1
|
Hancock JL, Kalimutho M, Straube J, Lim M, Gresshoff I, Saunus JM, Lee JS, Lakhani SR, Simpson KJ, Bush AI, Anderson RL, Khanna KK. COMMD3 loss drives invasive breast cancer growth by modulating copper homeostasis. J Exp Clin Cancer Res 2023; 42:90. [PMID: 37072858 PMCID: PMC10111822 DOI: 10.1186/s13046-023-02663-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/05/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Despite overall improvement in breast cancer patient outcomes from earlier diagnosis and personalised treatment approaches, some patients continue to experience recurrence and incurable metastases. It is therefore imperative to understand the molecular changes that allow transition from a non-aggressive state to a more aggressive phenotype. This transition is governed by a number of factors. METHODS As crosstalk with extracellular matrix (ECM) is critical for tumour cell growth and survival, we applied high throughput shRNA screening on a validated '3D on-top cellular assay' to identify novel growth suppressive mechanisms. RESULTS A number of novel candidate genes were identified. We focused on COMMD3, a previously poorly characterised gene that suppressed invasive growth of ER + breast cancer cells in the cellular assay. Analysis of published expression data suggested that COMMD3 is normally expressed in the mammary ducts and lobules, that expression is lost in some tumours and that loss is associated with lower survival probability. We performed immunohistochemical analysis of an independent tumour cohort to investigate relationships between COMMD3 protein expression, phenotypic markers and disease-specific survival. This revealed an association between COMMD3 loss and shorter survival in hormone-dependent breast cancers and in particularly luminal-A-like tumours (ER+/Ki67-low; 10-year survival probability 0.83 vs. 0.73 for COMMD3-positive and -negative cases, respectively). Expression of COMMD3 in luminal-A-like tumours was directly associated with markers of luminal differentiation: c-KIT, ELF5, androgen receptor and tubule formation (the extent of normal glandular architecture; p < 0.05). Consistent with this, depletion of COMMD3 induced invasive spheroid growth in ER + breast cancer cell lines in vitro, while Commd3 depletion in the relatively indolent 4T07 TNBC mouse cell line promoted tumour expansion in syngeneic Balb/c hosts. Notably, RNA sequencing revealed a role for COMMD3 in copper signalling, via regulation of the Na+/K+-ATPase subunit, ATP1B1. Treatment of COMMD3-depleted cells with the copper chelator, tetrathiomolybdate, significantly reduced invasive spheroid growth via induction of apoptosis. CONCLUSION Overall, we found that COMMD3 loss promoted aggressive behaviour in breast cancer cells.
Collapse
Affiliation(s)
- Janelle L Hancock
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD, 4006, Australia
| | - Murugan Kalimutho
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD, 4006, Australia
| | - Jasmin Straube
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD, 4006, Australia
| | - Malcolm Lim
- The University of Queensland Faculty of Medicine, UQ Centre for Clinical Research and Anatomical Pathology, Pathology Queensland, Herston, QLD, 4029, Australia
| | - Irma Gresshoff
- The University of Queensland Faculty of Medicine, UQ Centre for Clinical Research and Anatomical Pathology, Pathology Queensland, Herston, QLD, 4029, Australia
| | - Jodi M Saunus
- The University of Queensland Faculty of Medicine, UQ Centre for Clinical Research and Anatomical Pathology, Pathology Queensland, Herston, QLD, 4029, Australia
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, 4102, Australia
| | - Jason S Lee
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD, 4006, Australia
| | - Sunil R Lakhani
- The University of Queensland Faculty of Medicine, UQ Centre for Clinical Research and Anatomical Pathology, Pathology Queensland, Herston, QLD, 4029, Australia
| | - Kaylene J Simpson
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, 3010, Australia
- Sir Peter MacCallum Department of Oncology and the Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Ashley I Bush
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| | - Robin L Anderson
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia.
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD, 4006, Australia.
| |
Collapse
|
2
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
3
|
Cheng F, Peng G, Lu Y, Wang K, Ju Q, Ju Y, Ouyang M. Relationship between copper and immunity: The potential role of copper in tumor immunity. Front Oncol 2022; 12:1019153. [PMID: 36419894 PMCID: PMC9676660 DOI: 10.3389/fonc.2022.1019153] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/17/2022] [Indexed: 07/30/2023] Open
Abstract
Copper is an essential trace element in an organism, and changes in copper levels in vivo often indicate a diseased state. Copper and immunity have been discussed since the last century, with copper deficiency significantly affecting the development and function of the immune system, such as increased host susceptibility to various pathogens, decreased number and impaired function of neutrophils, reduced antibacterial activity of macrophages, decreased proliferation of splenocytes, impaired B cell ability to produce antibodies and impaired function of cytotoxic T lymphocyte and helper T cells. In the past 20 years, some studies have shown that copper ions are related to the development of many tumors, including lung cancer, acute lymphoid leukaemia, multiple myeloma and other tumors, wherein copper ion levels were significantly elevated, and current studies reveal that copper ions are involved in the development, growth and metastasis of tumors through various pathways. Moreover, recent studies have shown that copper ions can regulate the expression of PD-L1, thus, attention should be paid to the important role of copper in tumor immunity. By exploring and studying copper ions and tumor immunity, new insights into tumor immunity could be generated and novel therapeutic approaches to improve the clinical prognosis of patients can be provided.
Collapse
Affiliation(s)
- Fu Cheng
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Geng Peng
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan Lu
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Kang Wang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinuo Ju
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- Guangdong Country Garden School, Shunde, Foshan, Guangdong, China
| | - Yongle Ju
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Manzhao Ouyang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Merighi S, Nigro M, Travagli A, Gessi S. Microglia and Alzheimer's Disease. Int J Mol Sci 2022; 23:12990. [PMID: 36361780 PMCID: PMC9657945 DOI: 10.3390/ijms232112990] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 07/30/2023] Open
Abstract
There is a huge need for novel therapeutic and preventative approaches to Alzheimer's disease (AD) and neuroinflammation seems to be one of the most fascinating solutions. The primary cell type that performs immunosurveillance and helps clear out unwanted chemicals from the brain is the microglia. Microglia work to reestablish efficiency and stop further degeneration in the early stages of AD but mainly fail in the illness's later phases. This may be caused by a number of reasons, e.g., a protracted exposure to cytokines that induce inflammation and an inappropriate accumulation of amyloid beta (Aβ) peptide. Extracellular amyloid and/or intraneuronal phosphorylated tau in AD can both activate microglia. The activation of TLRs and scavenger receptors, inducing the activation of numerous inflammatory pathways, including the NF-kB, JAK-STAT, and NLRP3 inflammasome, facilitates microglial phagocytosis and activation in response to these mediators. Aβ/tau are taken up by microglia, and their removal from the extracellular space can also have protective effects, but if the illness worsens, an environment that is constantly inflamed and overexposed to an oxidative environment might encourage continuous microglial activation, which can lead to neuroinflammation, oxidative stress, iron overload, and neurotoxicity. The complexity and diversity of the roles that microglia play in health and disease necessitate the urgent development of new biomarkers that identify the activity of different microglia. It is imperative to comprehend the intricate mechanisms that result in microglial impairment to develop new immunomodulating therapies that primarily attempt to recover the physiological role of microglia, allowing them to carry out their core function of brain protection.
Collapse
Affiliation(s)
- Stefania Merighi
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| | | | | | | |
Collapse
|
5
|
Singh SK, Balendra V, Obaid AA, Esposto J, Tikhonova MA, Gautam NK, Poeggeler B. Copper-Mediated β-Amyloid Toxicity and its Chelation Therapy in Alzheimer's Disease. Metallomics 2022; 14:6554256. [PMID: 35333348 DOI: 10.1093/mtomcs/mfac018] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 03/08/2022] [Indexed: 01/10/2023]
Abstract
The link between bio-metals, Alzheimer's disease (AD), and its associated protein, amyloid-β (Aβ) is very complex and one of the most studied aspects currently. Alzheimer's disease, a progressive neurodegenerative disease, is proposed to occurs due to the misfolding and aggregation of Aβ. Dyshomeostasis of metal ions and their interaction with Aβ has largely been implicated in AD. Copper plays a crucial role in amyloid-β toxicity and AD development potentially occurs through direct interaction with the copper-binding motif of APP and different amino acid residues of Aβ. Previous reports suggest that high levels of copper accumulation in the AD brain result in modulation of toxic Aβ peptide levels, implicating the role of copper in the pathophysiology of AD. In this review, we explore the possible mode of copper ion interaction with Aβ which accelerates the kinetics of fibril formation and promote amyloid-β mediated cell toxicity in Alzheimer's disease and the potential use of various copper chelators in the prevention of copper-mediated Aβ toxicity.
