1
|
Ferretti S, Zanella I. The Underestimated Role of Iron in Frontotemporal Dementia: A Narrative Review. Int J Mol Sci 2024; 25:12987. [PMID: 39684697 DOI: 10.3390/ijms252312987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The term frontotemporal dementia (FTD) comprises a group of neurodegenerative disorders characterized by the progressive degeneration of the frontal and temporal lobes of the brain with language impairment and changes in cognitive, behavioral and executive functions, and in some cases motor manifestations. A high proportion of FTD cases are due to genetic mutations and inherited in an autosomal-dominant manner with variable penetrance depending on the implicated gene. Iron is a crucial microelement that is involved in several cellular essential functions in the whole body and plays additional specialized roles in the central nervous system (CNS) mainly through its redox-cycling properties. Such a feature may be harmful under aerobic conditions, since it may lead to the generation of highly reactive hydroxyl radicals. Dysfunctions of iron homeostasis in the CNS are indeed involved in several neurodegenerative disorders, although it is still challenging to determine whether the dyshomeostasis of this essential but harmful metal is a direct cause of neurodegeneration, a contributor factor or simply a consequence of other neurodegenerative mechanisms. Unlike many other neurodegenerative disorders, evidence of the dysfunction in brain iron homeostasis in FTD is still scarce; nonetheless, the recent literature intriguingly suggests its possible involvement. The present review aims to summarize what is currently known about the contribution of iron dyshomeostasis in FTD based on clinical, imaging, histological, biochemical and molecular studies, further suggesting new perspectives and offering new insights for future investigations on this underexplored field of research.
Collapse
Affiliation(s)
- Sara Ferretti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Medical Genetics Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| |
Collapse
|
2
|
Zhong G, Wang X, Li J, Xie Z, Wu Q, Chen J, Wang Y, Chen Z, Cao X, Li T, Liu J, Wang Q. Insights Into the Role of Copper in Neurodegenerative Diseases and the Therapeutic Potential of Natural Compounds. Curr Neuropharmacol 2024; 22:1650-1671. [PMID: 38037913 PMCID: PMC11284712 DOI: 10.2174/1570159x22666231103085859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/23/2023] [Accepted: 06/05/2023] [Indexed: 12/02/2023] Open
Abstract
Neurodegenerative diseases encompass a collection of neurological disorders originating from the progressive degeneration of neurons, resulting in the dysfunction of neurons. Unfortunately, effective therapeutic interventions for these diseases are presently lacking. Copper (Cu), a crucial trace element within the human body, assumes a pivotal role in various biological metabolic processes, including energy metabolism, antioxidant defense, and neurotransmission. These processes are vital for the sustenance, growth, and development of organisms. Mounting evidence suggests that disrupted copper homeostasis contributes to numerous age-related neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), Wilson's disease (WD), Menkes disease (MD), prion diseases, and multiple sclerosis (MS). This comprehensive review investigates the connection between the imbalance of copper homeostasis and neurodegenerative diseases, summarizing pertinent drugs and therapies that ameliorate neuropathological changes, motor deficits, and cognitive impairments in these conditions through the modulation of copper metabolism. These interventions include Metal-Protein Attenuating Compounds (MPACs), copper chelators, copper supplements, and zinc salts. Moreover, this review highlights the potential of active compounds derived from natural plant medicines to enhance neurodegenerative disease outcomes by regulating copper homeostasis. Among these compounds, polyphenols are particularly abundant. Consequently, this review holds significant implications for the future development of innovative drugs targeting the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Wang
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiaqi Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhouyuan Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiqing Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaxin Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiyun Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziying Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianyao Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinman Liu
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
3
|
Han J. Copper trafficking systems in cells: insights into coordination chemistry and toxicity. Dalton Trans 2023; 52:15277-15296. [PMID: 37702384 DOI: 10.1039/d3dt02166a] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Transition metal ions, such as copper, are indispensable components in the biological system. Copper ions which primarily exist in two major oxidation states Cu(I) and Cu(II) play crucial roles in various cellular processes including antioxidant defense, biosynthesis of neurotransmitters, and energy metabolism, owing to their inherent redox activity. The disturbance in copper homeostasis can contribute to the development of copper metabolism disorders, cancer, and neurodegenerative diseases, highlighting the significance of understanding the copper trafficking system in cellular environments. This review aims to offer a comprehensive overview of copper homeostatic machinery, with an emphasis on the coordination chemistry of copper transporters and trafficking proteins. While copper chaperones and the corresponding metalloenzymes are thoroughly discussed, we also explore the potential existence of low-molecular-mass metal complexes within cellular systems. Furthermore, we summarize the toxicity mechanisms originating from copper deficiency or accumulation, which include the dysregulation of oxidative stress, signaling pathways, signal transduction, and amyloidosis. This perspective review delves into the current knowledge regarding the intricate aspects of the copper trafficking system, providing valuable insights into potential treatment strategies from the standpoint of bioinorganic chemistry.
Collapse
Affiliation(s)
- Jiyeon Han
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea.
| |
Collapse
|
4
|
Cellular prion protein offers neuroprotection in astrocytes submitted to amyloid β oligomer toxicity. Mol Cell Biochem 2022:10.1007/s11010-022-04631-w. [PMID: 36576715 DOI: 10.1007/s11010-022-04631-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022]
Abstract
The cellular prion protein (PrPC), in its native conformation, performs numerous cellular and cognitive functions in brain tissue. However, despite the cellular prion research in recent years, there are still questions about its participation in oxidative and neurodegenerative processes. This study aims to elucidate the involvement of PrPC in the neuroprotection cascade in the presence of oxidative stressors. For that, astrocytes from wild-type mice and knockout to PrPC were subjected to the induction of oxidative stress with hydrogen peroxide (H2O2) and with the toxic oligomer of the amyloid β protein (AβO). We observed that the presence of PrPC showed resistance in the cell viability of astrocytes. It was also possible to monitor changes in basic levels of metals and associate them with an induced damage condition, indicating the precise role of PrPC in metal homeostasis, where the absence of PrPC leads to metallic unbalance, culminating in cellular vulnerability to oxidative stress. Increased caspase 3, p-Tau, p53, and Bcl2 may establish a relationship between a PrPC and an induced damage condition. Complementarily, it has been shown that PrPC prevents the internalization of AβO and promotes its degradation under oxidative stress induction, thus preventing protein aggregation in astrocytes. It was also observed that the presence of PrPC can be related to translocating SOD1 to cell nuclei under oxidative stress, probably controlling DNA damage. The results of this study suggest that PrPC acts against oxidative stress activating the cellular response and defense by displaying neuroprotection to neurons and ensuring the functionality of astrocytes.
Collapse
|
5
|
Studying Peptide-Metal Ion Complex Structures by Solution-State NMR. Int J Mol Sci 2022; 23:ijms232415957. [PMID: 36555599 PMCID: PMC9782655 DOI: 10.3390/ijms232415957] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Metal chelation can provide structural stability and form reactive centers in metalloproteins. Approximately one third of known protein structures are metalloproteins, and metal binding, or the lack thereof, is often implicated in disease, making it necessary to be able to study these systems in detail. Peptide-metal complexes are both present in nature and can provide a means to focus on the binding region of a protein and control experimental variables to a high degree. Structural studies of peptide complexes with metal ions by nuclear magnetic resonance (NMR) were surveyed for all the essential metal complexes and many non-essential metal complexes. The various methods used to study each metal ion are presented together with examples of recent research. Many of these metal systems have been individually reviewed and this current overview of NMR studies of metallopeptide complexes aims to provide a basis for inspiration from structural studies and methodology applied in the field.
Collapse
|
6
|
de Jesus JR, Galazzi RM, Lopes Júnior CA, Arruda MAZ. Trace element homeostasis in the neurological system after SARS-CoV-2 infection: Insight into potential biochemical mechanisms. J Trace Elem Med Biol 2022; 71:126964. [PMID: 35240553 PMCID: PMC8881805 DOI: 10.1016/j.jtemb.2022.126964] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/10/2022] [Accepted: 02/23/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Several studies have suggested that COVID-19 is a systemic disease that can affect several organs, including the brain. In the brain, specifically, viral infection can cause dyshomeostasis of some trace elements that promote complex biochemical reactions in specialized neurological functions. OBJECTIVE Understand the neurovirulence of SARS-CoV-2 and the relationship between trace elements and neurological disorders after infection, and provide new insights on the drug development for the treatment of SARS-CoV-2 infections. METHODS The main databases were used to search studies published up September 2021, focusing on the role of trace elements during viral infection and on the correct functioning of the brain. RESULTS The imbalance of important trace elements can accelerate SARS-CoV-2 neurovirulence and increase the neurotoxicity since many neurological processes can be associated with the homeostasis of metal and metalloproteins. Some studies involving animals and humans have suggested the synapse as a vulnerable region of the brain to neurological disorders after viral infection. Considering the combined evidence, some mechanisms have been suggested to understand the relationship between neurological disorders and imbalance of trace elements in the brain after viral infection. CONCLUSION Trace elements play important roles in viral infections, such as helping to activate immune cells, produce antibodies, and inhibit virus replication. However, the relationship between trace elements and virus infections is complex since the specific functions of several elements remain largely undefined. Therefore, there is still a lot to be explored to understand the biochemical mechanisms involved between trace elements and viral infections, especially in the brain.
Collapse
Affiliation(s)
- Jemmyson Romário de Jesus
- Research Laboratory in Bionanomaterials, LPbio, Brazil; Chemistry Department, Federal University of Viçosa, UFV, Viçosa, Minas Gerais, Brazil.
| | - Rodrigo Moretto Galazzi
- Analytical Instrumentation Division, Analytik Jena GmbH, an Endress & Hauser Company, São Paulo, SP 04029-901, Brazil.
| | - Cícero Alves Lopes Júnior
- Grupo de Estudos em Bioanalítica - GEBIO, Department of Chemistry, Federal University of Piauí, 64049-550 Teresina, PI, Brazil.
| | - Marco Aurélio Zezzi Arruda
- Spectrometry, Sample Preparation and Mechanization Group, GEPAM, Institute of Chemistry, University of Campinas, UNICAMP, Campinas, Brazil; National Institute of Science and Technology for Bioanalytics, Brazil.
| |
Collapse
|
7
|
Abstract
Amyloids are protein aggregates bearing a highly ordered cross β structural motif, which may be functional but are mostly pathogenic. Their formation, deposition in tissues and consequent organ dysfunction is the central event in amyloidogenic diseases. Such protein aggregation may be brought about by conformational changes, and much attention has been directed toward factors like metal binding, post-translational modifications, mutations of protein etc., which eventually affect the reactivity and cytotoxicity of the associated proteins. Over the past decade, a global effort from different groups working on these misfolded/unfolded proteins/peptides has revealed that the amino acid residues in the second coordination sphere of the active sites of amyloidogenic proteins/peptides cause changes in H-bonding pattern or protein-protein interactions, which dramatically alter the structure and reactivity of these proteins/peptides. These second sphere effects not only determine the binding of transition metals and cofactors, which define the pathology of some of these diseases, but also change the mechanism of redox reactions catalyzed by these proteins/peptides and form the basis of oxidative damage associated with these amyloidogenic diseases. The present review seeks to discuss such second sphere modifications and their ramifications in the etiopathology of some representative amyloidogenic diseases like Alzheimer's disease (AD), type 2 diabetes mellitus (T2Dm), Parkinson's disease (PD), Huntington's disease (HD), and prion diseases.
