1
|
Shang Y, Wu X, Luo H. A fluorescent probe based on pyrazoline with significant Stokes shifts for the detection of Cu 2+ ions and its applications. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024. [PMID: 39704273 DOI: 10.1039/d4ay01772b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
A simple pyrazoline derivative BBD has been synthesized for detecting Cu2+ in EtOH/HEPES (v/v = 1 : 1, pH = 7). The probe has high selectivity for Cu2+ by quenching the fluorescence intensity and was sensitive to pH. When excited at 398 nm, fluorescence is emitted at 520 nm with a Stokes shift of 122 nm, which is larger than that of other reported pyrazoline skeleton probes. The detection limit (LOD) of the probe for Cu2+ is 0.862 μM which is lower than the value of the WHO limit (31.5 μM) in water. The 2 : 1 binding mode and probable recognition mechanism of the probe with Cu2+ were confirmed by Job's plot, IR, ESI-MS, and TD-DFT analysis. Moreover, the probe has been successfully applied to actual water samples as well as cell and zebrafish imaging.
Collapse
Affiliation(s)
- Yajing Shang
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, China.
| | - Xinghu Wu
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, China.
| | - Haoting Luo
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, China.
| |
Collapse
|
2
|
Song F, Kovac V, Mohammadi B, Littau JL, Scharfenberg F, Matamoros Angles A, Vanni I, Shafiq M, Orge L, Galliciotti G, Djakkani S, Linsenmeier L, Černilec M, Hartman K, Jung S, Tatzelt J, Neumann JE, Damme M, Tschirner SK, Lichtenthaler SF, Ricklefs FL, Sauvigny T, Schmitz M, Zerr I, Puig B, Tolosa E, Ferrer I, Magnus T, Rupnik MS, Sepulveda-Falla D, Matschke J, Šmid LM, Bresjanac M, Andreoletti O, Krasemann S, Foliaki ST, Nonno R, Becker-Pauly C, Monzo C, Crozet C, Haigh CL, Glatzel M, Curin Serbec V, Altmeppen HC. Cleavage site-directed antibodies reveal the prion protein in humans is shed by ADAM10 at Y226 and associates with misfolded protein deposits in neurodegenerative diseases. Acta Neuropathol 2024; 148:2. [PMID: 38980441 PMCID: PMC11233397 DOI: 10.1007/s00401-024-02763-5] [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: 04/24/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
Proteolytic cell surface release ('shedding') of the prion protein (PrP), a broadly expressed GPI-anchored glycoprotein, by the metalloprotease ADAM10 impacts on neurodegenerative and other diseases in animal and in vitro models. Recent studies employing the latter also suggest shed PrP (sPrP) to be a ligand in intercellular communication and critically involved in PrP-associated physiological tasks. Although expectedly an evolutionary conserved event, and while soluble forms of PrP are present in human tissues and body fluids, for the human body neither proteolytic PrP shedding and its cleavage site nor involvement of ADAM10 or the biological relevance of this process have been demonstrated thus far. In this study, cleavage site prediction and generation (plus detailed characterization) of sPrP-specific antibodies enabled us to identify PrP cleaved at tyrosin 226 as the physiological and apparently strictly ADAM10-dependent shed form in humans. Using cell lines, neural stem cells and brain organoids, we show that shedding of human PrP can be stimulated by PrP-binding ligands without targeting the protease, which may open novel therapeutic perspectives. Site-specific antibodies directed against human sPrP also detect the shed form in brains of cattle, sheep and deer, hence in all most relevant species naturally affected by fatal and transmissible prion diseases. In human and animal prion diseases, but also in patients with Alzheimer`s disease, sPrP relocalizes from a physiological diffuse tissue pattern to intimately associate with extracellular aggregated deposits of misfolded proteins characteristic for the respective pathological condition. Findings and research tools presented here will accelerate novel insight into the roles of PrP shedding (as a process) and sPrP (as a released factor) in neurodegeneration and beyond.