Collapse
Affiliation(s)
- Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, Lucknow-226002, India
| | - Vyshnavy Balendra
- Saint James School of Medicine, Park Ridge, Illinois, United States of America 60068
| | - Ahmad A Obaid
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Josephine Esposto
- Department of Environmental and Life Sciences, Trent University, Peterborough, Ontario, CanadaK9L 0G2
| | - Maria A Tikhonova
- Laboratory of the Experimental Models of Neurodegenerative Processes, Scientific Research Institute of Neurosciences and Medicine; Timakov st., 4, Novosibirsk, 630117, Russia
| | - Naveen Kumar Gautam
- Department of Urology and Renal Transplantation, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Burkhard Poeggeler
- Johann-Friedrich-Blumenbach-Institute for Zoology & Anthropology, Faculty of Biology and Psychology, Georg-August-University of Göttingen, Am Türmchen 3,33332 Gütersloh, Germany
| |
Collapse
|
6
|
Eldamaty HS, Elbasiouny H, Elmoslemany AM, Abd El-Maoula LM, El-Desoky OI, Rehan M, Abd El Moneim D, Zedan A. Protective Effect of Wheat and Barley Grass Against the Acute Toxicological Effects of the Concurrent Administration of Excessive Heavy Metals in Drinking Water on the Rats Liver and Brain. APPLIED SCIENCES 2021; 11:5059. [DOI: 10.3390/app11115059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Heavy metal contaminated water is a great concern because of its high toxiciy, non-biodegradability, and bioaccumulation. Therefore, non-contaminated water is fundamental for a healthy life. Special attention is paid to the health-promoting ingredients of germinated whole cereal products. This study aimed to (1) examine the potentially harmful effects of Cu, Mn, and Zn on rat livers and brains, and (2) the potentially protective action of wheat and barley grasses against the expected harmful effects of these metals. The rats were treated with water contaminated by heavy metals (HMs) and germinated wheat and barley for 60 days. The rat liver functions and histopathological examinations were analyzed. Comet assay was evaluated to assess the damage in the DNA of rat livers and brains. The results indicated a significant alteration in liver functions in rats exposed to HMs; however, wheat and barley grasses at high doses decreased the harmful effects. An insignificant difference was noticed in total protein, albumin, and globulin of rats treated with HMs compared with the control. A significant increase in the serum and liver levels of HMs was recorded; however, they were reduced by wheat and barley grasses. Rat livers treated with HMs exhibited severe histological effects. The groups treated with wheat and barley grasses showed a normal liver architecture. A significant increase in DNA damage in the livers and brains was observed in rats treated with HMs, which was reduced when treated with wheat and barley grasses. Thus, using germinated seeds is promising to avoid damaging of HMs.
Collapse
|
7
|
Jouybari L, Kiani F, Islami F, Sanagoo A, Sayehmiri F, Hosnedlova B, Doşa MD, Kizek R, Chirumbolo S, Bjørklund G. Copper Concentrations in Breast Cancer: A Systematic Review and Meta-Analysis. Curr Med Chem 2021; 27:6373-6383. [PMID: 31533596 DOI: 10.2174/0929867326666190918120209] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/27/2019] [Accepted: 09/02/2019] [Indexed: 11/22/2022]
Abstract
Breast cancer is the most common neoplasm, comprising 16% of all women's cancers worldwide. Research of Copper (Cu) concentrations in various body specimens have suggested an association between Cu levels and breast cancer risks. This systematic review and meta-analysis summarize the results of published studies and examine this association. We searched the databases PubMed, Scopus, Web of Science, and Google Scholar and the reference lists of relevant publications. The Standardized Mean Differences (SMDs) between Cu levels in cancer cases and controls and corresponding Confidence Intervals (CIs), as well as I2 statistics, were calculated to examine heterogeneity. Following the specimens used in the original studies, the Cu concentrations were examined in three subgroups: serum or plasma, breast tissue, and scalp hair. We identified 1711 relevant studies published from 1984 to 2017. There was no statistically significant difference between breast cancer cases and controls for Cu levels assayed in any studied specimen; the SMD (95% CI) was -0.01 (-1.06 - 1.03; P = 0.98) for blood or serum, 0.51 (-0.70 - 1.73; P = 0.41) for breast tissue, and -0.88 (-3.42 - 1.65; P = 0.50) for hair samples. However, the heterogeneity between studies was very high (P < 0.001) in all subgroups. We did not find evidence for publication bias (P = 0.91). The results of this meta-analysis do not support an association between Cu levels and breast cancer. However, due to high heterogeneity in the results of original studies, this conclusion needs to be confirmed by well-designed prospective studies.
Collapse
Affiliation(s)
- Leila Jouybari
- Nursing Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Faezeh Kiani
- Student Research Committee, Ilam University of Medical Sciences, Ilam, Iran
| | - Farhad Islami
- Surveillance and Health Services Research, American Cancer Society, Atlanta, United States
| | - Akram Sanagoo
- Nursing Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fatemeh Sayehmiri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bozena Hosnedlova
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic,CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno-Bohunice, Brno, Czech Republic
| | - Monica Daniela Doşa
- Department of Pharmacology, Faculty of Medicine, Ovidius University, Constanta, Romania
| | - Rene Kizek
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic,CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno-Bohunice, Brno, Czech Republic
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy,CONEM Scientific Secretary, Verona, Italy
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| |
Collapse
|
8
|
Schilling KM, Tao L, Wu B, Kiblen JTM, Ubilla-Rodriguez NC, Pushie MJ, Britt RD, Roseman GP, Harris DA, Millhauser GL. Both N-Terminal and C-Terminal Histidine Residues of the Prion Protein Are Essential for Copper Coordination and Neuroprotective Self-Regulation. J Mol Biol 2020; 432:4408-4425. [PMID: 32473880 PMCID: PMC7387163 DOI: 10.1016/j.jmb.2020.05.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 01/19/2023]
Abstract
The cellular prion protein (PrPC) comprises two domains: a globular C-terminal domain and an unstructured N-terminal domain. Recently, copper has been observed to drive tertiary contact in PrPC, inducing a neuroprotective cis interaction that structurally links the protein's two domains. The location of this interaction on the C terminus overlaps with the sites of human pathogenic mutations and toxic antibody docking. Combined with recent evidence that the N terminus is a toxic effector regulated by the C terminus, there is an emerging consensus that this cis interaction serves a protective role, and that the disruption of this interaction by misfolded PrP oligomers may be a cause of toxicity in prion disease. We demonstrate here that two highly conserved histidines in the C-terminal domain of PrPC are essential for the protein's cis interaction, which helps to protect against neurotoxicity carried out by its N terminus. We show that simultaneous mutation of these histidines drastically weakens the cis interaction and enhances spontaneous cationic currents in cultured cells, the first C-terminal mutant to do so. Whereas previous studies suggested that Cu2+ coordination was localized solely to the protein's N-terminal domain, we find that both domains contribute equatorially coordinated histidine residue side-chains, resulting in a novel bridging interaction. We also find that extra N-terminal histidines in pathological familial mutations involving octarepeat expansions inhibit this interaction by sequestering copper from the C terminus. Our findings further establish a structural basis for PrPC's C-terminal regulation of its otherwise toxic N terminus.
Collapse
Affiliation(s)
- Kevin M Schilling
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Lizhi Tao
- Department of Chemistry, University of California, 1 Shields Ave., Davis, CA 95616, USA
| | - Bei Wu
- Department of Biochemistry, Boston University School of Medicine, 72 E. Concord St Silvio Conte., Boston, MA 02118, USA
| | - Joseph T M Kiblen
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Natalia C Ubilla-Rodriguez
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, CA 95064, USA
| | - M Jake Pushie
- Department of Surgery, College of Medicine, University of Saskatchewan, 107 Wiggins Rd B419, Saskatoon, SK S7N 5E5, Canada
| | - R David Britt
- Department of Chemistry, University of California, 1 Shields Ave., Davis, CA 95616, USA
| | - Graham P Roseman
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, CA 95064, USA
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, 72 E. Concord St Silvio Conte., Boston, MA 02118, USA.
| | - Glenn L Millhauser
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, CA 95064, USA.
| |
Collapse
|
9
|
Behzadfar L, Hassani S, Feizpour H, Abbasian N, Salek Maghsoudi A, Taghizadeh G, Pourahmad J, Sharifzadeh M. Effects of mercuric chloride on spatial memory deficit-induced by beta-amyloid and evaluation of mitochondrial function markers in the hippocampus of rats. Metallomics 2020; 12:144-153. [PMID: 31793599 DOI: 10.1039/c9mt00161a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mercury is a highly poisonous heavy metal abundantly found in the environment in its inorganic form. Although evidence have been provided about the possible role of inorganic mercury in the pathology of Alzheimer's disease (AD), its effect on cognitive and mitochondrial functions have not yet been completely understood. Thus, the purpose of the present study was to examine the effects of the chronic exposure to mercuric chloride (0.4, 0.8 and 1.6 mg kg-1 per day for 3 weeks) through drinking water (by gavage) on spatial learning and memory and hippocampal mitochondrial function in beta-amyloid treated rats (1 μg per μL per side, intrahippocampally). The acquisition and retention of spatial memory were evaluated by the Morris water maze (MWM) test. Several parameters of hippocampal mitochondrial function were also measured. The results indicated that mercury impaired spatial learning and memory as well as aggravated Aβ-induced memory impairments in a concentration-dependent manner. Furthermore, mercury exposure resulted in a significant increase in ROS generation, MMP collapse, mitochondrial swelling, glutathione oxidation, lipid peroxidation, and outer membrane damage. In addition, a reduced cytochrome c oxidase (complex IV) activity and elevated ADP/ATP ratio in the rats' hippocampus was also observed. The findings of the current study revealed that chronic mercury exposure led to mitochondrial dysfunction, which resulted in spatial memory impairments. The results also showed that mercury can exacerbate the toxic effects of Aβ on spatial memory and hippocampal mitochondrial function.