Collapse
Affiliation(s)
- Madhuparna Roy
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Arnab Kumar Nath
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
8
|
Daude N, Lau A, Vanni I, Kang SG, Castle AR, Wohlgemuth S, Dorosh L, Wille H, Stepanova M, Westaway D. Prion protein with a mutant N-terminal octarepeat region undergoes cobalamin-dependent assembly into high-molecular weight complexes. J Biol Chem 2022; 298:101770. [PMID: 35271850 PMCID: PMC9010764 DOI: 10.1016/j.jbc.2022.101770] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 11/28/2022] Open
Abstract
The cellular prion protein (PrPC) has a C-terminal globular domain and a disordered N-terminal region encompassing five octarepeats (ORs). Encounters between Cu(II) ions and four OR sites produce interchangeable binding geometries; however, the significance of Cu(II) binding to ORs in different combinations is unclear. To understand the impact of specific binding geometries, OR variants were designed that interact with multiple or single Cu(II) ions in specific locked coordinations. Unexpectedly, we found that one mutant produced detergent-insoluble, protease-resistant species in cells in the absence of exposure to the infectious prion protein isoform, scrapie-associated prion protein (PrPSc). Formation of these assemblies, visible as puncta, was reversible and dependent upon medium formulation. Cobalamin (Cbl), a dietary cofactor containing a corrin ring that coordinates a Co3+ ion, was identified as a key medium component, and its effect was validated by reconstitution experiments. Although we failed to find evidence that Cbl interacts with Cu-binding OR regions, we instead noted interactions of Cbl with the PrPC C-terminal domain. We found that some interactions occurred at a binding site of planar tetrapyrrole compounds on the isolated globular domain, but others did not, and N-terminal sequences additionally had a marked effect on their presence and position. Our studies define a conditional effect of Cbl wherein a mutant OR region can act in cis to destabilize a globular domain with a wild type sequence. The unexpected intersection between the properties of PrPSc's disordered region, Cbl, and conformational remodeling events may have implications for understanding sporadic prion disease that does not involve exposure to PrPSc.
Collapse
Affiliation(s)
- Nathalie Daude
- Centre for Prions and Protein Folding Diseases, University of Alberta, Canada
| | - Agnes Lau
- Centre for Prions and Protein Folding Diseases, University of Alberta, Canada
| | - Ilaria Vanni
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Sang-Gyun Kang
- Centre for Prions and Protein Folding Diseases, University of Alberta, Canada
| | - Andrew R Castle
- Centre for Prions and Protein Folding Diseases, University of Alberta, Canada
| | - Serene Wohlgemuth
- Centre for Prions and Protein Folding Diseases, University of Alberta, Canada
| | - Lyudmyla Dorosh
- Faculty of Engineering - Electrical & Computer Engineering Dept, University of Alberta, Canada
| | - Holger Wille
- Centre for Prions and Protein Folding Diseases, University of Alberta, Canada; Department of Biochemistry, University of Alberta, Canada
| | - Maria Stepanova
- Faculty of Engineering - Electrical & Computer Engineering Dept, University of Alberta, Canada
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, Canada.
| |
Collapse
|
9
|
Marques CMS, Pedron T, Batista BL, Cerchiaro G. Cellular prion protein activates Caspase 3 for apoptotic defense mechanism in astrocytes. Mol Cell Biochem 2021; 476:2149-2158. [PMID: 33547547 DOI: 10.1007/s11010-021-04078-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 01/25/2021] [Indexed: 12/31/2022]
Abstract
The cellular prion protein (PrPC) is anchored in the plasma membrane of cells, and it is highly present in cells of brain tissue, exerting numerous cellular and cognitive functions. The present study proves the importance of PrPC in the cellular defense mechanism and metal homeostasis in astrocytes cells. Through experimental studies using cell lines of immortalized mice astrocytes (wild type and knockout for PrPC), we showed that PrPc is involved in the apoptosis cell death process by the activation of Caspase 3, downregulation of p53, and cell cycle maintenance. Metal homeostasis was determined by inductively coupled plasma mass spectrometry technique, indicating the crucial role of PrPC to lower intracellular calcium. The lowered calcium concentration and the Caspase 3 downregulation in the PrPC-null astrocytes resulted in a faster growth rate in cells, comparing with PrPC wild-type one. The presence of PrPC shows to be essential to cell death and healthy growth. In conclusion, our results show for the first time that astrocyte knockout cells for the cellular prion protein could modulate apoptosis-dependent cell death pathways.
Collapse
Affiliation(s)
- Caroline M S Marques
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André, SP, 09210-580, Brazil
| | - Tatiana Pedron
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André, SP, 09210-580, Brazil
| | - Bruno L Batista
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André, SP, 09210-580, Brazil
| | - Giselle Cerchiaro
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André, SP, 09210-580, Brazil.
| |
Collapse
|
10
|
Li S, Kerman K. Electrochemical biosensors for biometal-protein interactions in neurodegenerative diseases. Biosens Bioelectron 2021; 179:113035. [PMID: 33578115 DOI: 10.1016/j.bios.2021.113035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/12/2021] [Accepted: 01/21/2021] [Indexed: 12/20/2022]
Abstract
Electrochemical biosensors have been adopted into a wide range of applications in the study of biometal-protein interactions in neurodegenerative diseases. Transition metals such as zinc, copper, and iron that are significant to biological functions have been shown to have strong implications in the progressive neural degeneration in Alzheimer's disease (AD), Parkinson's disease (PD), and prion protein diseases. This review presents a summative examination of the progress made in the design, fabrication, and applications of electrochemical biosensors in recent literature at understanding the metal-protein interactions in neurodegenerative diseases. The focus will be drawn on disease-causing biomarkers such as amyloid-β (Aβ) and tau proteins for AD, α-synuclein (α-syn) for PD, and prion proteins (PrP). Topics such as the use of electrochemical biosensing in monitoring biometal-induced conformational changes, elucidation of complexation motifs, production of reactive oxygen species (ROS) as well as the influence on downstream biomolecular interactions will be discussed. Major results and important concepts presented in these studies will be summarized in the hope to spark inspiration for the next generation of electrochemical sensors.
Collapse
Affiliation(s)
- Shaopei Li
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada
| | - Kagan Kerman
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada.
| |
Collapse
|
11
|
Mustazza C, Sbriccoli M, Minosi P, Raggi C. Small Molecules with Anti-Prion Activity. Curr Med Chem 2020; 27:5446-5479. [PMID: 31560283 DOI: 10.2174/0929867326666190927121744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 08/08/2019] [Accepted: 09/05/2019] [Indexed: 01/20/2023]
Abstract
Prion pathologies are fatal neurodegenerative diseases caused by the misfolding of the physiological Prion Protein (PrPC) into a β-structure-rich isoform called PrPSc. To date, there is no available cure for prion diseases and just a few clinical trials have been carried out. The initial approach in the search of anti-prion agents had PrPSc as a target, but the existence of different prion strains arising from alternative conformations of PrPSc, limited the efficacy of the ligands to a straindependent ability. That has shifted research to PrPC ligands, which either act as chaperones, by stabilizing the native conformation, or inhibit its interaction with PrPSc. The role of transition-metal mediated oxidation processes in prion misfolding has also been investigated. Another promising approach is the indirect action via other cellular targets, like membrane domains or the Protein- Folding Activity of Ribosomes (PFAR). Also, new prion-specific high throughput screening techniques have been developed. However, so far no substance has been found to be able to extend satisfactorily survival time in animal models of prion diseases. This review describes the main features of the Structure-Activity Relationship (SAR) of the various chemical classes of anti-prion agents.
Collapse
Affiliation(s)
- Carlo Mustazza
- National Centre for Control and Evaluation of Medicines, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marco Sbriccoli
- Department of Neurosciences, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Paola Minosi
- National Centre for Drug Research and Evaluation, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Carla Raggi
- National Centre for Control and Evaluation of Medicines, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
12
|
Yakubu UM, Catumbela CSG, Morales R, Morano KA. Understanding and exploiting interactions between cellular proteostasis pathways and infectious prion proteins for therapeutic benefit. Open Biol 2020; 10:200282. [PMID: 33234071 PMCID: PMC7729027 DOI: 10.1098/rsob.200282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Several neurodegenerative diseases of humans and animals are caused by the misfolded prion protein (PrPSc), a self-propagating protein infectious agent that aggregates into oligomeric, fibrillar structures and leads to cell death by incompletely understood mechanisms. Work in multiple biological model systems, from simple baker's yeast to transgenic mouse lines, as well as in vitro studies, has illuminated molecular and cellular modifiers of prion disease. In this review, we focus on intersections between PrP and the proteostasis network, including unfolded protein stress response pathways and roles played by the powerful regulators of protein folding known as protein chaperones. We close with analysis of promising therapeutic avenues for treatment enabled by these studies.
Collapse
Affiliation(s)
- Unekwu M Yakubu
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth, Houston, TX USA.,MD Anderson UTHealth Graduate School at UTHealth, Houston, TX USA
| | - Celso S G Catumbela
- MD Anderson UTHealth Graduate School at UTHealth, Houston, TX USA.,Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at UTHealth, Houston, TX USA
| | - Rodrigo Morales
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at UTHealth, Houston, TX USA.,Centro integrativo de biología y química aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Kevin A Morano
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth, Houston, TX USA
| |
Collapse
|
13
|
Freeman DM, O'Neal R, Zhang Q, Bouwer EJ, Wang Z. Manganese-induced Parkinsonism in mice is reduced using a novel contaminated water sediment exposure model. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 78:103399. [PMID: 32380377 DOI: 10.1016/j.etap.2020.103399] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 06/11/2023]
Abstract
Heavy metals enter the aquatic environment and accumulate within water sediments, but these metal-sediment interactions remain to be explored within toxicity studies. We developed an exposure model in mice that encapsulates the aquatic microenvironment of metals before exposure. Male and female C57/BL6 mice were exposed via their drinking water to manganese contaminated sediment (Sed_Mn) or to manganese without sediment interaction (Mn) for six weeks. Sediment interaction did not alter weekly manganese ingestion from water in males or females. We analyzed motor impairment, a common feature in manganese-induced Parkinsonism, using the beam traversal, cylinder, and accelerating rotarod tests. Sed_Mn mice performed better overall compared to Mn mice and males were more sensitive to manganese than females in both Sed_Mn and Mn treatment groups. Our study indicates that metal-sediment interactions may alter metal toxicity in mammals and introduces a new exposure model to test the toxicity of metal contaminants of drinking water.
