1
|
Zhu J, Chen H, Wu J, Li S, Lin W, Wang N, Bai L. Ferroptosis in Glaucoma: A Promising Avenue for Therapy. Adv Biol (Weinh) 2024; 8:e2300530. [PMID: 38411382 DOI: 10.1002/adbi.202300530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/08/2024] [Indexed: 02/28/2024]
Abstract
Glaucoma, a blind-leading disease largely since chronic pathological intraocular high pressure (ph-IOP). Hitherto, it is reckoned incurable for irreversible neural damage and challenges in managing IOP. Thus, it is significant to develop neuroprotective strategies. Ferroptosis, initially identified as an iron-dependent regulated death that triggers Fenton reactions and culminates in lipid peroxidation (LPO), has emerged as a focal point in multiple tumors and neurodegenerative diseases. Researches show that iron homeostasis play critical roles in the optic nerve (ON) and retinal ganglion cells (RGCs), suggesting targeted treatments could be effective. In glaucoma, apart from neural lesions, disrupted metal balance and increased oxidative stress in trabecular meshwork (TM) are observed. These disturbances lead to extracellular matrix excretion disorders, known as sclerotic mechanisms, resulting in refractory blockages. Importantly, oxidative stress, a significant downstream effect of ferroptosis, is also a key factor in cell senescence. It plays a crucial role in both the etiology and risk of glaucoma. Moreover, ferroptosis also induces non-infectious inflammation, which exacerbate glaucomatous injury. Therefore, the relevance of ferroptosis in glaucoma is extensive and multifaceted. In this review, the study delves into the current understanding of ferroptosis mechanisms in glaucoma, aiming to provide clues to inform clinical therapeutic practices.
Collapse
Affiliation(s)
- Jingyun Zhu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, No.1023-1063, Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Hui Chen
- Department of Geriatrics, Hospital of Traditional Chinese Medicine Affiliated to Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, Sichuan, 646000, China
| | - Jian Wu
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, No. 8, East Chongwenmennei Street, Dongcheng District, Beijing, 100005, China
| | - Sen Li
- Department of Spinal Surgery, Drum Tower Hospital, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing, Jiangsu, 210008, China
| | - Wanying Lin
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, No.1023-1063, Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Ningli Wang
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, No. 8, East Chongwenmennei Street, Dongcheng District, Beijing, 100005, China
| | - Lang Bai
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, No.1023-1063, Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
2
|
Zhong G, Wang X, Li J, Xie Z, Wu Q, Chen J, Wang Y, Chen Z, Cao X, Li T, Liu J, Wang Q. Insights Into the Role of Copper in Neurodegenerative Diseases and the Therapeutic Potential of Natural Compounds. Curr Neuropharmacol 2024; 22:1650-1671. [PMID: 38037913 PMCID: PMC11284712 DOI: 10.2174/1570159x22666231103085859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/23/2023] [Accepted: 06/05/2023] [Indexed: 12/02/2023] Open
Abstract
Neurodegenerative diseases encompass a collection of neurological disorders originating from the progressive degeneration of neurons, resulting in the dysfunction of neurons. Unfortunately, effective therapeutic interventions for these diseases are presently lacking. Copper (Cu), a crucial trace element within the human body, assumes a pivotal role in various biological metabolic processes, including energy metabolism, antioxidant defense, and neurotransmission. These processes are vital for the sustenance, growth, and development of organisms. Mounting evidence suggests that disrupted copper homeostasis contributes to numerous age-related neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), Wilson's disease (WD), Menkes disease (MD), prion diseases, and multiple sclerosis (MS). This comprehensive review investigates the connection between the imbalance of copper homeostasis and neurodegenerative diseases, summarizing pertinent drugs and therapies that ameliorate neuropathological changes, motor deficits, and cognitive impairments in these conditions through the modulation of copper metabolism. These interventions include Metal-Protein Attenuating Compounds (MPACs), copper chelators, copper supplements, and zinc salts. Moreover, this review highlights the potential of active compounds derived from natural plant medicines to enhance neurodegenerative disease outcomes by regulating copper homeostasis. Among these compounds, polyphenols are particularly abundant. Consequently, this review holds significant implications for the future development of innovative drugs targeting the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Wang
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiaqi Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhouyuan Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiqing Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaxin Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiyun Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziying Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianyao Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinman Liu
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
3
|
Hu S, Yang B, Li B, Fan Q, Wu T, Li S, Wang D, Yang T, Song Z. RNA-Seq Analysis Reveals Potential Neuroprotective Mechanisms of Pachymic Acid Toward Iron-Induced Oxidative Stress and Cell Death. Cell Transplant 2024; 33:9636897231218382. [PMID: 38314688 PMCID: PMC10845991 DOI: 10.1177/09636897231218382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/15/2023] [Accepted: 11/18/2023] [Indexed: 02/07/2024] Open
Abstract
Iron dysregulation is a crucial factor in the development of neurological diseases, leading to the accumulation of reactive oxygen species (ROS) and oxidative stress, triggering inflammatory responses, and ultimately causing neurological impairment. Pachymic acid (PA) is an active ingredient extracted from the medicinal fungus Poria cocos, which has been reported with multiple pharmacological effects, including anti-inflammatory, anti-ischemia/reperfusion, and anticancer actions. In this study, we test whether PA have neuroprotection effect aganist ferrous ions induced toxicity in SH-SY5Y cells. It was found that pre-treatment with PA reduced intracellular ROS levels, increased mitochondrial membrane potential, and protected cells from apoptotic death. RNA-seq and qRT-PCR results indicated that PA can regulate the key genes IL1B, CXCL8, CCL7, and LRP1 on the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, such as NF-κB signaling pathway, IL-17 signaling pathway, to prevent Fe2+-induced apoptotic cell death. Our research indicated that PA has potential therapeutic effects on the neuroprotection by regulating neuroinflammation and oxidative stress damage.