Collapse
Affiliation(s)
- Feizhi Song
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Valerija Kovac
- Centre for Immunology and Development, Blood Transfusion Centre of Slovenia (BTCS), Ljubljana, Slovenia
| | - Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jessica L Littau
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | | - Andreu Matamoros Angles
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ilaria Vanni
- Department of Food Safety and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Leonor Orge
- National Institute for Agricultural and Veterinary Research (INIAV), Oeiras, Portugal
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Giovanna Galliciotti
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Salma Djakkani
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Luise Linsenmeier
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Maja Černilec
- Centre for Immunology and Development, Blood Transfusion Centre of Slovenia (BTCS), Ljubljana, Slovenia
| | - Katrina Hartman
- Centre for Immunology and Development, Blood Transfusion Centre of Slovenia (BTCS), Ljubljana, Slovenia
| | - Sebastian Jung
- Department of Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Jörg Tatzelt
- Department of Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Cluster of Excellence RESOLV, Ruhr University Bochum, Bochum, Germany
| | - Julia E Neumann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- Center for Molecular Neurobiology Hamburg (ZMNH), UKE, Hamburg, Germany
| | - Markus Damme
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| | - Sarah K Tschirner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University Munich, 81675, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University Munich, 81675, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Thomas Sauvigny
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Matthias Schmitz
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI), UKE, Hamburg, Germany
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, IDIBELL, Hospitalet de Llobregat, Spain
| | - Tim Magnus
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI), UKE, Hamburg, Germany
| | - Marjan S Rupnik
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Diego Sepulveda-Falla
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jakob Matschke
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Lojze M Šmid
- LNPR, Institute of Pathophysiology and Prion Laboratory, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mara Bresjanac
- LNPR, Institute of Pathophysiology and Prion Laboratory, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Olivier Andreoletti
- UMR INRAE ENVT 1225, Interactions Hôtes-Agents Pathogènes, École Nationale Vétérinaire de Toulouse, Toulouse, France
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Simote T Foliaki
- Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, MT, USA
| | - Romolo Nonno
- Department of Food Safety and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | | | - Cecile Monzo
- Institute for Regenerative Medicine and Biotherapies (IRMB), Neural Stem Cell, MSC and Neurodegenerative Diseases, INSERM, Montpellier, France
| | - Carole Crozet
- Institute for Regenerative Medicine and Biotherapies (IRMB), Neural Stem Cell, MSC and Neurodegenerative Diseases, INSERM, Montpellier, France
| | - Cathryn L Haigh
- Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, MT, USA
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Vladka Curin Serbec
- Centre for Immunology and Development, Blood Transfusion Centre of Slovenia (BTCS), Ljubljana, Slovenia.
| | - Hermann C Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| |
Collapse
|
3
|
Liu J, Su G, Duan C, Sun Z, Xiao S, Zhou Y, Fang L. Porcine reproductive and respiratory syndrome virus infection activates ADAM17 to induce inflammatory responses. Vet Microbiol 2024; 292:110066. [PMID: 38555788 DOI: 10.1016/j.vetmic.2024.110066] [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: 01/23/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
Porcine reproductive and respiratory syndrome (PRRS), which has posed substantial threats to the swine industry worldwide, is primarily characterized by interstitial pneumonia. A disintegrin and metalloproteinase 17 (ADAM17) is a multifunctional sheddase involved in various inflammatory diseases. Herein, our study showed that PRRS virus (PRRSV) infection elevated ADAM17 activity, as demonstrated in primary porcine alveolar macrophages (PAMs), an immortalized PAM cell line (IPAM cells), and the lung tissues of PRRSV-infected piglets. We found that PRRSV infection promoted ADAM17 translocation from the endoplasmic reticulum to the Golgi by enhancing its interaction with inactive rhomboid protein 2 (iRhom2), a newly identified ADAM17 regulator, which in turn elevated ADAM17 activity. By screening for PRRSV-encoded structural proteins, viral envelope (E) and nucleocapsid (N) proteins were identified as the predominant ADAM17 activators. E and N proteins bind with both ADAM17 and iRhom2 to form ternary protein complexes, ultimately strengthening their interactions. Additionally, we demonstrated, using an ADAM17-knockout cell line, that ADAM17 augmented the shedding of soluble TNF-α, a pivotal inflammatory mediator. We also discovered that ADAM17-mediated cleavage of porcine TNF-α occurred between Arg-78 and Ser-79. By constructing a precision mutant cell line with Arg-78-Glu/Ser-79-Glu substitution mutations in TNF-α, we further revealed that the ADAM17-mediated production of soluble TNF-α contributed to the induction of inflammatory responses by PRRSV and its E and N proteins. Taken together, our results elucidate the mechanism by which PRRSV infection activates the iRhom2/ADAM17/TNF-α axis to enhance inflammatory responses, providing valuable insights into the elucidation of PRRSV pathogenesis.