Collapse
Affiliation(s)
- Dana M Freeman
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Rachel O'Neal
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Qiang Zhang
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Edward J Bouwer
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhibin Wang
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
14
|
Rokad D, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Exosomes as Mediators of Chemical-Induced Toxicity. Curr Environ Health Rep 2020; 6:73-79. [PMID: 31102182 DOI: 10.1007/s40572-019-00233-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW This review provides information regarding how exosomes may contribute to environmental chemical-induced pathogenesis of chronic diseases. In connecting exosome biology to environmental toxicology and disease pathogenesis, we address vital questions regarding what constitutes exosomal cargo, how toxicants influence exosomal cargo, and how environmental stimuli influence exosomal physiological and pathological functions. RECENT FINDINGS Recent studies in the field demonstrate that exosomal cargo changes depending on external stimuli, which has consequences for the microenvironment of recipient cells. Based on recent findings, it is evident that exosomal cargo comprises various biological molecules including proteins, nucleic acids, and lipid molecules. Misfolded proteins and miRNA are examples of exosomal cargo molecules that can be altered by toxicants, ultimately changing the microenvironment of recipient cells in ways that are conducive to pathological processes. It will be crucial to map out the key signaling pathways that toxicants target to modulate exosomal cargo and their release.
Collapse
Affiliation(s)
- Dharmin Rokad
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, 1222 Veterinary Medicine Building, Ames, IA, 50011, USA
| | - Huajun Jin
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, 1222 Veterinary Medicine Building, Ames, IA, 50011, USA
| | - Vellareddy Anantharam
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, 1222 Veterinary Medicine Building, Ames, IA, 50011, USA
| | - Arthi Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, 1222 Veterinary Medicine Building, Ames, IA, 50011, USA
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, 1222 Veterinary Medicine Building, Ames, IA, 50011, USA.
| |
Collapse
|
15
|
Nowakowski M, Czapla-Masztafiak J, Zhukov I, Zhukova L, Kozak M, Kwiatek WM. Electronic properties of a PrP C-Cu(ii) complex as a marker of 5-fold Cu(ii) coordination. Metallomics 2019; 11:632-642. [PMID: 30756103 DOI: 10.1039/c8mt00339d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Human prion protein is a subject of extensive study, related in particular to the molecular basis of neurodegenerative disease development and prevention. This protein has two main domains: the membrane C-terminal, structured domain as well as the unstructured N-terminal domain. While PrPC (23-231) has up to eight Cu(ii) binding sites in the N-terminal domain, it includes a characteristic, conservative octarepeat region PHGGGWGQ, which was studied by means of X-ray absorption near edge spectroscopy. The measurements were conducted at the SuperXAS beamline (SLS, PSI, Villigen). For the initial 1 : 1 protein-to-Cu(ii) ratio, the two main Cu(ii) binding modes were identified using linear combination fitting and ab initio FEFF calculations for X-ray spectra. Their electronic structures indicated that Cu(ii) coordinated by strong π-donors could effectively suppress the pre-edge structure due to the filling of empty Cu(ii) d-states. The suppression was correlated with the charge transfer effect and filling of the virtual electronic Cu(ii) states. What is more, we showed that the 1s → 4p + LMCT (Ligand-to-Metal-Charge-Transfer) multielectron transition relation with the main edge transition could be used as a marker for preliminary comparison of an unknown organic compound to a reference. The presented results permitted a possible explanation of the mechanism of choosing the preferred Cu(ii) modes in PrPC-Cu(ii) coordination processes and of the complex stability from the electronic point of view.
Collapse
Affiliation(s)
- Michał Nowakowski
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31-342 Krakow, Poland.
| | | | | | | | | | | |
Collapse
|
16
|
Structural Determinants of the Prion Protein N-Terminus and Its Adducts with Copper Ions. Int J Mol Sci 2018; 20:ijms20010018. [PMID: 30577569 PMCID: PMC6337743 DOI: 10.3390/ijms20010018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 12/24/2022] Open
Abstract
The N-terminus of the prion protein is a large intrinsically disordered region encompassing approximately 125 amino acids. In this paper, we review its structural and functional properties, with a particular emphasis on its binding to copper ions. The latter is exploited by the region’s conformational flexibility to yield a variety of biological functions. Disease-linked mutations and proteolytic processing of the protein can impact its copper-binding properties, with important structural and functional implications, both in health and disease progression.
Collapse
|
17
|
Harischandra DS, Ghaisas S, Rokad D, Kanthasamy AG. Exosomes in Toxicology: Relevance to Chemical Exposure and Pathogenesis of Environmentally Linked Diseases. Toxicol Sci 2018; 158:3-13. [PMID: 28505322 DOI: 10.1093/toxsci/kfx074] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chronic exposure to environmental toxins has been known to initiate or aggravate various neurological disorders, carcinomas and other adverse health effects. Uptake by naïve cells of pathogenic factors such as danger-associated molecules, mRNAs, miRNAs or aggregated proteins leads to disruption in cellular homeostasis further resulting in inflammation and disease propagation. Although early research tended to focus solely on exosomal removal of unwanted cellular contents, more recent reports indicate that these nano-vesicles play an active role in intercellular signaling. Not only is the exosomal cargo cell type-specific, but it also differs between healthy and dying cells. Moreover, following exosome uptake by naïve cells, the contents from these vesicles can alter the fate of recipient cells. Since exosomes can traverse long distances, they can influence distantly located cells and tissues. This review briefly explores the role played by environmental toxins in stimulating exosome release in the context of progressive neurodegenerative diseases such as Alzheimer's, Parkinson's, and Huntington's, as well as certain cancers such as lung, liver, ovarian, and tracheal carcinomas.
Collapse
Affiliation(s)
- Dilshan S Harischandra
- Parkinson Disorders Research Program, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Shivani Ghaisas
- Parkinson Disorders Research Program, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Dharmin Rokad
- Parkinson Disorders Research Program, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Program, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| |
Collapse
|
18
|
Langley MR, Ghaisas S, Ay M, Luo J, Palanisamy BN, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Manganese exposure exacerbates progressive motor deficits and neurodegeneration in the MitoPark mouse model of Parkinson's disease: Relevance to gene and environment interactions in metal neurotoxicity. Neurotoxicology 2018; 64:240-255. [PMID: 28595911 PMCID: PMC5736468 DOI: 10.1016/j.neuro.2017.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 10/19/2022]
Abstract
Parkinson's disease (PD) is now recognized as a neurodegenerative condition caused by a complex interplay of genetic and environmental influences. Chronic manganese (Mn) exposure has been implicated in the development of PD. Since mitochondrial dysfunction is associated with PD pathology as well as Mn neurotoxicity, we investigated whether Mn exposure augments mitochondrial dysfunction and neurodegeneration in the nigrostriatal dopaminergic system using a newly available mitochondrially defective transgenic mouse model of PD, the MitoPark mouse. This unique PD model recapitulates key features of the disease including progressive neurobehavioral changes and neuronal degeneration. We exposed MitoPark mice to a low dose of Mn (10mg/kg, p.o.) daily for 4 weeks starting at age 8 wks and then determined the behavioral, neurochemical and histological changes. Mn exposure accelerated the rate of progression of motor deficits in MitoPark mice when compared to the untreated MitoPark group. Mn also worsened olfactory function in this model. Most importantly, Mn exposure intensified the depletion of striatal dopamine and nigral TH neuronal loss in MitoPark mice. The neurodegenerative changes were accompanied by enhanced oxidative damage in the striatum and substantia nigra (SN) of MitoPark mice treated with Mn. Furthermore, Mn-treated MitoPark mice had significantly more oligomeric protein and IBA-1-immunoreactive microglia cells, suggesting Mn augments neuroinflammatory processes in the nigrostriatal pathway. To further confirm the direct effect of Mn on impaired mitochondrial function, we also generated a mitochondrially defective dopaminergic cell model by knocking out the TFAM transcription factor by using a CRISPR-Cas9 gene-editing method. Seahorse mitochondrial bioenergetic analysis revealed that Mn decreases mitochondrial basal and ATP-linked respiration in the TFAM KO cells. Collectively, our results reveal that Mn can augment mitochondrial dysfunction to exacerbate nigrostriatal neurodegeneration and PD-related behavioral symptoms. Our study also demonstrates that the MitoPark mouse is an excellent model to study the gene-environment interactions associated with mitochondrial defects in the nigral dopaminergic system as well as to evaluate the contribution of potential environmental toxicant interactions in a slowly progressive model of Parkinsonism.
Collapse
Affiliation(s)
- Monica R Langley
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Shivani Ghaisas
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Muhammet Ay
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Jie Luo
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Bharathi N Palanisamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Huajun Jin
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Vellareddy Anantharam
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Arthi Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
19
|
Nichols TA, Spraker TR, Gidlewski T, Cummings B, Hill D, Kong Q, Balachandran A, VerCauteren KC, Zabel MD. Dietary magnesium and copper affect survival time and neuroinflammation in chronic wasting disease. Prion 2017; 10:228-50. [PMID: 27216881 PMCID: PMC4981212 DOI: 10.1080/19336896.2016.1181249] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chronic wasting disease (CWD), the only known wildlife prion disease, affects deer, elk and moose. The disease is an ongoing and expanding problem in both wild and captive North American cervid populations and is difficult to control in part due to the extreme environmental persistence of prions, which can transmit disease years after initial contamination. The role of exogenous factors in CWD transmission and progression is largely unexplored. In an effort to understand the influence of environmental and dietary constituents on CWD, we collected and analyzed water and soil samples from CWD-negative and positive captive cervid facilities, as well as from wild CWD-endozootic areas. Our analysis revealed that, when compared with CWD-positive sites, CWD-negative sites had a significantly higher concentration of magnesium, and a higher magnesium/copper (Mg/Cu) ratio in the water than that from CWD-positive sites. When cevidized transgenic mice were fed a custom diet devoid of Mg and Cu and drinking water with varied Mg/Cu ratios, we found that higher Mg/Cu ratio resulted in significantly longer survival times after intracerebral CWD inoculation. We also detected reduced levels of inflammatory cytokine gene expression in mice fed a modified diet with a higher Mg/Cu ratio compared to those on a standard rodent diet. These findings indicate a role for dietary Mg and Cu in CWD pathogenesis through modulating inflammation in the brain.