Collapse
Affiliation(s)
- Shuyang Hu
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Baili Yang
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Binbin Li
- Department of Rehabilitation Medicine, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou, China
| | - Qianqian Fan
- Department of Rehabilitation Medicine, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou, China
| | - Tinglong Wu
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Shanshan Li
- College of Pharmacy, Hainan Medical University, Haikou, China
| | - Dong Wang
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Tao Yang
- College of Pharmacy, Hainan Medical University, Haikou, China
| | - Zhenghua Song
- Department of Rehabilitation Medicine, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou, China
| |
Collapse
|
4
|
Zhang B, Burke R. Copper homeostasis and the ubiquitin proteasome system. Metallomics 2023; 15:7055959. [PMID: 36822629 PMCID: PMC10022722 DOI: 10.1093/mtomcs/mfad010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/03/2023] [Indexed: 02/25/2023]
Abstract
Copper is involved in many physiological pathways and important biological processes as a cofactor of several copper-dependent enzymes. Given the requirement for copper and its potential toxicity, intracellular copper levels are tightly controlled. Disturbances of human copper homeostasis are characterized by disorders of copper overload (Wilson's disease) or copper deficiency (Menkes disease). The maintenance of cellular copper levels involves numerous copper transporters and copper chaperones. Recently, accumulating evidence has revealed that components of the ubiquitin proteasome system (UPS) participate in the posttranslational regulation of these proteins, suggesting that they might play a role in maintaining copper homeostasis. Cellular copper levels could also affect the activity of the UPS, indicating that copper homeostasis and the UPS are interdependent. Copper homeostasis and the UPS are essential to the integrity of normal brain function and while separate links between neurodegenerative diseases and UPS inhibition/copper dyshomeostasis have been extensively reported, there is growing evidence that these two networks might contribute synergistically to the occurrence of neurodegenerative diseases. Here, we review the role of copper and the UPS in the development of Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, and discuss the genetic interactions between copper transporters/chaperones and components of the UPS.
Collapse
Affiliation(s)
- Bichao Zhang
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Richard Burke
- School of Biological Sciences, Monash University, Clayton 3800, Victoria, Australia
| |
Collapse
|
5
|
Yang Y, Liu Y, Zhu J, Song S, Huang Y, Zhang W, Sun Y, Hao J, Yang X, Gao Q, Ma Z, Zhang J, Gu X. Neuroinflammation-mediated mitochondrial dysregulation involved in postoperative cognitive dysfunction. Free Radic Biol Med 2022; 178:134-146. [PMID: 34875338 DOI: 10.1016/j.freeradbiomed.2021.12.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/20/2021] [Accepted: 12/02/2021] [Indexed: 12/22/2022]
Abstract
Neuroinflammation following peripheral surgery is a pivotal pathogenic mechanism of postoperative cognitive dysfunction (POCD). However, the key site of inflammation-mediated neural damage remains unclear. Impaired mitochondrial function is a vital feature of degenerated neurons. Dynamin-related protein 1 (DRP1), a crucial regulator of mitochondrial dynamics, has been shown to play an essential role in synapse formation. Here, we designed experiments to assess whether Drp1-regulated mitochondrial dynamics and function are involved in the pathological processes of POCD and elucidate its relationship with neuroinflammation. Aged mice were subjected to experimental laparotomy under isoflurane anesthesia. Primary neurons and SH-SY5Y cells were exposed to tumor necrosis factor (TNF). We found an increase in Drp1 activation as well as mitochondrial fragmentation both in the hippocampus of mice after surgery and primary neurons after TNF exposure. Pretreatment with Mdivi-1, a Drp1 specific inhibitor, reduced this mitochondrial fragmentation. Drp1 knockdown with small interfering RNA blocked TNF-induced mitochondrial fragmentation in SH-SY5Y cells. However, the application of Mdivi-1 exhibited a negative impact on mitochondrial function and neurite growth in primary neurons. Calcineurin activity was increased in primary neurons after TNF exposure and contributed to the Drp1 activation. The calcineurin inhibitor FK506 exhibited a Drp1-independent function that mitigated mitochondrial dysfunction. Finally, we found that FK506 pretreatment ameliorated the neurite growth in neurons treated with TNF and the learning ability of mice after surgery. Overall, our research indicated a crucial role of mitochondrial function in the pathological processes of POCD, and neuronal metabolic modulation may represent a novel and important target for POCD.