Collapse
Affiliation(s)
- Ladan Behzadfar
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
O'Doherty C, Keenan J, Henry M, Meleady P, Sinkunaite I, Clynes M, O'Sullivan F, Horgan K, Murphy R. Characterisation and proteomic profiling of continuously exposed Cu-resistant variants of the Caco-2 cell line. Toxicol In Vitro 2020; 65:104773. [PMID: 31981602 DOI: 10.1016/j.tiv.2020.104773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/06/2020] [Accepted: 01/14/2020] [Indexed: 12/21/2022]
Abstract
Studies in hepatic systems identify multiple factors involved in the generation of copper resistance. As the intestine is the route of exposure to dietary copper, we wanted to understand how intestinal cells overcome the toxic effects of high copper and what mechanisms of resistance develop. Using the intestinal cell line Caco-2, resistance was developed by serial subculture in 50 μM copper in inorganic (CuSO4) or organic (Cu proteinate) forms. Caco-2 variants exhibited resistance to copper and retained the non-monotonic dose response while displaying stable phenotypes following repeated subculture in the absence of copper. Phenotypic changes on exposure to copper in parental Caco-2 cells included significantly increased total protein yield, ROS, SOD, metallothionein expression, GSH and total glutathione. These phenotypic changes were not replicated in resistant variants on a per cell basis. Quantitative label-free LC-MS/MS proteomic analysis identified 1113 differentially expressed proteins (DEPs) between parental Caco-2 and resistant cells. With some exceptions, most of the DEPs were overexpressed to a low level around 2-fold suggesting resistance was supported by multiple small changes in protein expression. These variants may be a useful tool in studying the toxicity of stress responses in further Cu-related studies.
Collapse
Affiliation(s)
- Charles O'Doherty
- National Institute for Cellular Biotechnology and SSPC-SFI, Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin D09 W6Y4, Ireland.
| | - Joanne Keenan
- National Institute for Cellular Biotechnology and SSPC-SFI, Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin D09 W6Y4, Ireland
| | - Michael Henry
- National Institute for Cellular Biotechnology and SSPC-SFI, Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin D09 W6Y4, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology and SSPC-SFI, Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin D09 W6Y4, Ireland
| | - Indre Sinkunaite
- Alltech Ireland, European Bioscience Centre, Summerhill Rd, Sarney, Dunboyne, Co. Meath, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology and SSPC-SFI, Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin D09 W6Y4, Ireland
| | - Finbarr O'Sullivan
- National Institute for Cellular Biotechnology and SSPC-SFI, Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin D09 W6Y4, Ireland
| | - Karina Horgan
- Alltech Ireland, European Bioscience Centre, Summerhill Rd, Sarney, Dunboyne, Co. Meath, Ireland
| | - Richard Murphy
- Alltech Ireland, European Bioscience Centre, Summerhill Rd, Sarney, Dunboyne, Co. Meath, Ireland
| |
Collapse
|
11
|
Wang Q, Jiang H, Wang L, Yi H, Li Z, Liu R. Vitegnoside Mitigates Neuronal Injury, Mitochondrial Apoptosis, and Inflammation in an Alzheimer’s Disease Cell Model via the p38 MAPK/JNK Pathway. J Alzheimers Dis 2019; 72:199-214. [DOI: 10.3233/jad-190640] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Qian Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Hailun Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Linlin Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Hong Yi
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Zhuorong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Rui Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
12
|
De Luca A, Barile A, Arciello M, Rossi L. Copper homeostasis as target of both consolidated and innovative strategies of anti-tumor therapy. J Trace Elem Med Biol 2019; 55:204-213. [PMID: 31345360 DOI: 10.1016/j.jtemb.2019.06.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/28/2019] [Accepted: 06/14/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Copper was reported to be involved in the onset and progression of cancer. Proteins in charge of copper uptake and distribution, as well as cuproenzymes, are altered in cancer. More recently, proteins involved in signaling cascades, regulating cell proliferation, and anti-apoptotic protein factors were found to interact with copper. Therefore, therapeutic strategies using copper complexing molecules have been proposed for cancer therapy and used in clinical trials. OBJECTIVES This review will focus on novel findings about the involvement of copper and cupro-proteins in cancer dissemination process, epithelium to mesenchymal transition and vascularization. Particularly, implication of well-established (e.g. lysil oxidase) or newly identified copper-binding proteins (e.g. MEMO1), as well as their interplay, will be discussed. Moreover, we will describe recently synthesized copper complexes, including plant-derived ones, and their efficacy in contrasting cancer development. CONCLUSIONS The research on the involvement of copper in cancer is still an open field. Further investigation is required to unveil the mechanisms involved in copper delivery to the novel copper-binding proteins, which may identify other possible gene and protein targets for cancer therapy.
Collapse
Affiliation(s)
| | - Anna Barile
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| | - Mario Arciello
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| | - Luisa Rossi
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
13
|
Hsu HW, Bondy SC, Kitazawa M. Environmental and Dietary Exposure to Copper and Its Cellular Mechanisms Linking to Alzheimer's Disease. Toxicol Sci 2019; 163:338-345. [PMID: 29409005 DOI: 10.1093/toxsci/kfy025] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Metals are commonly found in the environment, household, and workplaces in various forms, and a significant segment of the population is routinely exposed to the trace amount of metals from variety of sources. Exposure to metals, such as aluminum, lead, iron, and copper, from environment has long been debated as a potential environmental risk factor for Alzheimer's disease (AD) for decades, yet results from in vitro, in vivo, and human population remain controversial. In the case of copper, the neurotoxic mechanism of action was classically viewed as its strong affinity to amyloid-beta (Aβ) to help its aggregation and increase oxidative stress via Fenton reaction. Thus, it has been thought that accumulation of copper mediates neurotoxicity, and removing it from the brain prevents or reverse Aβ plaque burden. Recent evidence, however, suggests dyshomeostasis of copper and its valency in the body, instead of the accumulation and interaction with Aβ, are major determinants of its beneficial effects as an essential metal or its neurotoxic counterpart. This notion is also supported by the fact that genetic loss-of-function mutations on copper transporters lead to severe neurological symptoms. Along with its altered distribution, recent studies have also proposed novel mechanisms of copper neurotoxicity mediated by nonneuronal cell lineages in the brain, such as capillary endothelial cells, leading to development of AD neuropathology. This review covers recent findings of multifactorial toxic mechanisms of copper and discusses the risk of environmental exposure as a potential factor in accounting for the variability of AD incidence.
Collapse
Affiliation(s)
- Heng-Wei Hsu
- Department of Medicine, Center for Occupational and Environmental Health, University of California, Irvine, California 92617
| | - Stephen C Bondy
- Department of Medicine, Center for Occupational and Environmental Health, University of California, Irvine, California 92617
| | - Masashi Kitazawa
- Department of Medicine, Center for Occupational and Environmental Health, University of California, Irvine, California 92617
| |
Collapse
|
14
|
Young TR, Pukala TL, Cappai R, Wedd AG, Xiao Z. The Human Amyloid Precursor Protein Binds Copper Ions Dominated by a Picomolar-Affinity Site in the Helix-Rich E2 Domain. Biochemistry 2018; 57:4165-4176. [DOI: 10.1021/acs.biochem.8b00572] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Tessa R. Young
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tara L. Pukala
- Discipline of Chemistry, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Roberto Cappai
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anthony G. Wedd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Zhiguang Xiao
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
15
|
Ilyechova EY, Puchkova LV, Shavlovskii MM, Korzhevskii DE, Petrova ES, Tsymbalenko NV. Effect of Silver Ions on Copper Metabolism during Mammalian Ontogenesis. Russ J Dev Biol 2018. [DOI: 10.1134/s1062360418030037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Investigation of Trace Elements in the Hair and Nail of Patients with Stomach Cancer. Indian J Clin Biochem 2017; 33:450-455. [PMID: 30319192 DOI: 10.1007/s12291-017-0693-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/31/2017] [Indexed: 10/18/2022]
Abstract
Trace metals are beneficial nutrient materials that act as essential cofactors in physiological processes. Recent evidence suggests that increase or decrease in certain trace metals may be related with risk and development of chronic diseases such as cancer. This study analyzed some trace elements level in hair and nail of patients with stomach cancer, and compared with their level in healthy controls. Trace elements (Cu, Fe, K, Li, Mg, Mn, Na, P, Se, Sr and Zn) are estimated in hair and nail of the 73 cancer patients and 83 controls by atomic absorption spectrophotometric method. The levels of Cu, K, Li, P and Se in hair and nail samples, were significantly higher in cases than controls. Levels of Mg and Sr were significantly lower in cases than controls. Fe level in hair samples was significantly higher in cases than controls. The mean concentrations of Fe, Se and P significantly increased with increasing cancer stage in the hair of patients. The average concentration of k also significantly increased with increasing cancer stage in the nail of patients. The results of our study show that there is an association between the increase in Cu, K, Li, P, Se and Fe, and stomach cancer development. Our results reveal that the increase in the trace elements could be a potential diagnostic marker to predict cancer progression and its etiology.