Collapse
Affiliation(s)
- Tracy A Nichols
- a National Wildlife Research Center, US Department of Agriculture , Animal and Plant Health Inspection Service, Wildlife Services , Fort Collins , CO , USA
| | - Terry R Spraker
- b Colorado State University Diagnostic Laboratory, College of Veterinary Medicine, Colorado State University , Fort Collins , CO , USA;,c Department of Microbiology , Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University Prion Research Center , Fort Collins , CO , USA
| | - Thomas Gidlewski
- a National Wildlife Research Center, US Department of Agriculture , Animal and Plant Health Inspection Service, Wildlife Services , Fort Collins , CO , USA
| | - Bruce Cummings
- b Colorado State University Diagnostic Laboratory, College of Veterinary Medicine, Colorado State University , Fort Collins , CO , USA
| | - Dana Hill
- c Department of Microbiology , Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University Prion Research Center , Fort Collins , CO , USA
| | - Qingzhong Kong
- d Departments of Pathology and Neurology & National Center for Regenerative Medicine , Case Western Reserve University , Cleveland , OH , USA
| | - Aru Balachandran
- e National and OIE Reference Laboratory for Scrapie and CWD, Canadian Food Inspection Agency , Ottawa , Ontario , Canada
| | - Kurt C VerCauteren
- a National Wildlife Research Center, US Department of Agriculture , Animal and Plant Health Inspection Service, Wildlife Services , Fort Collins , CO , USA
| | - Mark D Zabel
- c Department of Microbiology , Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University Prion Research Center , Fort Collins , CO , USA
| |
Collapse
|
20
|
Yen CF, Harischandra DS, Kanthasamy A, Sivasankar S. Copper-induced structural conversion templates prion protein oligomerization and neurotoxicity. SCIENCE ADVANCES 2016; 2:e1600014. [PMID: 27419232 PMCID: PMC4942324 DOI: 10.1126/sciadv.1600014] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/27/2016] [Indexed: 05/26/2023]
Abstract
Prion protein (PrP) misfolding and oligomerization are key pathogenic events in prion disease. Copper exposure has been linked to prion pathogenesis; however, its mechanistic basis is unknown. We resolve, with single-molecule precision, the molecular mechanism of Cu(2+)-induced misfolding of PrP under physiological conditions. We also demonstrate that misfolded PrPs serve as seeds for templated formation of aggregates, which mediate inflammation and degeneration of neuronal tissue. Using a single-molecule fluorescence assay, we demonstrate that Cu(2+) induces PrP monomers to misfold before oligomer assembly; the disordered amino-terminal region mediates this structural change. Single-molecule force spectroscopy measurements show that the misfolded monomers have a 900-fold higher binding affinity compared to the native isoform, which promotes their oligomerization. Real-time quaking-induced conversion demonstrates that misfolded PrPs serve as seeds that template amyloid formation. Finally, organotypic slice cultures show that misfolded PrPs mediate inflammation and degeneration of neuronal tissue. Our study establishes a direct link, at the molecular level, between copper exposure and PrP neurotoxicity.
Collapse
Affiliation(s)
- Chi-Fu Yen
- Department of Electrical and Computer Engineering, Iowa State University, Ames, IA 50011, USA
| | - Dilshan S. Harischandra
- Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Anumantha Kanthasamy
- Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Sanjeevi Sivasankar
- Department of Electrical and Computer Engineering, Iowa State University, Ames, IA 50011, USA
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
21
|
Faburay B, Tark D, Kanthasamy AG, Richt JA. In vitro amplification of scrapie and chronic wasting disease PrP(res) using baculovirus-expressed recombinant PrP as substrate. Prion 2015; 8:393-403. [PMID: 25495764 DOI: 10.4161/19336896.2014.983753] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Protein misfolding cyclic amplification (PMCA) is an in vitro simulation of prion replication, which relies on the use of normal brain homogenate derived from host species as substrate for the specific amplification of abnormal prion protein, PrP(Sc). Studies showed that recombinant cellular PrP, PrP(C), expressed in Escherichia coli lacks N-glycosylation and an glycophosphatidyl inositol anchor (GPI) and therefore may not be the most suitable substrate in seeded PMCA reactions to recapitulate prion conversion in vitro. In this study, we expressed 2 PRNP genotypes of sheep, V136L141R154Q171 and A136F141R154Q171, and one genotype of white-tailed deer (Q95G96, X132,Y216) using the baculovirus expression system and evaluated their suitability as substrates in seeded-PMCA. It has been reported that host-encoded mammalian RNA molecules and divalent cations play a role in the pathogenesis of prion diseases, and RNA molecules have also been shown to improve the sensitivity of PMCA assays. Therefore, we also assessed the effect of co-factors, such as prion-specific mRNA molecules and a divalent cation, manganese, on protein conversion. Here, we report that baculovirus-expressed recombinant PrP(C) shows a glycoform and GPI-anchor profile similar to mammalian brain-derived PrP(C) and supports amplification of PrP(Sc) and PrP(CWD) derived from prion-affected animals in a single round of seeded PMCA in the absence of exogenous co-factors. Addition of species-specific in vitro transcribed PrP mRNA molecules stimulated the conversion efficiency resulting in increased PrP(Sc) or PrP(CWD) production. Addition of 2 to 20 μM of manganese chloride (MnCl2) to unseeded PMCA resulted in conversion of recombinant PrP(C) to protease-resistant PrP. Collectively, we demonstrate, for the first time, that baculovirus expressed sheep and deer PrP can serve as a substrate in protein misfolding cyclic amplification for sheep and deer prions in the absence of additional exogenous co-factors.
Collapse
Affiliation(s)
- Bonto Faburay
- a Department of Diagnostic Medicine and Pathobiology ; College of Veterinary Medicine ; Kansas State University ; Manhattan , KS USA
| | | | | | | |
Collapse
|
22
|
Harischandra DS, Kondru N, Martin DP, Kanthasamy A, Jin H, Anantharam V, Kanthasamy AG. Role of proteolytic activation of protein kinase Cδ in the pathogenesis of prion disease. Prion 2015; 8:143-53. [PMID: 24576946 DOI: 10.4161/pri.28369] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Prion diseases are infectious and inevitably fatal neurodegenerative diseases characterized by prion replication, widespread protein aggregation and spongiform degeneration of major brain regions controlling motor function. Oxidative stress has been implicated in prion-related neuronal degeneration, but the molecular mechanisms underlying prion-induced oxidative damage are not well understood. In this study, we evaluated the role of oxidative stress-sensitive, pro-apoptotic protein kinase Cδ (PKCδ) in prion-induced neuronal cell death using cerebellar organotypic slice cultures (COSC) and mouse models of prion diseases. We found a significant upregulation of PKCδ in RML scrapie-infected COSC, as evidenced by increased levels of both PKCδ protein and its mRNA. We also found an enhanced regulatory phosphorylation of PKCδ at its two regulatory sites, Thr505 in the activation loop and Tyr311 at the caspase-3 cleavage site. The prion infection also induced proteolytic activation of PKCδ in our COSC model. Immunohistochemical analysis of scrapie-infected COSC revealed loss of PKCδ positive Purkinje cells and enhanced astrocyte proliferation. Further examination of PKCδ signaling in the RML scrapie adopted in vivo mouse model showed increased proteolytic cleavage and Tyr 311 phosphorylation of the kinase. Notably, we observed a delayed onset of scrapie-induced motor symptoms in PKCδ knockout (PKCδ(-/-)) mice as compared with wild-type (PKCδ(+/+)) mice, further substantiating the role of PKCδ in prion disease. Collectively, these data suggest that PKCδ signaling likely plays a role in the neurodegenerative processes associated with prion diseases.
Collapse
|
23
|
Marchetti C. Interaction of metal ions with neurotransmitter receptors and potential role in neurodiseases. Biometals 2014; 27:1097-113. [PMID: 25224737 DOI: 10.1007/s10534-014-9791-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 09/05/2014] [Indexed: 12/25/2022]
Abstract
There is increasing evidence that toxic metals play a role in diseases of unknown etiology. Their action is often mediated by membrane proteins, and in particular neurotransmitter receptors. This brief review will describe recent findings on the direct interaction of metal ions with ionotropic γ-aminobutyric acid (GABAA) and glutamate receptors, the main inhibitory and excitatory neurotransmitter receptors in the mammalian central nervous system, respectively. Both hyper and hypo function of these receptors are involved in neurological and psychotic syndromes and modulation by metal ions is an important pharmacological issue. The focus will be on three xenobiotic metals, lead (Pb), cadmium (Cd) and nickel (Ni) that have no biological function and whose presence in living organisms is only detrimental, and two trace metals, zinc (Zn) and copper (Cu), which are essential for several enzymatic functions, but can mediate toxic actions if deregulated. Despite limited access to the brain and tight control by metalloproteins, exogenous metals interfere with receptor performances by mimicking physiological ions and occupying one or more modulatory sites on the protein. These interactions will be discussed as a potential cause of neuronal dysfunction.
Collapse
Affiliation(s)
- Carla Marchetti
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, via De Marini, 6, 16149, Genoa, Italy,
| |
Collapse
|
24
|
Singh N, Haldar S, Tripathi AK, McElwee MK, Horback K, Beserra A. Iron in neurodegenerative disorders of protein misfolding: a case of prion disorders and Parkinson's disease. Antioxid Redox Signal 2014; 21:471-84. [PMID: 24512387 PMCID: PMC4076993 DOI: 10.1089/ars.2014.5874] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Intracellular and extracellular aggregation of a specific protein or protein fragments is the principal pathological event in several neurodegenerative conditions. We describe two such conditions: sporadic Creutzfeldt-Jakob disease (sCJD), a rare but potentially infectious and invariably fatal human prion disorder, and Parkinson's disease (PD), a common neurodegenerative condition second only to Alzheimer's disease in prevalence. In sCJD, a cell surface glycoprotein known as the prion protein (PrP(C)) undergoes a conformational change to PrP-scrapie, a pathogenic and infectious isoform that accumulates in the brain parenchyma as insoluble aggregates. In PD, α-synuclein, a cytosolic protein, forms insoluble aggregates that accumulate in neurons of the substantia nigra and cause neurotoxicity. RECENT ADVANCES Although distinct processes are involved in the pathogenesis of sCJD and PD, both share brain iron dyshomeostasis as a common associated feature that is reflected in the cerebrospinal fluid in a disease-specific manner. CRITICAL ISSUES Since PrP(C) and α-synuclein play a significant role in maintaining cellular iron homeostasis, it is important to understand whether the aggregation of these proteins and iron dyshomeostasis are causally related. Here, we discuss recent information on the normal function of PrP(C) and α-synuclein in cellular iron metabolism and the cellular and biochemical processes that contribute to iron imbalance in sCJD and PD. FUTURE DIRECTIONS Improved understanding of the relationship between brain iron imbalance and protein aggregation is likely to help in the development of therapeutic strategies that can restore brain iron homeostasis and mitigate neurotoxicity.