Collapse
Affiliation(s)
- Jiao Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Guanning Su
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Chenrui Duan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Zheng Sun
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Shaobo Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Yanrong Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.
| | - Liurong Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.
| |
Collapse
|
4
|
Benarroch E. What Are the Roles of Cellular Prion Protein in Normal and Pathologic Conditions? Neurology 2024; 102:e209272. [PMID: 38484222 DOI: 10.1212/wnl.0000000000209272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 03/19/2024] Open
|
5
|
Srishti K, Negi O, Hota PK. Recent Development on Copper-Sensor and its Biological Applications: A Review. J Fluoresc 2024:10.1007/s10895-024-03587-y. [PMID: 38416283 DOI: 10.1007/s10895-024-03587-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/14/2024] [Indexed: 02/29/2024]
Abstract
Metal ion recognition is one of the most prospective research topics in the field of chemical sensors due to its wide range of clinical, biological and environmental applications. In this context, hydrazones are well known compounds that exhibit metal sensing and several biological properties due to the presence of N=CH- bond. Some of the biological properties includes anti-cancer, anti-tumor, anti-oxidant, anti-microbial activities. Hydrazones are also used as a ligand to detect metal ion as well as to generate metal complexes that exhibit medicinal properties. Thus, in recent years, many attempts were made to develop novel ligands with enhanced metal sensing and medicinal properties. In this review, some of the recent development on the hydrazones and their copper complexes are covered from the last few years from 2015-2023. These includes significance of copper ions, synthesis, biological properties, mechanism and metal sensing properties of some of the copper complexes were discussed.
Collapse
Affiliation(s)
- Km Srishti
- Department of Chemistry, School of Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Oseen Negi
- Department of Chemistry, School of Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Prasanta Kumar Hota
- Department of Chemistry, School of Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India.
| |
Collapse
|
6
|
Li Y, Wang L, Wang L, Zhu B, Ma J. A novel carbazole-based fluorometric and colorimetric sensor for the highly sensitive and specific detection of Cu 2+ in aqueous solution. RSC Adv 2023; 13:33276-33287. [PMID: 37964909 PMCID: PMC10641437 DOI: 10.1039/d3ra04571d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 11/02/2023] [Indexed: 11/16/2023] Open
Abstract
Based on the typical Suzuki coupling reaction and Schiff base reaction, a novel fluorescent molecular PCBW is synthesized and applied as a fluorescence and colorimetric sensor to detect Cu2+ in aqueous solution. The PCBW sensor presents the aggregation-caused quenching (ACQ) effect and at 1 × 10-5 mol L-1 it emits the strongest turquoise fluorescence in the DMSO-H2O system (fw = 40%). The sensor exhibits a 'turn-off' fluorescent characteristic by adding Cu2+, and its fluorescent intensity shows a reliable linear relationship with the Cu2+ concentration in the range of 0-6 × 10-6 mol L-1, with a detection limit of 1.19 × 10-8 mol L-1. Meanwhile, the PCBW sensor also exhibits the colorimetric sensing from colorless to light yellow. The sensor has good selectivity and anti-interference and its pH application range can be extended from 5 to 10. The intramolecular charge transfer (ICT) is speculated as the main fluorescence mechanism of PCBW. In addition, the sensor presents good reusability and is practicable to detect Cu2+ in diverse aqueous samples.