Collapse
|
17
|
Copper- and Zinc-Promoted Interdomain Structure in the Prion Protein: A Mechanism for Autoinhibition of the Neurotoxic N-Terminus. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:35-56. [PMID: 28838668 DOI: 10.1016/bs.pmbts.2017.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The function of the cellular prion protein (PrPC), while still poorly understood, is increasingly linked to its ability to bind physiological metal ions at the cell surface. PrPC binds divalent forms of both copper and zinc through its unstructured N-terminal domain, modulating interactions between PrPC and various receptors at the cell surface and ultimately tuning downstream cellular processes. In this chapter, we briefly discuss the molecular features of copper and zinc uptake by PrPC and summarize evidence implicating these metal ions in PrP-mediated physiology. We then focus our review on recent biophysical evidence revealing a physical interaction between the flexible N-terminal and globular C-terminal domains of PrPC. This interdomain cis interaction is electrostatic in nature and is promoted by the binding of Cu2+ and Zn2+ to the N-terminal octarepeat domain. These findings, along with recent cellular studies, suggest a mechanism whereby NC interactions serve to regulate the activity and/or toxicity of the PrPC N-terminus. We discuss this potential mechanism in relation to familial prion disease mutations, lethal deletions of the PrPC central region, and neurotoxicity induced by certain globular domain ligands, including bona fide prions and toxic amyloid-β oligomers.
Collapse
|
18
|
Dirksen K, Spee B, Penning LC, van den Ingh TSGAM, Burgener IA, Watson AL, Groot Koerkamp M, Rothuizen J, van Steenbeek FG, Fieten H. Gene expression patterns in the progression of canine copper-associated chronic hepatitis. PLoS One 2017; 12:e0176826. [PMID: 28459846 PMCID: PMC5411060 DOI: 10.1371/journal.pone.0176826] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 04/18/2017] [Indexed: 12/26/2022] Open
Abstract
Copper is an essential trace element, but can become toxic when present in abundance. The severe effects of copper-metabolism imbalance are illustrated by the inherited disorders Wilson disease and Menkes disease. The Labrador retriever dog breed is a novel non-rodent model for copper-storage disorders carrying mutations in genes known to be involved in copper transport. Besides disease initiation and progression of copper accumulation, the molecular mechanisms and pathways involved in progression towards copper-associated chronic hepatitis still remain unclear. Using expression levels of targeted candidate genes as well as transcriptome micro-arrays in liver tissue of Labrador retrievers in different stages of copper-associated hepatitis, pathways involved in progression of the disease were studied. At the initial phase of increased hepatic copper levels, transcriptomic alterations in livers mainly revealed enrichment for cell adhesion, developmental, inflammatory, and cytoskeleton pathways. Upregulation of targeted MT1A and COMMD1 mRNA shows the liver's first response to rising intrahepatic copper concentrations. In livers with copper-associated hepatitis mainly an activation of inflammatory pathways is detected. Once the hepatitis is in the chronic stage, transcriptional differences are found in cell adhesion adaptations and cytoskeleton remodelling. In view of the high similarities in copper-associated hepatopathies between men and dog extrapolation of these dog data into human biomedicine seems feasible.
Collapse
Affiliation(s)
- Karen Dirksen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Louis C. Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | | | - Iwan A. Burgener
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department für Kleintiere und Pferde, Veterinärmedizinische Universität Wien, Vienna, Austria
| | | | | | - Jan Rothuizen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Frank G. van Steenbeek
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Hille Fieten
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
19
|
Walls KL, Boulic M, Boddy JWD. The Built Environment-A Missing "Cause of the Causes" of Non-Communicable Diseases. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:E956. [PMID: 27690064 PMCID: PMC5086695 DOI: 10.3390/ijerph13100956] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 11/16/2022]
Abstract
The United Nations "25 × 25 Strategy" of decreasing non-communicable diseases (NCDs), including cardiovascular diseases, diabetes, cancer and chronic respiratory diseases, by 25% by 2025 does not appear to take into account all causes of NCDs. Its focus is on a few diseases, which are often linked with life-style factors with "voluntary" "modifiable behavioral risk factors" causes tending towards an over-simplification of the issues. We propose to add some aspects of our built environment related to hazardous building materials, and detailed form of the construction of infrastructure and buildings, which we think are some of the missing causes of NCDs. Some of these could be termed "involuntary causes", as they relate to factors that are beyond the control of the general public.
Collapse
Affiliation(s)
- Kelvin L Walls
- Building Code Consultants Ltd., P.O. Box 99613, Newmarket, Auckland 1149, New Zealand.
| | - Mikael Boulic
- School of Engineering and Advanced Technology, Massey University, Auckland 0745, New Zealand.
| | - John W D Boddy
- Urban Planning and Environmental Services, MWH Stantec, Level 3, 111 Carlton Gore Road, Auckland 0745, New Zealand.
| |
Collapse
|
20
|
Evans EGB, Pushie MJ, Markham KA, Lee HW, Millhauser GL. Interaction between Prion Protein's Copper-Bound Octarepeat Domain and a Charged C-Terminal Pocket Suggests a Mechanism for N-Terminal Regulation. Structure 2016; 24:1057-67. [PMID: 27265848 DOI: 10.1016/j.str.2016.04.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/19/2016] [Accepted: 04/20/2016] [Indexed: 11/29/2022]
Abstract
Copper plays a critical role in prion protein (PrP) physiology. Cu(2+) binds with high affinity to the PrP N-terminal octarepeat (OR) domain, and intracellular copper promotes PrP expression. The molecular details of copper coordination within the OR are now well characterized. Here we examine how Cu(2+) influences the interaction between the PrP N-terminal domain and the C-terminal globular domain. Using nuclear magnetic resonance and copper-nitroxide pulsed double electron-electron resonance, with molecular dynamics refinement, we localize the position of Cu(2+) in its high-affinity OR-bound state. Our results reveal an interdomain cis interaction that is stabilized by a conserved, negatively charged pocket of the globular domain. Interestingly, this interaction surface overlaps an epitope recognized by the POM1 antibody, the binding of which drives rapid cerebellar degeneration mediated by the PrP N terminus. The resulting structure suggests that the globular domain regulates the N-terminal domain by binding the Cu(2+)-occupied OR within a complementary pocket.
Collapse
Affiliation(s)
- Eric G B Evans
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - M Jake Pushie
- Department of Geological Sciences, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Kate A Markham
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Hsiau-Wei Lee
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Glenn L Millhauser
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
21
|
Hare DJ, Rembach A, Roberts BR. The Emerging Role of Metalloproteomics in Alzheimer's Disease Research. Methods Mol Biol 2016; 1303:379-89. [PMID: 26235079 DOI: 10.1007/978-1-4939-2627-5_22] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Metals are increasingly recognized to have an important role in molecular processes underlying Alzheimer's disease (AD). This chapter discusses the current role of metals in AD and expands on the development of metalloproteomics and how the recent advances in analytical technology will allow detailed investigation of metalloproteins. Investigation of individual metalloproteins will yield new mechanistic details about the role of metals in AD.
Collapse
Affiliation(s)
- Dominic J Hare
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | | | | |
Collapse
|
22
|
Gokhale A, Vrailas-Mortimer A, Larimore J, Comstra HS, Zlatic SA, Werner E, Manvich DF, Iuvone PM, Weinshenker D, Faundez V. Neuronal copper homeostasis susceptibility by genetic defects in dysbindin, a schizophrenia susceptibility factor. Hum Mol Genet 2015. [PMID: 26199316 DOI: 10.1093/hmg/ddv282] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Environmental factors and susceptible genomes interact to determine the risk of neurodevelopmental disorders. Although few genes and environmental factors have been linked, the intervening cellular and molecular mechanisms connecting a disorder susceptibility gene with environmental factors remain mostly unexplored. Here we focus on the schizophrenia susceptibility gene DTNBP1 and its product dysbindin, a subunit of the BLOC-1 complex, and describe a neuronal pathway modulating copper metabolism via ATP7A. Mutations in ATP7A result in Menkes disease, a disorder of copper metabolism. Dysbindin/BLOC-1 and ATP7A genetically and biochemically interact. Furthermore, disruption of this pathway causes alteration in the transcriptional profile of copper-regulatory and dependent factors in the hippocampus of dysbindin/BLOC-1-null mice. Dysbindin/BLOC-1 loss-of-function alleles do not affect cell and tissue copper content, yet they alter the susceptibility to toxic copper challenges in both mammalian cells and Drosophila. Our results demonstrate that perturbations downstream of the schizophrenia susceptibility gene DTNBP1 confer susceptibility to copper, a metal that in excess is a neurotoxin and whose depletion constitutes a micronutrient deficiency.