Collapse
Affiliation(s)
- Yan Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Yue Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Jixiang Zhu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Shiyu Song
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu Province, 210093, China
| | - Yulin Huang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Wei Zhang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Yu'e Sun
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Jing Hao
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Xuli Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu Province, 210093, China
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China.
| | - Juan Zhang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China.
| |
Collapse
|
6
|
Cheng H, Yang B, Ke T, Li S, Yang X, Aschner M, Chen P. Mechanisms of Metal-Induced Mitochondrial Dysfunction in Neurological Disorders. TOXICS 2021; 9:142. [PMID: 34204190 PMCID: PMC8235163 DOI: 10.3390/toxics9060142] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 01/31/2023]
Abstract
Metals are actively involved in multiple catalytic physiological activities. However, metal overload may result in neurotoxicity as it increases formation of reactive oxygen species (ROS) and elevates oxidative stress in the nervous system. Mitochondria are a key target of metal-induced toxicity, given their role in energy production. As the brain consumes a large amount of energy, mitochondrial dysfunction and the subsequent decrease in levels of ATP may significantly disrupt brain function, resulting in neuronal cell death and ensuing neurological disorders. Here, we address contemporary studies on metal-induced mitochondrial dysfunction and its impact on the nervous system.
Collapse
Affiliation(s)
- Hong Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China; (H.C.); (X.Y.)
| | - Bobo Yang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China;
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China; (H.C.); (X.Y.)
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou 545006, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| |
Collapse
|
7
|
Hoffman JF, Kalinich JF. Effects of Incubation of Human Brain Microvascular Endothelial Cells and Astrocytes with Pyridostigmine Bromide, DEET, or Permethrin in the Absence or Presence of Metal Salts. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17228336. [PMID: 33187257 PMCID: PMC7696739 DOI: 10.3390/ijerph17228336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022]
Abstract
Gulf War Illness (GWI) is a chronic, multi-symptom illness suffered by over one-third of American military veterans who served in the Persian Gulf War between 1990 and 1991. No current single-exposure scenario accounts for all the symptoms observed in GWI, and instead may be due to a multi-exposure scenario. As a larger effort to understand how one category of multi-exposure scenarios of organic compounds such as nerve gas prophylactic pyridostigmine bromide, or insecticides/pesticides such as N,N-diethyl-m-toluamide (DEET) and permethrin, plus heavy metals found in inhaled dust particles (Al, Fe, Ni, Sr, DU, Co, Cu, Mn, and Zn) might play a role in neural aspects of GWI, we begin this initial study to examine the toxicity and oxidative damage markers of human brain endothelial cell and human astrocyte cell cultures in response to these compounds. A battery of cytotoxicity assessments, including the MTT assay, Neutral Red uptake, and direct microscopic observation, was used to determine a non-toxic dose of the test compounds. After testing a wide range of doses of each compound, we chose a sub-toxic dose of 10 µM for the three organic compounds and 1 µM for the nine metals of interest for co-exposure experiments on cell cultures and examined an array of oxidative stress-response markers including nitric oxide production, formation of protein carbonyls, production of thiobarbituric acid-reactive substances, and expression of proteins involved in oxidative stress and cell damage. Many markers were not significantly altered, but we report a significant increase in nitric oxide after exposure to any of the three compounds in conjunction with depleted uranium.
Collapse
|
8
|
Abstract
This review focuses on recent progress in understanding the role of mitochondrial markers in the context of mitochondrial dysfunction in glaucoma and discussing new therapeutic approaches to modulate mitochondrial function and potentially lead to improved outcomes in glaucoma.
Collapse
|
9
|
Yang GJ, Liu H, Ma DL, Leung CH. Rebalancing metal dyshomeostasis for Alzheimer's disease therapy. J Biol Inorg Chem 2019; 24:1159-1170. [PMID: 31486954 DOI: 10.1007/s00775-019-01712-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/29/2019] [Indexed: 12/26/2022]
Abstract
Alzheimer's disease (AD) is a type of neurodegenerative malady that is associated with the accumulation of amyloid plaques. Metal ions are critical for the development and upkeep of brain activity, but metal dyshomeostasis can contribute to the development of neurodegenerative diseases, including AD. This review highlights the association between metal dyshomeostasis and AD pathology, the feasibility of rebalancing metal homeostasis as a therapeutic strategy for AD, and a survey of current drugs that action via rebalancing metal homeostasis. Finally, we discuss the challenges that should be overcome by researchers in the future to enable the practical use of metal homeostasis rebalancing agents for clinical application.