Collapse
Affiliation(s)
- Yiduo Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology Shanghai 200093 P. R. China
| | - Luyue Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology Shanghai 200093 P. R. China
| | - Liqiang Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology Shanghai 200093 P. R. China
| | - Baokun Zhu
- School of Materials and Chemistry, University of Shanghai for Science and Technology Shanghai 200093 P. R. China
| | - Jie Ma
- School of Materials and Chemistry, University of Shanghai for Science and Technology Shanghai 200093 P. R. China
| |
Collapse
|
7
|
Castle AR, Kang SG, Eskandari-Sedighi G, Wohlgemuth S, Nguyen MA, Drucker DJ, Mulvihill EE, Westaway D. Beta-endoproteolysis of the cellular prion protein by dipeptidyl peptidase-4 and fibroblast activation protein. Proc Natl Acad Sci U S A 2023; 120:e2209815120. [PMID: 36574660 PMCID: PMC9910601 DOI: 10.1073/pnas.2209815120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/18/2022] [Indexed: 12/29/2022] Open
Abstract
The cellular prion protein (PrPC) converts to alternatively folded pathogenic conformations (PrPSc) in prion infections and binds neurotoxic oligomers formed by amyloid-β α-synuclein, and tau. β-Endoproteolysis, which splits PrPC into N- and C-terminal fragments (N2 and C2, respectively), is of interest because a protease-resistant, C2-sized fragment (C2Sc) accumulates in the brain during prion infections, seemingly comprising the majority of PrPSc at disease endpoint in mice. However, candidates for the underlying proteolytic mechanism(s) remain unconfirmed in vivo. Here, a cell-based screen of protease inhibitors unexpectedly linked type II membrane proteins of the S9B serine peptidase subfamily to PrPC β-cleavage. Overexpression experiments in cells and assays with recombinant proteins confirmed that fibroblast activation protein (FAP) and its paralog, dipeptidyl peptidase-4 (DPP4), cleave directly at multiple sites within PrPC's N-terminal domain. For wild-type mouse and human PrPC substrates expressed in cells, the rank orders of activity were human FAP ~ mouse FAP > mouse DPP4 > human DPP4 and human FAP > mouse FAP > mouse DPP4 >> human DPP4, respectively. C2 levels relative to total PrPC were reduced in several tissues from FAP-null mice, and, while knockout of DPP4 lacked an analogous effect, the combined DPP4/FAP inhibitor linagliptin, but not the FAP-specific inhibitor SP-13786, reduced C2Sc and total PrPSc levels in two murine cell-based models of prion infections. Thus, the net activity of the S9B peptidases FAP and DPP4 and their cognate inhibitors/modulators affect the physiology and pathogenic potential of PrPC.