Collapse
Affiliation(s)
- Avanti Gokhale
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | | | | | - Heather S Comstra
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | | | - Erica Werner
- Department of Biochemistry, Emory University, Atlanta, GA 30322, USA
| | - Daniel F Manvich
- Department of Human Genetics, Emory University, Atlanta, GA 30322, USA
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University, Atlanta, GA 30322, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA 30322, USA
| | - Victor Faundez
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA, Center for Social Translational Neuroscience, Emory University, Atlanta, GA 30322, USA,
| |
Collapse
|
23
|
Bellingham SA, Guo B, Hill AF. The secret life of extracellular vesicles in metal homeostasis and neurodegeneration. Biol Cell 2015; 107:389-418. [PMID: 26032945 DOI: 10.1111/boc.201500030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/27/2015] [Indexed: 12/21/2022]
Abstract
Biologically active metals such as copper, zinc and iron are fundamental for sustaining life in different organisms with the regulation of cellular metal homeostasis tightly controlled through proteins that coordinate metal uptake, efflux and detoxification. Many of the proteins involved in either uptake or efflux of metals are localised and function on the plasma membrane, traffic between intracellular compartments depending upon the cellular metal environment and can undergo recycling via the endosomal pathway. The biogenesis of exosomes also occurs within the endosomal system, with several major neurodegenerative disease proteins shown to be released in association with these vesicles, including the amyloid-β (Aβ) peptide in Alzheimer's disease and the infectious prion protein involved in Prion diseases. Aβ peptide and the prion protein also bind biologically active metals and are postulated to play important roles in metal homeostasis. In this review, we will discuss the role of extracellular vesicles in Alzheimer's and Prion diseases and explore their potential contribution to metal homeostasis.
Collapse
Affiliation(s)
- Shayne A Bellingham
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Belinda Guo
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Andrew F Hill
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Transcriptional response to copper excess and identification of genes involved in heavy metal tolerance in the extremophilic microalga Chlamydomonas acidophila. Extremophiles 2015; 19:657-72. [DOI: 10.1007/s00792-015-0746-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/23/2015] [Indexed: 01/05/2023]
|
25
|
Quiroz N, Rivas N, del Pozo T, Burkhead J, Suazo M, González M, Latorre M. Transcriptional activation of glutathione pathways and role of glucose homeostasis during copper imbalance. Biometals 2015; 28:321-8. [DOI: 10.1007/s10534-015-9834-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 02/05/2015] [Indexed: 12/18/2022]
|
26
|
Hou P, Liu G, Zhao Y, Shi Z, Zheng Q, Bu G, Xu H, Zhang YW. Role of copper and the copper-related protein CUTA in mediating APP processing and Aβ generation. Neurobiol Aging 2014; 36:1310-5. [PMID: 25557959 DOI: 10.1016/j.neurobiolaging.2014.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 11/11/2014] [Accepted: 12/04/2014] [Indexed: 11/16/2022]
Abstract
One major pathologic hallmark and trigger of Alzheimer's disease (AD) is overproduction and accumulation of β-amyloid (Aβ) species in the brain. Aβ is derived from β-amyloid precursor protein (APP) through sequential cleavages by β- and γ-secretases. Abnormal copper homeostasis also contributes to AD pathogenesis. Recently, we find that a copper-related protein, CutA divalent cation tolerance homolog of Escherichia coli (CUTA), interacts with the β-secretase β-site APP cleaving enzyme 1 (BACE1) and inhibits APP β-processing and Aβ generation. Herein, we further found that overexpression of CUTA increases intracellular copper level, whereas copper treatments promote CUTA expression. We also confirmed that copper treatments promote APP expression and Aβ secretion. In addition, copper treatments promoted the increase of Aβ secretion induced by CUTA downregulation but had no effect on CUTA-β-site APP cleaving enzyme 1 interaction. On the other hand, CUTA overexpression ameliorated copper-induced Aβ secretion but had no effect on APP expression. Moreover, we found that Aβ treatments can reduce both CUTA and copper levels in mouse primary neurons. Consistently, both CUTA and copper levels were decreased in the hippocampus of APP/PS1 AD mouse brain. Together, our results reveal a reciprocal modulation of copper and CUTA and suggest that both regulate Aβ generation through different mechanisms, although Aβ mutually affects copper and CUTA levels.
Collapse
Affiliation(s)
- Ping Hou
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, China; School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Guiying Liu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, China
| | - Yingjun Zhao
- Degenerative Disease Research Program, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Zhun Shi
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, China
| | - Qiuyang Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, China
| | - Guojun Bu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, China; Degenerative Disease Research Program, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Yun-wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
27
|
Gough M, Blanthorn-Hazell S, Delury C, Parkin E. The E1 copper binding domain of full-length amyloid precursor protein mitigates copper-induced growth inhibition in brain metastatic prostate cancer DU145 cells. Biochem Biophys Res Commun 2014; 453:741-7. [PMID: 25305487 PMCID: PMC4256156 DOI: 10.1016/j.bbrc.2014.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/02/2014] [Indexed: 02/04/2023]
Abstract
Copper plays an important role in the aetiology and growth of tumours and levels of the metal are increased in the serum and tumour tissue of patients affected by a range of cancers including prostate cancer (PCa). The molecular mechanisms that enable cancer cells to proliferate in the presence of elevated copper levels are, therefore, of key importance in our understanding of tumour growth progression. In the current study, we have examined the role played by the amyloid precursor protein (APP) in mitigating copper-induced growth inhibition of the PCa cell line, DU145. A range of APP molecular constructs were stably over-expressed in DU145 cells and their effects on cell proliferation in the presence of copper were monitored. Our results show that endogenous APP expression was induced by sub-toxic copper concentrations in DU145 cells and over-expression of the wild-type protein was able to mitigate copper-induced growth inhibition via a mechanism involving the cytosolic and E1 copper binding domains of the full-length protein. APP likely represents one of a range of copper binding proteins that PCa cells employ in order to ensure efficient proliferation despite elevated concentrations of the metal within the tumour microenvironment. Targeting the expression of such proteins may contribute to therapeutic strategies for the treatment of cancers.
Collapse
Affiliation(s)
- Mallory Gough
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK.
| | - Sophee Blanthorn-Hazell
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK.
| | - Craig Delury
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK.
| | - Edward Parkin
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK.
| |
Collapse
|
28
|
Karimi A, Madjd Z, Habibi L, Akrami SM. Evaluating the extent of LINE-1 mobility following exposure to heavy metals in HepG2 cells. Biol Trace Elem Res 2014; 160:143-51. [PMID: 24894828 DOI: 10.1007/s12011-014-0015-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 05/09/2014] [Indexed: 12/23/2022]
Abstract
The long interspersed elements-1 (LINE1 or L1 retrotransposon) constitute 17% of the human genome and retain mobility properties within the genome. At present, 80-100 human L1 elements are thought to be active in the genome. The mobilization of these active elements may be influenced upon exposure to the heavy metals. In the present study, we evaluated the association of aluminum, lead, and copper exposure with L1 retrotransposition in human hepatocellular carcinoma (HepG2) cell line. An in vitro retrotransposition assay using an enhanced green fluorescent protein (EGFP)-tagged L1RP cassette was established to track EGFP shining as the mark of retrotransposition. Following determination of noncytotoxic concentrations of these metals, pL1RP-EGFP-transfected HepG2 cells were subjected to long-term treatment. Flow cytometry analysis of cells treated with various concentrations of these metals along with quantitative real-time PCR was used to quantify L1 retrotransposition frequencies. Aluminum significantly increased L1 retrotransposition frequency, while no significant association was found concerning lead exposure and L1 retrotransposition. Copper treatment downregulated L1 retrotransposition as a result of EGFP-tagged L1RP expression. Our findings suggest that aluminum might have the potential to cause genomic instability by the enhancement of L1 mobilization. Thus, the risk of induced L1 retrotransposition should be considered during drug safety evaluation and risk assessments of exposure to toxic environmental agents. Further studies are needed for a more robust assay to evaluate any associations between long-term lead exposure and L1 mobility in cell culture assay.
Collapse
Affiliation(s)
- Abbas Karimi
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine (FATiM), Iran University of Medical Sciences, Tehran, Iran
| | | | | | | |
Collapse
|
29
|
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
|
30
|
Wong BX, Hung YH, Bush AI, Duce JA. Metals and cholesterol: two sides of the same coin in Alzheimer's disease pathology. Front Aging Neurosci 2014; 6:91. [PMID: 24860500 PMCID: PMC4030154 DOI: 10.3389/fnagi.2014.00091] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/28/2014] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease. It begins years prior to the onset of clinical symptoms, such as memory loss and cognitive decline. Pathological hallmarks of AD include the accumulation of β-amyloid in plaques and hyperphosphorylated tau in neurofibrillary tangles. Copper, iron, and zinc are abnormally accumulated and distributed in the aging brain. These metal ions can adversely contribute to the progression of AD. Dysregulation of cholesterol metabolism has also been implicated in the development of AD pathology. To date, large bodies of research have been carried out independently to elucidate the role of metals or cholesterol on AD pathology. Interestingly, metals and cholesterol affect parallel molecular and biochemical pathways involved in AD pathology. The possible links between metal dyshomeostasis and altered brain cholesterol metabolism in AD are reviewed.