Collapse
Affiliation(s)
- Neena Singh
- 1 Department of Pathology, Case Western Reserve University , Cleveland, Ohio
| | | | | | | | | | | |
Collapse
|
25
|
Evolutionary implications of metal binding features in different species' prion protein: an inorganic point of view. Biomolecules 2014; 4:546-65. [PMID: 24970230 PMCID: PMC4101497 DOI: 10.3390/biom4020546] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/29/2014] [Accepted: 05/06/2014] [Indexed: 12/21/2022] Open
Abstract
Prion disorders are a group of fatal neurodegenerative conditions of mammals. The key molecular event in the pathogenesis of such diseases is the conformational conversion of prion protein, PrPC, into a misfolded form rich in β-sheet structure, PrPSc, but the detailed mechanistic aspects of prion protein conversion remain enigmatic. There is uncertainty on the precise physiological function of PrPC in healthy individuals. Several evidences support the notion of its role in copper homeostasis. PrPC binds Cu2+ mainly through a domain composed by four to five repeats of eight amino acids. In addition to mammals, PrP homologues have also been identified in birds, reptiles, amphibians and fish. The globular domain of protein is retained in the different species, suggesting that the protein carries out an essential common function. However, the comparison of amino acid sequences indicates that prion protein has evolved differently in each vertebrate class. The primary sequences are strongly conserved in each group, but these exhibit a low similarity with those of mammals. The N-terminal domain of different prions shows tandem amino acid repeats with an increasing amount of histidine residues going from amphibians to mammals. The difference in the sequence affects the number of copper binding sites, the affinity and the coordination environment of metal ions, suggesting that the involvement of prion in metal homeostasis may be a specific characteristic of mammalian prion protein. In this review, we describe the similarities and the differences in the metal binding of different species' prion protein, as revealed by studies carried out on the entire protein and related peptide fragments.
Collapse
|
26
|
Mot AI, Wedd AG, Sinclair L, Brown DR, Collins SJ, Brazier MW. Metal attenuating therapies in neurodegenerative disease. Expert Rev Neurother 2014; 11:1717-45. [DOI: 10.1586/ern.11.170] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
27
|
Singh A, Haldar S, Horback K, Tom C, Zhou L, Meyerson H, Singh N. Prion protein regulates iron transport by functioning as a ferrireductase. J Alzheimers Dis 2013; 35:541-52. [PMID: 23478311 DOI: 10.3233/jad-130218] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Prion protein (PrPC) is implicated in the pathogenesis of prion disorders, but its normal function is unclear. We demonstrate that PrPC is a ferrireductase (FR), and its absence causes systemic iron deficiency in PrP knock-out mice (PrP-/-). When exposed to non-transferrin-bound (NTB) radioactive-iron (59FeCl3) by gastric-gavage, PrP-/- mice absorb significantly more 59Fe from the intestinal lumen relative to controls, indicating appropriate systemic response to the iron deficiency. Chronic exposure to excess dietary iron corrects this deficiency, but unlike wild-type (PrP+/+) controls that remain iron over-loaded, PrP-/- mice revert back to the iron deficient phenotype after 5 months of chase on normal diet. Bone marrow (BM) preparations of PrP-/- mice on normal diet show relatively less stainable iron, and this phenotype is only partially corrected by intraperitoneal administration of excess iron-dextran. Cultured PrP-/- BM-macrophages incorporate significantly less NTB-59Fe in the absence or presence of excess extracellular iron, indicating reduced uptake and/or storage of available iron in the absence of PrPC. When expressed in neuroblastoma cells, PrPC exhibits NAD(P)H-dependent cell-surface and intracellular FR activity that requires the copper-binding octa-peptide-repeat region and linkage to the plasma membrane for optimal function. Incorporation of NTB-59Fe by neuroblastoma cells correlates with FR activity of PrPC, implicating PrPC in cellular iron uptake and metabolism. These observations explain the correlation between PrPC expression and cellular iron levels, and the cause of iron imbalance in sporadic-Creutzfeldt-Jakob-disease brains where PrPC accumulates as insoluble aggregates.
Collapse
Affiliation(s)
- Ajay Singh
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Yuan F, Yang L, Zhang Z, Wu W, Zhou X, Yin X, Zhao D. Cellular prion protein (PrPC) of the neuron cell transformed to a PK-resistant protein under oxidative stress, comprising main mitochondrial damage in prion diseases. J Mol Neurosci 2013; 51:219-24. [PMID: 23715697 PMCID: PMC3739867 DOI: 10.1007/s12031-013-0008-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 03/18/2013] [Indexed: 11/30/2022]
Abstract
Prion diseases characterize a category of fatal neurodegenerative diseases. Although reports have increasingly shown that oxidative stress plays an important role in the progression of prion diseases, little is known about whether oxidative stress is a cause or a consequence of a prion disease. The mechanism of prion disease development also remains unclear. The purpose of this study was to investigate three things: the possible mechanisms of neuron cell damage, the conformation of anti-protease K (PK) PrPSc, and the role of oxidative stress in the progression of prion diseases. The study results demonstrated that normal PrPC transformed into a PK-resistant protein under oxidative stress in the presence of PrP106–126. Further, the protein misfolding cyclic amplification procedure may have accelerated this process. Mitochondrial damage and dysfunction in prion disease progression were also observed in this study. Our results suggested that neuron cell damage, and particularly mitochondrial damage, was induced by oxidative stress. This damage may be the initial cause of a given prion disease.
Collapse
Affiliation(s)
- Fangzhong Yuan
- State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 China
| | - Lifeng Yang
- State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 China
| | - Zhuming Zhang
- State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 China
| | - Wenyu Wu
- State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 China
| | - Xiangmei Zhou
- State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 China
| | - Xiaomin Yin
- State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 China
| | - Deming Zhao
- State Key Laboratories for Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 China
| |
Collapse
|
29
|
Peana M, Medici S, Nurchi VM, Crisponi G, Lachowicz JI, Zoroddu MA. Manganese and cobalt binding in a multi-histidinic fragment. Dalton Trans 2013; 42:16293-301. [DOI: 10.1039/c3dt51091c] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
30
|
Choi S, Woo HJ, Lee J. Sequence variations of the bovine prion protein gene (PRNP) in native Korean Hanwoo cattle. J Vet Sci 2012; 13:127-37. [PMID: 22705734 PMCID: PMC3386337 DOI: 10.4142/jvs.2012.13.2.127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Bovine spongiform encephalopathy (BSE) is one of the fatal neurodegenerative diseases known as transmissible spongiform encephalopathies (TSEs) caused by infectious prion proteins. Genetic variations correlated with susceptibility or resistance to TSE in humans and sheep have not been reported for bovine strains including those from Holstein, Jersey, and Japanese Black cattle. Here, we investigated bovine prion protein gene (PRNP) variations in Hanwoo cattle [Bos (B.) taurus coreanae], a native breed in Korea. We identified mutations and polymorphisms in the coding region of PRNP, determined their frequency, and evaluated their significance. We identified four synonymous polymorphisms and two non-synonymous mutations in PRNP, but found no novel polymorphisms. The sequence and number of octapeptide repeats were completely conserved, and the haplotype frequency of the coding region was similar to that of other B. taurus strains. When we examined the 23-bp and 12-bp insertion/deletion (indel) polymorphisms in the non-coding region of PRNP, Hanwoo cattle had a lower deletion allele and 23-bp del/12-bp del haplotype frequency than healthy and BSE-affected animals of other strains. Thus, Hanwoo are seemingly less susceptible to BSE than other strains due to the 23-bp and 12-bp indel polymorphisms.
Collapse
Affiliation(s)
- Sangho Choi
- Laboratory of Immunology, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea
| | | | | |
Collapse
|
31
|
Arena G, La Mendola D, Pappalardo G, Sóvágó I, Rizzarelli E. Interactions of Cu2+ with prion family peptide fragments: Considerations on affinity, speciation and coordination. Coord Chem Rev 2012. [DOI: 10.1016/j.ccr.2012.03.038] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
32
|
Coordination of zinc ions to the key proteins of neurodegenerative diseases: Aβ, APP, α-synuclein and PrP. Coord Chem Rev 2012. [DOI: 10.1016/j.ccr.2011.12.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
33
|
Singh A, Qing L, Kong Q, Singh N. Change in the characteristics of ferritin induces iron imbalance in prion disease affected brains. Neurobiol Dis 2012; 45:930-8. [PMID: 22182691 PMCID: PMC3286598 DOI: 10.1016/j.nbd.2011.12.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 10/20/2011] [Accepted: 12/04/2011] [Indexed: 01/28/2023] Open
Abstract
Prion disease associated neurotoxicity is mainly attributed to PrP-scrapie (PrP(Sc)), the disease associated isoform of a normal protein, the prion protein (PrP(C)). Participation of other proteins and processes is suspected, but their identity and contribution to the pathogenic process is unclear. Emerging evidence implicates imbalance of brain iron homeostasis as a significant cause of prion disease-associated neurotoxicity. The underlying cause of this change, however, remains unclear. We demonstrate that iron is sequestered in heat and SDS-stable protein complexes in sporadic-Creutzfeldt-Jakob-disease (sCJD) brains, creating a phenotype of iron deficiency. The underlying cause is change in the characteristics of ferritin, an iron storage protein that becomes aggregated, detergent-insoluble, and partitions with denatured ferritin using conventional methods of ferritin purification. A similar phenotype of iron deficiency is noted in the lumbar spinal cord (SC) tissue of scrapie infected hamsters, a site unlikely to be affected by massive neuronal death and non-specific iron deposition. As a result, the iron uptake protein transferrin (Tf) is upregulated in scrapie infected SC tissue, and increases with disease progression. A direct correlation between Tf and PrP(Sc) suggests sequestration of iron in dysfunctional ferritin that either co-aggregates with PrP(Sc) or is rendered dysfunctional by PrP(Sc) through an indirect process. Surprisingly, amplification of PrP(Sc)in vitro by the protein-misfolding-cyclic-amplification (PMCA) reaction using normal brain homogenate as substrate does not increase the heat and SDS-stable pool of iron even though both PrP(Sc) and ferritin aggregate by this procedure. These observations highlight important differences between PrP(Sc)-protein complexes generated in vivo during disease progression and in vitro by the PMCA reaction, and the significance of these complexes in PrP(Sc)-associated neurotoxicity.