Collapse
Affiliation(s)
- Andrew R. Castle
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, ABT6G 2M8, Canada
- Department of Medicine, University of Alberta, Edmonton, ABT6G 2G3, Canada
| | - Sang-Gyun Kang
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, ABT6G 2M8, Canada
- Department of Medicine, University of Alberta, Edmonton, ABT6G 2G3, Canada
| | - Ghazaleh Eskandari-Sedighi
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, ABT6G 2M8, Canada
- Department of Biochemistry, University of Alberta, Edmonton, ABT6G 2H7, Canada
| | - Serene Wohlgemuth
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, ABT6G 2M8, Canada
- Department of Medicine, University of Alberta, Edmonton, ABT6G 2G3, Canada
| | - My-Anh Nguyen
- University of Ottawa Heart Institute, Ottawa, ONK1Y 4W7, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ONK1H 8M5, Canada
| | - Daniel J. Drucker
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ONM5G 1X5, Canada
- Department of Medicine, University of Toronto, Toronto, ONM5S 2J7, Canada
| | - Erin E. Mulvihill
- University of Ottawa Heart Institute, Ottawa, ONK1Y 4W7, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ONK1H 8M5, Canada
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, ABT6G 2M8, Canada
- Department of Medicine, University of Alberta, Edmonton, ABT6G 2G3, Canada
- Department of Biochemistry, University of Alberta, Edmonton, ABT6G 2H7, Canada
| |
Collapse
|
8
|
Vanni I, Iacobone F, D’Agostino C, Giovannelli M, Pirisinu L, Altmeppen HC, Castilla J, Torres JM, Agrimi U, Nonno R. An optimized Western blot assay provides a comprehensive assessment of the physiological endoproteolytic processing of the prion protein. J Biol Chem 2022; 299:102823. [PMID: 36565989 PMCID: PMC9867980 DOI: 10.1016/j.jbc.2022.102823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
The prion protein (PrPC) is subjected to several conserved endoproteolytic events producing bioactive fragments that are of increasing interest for their physiological functions and their implication in the pathogenesis of prion diseases and other neurodegenerative diseases. However, systematic and comprehensive investigations on the full spectrum of PrPC proteoforms have been hampered by the lack of methods able to identify all PrPC-derived proteoforms. Building on previous knowledge of PrPC endoproteolytic processing, we thus developed an optimized Western blot assay able to obtain the maximum information about PrPC constitutive processing and the relative abundance of PrPC proteoforms in a complex biological sample. This approach led to the concurrent identification of the whole spectrum of known endoproteolytic-derived PrPC proteoforms in brain homogenates, including C-terminal, N-terminal and, most importantly, shed PrPC-derived fragments. Endoproteolytic processing of PrPC was remarkably similar in the brain of widely used wild type and transgenic rodent models, with α-cleavage-derived C1 representing the most abundant proteoform and ADAM10-mediated shedding being an unexpectedly prominent proteolytic event. Interestingly, the relative amount of shed PrPC was higher in WT mice than in most other models. Our results indicate that constitutive endoproteolytic processing of PrPC is not affected by PrPC overexpression or host factors other than PrPC but can be impacted by PrPC primary structure. Finally, this method represents a crucial step in gaining insight into pathophysiological roles, biomarker suitability, and therapeutic potential of shed PrPC and for a comprehensive appraisal of PrPC proteoforms in therapies, drug screening, or in the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ilaria Vanni
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy.