Collapse
Affiliation(s)
- Guan-Jun Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau SAR, China
| | - Hao Liu
- Department of Chemistry, Hong Kong Baptist University, Kowloon, 999077, Hong Kong SAR, China
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon, 999077, Hong Kong SAR, China.
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau SAR, China.
| |
Collapse
|
10
|
de Carvalho LP, de Melo EJT. Autophagic elimination of Trypanosoma cruzi in the presence of metals. J Microbiol 2019; 57:918-926. [PMID: 31463789 DOI: 10.1007/s12275-019-9018-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 06/14/2019] [Accepted: 07/01/2019] [Indexed: 10/26/2022]
Abstract
Trypanosoma cruzi is an obligate intracellular parasite transmitted to vertebrate hosts by blood-sucking insects. Molecules present in parasites and mammalian cells allow the recognition and parasite internalization. Metallic ions play an essential role in the establishment and maintenance of host-parasite interaction. However, little is known about how parasites handle with essential and nonessential metal quotas. This study aimed to investigate the influence of metal ions on the biological processes of T. cruzi infected cells. Infected cells were incubated with ZnCl2, CdCl2, and HgCl2 for 12 h and labeled with different specific dyes to investigate the cellular events related to intracellular parasite death and elimination. Infected host cells and parasite's mitochondria underwent functional and structural disorders, in addition to parasite's DNA condensation and pH decrease on host cells, which led to parasite death. Further investigations suggested that lysosomes were involved in pH decrease and the double membrane of the endoplasmic reticulum formed vacuoles surrounding damaged parasites, which indicate the occurrence of autophagy for parasite elimination. In conclusion, low concentrations of nonessential and essential metals cause a series of damage to Trypanosoma cruzi organelles, leading to its loss of viability, death, and elimination, with no removal of the host cells.
Collapse
Affiliation(s)
- Laís Pessanha de Carvalho
- Laboratory of Tissue and Cell Biology, State University of North Fluminense - Darcy Ribeiro, Campos dos Goytacazes, 28013-602, Rio de Janeiro, Brazil
| | - Edésio José Tenório de Melo
- Laboratory of Tissue and Cell Biology, State University of North Fluminense - Darcy Ribeiro, Campos dos Goytacazes, 28013-602, Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
Oxidative stress and neurodegeneration: the involvement of iron. Biometals 2018; 31:715-735. [PMID: 30014355 DOI: 10.1007/s10534-018-0126-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022]
Abstract
Many evidences indicate that oxidative stress plays a significant role in a variety of human disease states, including neurodegenerative diseases. Iron is an essential metal for almost all living organisms due to its involvement in a large number of iron-containing proteins and enzymes, though it could be also toxic. Actually, free iron excess generates oxidative stress, particularly in brain, where anti-oxidative defences are relatively low. Its accumulation in specific regions is associated with pathogenesis in a variety of neurodegenerative diseases (i.e., Parkinson's disease, Alzheimer's disease, Huntington's chorea, Amyotrophic Lateral Sclerosis and Neurodegeneration with Brain Iron Accumulation). Anyway, the extent of toxicity is dictated, in part, by the localization of the iron complex within the cell (cytosolic, lysosomal and mitochondrial), its biochemical form, i.e., ferritin or hemosiderin, as well as the ability of the cell to prevent the generation and propagation of free radical by the wide range of antioxidants and cytoprotective enzymes in the cell. Particularly, ferrous iron can act as a catalyst in the Fenton reaction that potentiates oxygen toxicity by generating a wide range of free radical species, including hydroxyl radicals (·OH). The observation that patients with neurodegenerative diseases show a dramatic increase in their brain iron content, correlated with the production of reactive oxigen species in these areas of the brain, conceivably suggests that disturbances in brain iron homeostasis may contribute to the pathogenesis of these disorders. The aim of this review is to describe the chemical features of iron in human beings and iron induced toxicity in neurodegenerative diseases. Furthermore, the attention is focused on metal chelating drugs therapeutic strategies.
Collapse
|
12
|
Further aspects of Toxoplasma gondii elimination in the presence of metals. Parasitol Res 2018; 117:1245-1256. [PMID: 29455419 DOI: 10.1007/s00436-018-5806-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/08/2018] [Indexed: 01/06/2023]
Abstract
Toxoplasma gondii, the etiological agent of toxoplasmosis, infects nucleated cells and then resides and multiplies within a parasitophorous vacuole. For this purpose, the parasite secretes many virulence factors for the purpose of invading and subverting the host microbicidal defenses in order to facilitate its survival in the intracellular milieu. Essential metals are structural components of proteins and enzymes or cofactors of enzymatic reactions responsible for these parasitic survival mechanisms. However, an excess of non-essential or essential metals can lead to parasite death. Thus, infected host cells were incubated with 20 μM ZnCl2 in conjunction with 3 μM CdCl2 or HgCl2 for 12 h in order to investigate cellular events and organelle damage related to intracellular parasite death and elimination. In the presence of these metals, the tachyzoites undergo lipid uptake and transport impairment, functional and structural mitochondrial disorders, DNA condensation, and acidification of the parasitophorous vacuole, thus leading to parasite death. Additional research has suggested that lysosome-vacuole fusion was involved in parasite elimination since acid phosphatases were found inside the parasitophorous vacuole, and vacuoles containing parasites were also positive for autophagy. In conclusion, low concentrations of CdCl2, HgCl2, and ZnCl2 can cause damage to Toxoplasma gondii organelles, leading to loss of viability, organelle death, and elimination without causing toxic effects to host cells.