Collapse
Affiliation(s)
- Ajay Singh
- Departments of Pathology and Neurology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106
| | - Liuting Qing
- Departments of Pathology and Neurology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106
| | - Qingzhong Kong
- Departments of Pathology and Neurology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106
| | - Neena Singh
- Departments of Pathology and Neurology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106
| |
Collapse
|
34
|
Li B, Qing L, Yan J, Kong Q. Instability of the octarepeat region of the human prion protein gene. PLoS One 2011; 6:e26635. [PMID: 22028931 PMCID: PMC3197570 DOI: 10.1371/journal.pone.0026635] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 09/30/2011] [Indexed: 01/24/2023] Open
Abstract
Prion diseases are a family of unique fatal transmissible neurodegenerative diseases that affect humans and many animals. Sporadic Creutzfeldt-Jakob disease (sCJD) is the most common prion disease in humans, accounting for 85–90% of all human prion cases, and exhibits a high degree of diversity in phenotypes. The etiology of sCJD remains to be elucidated. The human prion protein gene has an octapeptide repeat region (octarepeats) that normally contains 5 repeats of 24–27 bp (1 nonapeptide and 4 octapeptide coding sequences). An increase of the octarepeat numbers to six or more or a decrease of the octarepeat number to three is linked to genetic prion diseases with heterogeneous phenotypes in humans. Here we report that the human octarepeat region is prone to either contraction or expansion when subjected to PCR amplification in vitro using Taq or Pwo polymerase and when replicated in wild type E. coli cells. Octarepeat insertion mutants were even less stable, and the mutation rate for the wild type octarepeats was much higher when replicated in DNA mismatch repair-deficient E.coli cells. All observed octarepeat mutants resulting from DNA replication in E.coli were contained in head-to-head plasmid dimers and DNA mfold analysis (http://mfold.rna.albany.edu/?q=mfold/DNA-Folding-Form) indicates that both DNA strands of the octarepeat region would likely form multiple stable hairpin structures, suggesting that the octarepeat sequence may form stable hairpin structures during DNA replication or repair to cause octarepeat instability. These results provide the first evidence supporting a somatic octarepeat mutation-based model for human sCJD etiology: 1) the instability of the octarepeat region leads to accumulation of somatic octarepeat mutations in brain cells during development and aging, 2) this instability is augmented by compromised DNA mismatch repair in aged cells, and 3) eventually some of the octarepeat mutation-containing brain cells start spontaneous de novo prion formation and replication to initiate sCJD.
Collapse
Affiliation(s)
- Baiya Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | | | | | | |
Collapse
|
35
|
Martin DP, Anantharam V, Jin H, Witte T, Houk R, Kanthasamy A, Kanthasamy AG. Infectious prion protein alters manganese transport and neurotoxicity in a cell culture model of prion disease. Neurotoxicology 2011; 32:554-62. [PMID: 21871919 DOI: 10.1016/j.neuro.2011.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 07/20/2011] [Accepted: 07/21/2011] [Indexed: 01/26/2023]
Abstract
Protein misfolding and aggregation are considered key features of many neurodegenerative diseases, but biochemical mechanisms underlying protein misfolding and the propagation of protein aggregates are not well understood. Prion disease is a classical neurodegenerative disorder resulting from the misfolding of endogenously expressed normal cellular prion protein (PrP(C)). Although the exact function of PrP(C) has not been fully elucidated, studies have suggested that it can function as a metal binding protein. Interestingly, increased brain manganese (Mn) levels have been reported in various prion diseases indicating divalent metals also may play a role in the disease process. Recently, we reported that PrP(C) protects against Mn-induced cytotoxicity in a neural cell culture model. To further understand the role of Mn in prion diseases, we examined Mn neurotoxicity in an infectious cell culture model of prion disease. Our results show CAD5 scrapie-infected cells were more resistant to Mn neurotoxicity as compared to uninfected cells (EC(50)=428.8 μM for CAD5 infected cells vs. 211.6 μM for uninfected cells). Additionally, treatment with 300 μM Mn in persistently infected CAD5 cells showed a reduction in mitochondrial impairment, caspase-3 activation, and DNA fragmentation when compared to uninfected cells. Scrapie-infected cells also showed significantly reduced Mn uptake as measured by inductively coupled plasma-mass spectrometry (ICP-MS), and altered expression of metal transporting proteins DMT1 and transferrin. Together, our data indicate that conversion of PrP to the pathogenic isoform enhances its ability to regulate Mn homeostasis, and suggest that understanding the interaction of metals with disease-specific proteins may provide further insight to protein aggregation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Dustin P Martin
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicity, Ames, IA 50011, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Zhang Y, Wang X, Ding L. Synthesis and DNA binding studies of Mg(II) complex of Schiff base derived from vanillin and L-tryptophan. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2011; 30:49-62. [PMID: 21259163 DOI: 10.1080/15257770.2010.543117] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The Mg(II) complex of Schiff base (K[HL]) derived from vanillin and L-tryptophan could bind with herring sperm DNA. The binding behaviors between them in physiological pH environment (pH 7.40) have been studied by spectroscopy, cyclic voltammetry and viscosity methods. Binding ratios of n(Mg(II)): n(K[HL]) = 1:1 and n(Mg(II)L): n(DNA) = 5:1 were confirmed. The obtained thermodynamic parameters suggest that the interaction between Mg(II)L and DNA is driven mainly by entropy. Combined with fluorimeteric studies, cyclic voltammetry, CD spectroscopy and viscosity methods, the results indicate the interaction modes between Mg(II)L and DNA are mainly with intercalation and involving some electrostatic interaction.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Chemistry, School of Materials Science and Engineering, Southwest University of Science and Technology, Mianyang, P R China
| | | | | |
Collapse
|
37
|
Kahn RE, Clouser DF, Richt JA. Emerging infections: a tribute to the one medicine, one health concept. Zoonoses Public Health 2011; 56:407-28. [PMID: 19486315 DOI: 10.1111/j.1863-2378.2009.01255.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Events in the last decade have taught us that we are now, more than ever, vulnerable to fatal zoonotic diseases such as those caused by haemorrhagic fever viruses, influenza, rabies and BSE/vCJD. Future research activities should focus on solutions to these problems arising at the interface between animals and humans. A 4-fold classification of emerging zoonoses was proposed: Type 1: from wild animals to humans (Hanta); Type 1 plus: from wild animals to humans with further human-to-human transmission (AIDS); Type 2: from wild animals to domestic animals to humans (Avian flu) and Type 2 plus: from wild animals to domestic animals to humans, with further human-to-human transmission (Severe Acute Respiratory Syndrome, SARS). The resulting holistic approach to emerging infections links microbiology, veterinary medicine, human medicine, ecology, public health and epidemiology. As emerging 'new' respiratory viruses are identified in many wild and domestic animals, issues of interspecies transmission have become of increasing concern. The development of safe and effective human and veterinary vaccines is a priority. For example, the spread of different influenza viruses has stimulated influenza vaccine development, just as the spread of Ebola and Marburg viruses has led to new approaches to filovirus vaccines. Interdisciplinary collaboration has become essential because of the convergence of human disease, animal disease and a common approach to biosecurity. High containment pathogens pose a significant threat to public health systems, as well as a major research challenge, because of limited experience in case management, lack of appropriate resources in affected areas and a limited number of animal research facilities in developed countries. Animal models that mimic certain diseases are key elements for understanding the underlying mechanisms of disease pathogenesis, as well as for the development and efficacy testing of therapeutics and vaccines. An updated veterinary curriculum is essential to empower future graduates to work in an international environment, applying international standards for disease surveillance, veterinary public health, food safety and animal welfare.
Collapse
Affiliation(s)
- R E Kahn
- Avian Flu Action, Warrington, Cheshire, UK
| | | | | |
Collapse
|
38
|
David Walter W, Walsh DP, Farnsworth ML, Winkelman DL, Miller MW. Soil clay content underlies prion infection odds. Nat Commun 2011; 2:200. [PMID: 21326232 PMCID: PMC3105318 DOI: 10.1038/ncomms1203] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 01/19/2011] [Indexed: 11/09/2022] Open
Abstract
Environmental factors—especially soil properties—have been suggested as potentially important in the transmission of infectious prion diseases. Because binding to montmorillonite (an aluminosilicate clay mineral) or clay-enriched soils had been shown to enhance experimental prion transmissibility, we hypothesized that prion transmission among mule deer might also be enhanced in ranges with relatively high soil clay content. In this study, we report apparent influences of soil clay content on the odds of prion infection in free-ranging deer. Analysis of data from prion-infected deer herds in northern Colorado, USA, revealed that a 1% increase in the clay-sized particle content in soils within the approximate home range of an individual deer increased its odds of infection by up to 8.9%. Our findings suggest that soil clay content and related environmental properties deserve greater attention in assessing risks of prion disease outbreaks and prospects for their control in both natural and production settings. The infectious prion diseases affect numerous hoofed animal species, and it has been suggested that the properties of the local soil affect transmission of these diseases. Here, the authors studied two North American locations and demonstrate that soil clay content can influence the infection rate in deer.
Collapse
Affiliation(s)
- W David Walter
- United States Department of the Interior, United States Geological Survey, Colorado Cooperative Fish and Wildlife Research Unit, Fort Collins, Colorado 80523-1484, USA
| | | | | | | | | |
Collapse
|
39
|
Soto C, Satani N. The intricate mechanisms of neurodegeneration in prion diseases. Trends Mol Med 2011; 17:14-24. [PMID: 20889378 PMCID: PMC3056171 DOI: 10.1016/j.molmed.2010.09.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 08/27/2010] [Accepted: 09/01/2010] [Indexed: 12/20/2022]
Abstract
Prion diseases are a group of infectious neurodegenerative diseases with an entirely novel mechanism of transmission, involving a protein-only infectious agent that propagates the disease by transmitting protein conformational changes. The disease results from extensive and progressive brain degeneration. The molecular mechanisms involved in neurodegeneration are not entirely known but involve multiple processes operating simultaneously and synergistically in the brain, including spongiform degeneration, synaptic alterations, brain inflammation, neuronal death and the accumulation of protein aggregates. Here, we review the pathways implicated in prion-induced brain damage and put the pieces together into a possible model of neurodegeneration in prion disorders. A more comprehensive understanding of the molecular basis of brain degeneration is essential to develop a much needed therapy for these devastating diseases.