Collapse
Affiliation(s)
- Bruce X Wong
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - Ya Hui Hung
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - Ashley I Bush
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - James A Duce
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia ; School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds Leeds, North Yorkshire, UK
| |
Collapse
|
31
|
Could Intracrine Biology Play a Role in the Pathogenesis of Transmissable Spongiform Encephalopathies Alzheimer’s Disease and Other Neurodegenerative Diseases? Am J Med Sci 2014; 347:312-20. [DOI: 10.1097/maj.0b013e3182a28af3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
32
|
Song JW, Choi BS. Mercury induced the Accumulation of Amyloid Beta (Aβ) in PC12 Cells: The Role of Production and Degradation of Aβ. Toxicol Res 2014; 29:235-40. [PMID: 24578793 PMCID: PMC3936175 DOI: 10.5487/tr.2013.29.4.235] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 12/16/2013] [Accepted: 12/17/2013] [Indexed: 01/15/2023] Open
Abstract
Extracellular accumulation of amyloid beta protein (Aβ) plays a central role in Alzheimer’s disease (AD). Some metals, such as copper, lead, and aluminum can affect the Aβ accumulation in the brain. However, the effect of mercury on Aβ accumulation in the brain is not clear. Thus, this study was proposed to estimate whether mercury concentration affects Aβ accumulation in PC12 cells. We treated 10, 100, and 1000 nM HgCl2 (Hg) or CH3HgCl2 (MeHg) for 48 hr in PC12 cells. After treatment, Aβ40 in culture medium increased in a dose- and time-dependent manner. Hg and MeHg increased amyloid precursor protein (APP), which is related to Aβ production. Neprilysin (NEP) levels in PC12 cells were decreased by Hg and MeHg treatment. These results suggested that Hg induced Aβ accumulation through APP overproduction and reduction of NEP.
Collapse
Affiliation(s)
- Ji-Won Song
- Department of Preventive Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Byung-Sun Choi
- Department of Preventive Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| |
Collapse
|
33
|
Gutiérrez-García R, del Pozo T, Suazo M, Cambiazo V, González M. Physiological copper exposure in Jurkat cells induces changes in the expression of genes encoding cholesterol biosynthesis proteins. Biometals 2013; 26:1033-40. [PMID: 24170205 DOI: 10.1007/s10534-013-9680-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 10/17/2013] [Indexed: 10/26/2022]
Abstract
Copper is an essential micronutrient that functions as an enzymatic cofactor in a wide range of cellular processes. Although adequate Cu levels are essential for normal metabolism, excess Cu can be toxic to cells. Cellular responses to copper deficiency and overload involve changes in the expression of genes directly and indirectly involved in copper metabolism. However little is known on the effect of physiological copper concentration on gene expression changes. In the current study we aimed to establish whether the expression of genes encoding enzymes related to cholesterol (hmgcs1, hmgcr, fdft) and fatty acid biosynthesis and LDL receptor can be induced by an iso-physiological copper concentration. The iso-physiological copper concentration was determined as the bioavailable plasmatic copper in a healthy adult population. In doing so, two blood cell lines (Jurkat and THP-1) were exposed for 6 or 24 h to iso- or supraphysiological copper concentrations. Our results indicated that in cells exposed to an iso-physiological copper concentration the early induction of genes involved in lipid metabolism was not mediated by copper itself but by the modification of the cellular redox status. Thus our results contributed to understand the involvement of copper in the regulation of cholesterol metabolism under physiological conditions.
Collapse
Affiliation(s)
- Ricardo Gutiérrez-García
- Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, El Líbano 5524, Macul, Santiago, Chile
| | | | | | | | | |
Collapse
|
34
|
Hung YH, Bush AI, La Fontaine S. Links between copper and cholesterol in Alzheimer's disease. Front Physiol 2013; 4:111. [PMID: 23720634 PMCID: PMC3655288 DOI: 10.3389/fphys.2013.00111] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 04/30/2013] [Indexed: 01/01/2023] Open
Abstract
Altered copper homeostasis and hypercholesterolemia have been identified independently as risk factors for Alzheimer's disease (AD). Abnormal copper and cholesterol metabolism are implicated in the genesis of amyloid plaques and neurofibrillary tangles (NFT), which are two key pathological signatures of AD. Amyloidogenic processing of a sub-population of amyloid precursor protein (APP) that produces Aβ occurs in cholesterol-rich lipid rafts in copper deficient AD brains. Co-localization of Aβ and a paradoxical high concentration of copper in lipid rafts fosters the formation of neurotoxic Aβ:copper complexes. These complexes can catalytically oxidize cholesterol to generate H2O2, oxysterols and other lipid peroxidation products that accumulate in brains of AD cases and transgenic mouse models. Tau, the core protein component of NFTs, is sensitive to interactions with copper and cholesterol, which trigger a cascade of hyperphosphorylation and aggregation preceding the generation of NFTs. Here we present an overview of copper and cholesterol metabolism in the brain, and how their integrated failure contributes to development of AD.
Collapse
Affiliation(s)
- Ya Hui Hung
- Oxidation Biology Laboratory, Florey Institute of Neuroscience and Mental Health Parkville, VIC, Australia ; Centre for Neuroscience Research, The University of Melbourne Parkville, VIC, Australia
| | | | | |
Collapse
|
35
|
Greenough MA, Camakaris J, Bush AI. Metal dyshomeostasis and oxidative stress in Alzheimer’s disease. Neurochem Int 2013; 62:540-55. [DOI: 10.1016/j.neuint.2012.08.014] [Citation(s) in RCA: 266] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/13/2012] [Accepted: 08/30/2012] [Indexed: 01/21/2023]
|
36
|
Lee BH, Kim JH, Kim JM, Heo SH, Kang M, Kim GH, Choi JH, Yoo HW. The early molecular processes underlying the neurological manifestations of an animal model of Wilson's disease. Metallomics 2013; 5:532-40. [PMID: 23519153 DOI: 10.1039/c3mt20243g] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The Long-Evans Cinnamon (LEC) rat shows age-dependent hepatic manifestations that are similar to those of Wilson's disease (WD). The pathogenic process in the brain has, however, not been evaluated in detail due to the rarity of the neurological symptoms. However, copper accumulation is noted in LEC rat brain tissue from 24 weeks of age, which results in oxidative injuries. The current study investigated the gene expression profiles of LEC rat brains at 24 weeks of age in order to identify the important early molecular changes that underlie the development of neurological symptoms in WD. Biological ontology-based analysis revealed diverse altered expressions of the genes related to copper accumulation. Of particular interest, we found altered expression of genes connected to mitochondrial respiration (Sdhaf2 and Ndufb7), calcineurin-mediated cellular processes (Ppp3ca, Ppp3cb, and Camk2a), amyloid precursor protein (Anks1b and A2m) and alpha-synuclein (Snca). In addition to copper-related changes, compensatory upregulations of Cp and Hamp reflect iron-mediated neurotoxicity. Of note, reciprocal expression of Asmt and Bhmt is an important clue that altered S-adenosylhomocysteine metabolism underlies brain injury in WD, which is directly correlated to the decreased expression of S-adenosylhomocysteine hydrolase in hepatic tissue in LEC rats. In conclusion, our study indicates that diverse molecular changes, both variable and complex, underlie the development of neurological manifestations in WD. Copper-related injuries were found to be the principal pathogenic process, but Fe- or adenosylhomocysteine-related injuries were also implicated. Investigations using other animal models or accessible human samples will be required to confirm our observations.
Collapse
Affiliation(s)
- Beom Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Linder MC. The relationship of copper to DNA damage and damage prevention in humans. Mutat Res 2013; 733:83-91. [PMID: 23463874 DOI: 10.1016/j.mrfmmm.2012.03.010] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Copper ions are well suited to facilitate formation of reactive oxygen species (ROS) that can damage biomolecules, including DNA and chromatin. That this can occur in vitro with isolated DNA or chromatin,or by exposure of cultured mammalian cells to copper complexed with various agents, has been well demonstrated. Whether that is likely to occur in vivo is not as clear. This review addresses the question of whether and how copper ions or complexes – in forms that could be present in vivo, damage DNA and chromosome structure and/or promote epigenetic changes that can lead to pathology and diseases, including cancer and neurological conditions such as Alzheimer's disease, Lewy body dementias, and spongiform encephalopathies. This question is considered in light of our knowledge that copper-dependent enzymes are important contributors to antioxidant defense, and that the mammalian organism has robust mechanisms for maintaining constant levels of copper not only in body fluids but in its major organs. Overall,and except in unusual genetic states that lead to copper overload in specific cells (particularly those in liver), it appears that excessive intake of copper is not a significant factor in the development of disease states.