Collapse
|
13
|
Huuskonen MT, Tuo QZ, Loppi S, Dhungana H, Korhonen P, McInnes LE, Donnelly PS, Grubman A, Wojciechowski S, Lejavova K, Pomeshchik Y, Periviita L, Kosonen L, Giordano M, Walker FR, Liu R, Bush AI, Koistinaho J, Malm T, White AR, Lei P, Kanninen KM. The Copper bis(thiosemicarbazone) Complex Cu II(atsm) Is Protective Against Cerebral Ischemia Through Modulation of the Inflammatory Milieu. Neurotherapeutics 2017; 14:519-532. [PMID: 28050710 PMCID: PMC5398983 DOI: 10.1007/s13311-016-0504-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Developing new therapies for stroke is urgently needed, as this disease is the leading cause of death and disability worldwide, and the existing treatment is only available for a small subset of patients. The interruption of blood flow to the brain during ischemic stroke launches multiple immune responses, characterized by infiltration of peripheral immune cells, the activation of brain microglial cells, and the accumulation of immune mediators. Copper is an essential trace element that is required for many critical processes in the brain. Copper homeostasis is disturbed in chronic neurodegenerative diseases and altered in stroke patients, and targeted copper delivery has been shown to be protective against chronic neurodegeneration. This study was undertaken to assess whether the copper bis(thiosemicarbazone) complex, CuII(atsm), is beneficial in acute brain injury, in preclinical mouse models of ischemic stroke. We demonstrate that the copper complex CuII(atsm) protects neurons from excitotoxicity and N2a cells from OGD in vitro, and is protective in permanent and transient ischemia models in mice as measured by functional outcome and lesion size. Copper delivery in the ischemic brains modulates the inflammatory response, specifically affecting the myeloid cells. It reduces CD45 and Iba1 immunoreactivity, and alters the morphology of Iba1 positive cells in the ischemic brain. CuII(atsm) also protects endogenous microglia against ischemic insult and reduces the proportion of invading monocytes. These results demonstrate that the copper complex CuII(atsm) is an inflammation-modulating compound with high therapeutic potential in stroke and is a strong candidate for the development of therapies for acute brain injury.
Collapse
Affiliation(s)
- Mikko T. Huuskonen
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Qing-zhang Tuo
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria Australia
| | - Sanna Loppi
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Hiramani Dhungana
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Paula Korhonen
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lachlan E. McInnes
- School of Chemistry and Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria Australia
| | - Paul S. Donnelly
- School of Chemistry and Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria Australia
| | - Alexandra Grubman
- Department of Pathology, The University of Melbourne, Parkville, Victoria Australia
| | - Sara Wojciechowski
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Katarina Lejavova
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Yuriy Pomeshchik
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Laura Periviita
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lotta Kosonen
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Martina Giordano
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Frederick R. Walker
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW Australia
| | - Rong Liu
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ashley I. Bush
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria Australia
| | - Jari Koistinaho
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tarja Malm
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Anthony R. White
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria Australia
- Department of Pathology, The University of Melbourne, Parkville, Victoria Australia
- Present Address: QIMR Berghofer Medical Research Institute, Herston, Queensland Australia
- Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Locked Bag 2000, Herston, QLD 4029 Australia
| | - Peng Lei
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria Australia
- Department of Neurology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan China
| | - Katja M. Kanninen
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
14
|
Braidy N, Poljak A, Marjo C, Rutlidge H, Rich A, Jugder BE, Jayasena T, Inestrosa NC, Sachdev PS. Identification of Cerebral Metal Ion Imbalance in the Brain of Aging Octodon degus. Front Aging Neurosci 2017; 9:66. [PMID: 28405187 PMCID: PMC5370394 DOI: 10.3389/fnagi.2017.00066] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 03/03/2017] [Indexed: 01/18/2023] Open
Abstract
The accumulation of redox-active transition metals in the brain and metal dyshomeostasis are thought to be associated with the etiology and pathogenesis of several neurodegenerative diseases, and Alzheimer’s disease (AD) in particular. As well, distinct biometal imaging and role of metal uptake transporters are central to understanding AD pathogenesis and aging but remain elusive, due inappropriate detection methods. We therefore hypothesized that Octodon degus develop neuropathological abnormalities in the distribution of redox active biometals, and this effect may be due to alterations in the expression of lysosomal protein, major Fe/Cu transporters, and selected Zn transporters (ZnTs and ZIPs). Herein, we report the distribution profile of biometals in the aged brain of the endemic Chilean rodent O. degus—a natural model to investigate the role of metals on the onset and progression of AD. Using laser ablation inductively coupled plasma mass spectrometry, our quantitative images of biometals (Fe, Ca, Zn, Cu, and Al) appear significantly elevated in the aged O. degus and show an age-dependent rise. The metals Fe, Ca, Zn, and Cu were specifically enriched in the cortex and hippocampus, which are the regions where amyloid plaques, tau phosphorylation and glial alterations are most commonly reported, whilst Al was enriched in the hippocampus alone. Using whole brain extracts, age-related deregulation of metal trafficking pathways was also observed in O. degus. More specifically, we observed impaired lysosomal function, demonstrated by increased cathepsin D protein expression. An age-related reduction in the expression of subunit B2 of V-ATPase, and significant increases in amyloid beta peptide 42 (Aβ42), and the metal transporter ATP13a2 were also observed. Although the protein expression levels of the zinc transporters, ZnT (1,3,4,6, and 7), and ZIP7,8 and ZIP14 increased in the brain of aged O. degus, ZnT10, decreased. Although no significant age-related change was observed for the major iron/copper regulator IRP2, we did find a significant increase in the expression of DMT1, a major transporter of divalent metal species, 5′-aminolevulinate synthase 2 (ALAS2), and the proto-oncogene, FOS. Collectively, our data indicate that transition metals may be enriched with age in the brains of O. degus, and metal dyshomeostasis in specific brain regions is age-related.