Collapse
Affiliation(s)
- Claudio Soto
- Mitchell Center for Alzheimer's disease and related Brain disorders, Dept of Neurology, University of Texas Houston Medical School, Houston, TX 77030, USA
| | - Nikunj Satani
- Mitchell Center for Alzheimer's disease and related Brain disorders, Dept of Neurology, University of Texas Houston Medical School, Houston, TX 77030, USA
| |
Collapse
|
40
|
Capellari S, Strammiello R, Saverioni D, Kretzschmar H, Parchi P. Genetic Creutzfeldt-Jakob disease and fatal familial insomnia: insights into phenotypic variability and disease pathogenesis. Acta Neuropathol 2011; 121:21-37. [PMID: 20978903 DOI: 10.1007/s00401-010-0760-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 09/17/2010] [Accepted: 10/11/2010] [Indexed: 01/18/2023]
Abstract
Human prion diseases are a group of rare neurodegenerative disorders characterized by the conversion of the constitutively expressed prion protein, PrP(C), into an abnormally aggregated isoform, called PrP(Sc). While most people who develop a prion disease have no identifiable cause and a few acquire the disease through an identified source of infection, about 10-15% of patients are affected by a genetic form and carry either a point mutation or an insertion of octapeptide repeats in the prion protein gene. Prion diseases show the highest extent of phenotypic heterogeneity among neurodegenerative disorders and comprise three major disease entities with variable though overlapping phenotypic features: Creutzfeldt-Jakob disease (CJD), fatal insomnia and the Gerstmann-Sträussler-Scheinker syndrome. Both CJD and fatal insomnia are fully transmissible diseases, a feature that led to the isolation and characterization of different strains of the agent or prion showing distinctive clinical and neuropathological features after transmission to syngenic animals. Here, we review the current knowledge of the effects of the pathogenic mutations linked to genetic CJD and fatal familial insomnia on the prion protein metabolism and physicochemical properties, the disease phenotype and the strain characteristics. The data derived from studies in vitro and from those using cell and animal models are compared with those obtained from the analyses of the naturally occurring disease. The extent of phenotypic variation in genetic prion disease is analyzed in comparison to that of the sporadic disease, which has recently been the topic of a systematic and detailed characterization.
Collapse
|
41
|
Pan YH, Wang YC, Zhang LM, Duan SR. Protective effect of edaravone against PrP106-126-induced PC12 cell death. J Biochem Mol Toxicol 2010; 24:235-41. [PMID: 20806394 DOI: 10.1002/jbt.20330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The prion protein peptide PrP106-126 induces cell apoptosis through mechanisms involving production of intracellular reactive oxygen species. The present study investigated the effects of edaravone, a potent free radical scavenger in clinical use, on cell cytotoxicity induced by PrP106-126. Results showed that PrP106-126 decreased PC12 cell viability in a dose- and time-dependent manner. Edaravone significantly antagonized the cytotoxic effects of PrP106-126. Mechanistically, PrP106-126 decreased PC 12 intracellular glutathione (GSH) concentrations, decreased superoxide dismutase (SOD) enzyme activity, increased concentrations of the oxidation end product malondialdehyde (MDA), depolarized the mitochondrial membrane, and increased caspase-3 activity. Edaravone alone did not affect GSH, SOD, or MDA but did effectively reverse all of the intracellular prooxidant effects induced by PrP106-126 and inhibit induced apoptosis in PC12 cells. In conclusion, edaravone may be a viable candidate for the treatment of oxidative stress-induced neurodegenerative disease.
Collapse
Affiliation(s)
- Yong-Hui Pan
- Department of Neurology, First Clinical Medical College of Harbin Medical University, Harbin 150001, People's Republic of China
| | | | | | | |
Collapse
|
42
|
Kell DB. Towards a unifying, systems biology understanding of large-scale cellular death and destruction caused by poorly liganded iron: Parkinson's, Huntington's, Alzheimer's, prions, bactericides, chemical toxicology and others as examples. Arch Toxicol 2010; 84:825-89. [PMID: 20967426 PMCID: PMC2988997 DOI: 10.1007/s00204-010-0577-x] [Citation(s) in RCA: 266] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 07/14/2010] [Indexed: 12/11/2022]
Abstract
Exposure to a variety of toxins and/or infectious agents leads to disease, degeneration and death, often characterised by circumstances in which cells or tissues do not merely die and cease to function but may be more or less entirely obliterated. It is then legitimate to ask the question as to whether, despite the many kinds of agent involved, there may be at least some unifying mechanisms of such cell death and destruction. I summarise the evidence that in a great many cases, one underlying mechanism, providing major stresses of this type, entails continuing and autocatalytic production (based on positive feedback mechanisms) of hydroxyl radicals via Fenton chemistry involving poorly liganded iron, leading to cell death via apoptosis (probably including via pathways induced by changes in the NF-κB system). While every pathway is in some sense connected to every other one, I highlight the literature evidence suggesting that the degenerative effects of many diseases and toxicological insults converge on iron dysregulation. This highlights specifically the role of iron metabolism, and the detailed speciation of iron, in chemical and other toxicology, and has significant implications for the use of iron chelating substances (probably in partnership with appropriate anti-oxidants) as nutritional or therapeutic agents in inhibiting both the progression of these mainly degenerative diseases and the sequelae of both chronic and acute toxin exposure. The complexity of biochemical networks, especially those involving autocatalytic behaviour and positive feedbacks, means that multiple interventions (e.g. of iron chelators plus antioxidants) are likely to prove most effective. A variety of systems biology approaches, that I summarise, can predict both the mechanisms involved in these cell death pathways and the optimal sites of action for nutritional or pharmacological interventions.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry and the Manchester Interdisciplinary Biocentre, The University of Manchester, Manchester M1 7DN, UK.
| |
Collapse
|
43
|
Williams BB, Kwakye GF, Wegrzynowicz M, Li D, Aschner M, Erikson KM, Bowman AB. Altered manganese homeostasis and manganese toxicity in a Huntington's disease striatal cell model are not explained by defects in the iron transport system. Toxicol Sci 2010; 117:169-79. [PMID: 20547568 PMCID: PMC2923282 DOI: 10.1093/toxsci/kfq174] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Accepted: 06/07/2010] [Indexed: 01/19/2023] Open
Abstract
Expansion of a polyglutamine tract in Huntingtin (Htt) leads to the degeneration of medium spiny neurons in Huntington's disease (HD). Furthermore, the HTT gene has been functionally linked to iron (Fe) metabolism, and HD patients show alterations in brain and peripheral Fe homeostasis. Recently, we discovered that expression of mutant HTT is associated with impaired manganese (Mn) uptake following overexposure in a striatal neuronal cell line and mouse model of HD. Here we test the hypothesis that the transferrin receptor (TfR)-mediated Fe uptake pathway is responsible for the HD-associated defects in Mn uptake. Western blot analysis showed that TfR levels are reduced in the mutant STHdh(Q111/Q111) striatal cell line, whereas levels of the Fe and Mn transporter, divalent metal transporter 1 (DMT1), are unchanged. To stress the Fe transport system, we exposed mutant and wild-type cells to elevated Fe(III), which revealed a subtle impairment in net Fe uptake only at the highest Fe exposures. In contrast, the HD mutant line exhibited substantial deficits in net Mn uptake, even under basal conditions. Finally, to functionally evaluate a role for Fe transporters in the Mn uptake deficit, we examined Mn toxicity in the presence of saturating Fe(III) levels. Although Fe(III) exposure decreased Mn neurotoxicity, it did so equally for wild-type and mutant cells. Therefore, although Fe transporters contribute to Mn uptake and toxicity in the striatal cell lines, functional alterations in this pathway are insufficient to explain the strong Mn resistance phenotype of this HD cell model.
Collapse
Affiliation(s)
- B. Blairanne Williams
- Neuroscience Graduate Program
- Department of Neurology
- Vanderbilt Kennedy Center for Research on Human Development
- Vanderbilt Brain Institute
- Center in Molecular Toxicology
| | - Gunnar F. Kwakye
- Neuroscience Graduate Program
- Department of Neurology
- Vanderbilt Kennedy Center for Research on Human Development
- Vanderbilt Brain Institute
- Center in Molecular Toxicology
| | | | - Daphne Li
- Department of Neurology
- Vanderbilt Kennedy Center for Research on Human Development
| | - Michael Aschner
- Vanderbilt Kennedy Center for Research on Human Development
- Vanderbilt Brain Institute
- Center in Molecular Toxicology
- Department of Pediatrics
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Keith M. Erikson
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, North Carolina 27402-6107
| | - Aaron B. Bowman
- Department of Neurology
- Vanderbilt Kennedy Center for Research on Human Development
- Vanderbilt Brain Institute
- Center in Molecular Toxicology
| |
Collapse
|
44
|
Fitsanakis VA, Zhang N, Garcia S, Aschner M. Manganese (Mn) and iron (Fe): interdependency of transport and regulation. Neurotox Res 2010; 18:124-31. [PMID: 19921534 PMCID: PMC7909719 DOI: 10.1007/s12640-009-9130-1] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 10/28/2009] [Accepted: 11/02/2009] [Indexed: 12/25/2022]
Abstract
Manganese (Mn) and iron (Fe) are transition metals that are crucial to the appropriate growth, development, function, and maintenance of biological organisms. Because of their chemical similarity, in organisms ranging from bacteria to mammals they share and compete for many protein transporters, such as the divalent metal transporter-1. As such, during conditions of low Fe, abnormal Mn accumulation occurs. Conversely, when Mn concentrations are altered, the homeostasis and deposition of Fe and other transition metals are disrupted. Our lab has undertaken a series of studies in rats involving pregnant dams, neo- and perinatal pups, and adult animals. Animals were exposed to various concentrations of dietary Fe and/or Mn, and protein transporter expression, blood Mn and Fe concentrations, brain transition metal concentrations, and temporal brain deposition patterns were examined. As a result, we have demonstrated the importance of the interdependence of the transport of Mn and Fe, and established brain metal concentrations in several longitudinal studies. The purpose of this review is to examine these studies in their entirety and highlight the importance of monitoring the deposition and accumulation of both Mn and Fe when designing future studies related to either dietary or environmental changes in transition metal levels. Finally, this review will provide information about various transport proteins currently under investigation in the research community related to Fe and Mn regulation and transport.
