development of AD. Therefore, iron-targeted therapeutic strategies have become a new direction. Iron chelators, such as desferoxamine, deferiprone, deferasirox, and clioquinol, have received a great deal of attention and have obtained good results in scientific experiments and some clinical trials. Given the limitations and side effects of the long-term application of traditional iron chelators, alpha-lipoic acid and lactoferrin, as self-synthesized naturally small molecules, have shown very intriguing biological activities in blocking Aβ-aggregation, tauopathy and neuronal damage. Despite a lack of evidence for any clinical benefits, the conjecture that therapeutic chelation, with a special focus on iron ions, is a valuable approach for treating AD remains widespread.
Collapse
Affiliation(s)
- Jun-Lin Liu
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yong-Gang Fan
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Zheng-Sheng Yang
- Department of Dermatology, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, China.,Key Laboratory of Medical Cell Biology of Ministry of Education, Institute of Health Sciences, China Medical University, Shenyang, China
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
47
|
Sadakane Y, Kawahara M. Implications of Metal Binding and Asparagine Deamidation for Amyloid Formation. Int J Mol Sci 2018; 19:ijms19082449. [PMID: 30126231 PMCID: PMC6121660 DOI: 10.3390/ijms19082449] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 12/17/2022] Open
Abstract
Increasing evidence suggests that amyloid formation, i.e., self-assembly of proteins and the resulting conformational changes, is linked with the pathogenesis of various neurodegenerative disorders such as Alzheimer’s disease, prion diseases, and Lewy body diseases. Among the factors that accelerate or inhibit oligomerization, we focus here on two non-genetic and common characteristics of many amyloidogenic proteins: metal binding and asparagine deamidation. Both reflect the aging process and occur in most amyloidogenic proteins. All of the amyloidogenic proteins, such as Alzheimer’s β-amyloid protein, prion protein, and α-synuclein, are metal-binding proteins and are involved in the regulation of metal homeostasis. It is widely accepted that these proteins are susceptible to non-enzymatic posttranslational modifications, and many asparagine residues of these proteins are deamidated. Moreover, these two factors can combine because asparagine residues can bind metals. We review the current understanding of these two common properties and their implications in the pathogenesis of these neurodegenerative diseases.
Collapse
Affiliation(s)
- Yutaka Sadakane
- Graduate School of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka 513-8670, Japan.
| | - Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo, Tokyo 202-8585, Japan.
| |
Collapse
|
48
|
Zhang W, Liu Y, Hureau C, Robert A, Meunier B. N 4 -Tetradentate Chelators Efficiently Regulate Copper Homeostasis and Prevent ROS Production Induced by Copper-Amyloid-β 1-16. Chemistry 2018; 24:7825-7829. [PMID: 29687932 DOI: 10.1002/chem.201801387] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Indexed: 01/15/2023]
Abstract
The disruption of copper homeostasis and the oxidative stress induced by Cu-amyloids are crucial features of Alzheimer's disease pathology. The copper specific N4 -tetradendate ligands TDMQ20 and 1 are able to fully inhibit in vitro the aerobic oxidation of ascorbate induced by Cu-Aβ1-16 , even in the presence of 100 molar equivalents of ZnII with respect to CuII , whereas other ligands with N2 O2 or N3 O2 coordination spheres failed to do so. This essential result indicates that, in addition to metal selectivity, the coordination sphere of copper chelators should exhibit a N4 -tetradendate motif to be able to reduce an oxidative stress in the zinc-rich physiological environment of brain. The N4 -scaffolds of these two aminoquinoline-based ligands, TDMQ20 or 1, suitable for a square-planar coordination of copper(II), allowed them to enhance both the selectivity for copper and the ability to reduce the oxidative stress induced by copper-amyloid in a zinc-rich environment.