Collapse
Affiliation(s)
- Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales Sydney, NSW, Australia
| | - Anne Poljak
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South WalesSydney, NSW, Australia; Mark Wainwright Analytical Centre, University of New South WalesSydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine, University of New South WalesSydney, NSW, Australia
| | - Chris Marjo
- Mark Wainwright Analytical Centre, University of New South Wales Sydney, NSW, Australia
| | - Helen Rutlidge
- Mark Wainwright Analytical Centre, University of New South Wales Sydney, NSW, Australia
| | - Anne Rich
- Mark Wainwright Analytical Centre, University of New South Wales Sydney, NSW, Australia
| | - Bat-Erdene Jugder
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales Sydney, NSW, Australia
| | - Tharusha Jayasena
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales Sydney, NSW, Australia
| | - Nibaldo C Inestrosa
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South WalesSydney, NSW, Australia; Centre for Ageing and Regeneration, Faculty of Biological Sciences, Pontifical Catholic University of ChileSantiago, Chile
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South WalesSydney, NSW, Australia; Neuropsychiatric Institute, Euroa Centre, Prince of Wales HospitalSydney, NSW, Australia
| |
Collapse
|
15
|
Alta RYP, Vitorino HA, Goswami D, Liria CW, Wisnovsky SP, Kelley SO, Machini MT, Espósito BP. Mitochondria-penetrating peptides conjugated to desferrioxamine as chelators for mitochondrial labile iron. PLoS One 2017; 12:e0171729. [PMID: 28178347 PMCID: PMC5298241 DOI: 10.1371/journal.pone.0171729] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/24/2017] [Indexed: 12/16/2022] Open
Abstract
Desferrioxamine (DFO) is a bacterial siderophore with a high affinity for iron, but low cell penetration. As part of our ongoing project focused on DFO-conjugates, we synthesized, purified, characterized and studied new mtDFOs (DFO conjugated to the Mitochondria Penetrating Peptides TAT49-57, 1A, SS02 and SS20) using a succinic linker. These new conjugates retained their strong iron binding ability and antioxidant capacity. They were relatively non toxic to A2780 cells (IC50 40–100 μM) and had good mitochondrial localization (Rr +0.45 –+0.68) as observed when labeled with carboxy-tetramethylrhodamine (TAMRA) In general, mtDFO caused only modest levels of mitochondrial DNA (mtDNA) damage. DFO-SS02 retained the antioxidant ability of the parent peptide, shown by the inhibition of mitochondrial superoxide formation. None of the compounds displayed cell cycle arrest or enhanced apoptosis. Taken together, these results indicate that mtDFO could be promising compounds for amelioration of the disease symptoms of iron overload in mitochondria.
Collapse
Affiliation(s)
- Roxana Y. P. Alta
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Hector A. Vitorino
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Cleber W. Liria
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Simon P. Wisnovsky
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Shana O. Kelley
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - M. Terêsa Machini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (MTM); (BPE)
| | - Breno P. Espósito
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (MTM); (BPE)
| |
Collapse
|
16
|
González-Villalva A, Colín-Barenque L, Bizarro-Nevares P, Rojas-Lemus M, Rodríguez-Lara V, García-Pelaez I, Ustarroz-Cano M, López-Valdez N, Albarrán-Alonso JC, Fortoul TI. Pollution by metals: Is there a relationship in glycemic control? ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 46:337-343. [PMID: 27552445 DOI: 10.1016/j.etap.2016.06.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 06/06/2023]
Abstract
There are evidences of environmental pollution and health effects. Metals are pollutants implicated in systemic toxicity. One of the least studied effects, but which is currently becoming more important, is the effect of metals on glycemic control. Metals have been implicated as causes of chronic inflammation and oxidative stress and are associated to obesity, hyperglycemia and even diabetes. Arsenic, iron, mercury, lead, cadmium and nickel have been studied as a risk factor for hyperglycemia and diabetes. There is another group of metals that causes hypoglycemia such as vanadium, chromium, zinc and magnesium by different mechanisms. Zinc, magnesium and chromium deficiency is associated with increased risk of diabetes. This review summarizes some metals involved in glycemic control and pretends to alert health professionals about considering environmental metals as an important factor that could explain the poor glycemic control in patients. Further studies are needed to understand this poorly assessed problem.