| | - Floriana Iacobone
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Claudia D’Agostino
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Matteo Giovannelli
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Pirisinu
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | | | - Joaquin Castilla
- Basque Research and Technology Alliance (BRTA) - CIC BioGUNE & IKERBasque, Bizkaia, Spain,Centro de Investigación Biomédica en Red de Enfermedades infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Maria Torres
- Centro de Investigación en Sanidad Animal (CISA-INIA-CSIC), Valdeolmos, Madrid, Spain
| | - Umberto Agrimi
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Romolo Nonno
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
9
|
Mays CE, Trinh THT, Telling G, Kang HE, Ryou C. Endoproteolysis of cellular prion protein by plasmin hinders propagation of prions. Front Mol Neurosci 2022; 15:990136. [PMID: 36117913 PMCID: PMC9478470 DOI: 10.3389/fnmol.2022.990136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/15/2022] [Indexed: 01/21/2023] Open
Abstract
Many questions surround the underlying mechanism for the differential metabolic processing observed for the prion protein (PrP) in healthy and prion-infected mammals. Foremost, the physiological α-cleavage of PrP interrupts a region critical for both toxicity and conversion of cellular PrP (PrP C ) into its misfolded pathogenic isoform (PrP Sc ) by generating a glycosylphosphatidylinositol (GPI)-anchored C1 fragment. During prion diseases, alternative β-cleavage of PrP becomes prominent, producing a GPI-anchored C2 fragment with this particular region intact. It remains unexplored whether physical up-regulation of α-cleavage can inhibit disease progression. Furthermore, several pieces of evidence indicate that a disintegrin and metalloproteinase (ADAM) 10 and ADAM17 play a much smaller role in the α-cleavage of PrP C than originally believed, thus presenting the need to identify the primary protease(s) responsible. For this purpose, we characterized the ability of plasmin to perform PrP α-cleavage. Then, we conducted functional assays using protein misfolding cyclic amplification (PMCA) and prion-infected cell lines to clarify the role of plasmin-mediated α-cleavage during prion propagation. Here, we demonstrated an inhibitory role of plasmin for PrP Sc formation through PrP α-cleavage that increased C1 fragments resulting in reduced prion conversion compared with non-treated PMCA and cell cultures. The reduction of prion infectious titer in the bioassay of plasmin-treated PMCA material also supported the inhibitory role of plasmin on PrP Sc replication. Our results suggest that plasmin-mediated endoproteolytic cleavage of PrP may be an important event to prevent prion propagation.
Collapse
Affiliation(s)
- Charles E. Mays
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Trang H. T. Trinh
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, South Korea,Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | - Glenn Telling
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, United States,Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States,Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Hae-Eun Kang
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States,Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States,Reference Laboratory for Chronic Wasting Disease (CWD), Foreign Animal Disease Division, Animal and Plant Quarantine Agency, Gimcheon, South Korea,Hae-Eun Kang,
| | - Chongsuk Ryou
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, United States,Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, South Korea,Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea,Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States,*Correspondence: Chongsuk Ryou,
| |
Collapse
|
10
|
Shafiq M, Da Vela S, Amin L, Younas N, Harris DA, Zerr I, Altmeppen HC, Svergun D, Glatzel M. The prion protein and its ligands: Insights into structure-function relationships. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119240. [PMID: 35192891 DOI: 10.1016/j.bbamcr.2022.119240] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/23/2022] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
The prion protein is a multifunctional protein that exists in at least two different folding states. It is subject to diverse proteolytic processing steps that lead to prion protein fragments some of which are membrane-bound whereas others are soluble. A multitude of ligands bind to the prion protein and besides proteinaceous binding partners, interaction with metal ions and nucleic acids occurs. Although of great importance, information on structural and functional consequences of prion protein binding to its partners is limited. Here, we will reflect on the structure-function relationship of the prion protein and its binding partners considering the different folding states and prion protein fragments.