Collapse
|
45
|
The depletion of α and β PrP from complex mixtures. J Virol Methods 2010; 169:253-8. [PMID: 20603150 DOI: 10.1016/j.jviromet.2010.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 06/21/2010] [Accepted: 06/28/2010] [Indexed: 11/20/2022]
Abstract
Prion disorders occur when endogenous prion protein (PrP(C)) undergoes a conformational change from a predominantly α-helix-rich structure to an insoluble β-sheet-rich structure (PrP(Sc)). The resulting PrP(Sc) then in some way facilitates the progressive transformation of nearby PrP(C) to PrP(Sc). In time this results in the deposition of insoluble PrP(Sc) aggregates in the brain; aggregate deposition is irreversible and is ultimately fatal. Prion diseases are transmissible orally or through transplantation (including blood transfusion). Current diagnostic methods are limited in that they lack the ability to distinguish qualitatively between PrP(C) and PrP(Sc). PrP has been shown to bind divalent cations including copper and zinc, these cations are toxic and thus of limited use in the removal of PrP from solutions destined for administration to subjects. We have immobilised Fe(3+) to an inert Sepharose resin; this resin was capable of quantitatively removing endogenous and recombinant PrP(C) and recombinant β PrP from complex solutions. The low toxicity of Fe(3+) suggests that the resin described in this report may be of practical use in the depletion of PrP from blood products destined for human use.
Collapse
|
46
|
Das D, Luo X, Singh A, Gu Y, Ghosh S, Mukhopadhyay CK, Chen SG, Sy MS, Kong Q, Singh N. Paradoxical role of prion protein aggregates in redox-iron induced toxicity. PLoS One 2010; 5:e11420. [PMID: 20625431 PMCID: PMC2897850 DOI: 10.1371/journal.pone.0011420] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 06/07/2010] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Imbalance of iron homeostasis has been reported in sporadic Creutzfeldt-Jakob-disease (sCJD) affected human and scrapie infected animal brains, but the contribution of this phenotype to disease associated neurotoxicity is unclear. METHODOLOGY/PRINCIPAL FINDINGS Using cell models of familial prion disorders, we demonstrate that exposure of cells expressing normal prion protein (PrP(C)) or mutant PrP forms to a source of redox-iron induces aggregation of PrP(C) and specific mutant PrP forms. Initially this response is cytoprotective, but becomes increasingly toxic with time due to accumulation of PrP-ferritin aggregates. Mutant PrP forms that do not aggregate are not cytoprotective, and cells show signs of acute toxicity. Intracellular PrP-ferritin aggregates induce the expression of LC3-II, indicating stimulation of autophagy in these cells. Similar observations are noted in sCJD and scrapie infected hamster brains, lending credence to these results. Furthermore, phagocytosis of PrP-ferritin aggregates by astrocytes is cytoprotective, while culture in astrocyte conditioned medium (CM) shows no measurable effect. Exposure to H(2)O(2), on the other hand, does not cause aggregation of PrP, and cells show acute toxicity that is alleviated by CM. CONCLUSIONS/SIGNIFICANCE These observations suggest that aggregation of PrP in response to redox-iron is cytoprotective. However, subsequent co-aggregation of PrP with ferritin induces intracellular toxicity unless the aggregates are degraded by autophagosomes or phagocytosed by adjacent scavenger cells. H(2)O(2), on the other hand, does not cause aggregation of PrP, and induces toxicity through extra-cellular free radicals. Together with previous observations demonstrating imbalance of iron homeostasis in prion disease affected brains, these observations provide insight into the mechanism of neurotoxicity by redox-iron, and the role of PrP in this process.
Collapse
Affiliation(s)
- Dola Das
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Xiu Luo
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Ajay Singh
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Yaping Gu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Soumya Ghosh
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | | | - Shu G. Chen
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Man-Sun Sy
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Qingzhong Kong
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Neena Singh
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| |
Collapse
|
47
|
Choi CJ, Anantharam V, Martin DP, Nicholson EM, Richt JA, Kanthasamy A, Kanthasamy AG. Manganese upregulates cellular prion protein and contributes to altered stabilization and proteolysis: relevance to role of metals in pathogenesis of prion disease. Toxicol Sci 2010; 115:535-46. [PMID: 20176619 PMCID: PMC2871751 DOI: 10.1093/toxsci/kfq049] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 02/05/2010] [Indexed: 11/14/2022] Open
Abstract
Prion diseases are fatal neurodegenerative diseases resulting from misfolding of normal cellular prion (PrP(C)) into an abnormal form of scrapie prion (PrP(Sc)). The cellular mechanisms underlying the misfolding of PrP(C) are not well understood. Since cellular prion proteins harbor divalent metal-binding sites in the N-terminal region, we examined the effect of manganese on PrP(C) processing in in vitro models of prion disease. Exposure to manganese significantly increased PrP(C) levels both in cytosolic and in membrane-rich fractions in a time-dependent manner. Manganese-induced PrP(C) upregulation was independent of messenger RNA transcription or stability. Additionally, manganese treatment did not alter the PrP(C) degradation by either proteasomal or lysosomal pathways. Interestingly, pulse-chase analysis showed that the PrP(C) turnover rate was significantly altered with manganese treatment, indicating increased stability of PrP(C) with the metal exposure. Limited proteolysis studies with proteinase-K further supported that manganese increases the stability of PrP(C). Incubation of mouse brain slice cultures with manganese also resulted in increased prion protein levels and higher intracellular manganese accumulation. Furthermore, exposure of manganese to an infectious prion cell model, mouse Rocky Mountain Laboratory-infected CAD5 cells, significantly increased prion protein levels. Collectively, our results demonstrate for the first time that divalent metal manganese can alter the stability of prion proteins and suggest that manganese-induced stabilization of prion protein may play a role in prion protein misfolding and prion disease pathogenesis.
Collapse
Affiliation(s)
- Christopher J. Choi
- Department of Biomedical Sciences, Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Dustin P. Martin
- Department of Biomedical Sciences, Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Eric M. Nicholson
- Viruses and Prion Diseases of Livestock Research Unit, National Animal Disease Center, USDA, ARS, Ames, Iowa 50011
| | - Jürgen A. Richt
- Department of Biomedical Sciences, Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506-5601
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Anumantha G. Kanthasamy
- Department of Biomedical Sciences, Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| |
Collapse
|
48
|
Singh N, Singh A, Das D, Mohan ML. Redox control of prion and disease pathogenesis. Antioxid Redox Signal 2010; 12:1271-94. [PMID: 19803746 PMCID: PMC2864664 DOI: 10.1089/ars.2009.2628] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 09/22/2009] [Accepted: 10/03/2009] [Indexed: 11/12/2022]
Abstract
Imbalance of brain metal homeostasis and associated oxidative stress by redox-active metals like iron and copper is an important trigger of neurotoxicity in several neurodegenerative conditions, including prion disorders. Whereas some reports attribute this to end-stage disease, others provide evidence for specific mechanisms leading to brain metal dyshomeostasis during disease progression. In prion disorders, imbalance of brain-iron homeostasis is observed before end-stage disease and worsens with disease progression, implicating iron-induced oxidative stress in disease pathogenesis. This is an unexpected observation, because the underlying cause of brain pathology in all prion disorders is PrP-scrapie (PrP(Sc)), a beta-sheet-rich conformation of a normal glycoprotein, the prion protein (PrP(C)). Whether brain-iron dyshomeostasis occurs because of gain of toxic function by PrP(Sc) or loss of normal function of PrP(C) remains unclear. In this review, we summarize available evidence suggesting the involvement of oxidative stress in prion-disease pathogenesis. Subsequently, we review the biology of PrP(C) to highlight its possible role in maintaining brain metal homeostasis during health and the contribution of PrP(Sc) in inducing brain metal imbalance with disease progression. Finally, we discuss possible therapeutic avenues directed at restoring brain metal homeostasis and alleviating metal-induced oxidative stress in prion disorders.
Collapse
Affiliation(s)
- Neena Singh
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
49
|
Abstract
The crucial event in the development of transmissible spongiform encephalopathies (TSEs) is the conformational change of a host-encoded membrane protein - the cellular PrPC - into a disease associated, fibril-forming isoform PrPSc. This conformational transition from the α-helix-rich cellular form into the mainly β-sheet containing counterpart initiates an ‘autocatalytic’ reaction which leads to the accumulation of amyloid fibrils in the central nervous system (CNS) and to neurodegeneration, a hallmark of TSEs. The exact molecular mechanisms which lead to the conformational change are still unknown. It also remains to be brought to light how a polypeptide chain can adopt at least two stable conformations. This review focuses on structural aspects of the prion protein with regard to protein-protein interactions and the initiation of prion protein misfolding. It therefore highlights parts of the protein which might play a notable role in the conformational transition from PrPC to PrPSc and consequently in inducing a fatal chain reaction of protein misfolding. Furthermore, features of different proteins, which are able to adopt insoluble fibrillar states under certain circumstances, are compared to PrP in an attempt to understand the unique characteristics of prion diseases.
Collapse
Affiliation(s)
- L Kupfer
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | | | | |
Collapse
|
50
|
Pandey KK, Snyder JP, Liotta DC, Musaev DG. Computational Studies of Transition Metal Selectivity of Octapeptide Repeat Region of Prion Protein (PrP). J Phys Chem B 2009; 114:1127-35. [PMID: 20020721 DOI: 10.1021/jp909945e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Krishna K. Pandey
- Cherry L. Emerson Center for Scientific Computation and Department of Chemistry, Emory University, Atlanta, Georgia 30322, and School of Chemical Sciences, Devi Ahilya University Indore, Indore 452001, India
| | - James P. Snyder
- Cherry L. Emerson Center for Scientific Computation and Department of Chemistry, Emory University, Atlanta, Georgia 30322, and School of Chemical Sciences, Devi Ahilya University Indore, Indore 452001, India
| | - Dennis C. Liotta
- Cherry L. Emerson Center for Scientific Computation and Department of Chemistry, Emory University, Atlanta, Georgia 30322, and School of Chemical Sciences, Devi Ahilya University Indore, Indore 452001, India
| | - Djamaladdin G. Musaev
- Cherry L. Emerson Center for Scientific Computation and Department of Chemistry, Emory University, Atlanta, Georgia 30322, and School of Chemical Sciences, Devi Ahilya University Indore, Indore 452001, India
| |
Collapse
|