Collapse
Affiliation(s)
- Adriana González-Villalva
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Laura Colín-Barenque
- Laboratorio de Neuromorfología, FES Iztacala, UNAM CP 54090 Edo. de México, Mexico.
| | - Patricia Bizarro-Nevares
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Marcela Rojas-Lemus
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Vianey Rodríguez-Lara
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Isabel García-Pelaez
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Martha Ustarroz-Cano
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Nelly López-Valdez
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Juan Carlos Albarrán-Alonso
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Teresa I Fortoul
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| |
Collapse
|
17
|
Davidson SM, Lopaschuk GD, Spedding M, Beart PM. Mitochondrial pharmacology: energy, injury and beyond. Br J Pharmacol 2014; 171:1795-7. [PMID: 24684388 DOI: 10.1111/bph.12679] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
While the mitochondrion has long fascinated biologists and the sheer diversity of druggable targets has made it attractive for potential drug development, there has been little success translatable to the clinic. Given the diversity of inborn errors of metabolism and mitochondrial diseases, mitochondrially mediated oxidative stress (myopathies, reperfusion injury, Parkinson's disease, ageing) and the consequences of disturbed energetics (circulatory shock, diabetes, cancer), the potential for meaningful gain with novel drugs targeting mitochondrial mechanisms is huge both in terms of patient quality of life and health care costs. In this themed issue of the British Journal of Pharmacology, we highlight the key directions of the contemporary advances in the field of mitochondrial biology, emerging drug targets and new molecules which are close to clinical application. Authors' contributions are diverse both in terms of species and organs in which the mitochondrially related studies are performed, and from the perspectives of mechanisms under study. Defined roles of mitochondria in disease are updated and previously unknown contributions to disease are described in terms of the interface between basic science and pathological relevance.
Collapse
Affiliation(s)
- S M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | | | | | | |
Collapse
|
18
|
Braidy N, Poljak A, Marjo C, Rutlidge H, Rich A, Jayasena T, Inestrosa NC, Sachdev P. Metal and complementary molecular bioimaging in Alzheimer's disease. Front Aging Neurosci 2014; 6:138. [PMID: 25076902 PMCID: PMC4098123 DOI: 10.3389/fnagi.2014.00138] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 06/09/2014] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly, affecting over 27 million people worldwide. AD represents a complex neurological disorder which is best understood as the consequence of a number of interconnected genetic and lifestyle variables, which culminate in multiple changes to brain structure and function. These can be observed on a gross anatomical level in brain atrophy, microscopically in extracellular amyloid plaque and neurofibrillary tangle formation, and at a functional level as alterations of metabolic activity. At a molecular level, metal dyshomeostasis is frequently observed in AD due to anomalous binding of metals such as Iron (Fe), Copper (Cu), and Zinc (Zn), or impaired regulation of redox-active metals which can induce the formation of cytotoxic reactive oxygen species and neuronal damage. Metal chelators have been administered therapeutically in transgenic mice models for AD and in clinical human AD studies, with positive outcomes. As a result, neuroimaging of metals in a variety of intact brain cells and tissues is emerging as an important tool for increasing our understanding of the role of metal dysregulation in AD. Several imaging techniques have been used to study the cerebral metallo-architecture in biological specimens to obtain spatially resolved data on chemical elements present in a sample. Hyperspectral techniques, such as particle-induced X-ray emission (PIXE), energy dispersive X-ray spectroscopy (EDS), X-ray fluorescence microscopy (XFM), synchrotron X-ray fluorescence (SXRF), secondary ion mass spectrometry (SIMS), and laser ablation inductively coupled mass spectrometry (LA-ICPMS) can reveal relative intensities and even semi-quantitative concentrations of a large set of elements with differing spatial resolution and detection sensitivities. Other mass spectrometric and spectroscopy imaging techniques such as laser ablation electrospray ionization mass spectrometry (LA ESI-MS), MALDI imaging mass spectrometry (MALDI-IMS), and Fourier transform infrared spectroscopy (FTIR) can be used to correlate changes in elemental distribution with the underlying pathology in AD brain specimens. Taken together, these techniques provide new techniques to probe the pathobiology of AD and pave the way for identifying new therapeutic targets. The current review aims to discuss the advantages and challenges of using these emerging elemental and molecular imaging techniques, and highlight clinical achievements in AD research using bioimaging techniques.