Collapse
Affiliation(s)
- Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany
| | - Stefano Da Vela
- European Molecular Biology Laboratory (EMBL), Hamburg c/o German Electron Synchrotron (DESY), Notkestraße 85, 22607 Hamburg, Germany
| | - Ladan Amin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Neelam Younas
- Department of Neurology, University Medical Center Goettingen, Robert-Koch-str. 40, 37075 Goettingen, Germany
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Inga Zerr
- Department of Neurology, University Medical Center Goettingen, Robert-Koch-str. 40, 37075 Goettingen, Germany
| | - Hermann C Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany
| | - Dmitri Svergun
- European Molecular Biology Laboratory (EMBL), Hamburg c/o German Electron Synchrotron (DESY), Notkestraße 85, 22607 Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| |
Collapse
|
11
|
AbhijnaKrishna R, Velmathi S. A review on fluorimetric and colorimetric detection of metal ions by chemodosimetric approach 2013–2021. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214401] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
12
|
|
13
|
Srisuwan P, Sappasombut A, Thongyod W, Jantarat T, Tipmanee V, Leesakul N, Sooksawat D. Highly sensitive and selective coumarin-based fluorescent chemosensor for Cu2+ detection. J Photochem Photobiol A Chem 2022. [DOI: 10.1016/j.jphotochem.2022.113841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
14
|
Mohammadi B, Song F, Matamoros-Angles A, Shafiq M, Damme M, Puig B, Glatzel M, Altmeppen HC. Anchorless risk or released benefit? An updated view on the ADAM10-mediated shedding of the prion protein. Cell Tissue Res 2022; 392:215-234. [PMID: 35084572 PMCID: PMC10113312 DOI: 10.1007/s00441-022-03582-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/12/2022] [Indexed: 11/24/2022]
Abstract
The prion protein (PrP) is a broadly expressed glycoprotein linked with a multitude of (suggested) biological and pathological implications. Some of these roles seem to be due to constitutively generated proteolytic fragments of the protein. Among them is a soluble PrP form, which is released from the surface of neurons and other cell types by action of the metalloprotease ADAM10 in a process termed 'shedding'. The latter aspect is the focus of this review, which aims to provide a comprehensive overview on (i) the relevance of proteolytic processing in regulating cellular PrP functions, (ii) currently described involvement of shed PrP in neurodegenerative diseases (including prion diseases and Alzheimer's disease), (iii) shed PrP's expected roles in intercellular communication in many more (patho)physiological conditions (such as stroke, cancer or immune responses), (iv) and the need for improved research tools in respective (future) studies. Deeper mechanistic insight into roles played by PrP shedding and its resulting fragment may pave the way for improved diagnostics and future therapeutic approaches in diseases of the brain and beyond.
Collapse
Affiliation(s)
- Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- Working Group for Interdisciplinary Neurobiology and Immunology (INI Research), Hamburg, Germany
| | - Feizhi Song
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Andreu Matamoros-Angles
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Damme
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | |
Collapse
|
15
|
Kovač V, Čurin Šerbec V. Prion Protein: The Molecule of Many Forms and Faces. Int J Mol Sci 2022; 23:ijms23031232. [PMID: 35163156 PMCID: PMC8835406 DOI: 10.3390/ijms23031232] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/10/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Cellular prion protein (PrPC) is a glycosylphosphatidylinositol (GPI)-anchored protein most abundantly found in the outer membrane of neurons. Due to structural characteristics (a flexible tail and structured core), PrPC interacts with a wide range of partners. Although PrPC has been proposed to be involved in many physiological functions, only peripheral nerve myelination homeostasis has been confirmed as a bona fide function thus far. PrPC misfolding causes prion diseases and PrPC has been shown to mediate β-rich oligomer-induced neurotoxicity in Alzheimer’s and Parkinson’s disease as well as neuroprotection in ischemia. Upon proteolytic cleavage, PrPC is transformed into released and attached forms of PrP that can, depending on the contained structural characteristics of PrPC, display protective or toxic properties. In this review, we will outline prion protein and prion protein fragment properties as well as overview their involvement with interacting partners and signal pathways in myelination, neuroprotection and neurodegenerative diseases.