Collapse
Affiliation(s)
- Weixin Zhang
- School of Chemical Engineering and Light Industry, Guangdong University of Technology (GDUT), Higher Education Mega Center, 100 Waihuan Xi road, Panyu District, Guangzhou, 510006, P. R. China.,Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, BP 44099, 31077, Toulouse cedex 4, France.,Université de Toulouse, 31077, Toulouse Cedex 4, France
| | - Yan Liu
- School of Chemical Engineering and Light Industry, Guangdong University of Technology (GDUT), Higher Education Mega Center, 100 Waihuan Xi road, Panyu District, Guangzhou, 510006, P. R. China
| | - Christelle Hureau
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, BP 44099, 31077, Toulouse cedex 4, France.,Université de Toulouse, 31077, Toulouse Cedex 4, France
| | - Anne Robert
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, BP 44099, 31077, Toulouse cedex 4, France.,Université de Toulouse, 31077, Toulouse Cedex 4, France
| | - Bernard Meunier
- School of Chemical Engineering and Light Industry, Guangdong University of Technology (GDUT), Higher Education Mega Center, 100 Waihuan Xi road, Panyu District, Guangzhou, 510006, P. R. China.,Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, BP 44099, 31077, Toulouse cedex 4, France.,Université de Toulouse, 31077, Toulouse Cedex 4, France
| |
Collapse
|
49
|
Kawahara M, Tanaka KI, Kato-Negishi M. Zinc, Carnosine, and Neurodegenerative Diseases. Nutrients 2018; 10:E147. [PMID: 29382141 PMCID: PMC5852723 DOI: 10.3390/nu10020147] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 01/02/2023] Open
Abstract
Zinc (Zn) is abundantly present in the brain, and accumulates in the synaptic vesicles. Synaptic Zn is released with neuronal excitation, and plays essential roles in learning and memory. Increasing evidence suggests that the disruption of Zn homeostasis is involved in various neurodegenerative diseases including Alzheimer's disease, a vascular type of dementia, and prion diseases. Our and other numerous studies suggest that carnosine (β-alanyl histidine) is protective against these neurodegenerative diseases. Carnosine is an endogenous dipeptide abundantly present in the skeletal muscles and in the brain, and has numerous beneficial effects such as antioxidant, metal chelating, anti-crosslinking, and anti-glycation activities. The complex of carnosine and Zn, termed polaprezinc, is widely used for Zn supplementation therapy and for the treatment of ulcers. Here, we review the link between Zn and these neurodegenerative diseases, and focus on the neuroprotective effects of carnosine. We also discuss the carnosine level in various foodstuffs and beneficial effects of dietary supplementation of carnosine.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan.
| | - Ken-Ichiro Tanaka
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan.
| | - Midori Kato-Negishi
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan.
| |
Collapse
|
50
|
Hohberger B, Chaudhri MA, Michalke B, Lucio M, Nowomiejska K, Schlötzer-Schrehardt U, Grieb P, Rejdak R, Jünemann AGM. Levels of aqueous humor trace elements in patients with open-angle glaucoma. J Trace Elem Med Biol 2018; 45:150-155. [PMID: 29173472 DOI: 10.1016/j.jtemb.2017.10.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/22/2017] [Accepted: 10/10/2017] [Indexed: 01/02/2023]
Abstract
PURPOSE Trace elements might play a role in the complex multifactorial pathogenesis of open-angle glaucoma. The aim of this study was to analyze concentrations of trace elements in aqueous humor samples of patients with primary open-angle glaucoma (POAG) and pseudoexfoliation glaucoma (PEXG). PATIENTS AND METHODS Thirty-three aqueous humor samples were obtained from patients undergoing cataract surgery: 12 patients with POAG (age 65.3±10.50, female 8, male 4), 10 patients with PEXG (age 65.9±11.27, female 6, male 4) and 11 patients without glaucoma (age 69.5±13.70, female 7, male 4) serving as controls. Aqueous humor levels of cadmium, iron, manganese, cobalt, copper and zinc were measured by Flow-Injection-Inductively-Coupled-Plasma-Mass-Spectrometry (FI-ICP-MS). RESULTS From the statistical evaluation, we observed that patients with POAG had significantly higher aqueous humor levels of zinc (p=0.006) compared to controls. Increased aqueous humor levels of zinc were also observed in patients with PEXG in relation to control (p=0.0006). For iron we observed a significantly reduction in PEXG compared to control (p=0.002) and a significant difference between POAG and PEXG (p=0.0091). No significant differences were observed in aqueous humor levels of manganese, cobalt, copper, cadmium between glaucoma and control patients. No differences were seen for iron (POAG vs. controls). Analysis of trace element ratios was added. CONCLUSION Significant differences in aqueous humor levels of zinc and iron between glaucoma and control patients support the hypothesis that these trace elements are involved in the pathogenesis of open-angle glaucoma.
Collapse
Affiliation(s)
- Bettina Hohberger
- Department of Ophthalmology, University of Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany,.
| | - M Anwar Chaudhri
- Institute of Surface Science and Corrosion (LKO), Department of Materials Science and Engineering, Technische Fakultät, Friedrich-Alexander Universität Erlangen-Nürnberg, Martensstr. 7, 91058 Erlangen, Germany,.
| | - Bernhard Michalke
- Helmholtz Zentrum München - German Research Center for Environmental Health, Research Unit Analytical BioGeoChemistry, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany,.
| | - Marianna Lucio
- Helmholtz Zentrum München - German Research Center for Environmental Health, Research Unit Analytical BioGeoChemistry, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany,.
| | - Katarzyna Nowomiejska
- Department of General Ophthalmology, Medical University of Lublin, Aleje Racławickie 1, 20-950 Lublin, Poland,.
| | | | - Pawel Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland,.
| | - Robert Rejdak
- Department of General Ophthalmology, Medical University of Lublin, Aleje Racławickie 1, 20-950 Lublin, Poland,.
| | - Anselm G M Jünemann
- Department of Ophthalmology, University of Rostock, Doberaner Straße 140, 18057 Rostock, Germany,.
| |
Collapse
|