Collapse
Affiliation(s)
- Nady Braidy
- Faculty of Medicine, Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Sydney, NSW, Australia
| | - Anne Poljak
- Faculty of Medicine, Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Sydney, NSW, Australia ; Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales Sydney, NSW, Australia ; Faculty of Medicine, School of Medical Sciences, University of New South Wales Sydney, NSW, Australia
| | - Christopher Marjo
- Solid State and Elemental Analysis Unit, Mark Wainwright Analytical Centre, University of New South Wales Sydney, NSW, Australia
| | - Helen Rutlidge
- Solid State and Elemental Analysis Unit, Mark Wainwright Analytical Centre, University of New South Wales Sydney, NSW, Australia
| | - Anne Rich
- Solid State and Elemental Analysis Unit, Mark Wainwright Analytical Centre, University of New South Wales Sydney, NSW, Australia
| | - Tharusha Jayasena
- Faculty of Medicine, Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Sydney, NSW, Australia
| | - Nibaldo C Inestrosa
- Faculty of Biological Sciences, Centre for Ageing and Regeneration, P. Catholic University of Chile Santiago, Chile
| | - Perminder Sachdev
- Faculty of Medicine, Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Sydney, NSW, Australia ; Euroa Centre, Neuropsychiatric Institute, Prince of Wales Hospital Sydney, NSW, Australia
| |
Collapse
|
19
|
Abstract
Copper is an essential element in many biological processes. The critical functions associated with copper have resulted from evolutionary harnessing of its potent redox activity. This same property also places copper in a unique role as a key modulator of cell signal transduction pathways. These pathways are the complex sequence of molecular interactions that drive all cellular mechanisms and are often associated with the interplay of key enzymes including kinases and phosphatases but also including intracellular changes in pools of smaller molecules. A growing body of evidence is beginning to delineate the how, when and where of copper-mediated control over cell signal transduction. This has been driven by research demonstrating critical changes to copper homeostasis in many disorders including cancer and neurodegeneration and therapeutic potential through control of disease-associated cell signalling changes by modulation of copper-protein interactions. This timely review brings together for the first time the diverse actions of copper as a key regulator of cell signalling pathways and discusses the potential strategies for controlling disease-associated signalling processes using copper modulators. It is hoped that this review will provide a valuable insight into copper as a key signal regulator and stimulate further research to promote our understanding of copper in disease and therapy.
Collapse
|
20
|
Urrutia PJ, Mena NP, Núñez MT. The interplay between iron accumulation, mitochondrial dysfunction, and inflammation during the execution step of neurodegenerative disorders. Front Pharmacol 2014; 5:38. [PMID: 24653700 PMCID: PMC3948003 DOI: 10.3389/fphar.2014.00038] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 02/19/2014] [Indexed: 12/21/2022] Open
Abstract
A growing set of observations points to mitochondrial dysfunction, iron accumulation, oxidative damage and chronic inflammation as common pathognomonic signs of a number of neurodegenerative diseases that includes Alzheimer’s disease, Huntington disease, amyotrophic lateral sclerosis, Friedrich’s ataxia and Parkinson’s disease. Particularly relevant for neurodegenerative processes is the relationship between mitochondria and iron. The mitochondrion upholds the synthesis of iron–sulfur clusters and heme, the most abundant iron-containing prosthetic groups in a large variety of proteins, so a fraction of incoming iron must go through this organelle before reaching its final destination. In turn, the mitochondrial respiratory chain is the source of reactive oxygen species (ROS) derived from leaks in the electron transport chain. The co-existence of both iron and ROS in the secluded space of the mitochondrion makes this organelle particularly prone to hydroxyl radical-mediated damage. In addition, a connection between the loss of iron homeostasis and inflammation is starting to emerge; thus, inflammatory cytokines like TNF-alpha and IL-6 induce the synthesis of the divalent metal transporter 1 and promote iron accumulation in neurons and microglia. Here, we review the recent literature on mitochondrial iron homeostasis and the role of inflammation on mitochondria dysfunction and iron accumulation on the neurodegenerative process that lead to cell death in Parkinson’s disease. We also put forward the hypothesis that mitochondrial dysfunction, iron accumulation and inflammation are part of a synergistic self-feeding cycle that ends in apoptotic cell death, once the antioxidant cellular defense systems are finally overwhelmed.
Collapse
Affiliation(s)
- Pamela J Urrutia
- Department of Biology and Research Ring on Oxidative Stress in the Nervous System, Faculty of Sciences, University of Chile Santiago, Chile
| | - Natalia P Mena
- Department of Biology and Research Ring on Oxidative Stress in the Nervous System, Faculty of Sciences, University of Chile Santiago, Chile
| | - Marco T Núñez
- Department of Biology and Research Ring on Oxidative Stress in the Nervous System, Faculty of Sciences, University of Chile Santiago, Chile
| |
Collapse
|