Collapse
Affiliation(s)
- Maria C Linder
- Department of Chemistry and Biochemistry, California State University, Fullerton, CA 92834-6866, USA.
| |
Collapse
|
38
|
Copper, zinc and iron in neurodegenerative diseases (Alzheimer's, Parkinson's and prion diseases). Coord Chem Rev 2012. [DOI: 10.1016/j.ccr.2012.03.013] [Citation(s) in RCA: 306] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
39
|
Martin GR, Alvarez AL, Bashashati M, Keenan CM, Jirik FR, Sharkey KA. Endogenous cellular prion protein regulates contractility of the mouse ileum. Neurogastroenterol Motil 2012; 24:e412-24. [PMID: 22762267 DOI: 10.1111/j.1365-2982.2012.01970.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Cellular prion protein (PrP(C) ) is expressed in the enteric nervous system (ENS), however, its physiological role has not been identified. Studies suggest that PrP(C) can function as a metal-binding protein, as absence of the protein has been linked to altered copper metabolism and atypical synaptic activity. Because copper is known to modulate smooth muscle relaxation, we tested the hypothesis that PrP(C) deficiency would alter intestinal contractility. METHODS We examined electrically evoked ileal contractility in Prnp(-/-) or wild type littermate mice and the effects of copper or copper chelation. PrP(C) expression was studied in whole mount ileal preparations of mice and guinea pigs by immunohistochemistry. KEY RESULTS Relative to wild type mice, ileal tissues of Prnp(-/-) mice exhibited reduced electrical field stimulation (EFS)-evoked contractility. Furthermore, EFS-induced relaxation, as a percentage of that induced by a nitric oxide donor, was enhanced. Addition of a copper donor to the organ bath increased, whereas the addition of a copper chelator inhibited, nitric oxide donor-induced ileal relaxation in Prnp(-/-) mice. PrP(C) was expressed on nerve fibers or terminals, and some cell bodies in the myenteric and submucosal plexuses of wild type mice. PrP(C) colocalized with a neuron-specific ectonucleotidase, nucleoside triphosphate diphosphohydrolase 3 (NTPDase3), but to only a limited extent with GFAP, a marker of enteric glia. Guinea pigs expressed PrP(C) in nerve fibers or terminals and enteric glia in the myenteric and submucosal plexuses. CONCLUSIONS & INFERENCES Our findings suggest that PrP(C) , which is abundant in the ENS, has a role in the regulation of ileal contractility.
Collapse
Affiliation(s)
- G R Martin
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| | | | | | | | | | | |
Collapse
|
40
|
Disruption of copper homeostasis due to a mutation of Atp7a delays the onset of prion disease. Proc Natl Acad Sci U S A 2012; 109:13733-8. [PMID: 22869751 DOI: 10.1073/pnas.1211499109] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Copper influences the pathogenesis of prion disease, but whether it is beneficial or detrimental remains controversial. Copper homeostasis is also essential for normal physiology, as highlighted by the spectrum of diseases caused by disruption of the copper transporting enzymes ATP7A and ATP7B. Here, by using a forward genetics approach in mice, we describe the isolation of three alleles of Atp7a, each with different phenotypic consequences. The mildest of the three, Atp7a(brown), was insufficient to cause lethality in hemizygotes or mottling of the coat in heterozygotes, but did lead to coat hypopigmentation and reduced copper content in the brains of hemizygous males. When challenged with Rocky Mountain Laboratory scrapie, the onset of prion disease was delayed in Atp7a(brown) mice, and significantly less proteinase-resistant prion protein was found in the brains of moribund Atp7a(brown) mice compared with WT littermates. Our results establish that ATP7A-mediated copper homeostasis is important for the formation of pathogenic proteinase-resistant prion protein.
Collapse
|
41
|
Urso E, Manno D, Serra A, Buccolieri A, Rizzello A, Danieli A, Acierno R, Salvato B, Maffia M. Role of the cellular prion protein in the neuron adaptation strategy to copper deficiency. Cell Mol Neurobiol 2012; 32:989-1001. [PMID: 22362149 DOI: 10.1007/s10571-012-9815-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 02/07/2012] [Indexed: 01/15/2023]
Abstract
Copper transporter 1 (CTR1), cellular prion protein (PrP(C)), natural resistance-associated macrophage protein 2 (NRAMP2) and ATP7A proteins control the cell absorption and efflux of copper (Cu) ions in nervous tissues upon physiological conditions. Little is known about their regulation under reduced Cu availability, a condition underlying the onset of diffused neurodegenerative disorders. In this study, rat neuron-like cells were exposed to Cu starvation for 48 h. The activation of Caspase-3 enzymes and the impairment of Cu,Zn superoxide dismutase (Cu,Zn SOD) activity depicted the initiation of a pro-apoptotic program, preliminary to the appearance of the morphological signs of apoptosis. The transcriptional response related to Cu transport proteins has been investigated. Notably, PrP(C) transcript and protein levels were consistently elevated upon Cu deficiency. The CTR1 protein amount was stable, despite a two-fold increase in the transcript amount, meaning the activation of post-translational regulatory mechanisms. NRAMP2 and ATP7A expressions were unvaried. The up-regulated PrP(C) has been demonstrated to enhance the cell Cu uptake ability by about 50% with respect to the basal transport, and so sustain the Cu delivery to the Cu,Zn SOD cuproenzymes. Conclusively, the study suggests a pivotal role for PrP(C) in the cell adaptation to Cu limitation through a direct activity of ion uptake. In this view, the PrP(C) accumulation observed in several cancer cell lines could be interpreted as a molecular marker of cell Cu deficiency and a potential target of therapeutic interventions against disorders caused by metal imbalances.
Collapse
Affiliation(s)
- Emanuela Urso
- Department of Biological and Environmental Sciences and Technologies, University of Salento, SP Lecce-Monteroni, Lecce, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Eskici G, Axelsen PH. Copper and Oxidative Stress in the Pathogenesis of Alzheimer’s Disease. Biochemistry 2012; 51:6289-311. [DOI: 10.1021/bi3006169] [Citation(s) in RCA: 204] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Gözde Eskici
- Departments of Pharmacology, Biochemistry and Biophysics,
and Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United
States
| | - Paul H. Axelsen
- Departments of Pharmacology, Biochemistry and Biophysics,
and Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United
States
| |
Collapse
|
43
|
Spoerri L, Vella LJ, Pham CLL, Barnham KJ, Cappai R. The amyloid precursor protein copper binding domain histidine residues 149 and 151 mediate APP stability and metabolism. J Biol Chem 2012; 287:26840-53. [PMID: 22685292 DOI: 10.1074/jbc.m112.355743] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
One of the key pathological hallmarks of Alzheimer disease (AD) is the accumulation of the APP-derived amyloid β peptide (Aβ) in the brain. Altered copper homeostasis has also been reported in AD patients and is thought to increase oxidative stress and to contribute to toxic Aβ accumulation and regulate APP metabolism. The potential involvement of the N-terminal APP copper binding domain (CuBD) in these events has not been investigated. Based on the tertiary structure of the APP CuBD, we examined the histidine residues of the copper binding site (His(147), His(149), and His(151)). We report that histidines 149 and 151 are crucial for CuBD stability and APP metabolism. Co-mutation of the APP CuBD His(149) and His(151) to asparagine decreased APP proteolytic processing, impaired APP endoplasmic reticulum-to-Golgi trafficking, and promoted aberrant APP oligomerization in HEK293 cells. Expression of the triple H147N/H149N/H151N-APP mutant led to up-regulation of the unfolded protein response. Using recombinant protein encompassing the APP CuBD, we found that insertion of asparagines at positions 149 and 151 altered the secondary structure of the domain. This study identifies two APP CuBD residues that are crucial for APP metabolism and suggests an additional role of this domain in APP folding and stability besides its previously identified copper binding activity. These findings are of major significance for the design of novel AD therapeutic drugs targeting this APP domain.
Collapse
Affiliation(s)
- Loredana Spoerri
- Department of Pathology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | | | | | | | | |
Collapse
|
44
|
Vázquez MC, del Pozo T, Robledo FA, Carrasco G, Pavez L, Olivares F, González M, Zanlungo S. Alteration of gene expression profile in Niemann-Pick type C mice correlates with tissue damage and oxidative stress. PLoS One 2011; 6:e28777. [PMID: 22216111 PMCID: PMC3245218 DOI: 10.1371/journal.pone.0028777] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 11/15/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Niemann-Pick type C disease (NPC) is a neurovisceral lipid storage disorder mainly characterized by unesterified cholesterol accumulation in lysosomal/late endosomal compartments, although there is also an important storage for several other kind of lipids. The main tissues affected by the disease are the liver and the cerebellum. Oxidative stress has been described in various NPC cells and tissues, such as liver and cerebellum. Although considerable alterations occur in the liver, the pathological mechanisms involved in hepatocyte damage and death have not been clearly defined. Here, we assessed hepatic tissue integrity, biochemical and oxidative stress parameters of wild-type control (Npc1(+/+); WT) and homozygous-mutant (Npc1(-/-); NPC) mice. In addition, the mRNA abundance of genes encoding proteins associated with oxidative stress, copper metabolism, fibrosis, inflammation and cholesterol metabolism were analyzed in livers and cerebella of WT and NPC mice. METHODOLOGY/PRINCIPAL FINDINGS We analyzed various oxidative stress parameters in the liver and hepatic and cerebellum gene expression in 7-week-old NPC1-deficient mice compared with control animals. We found signs of inflammation and fibrosis in NPC livers upon histological examination. These signs were correlated with increased levels of carbonylated proteins, diminished total glutathione content and significantly increased total copper levels in liver tissue. Finally, we analyzed liver and cerebellum gene expression patterns by qPCR and microarray assays. We found a correlation between fibrotic tissue and differential expression of hepatic as well as cerebellar genes associated with oxidative stress, fibrosis and inflammation in NPC mice. CONCLUSIONS/SIGNIFICANCE In NPC mice, liver disease is characterized by an increase in fibrosis and in markers associated with oxidative stress. NPC is also correlated with altered gene expression, mainly of genes involved in oxidative stress and fibrosis. These findings correlate with similar parameters in cerebellum, as has been previously reported in the NPC mice model.
Collapse
Affiliation(s)
- Mary C. Vázquez
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Talía del Pozo
- Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, Santiago, Chile
- FONDAP-Center of Genome Regulation (CGR), Santiago, Chile
| | - Fermín A. Robledo
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Leonardo Pavez
- Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, Santiago, Chile
| | - Felipe Olivares
- Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, Santiago, Chile
| | - Mauricio González
- Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, Santiago, Chile
- Laboratorio de Bioinformática y Matemáticas del Genoma, Centro de Modelamiento Matemático (CMM), Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Santiago, Chile
- FONDAP-Center of Genome Regulation (CGR), Santiago, Chile
| | - Silvana Zanlungo
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- FONDAP-Center of Genome Regulation (CGR), Santiago, Chile
- * E-mail:
| |
Collapse
|
45
|
Canello T, Friedman-Levi Y, Mizrahi M, Binyamin O, Cohen E, Frid K, Gabizon R. Copper is toxic to PrP-ablated mice and exacerbates disease in a mouse model of E200K genetic prion disease. Neurobiol Dis 2011; 45:1010-7. [PMID: 22198568 DOI: 10.1016/j.nbd.2011.12.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 11/04/2011] [Accepted: 12/04/2011] [Indexed: 01/10/2023] Open
Abstract
The pathogenesis of the diverse forms of prion disease was attributed solely to the accumulation of the misfolded PrP forms, and not to the potential loss of normal PrP(C) function during disease propagation. In this respect, it was also not established whether mutant PrPs linked to genetic prion diseases, as is the case for E200K PrP, preserve the function of PrP(C). We now show that fibroblasts generated from both PrP-ablated mice and TgMHu2ME199K, a transgenic mouse line mimicking E200KCJD, were significantly more sensitive to copper toxicity than wt fibroblasts. Long-term administration of copper significantly accelerated the onset and progression of spontaneous prion disease in TgMHu2ME199K mice and caused marked irritability and cerebellar associated tip-toe walking in PrP(0/0) mice, while wt mice were not affected. Our results are consistent with the hypothesis that a functional PrP(C) is required to protect cells from high levels of copper, and that its substitution for a nonfunctional mutant PrP may accelerate the onset of genetic prion disease during oxidative insults.
Collapse
Affiliation(s)
- Tamar Canello
- Department of Neurology, Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital, 91120 Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
46
|
Duce JA, Bush AI, Adlard PA. Role of amyloid-β–metal interactions in Alzheimer’s disease. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.11.43] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
There is an evolving field of metallobiology that has begun to describe a key role for bioavailable metals (particularly copper, zinc and iron) in the pathogenesis of Alzheimer’s disease (AD). In particular, there is an apparent failure in metal ion homeostasis, potentially caused by a pathological mislocalization of the metals in the brain, which appears to be an obligatory step in both the precipitation and potentiation of the disease. A number of both preclinical and clinical studies have also provided a strong burden of proof that normalizing metal ion homeostasis represents a valid therapeutic target, and may indeed represent the first disease-modifying strategy for AD. The role of metals in the pathophysiology of AD will be discussed in this article.
Collapse
Affiliation(s)
- James A Duce
- The Mental Health Research Institute, 155 Oak Street, Parkville, Victoria 3052, Australia
- Center for Neuroscience, The University of Melbourne, Victoria 3010, Australia
| | - Ashley I Bush
- The Mental Health Research Institute, 155 Oak Street, Parkville, Victoria 3052, Australia
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
| | | |
Collapse
|
47
|
Abstract
Fe and Cu could represent dietary risk factors for Alzheimer's disease (AD), which has become a global health concern. To establish the relationship between diets high in Cu and Fe and cognitive decline or AD, we have conducted a systematic review of the literature (up to January 2011). We identified two meta-analyses, two systematic reviews, eleven placebo-controlled trials, five observational studies, forty-five case-control studies, thirty autopsy and five uncontrolled studies, and one case report. There were eleven interventional trials that tried to either supplement or deplete Fe and Cu, but none of them provided clear evidence of a beneficial effect on cognitive performance in patients with AD. The prospective studies revealed an association between a diet simultaneously high in SFA and Cu and cognitive decline. Case-control and autopsy studies showed elevated Fe levels in the brains of AD patients, whereas the evidence was less consistent for Cu. In most of the studies, Cu concentrations were unchanged in the cerebrospinal fluid and the brain but increased in the serum. In conclusion, the existing data suggest that diets excessive in Fe or Cu, together with a high intake of SFA, should be avoided in the elderly who are not at risk of anaemia. Basic studies and, building on this, clinical investigations are needed to further elucidate in which dietary patterns and in which patient groups an Fe- and Cu-rich diet might foster the risk of developing AD.
Collapse
|
48
|
Acevedo KM, Hung YH, Dalziel AH, Li QX, Laughton K, Wikhe K, Rembach A, Roberts B, Masters CL, Bush AI, Camakaris J. Copper promotes the trafficking of the amyloid precursor protein. J Biol Chem 2010; 286:8252-8262. [PMID: 21177866 DOI: 10.1074/jbc.m110.128512] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Accumulation of the amyloid β peptide in the cortical and hippocampal regions of the brain is a major pathological feature of Alzheimer disease. Amyloid β peptide is generated from the sequential protease cleavage of the amyloid precursor protein (APP). We reported previously that copper increases the level of APP at the cell surface. Here we report that copper, but not iron or zinc, promotes APP trafficking in cultured polarized epithelial cells and neuronal cells. In SH-SY5Y neuronal cells and primary cortical neurons, copper promoted a redistribution of APP from a perinuclear localization to a wider distribution, including neurites. Importantly, a change in APP localization was not attributed to an up-regulation of APP protein synthesis. Using live cell imaging and endocytosis assays, we found that copper promotes an increase in cell surface APP by increasing its exocytosis and reducing its endocytosis, respectively. This study identifies a novel mechanism by which copper regulates the localization and presumably the function of APP, which is of major significance for understanding the role of APP in copper homeostasis and the role of copper in Alzheimer disease.
Collapse
Affiliation(s)
| | - Ya Hui Hung
- the Centre for Neuroscience, and; the Mental Health Research Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and
| | | | - Qiao-Xin Li
- the Mental Health Research Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and; the Department of Pathology
| | - Katrina Laughton
- the Mental Health Research Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and; the Department of Pathology
| | - Krutika Wikhe
- the Mental Health Research Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and
| | - Alan Rembach
- the Mental Health Research Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and; Commonwealth Scientific and Research Organization (CSIRO) Molecular and Health Technologies, Parkville, Victoria 3052, Australia
| | - Blaine Roberts
- the Mental Health Research Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and
| | - Colin L Masters
- the Centre for Neuroscience, and; the Mental Health Research Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and
| | - Ashley I Bush
- the Mental Health Research Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and
| | | |
Collapse
|
49
|
Lutsenko S, Bhattacharjee A, Hubbard AL. Copper handling machinery of the brain. Metallomics 2010; 2:596-608. [DOI: 10.1039/c0mt00006j] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
50
|
Wright JA, McHugh PC, Stockbridge M, Lane S, Kralovicova S, Brown DR. Activation and repression of prion protein expression by key regions of intron 1. Cell Mol Life Sci 2009; 66:3809-20. [PMID: 19756378 PMCID: PMC11115799 DOI: 10.1007/s00018-009-0154-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 09/01/2009] [Accepted: 09/02/2009] [Indexed: 10/20/2022]
Abstract
Expression of the prion protein is necessary for infection with prion diseases. Altered expression levels may play an important role in susceptibility to infection. Therefore, understanding the mechanisms that regulate prion protein expression is of great importance. It was previously shown that expression of the prion protein is to some degree regulated by an alternative promoter within intron 1. Studies using GFP and luciferase reporter systems were undertaken to determine key sites for the repression and activation of expression of the prion protein driven by intron 1. We identified a region within intron 1 sufficient to drive prion protein expression. Our findings highlight two potential repressor regions. Both regions have binding sites for the known repressor Hes-1. Hes-1 overexpression caused a dramatic decrease in PrP protein expression. Additionally, we have identified Atox-1 as a transcription factor that upregulates prion protein expression. These findings clearly indicate that intron 1 plays a key role in regulation of prion protein expression levels.
Collapse
Affiliation(s)
- Josephine A. Wright
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY UK
| | - Patrick C. McHugh
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY UK
| | - Mark Stockbridge
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY UK
| | - Samantha Lane
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY UK
| | - Silvia Kralovicova
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY UK
| | - David R. Brown
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY UK
| |
Collapse
|