Collapse
|
16
|
Loh D, Reiter RJ. Melatonin: Regulation of Prion Protein Phase Separation in Cancer Multidrug Resistance. Molecules 2022; 27:705. [PMID: 35163973 PMCID: PMC8839844 DOI: 10.3390/molecules27030705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
The unique ability to adapt and thrive in inhospitable, stressful tumor microenvironments (TME) also renders cancer cells resistant to traditional chemotherapeutic treatments and/or novel pharmaceuticals. Cancer cells exhibit extensive metabolic alterations involving hypoxia, accelerated glycolysis, oxidative stress, and increased extracellular ATP that may activate ancient, conserved prion adaptive response strategies that exacerbate multidrug resistance (MDR) by exploiting cellular stress to increase cancer metastatic potential and stemness, balance proliferation and differentiation, and amplify resistance to apoptosis. The regulation of prions in MDR is further complicated by important, putative physiological functions of ligand-binding and signal transduction. Melatonin is capable of both enhancing physiological functions and inhibiting oncogenic properties of prion proteins. Through regulation of phase separation of the prion N-terminal domain which targets and interacts with lipid rafts, melatonin may prevent conformational changes that can result in aggregation and/or conversion to pathological, infectious isoforms. As a cancer therapy adjuvant, melatonin could modulate TME oxidative stress levels and hypoxia, reverse pH gradient changes, reduce lipid peroxidation, and protect lipid raft compositions to suppress prion-mediated, non-Mendelian, heritable, but often reversible epigenetic adaptations that facilitate cancer heterogeneity, stemness, metastasis, and drug resistance. This review examines some of the mechanisms that may balance physiological and pathological effects of prions and prion-like proteins achieved through the synergistic use of melatonin to ameliorate MDR, which remains a challenge in cancer treatment.
Collapse
Affiliation(s)
- Doris Loh
- Independent Researcher, Marble Falls, TX 78654, USA
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, UT Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
17
|
Mantuano E, Azmoon P, Banki MA, Sigurdson CJ, Campana WM, Gonias SL. A Soluble PrP C Derivative and Membrane-Anchored PrP C in Extracellular Vesicles Attenuate Innate Immunity by Engaging the NMDA-R/LRP1 Receptor Complex. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:85-96. [PMID: 34810220 PMCID: PMC8702456 DOI: 10.4049/jimmunol.2100412] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/19/2021] [Indexed: 01/03/2023]
Abstract
Nonpathogenic cellular prion protein (PrPC) demonstrates anti-inflammatory activity; however, the responsible mechanisms are incompletely defined. PrPC exists as a GPI-anchored membrane protein in diverse cells; however, PrPC may be released from cells by ADAM proteases or when packaged into extracellular vesicles (EVs). In this study, we show that a soluble derivative of PrPC (S-PrP) counteracts inflammatory responses triggered by pattern recognition receptors in macrophages, including TLR2, TLR4, TLR7, TLR9, NOD1, and NOD2. S-PrP also significantly attenuates the toxicity of LPS in mice. The response of macrophages to S-PrP is mediated by a receptor assembly that includes the N-methyl-d-aspartate receptor (NMDA-R) and low-density lipoprotein receptor-related protein-1 (LRP1). PrPC was identified in EVs isolated from human plasma. These EVs replicated the activity of S-PrP, inhibiting cytokine expression and IκBα phosphorylation in LPS-treated macrophages. The effects of plasma EVs on LPS-treated macrophages were blocked by PrPC-specific Ab, by antagonists of LRP1 and the NMDA-R, by deleting Lrp1 in macrophages, and by inhibiting Src family kinases. Phosphatidylinositol-specific phospholipase C dissociated the LPS-regulatory activity from EVs, rendering the EVs inactive as LPS inhibitors. The LPS-regulatory activity that was lost from phosphatidylinositol-specific phospholipase C-treated EVs was recovered in solution. Collectively, these results demonstrate that GPI-anchored PrPC is the essential EV component required for the observed immune regulatory activity of human plasma EVs. S-PrP and EV-associated PrPC regulate innate immunity by engaging the NMDA-R/LRP1 receptor system in macrophages. The scope of pattern recognition receptors antagonized by S-PrP suggests that released forms of PrPC may have broad anti-inflammatory activity.
Collapse
Affiliation(s)
| | - Pardis Azmoon
- Department of Pathology, University of California San Diego, La Jolla, CA
| | - Michael A Banki
- Department of Pathology, University of California San Diego, La Jolla, CA
| | | | - Wendy M Campana
- Department of Anesthesiology and Program in Neurosciences, University of California San Diego, La Jolla, CA; and
- Veterans Administration San Diego Healthcare System, San Diego, CA
| | - Steven L Gonias
- Department of Pathology, University of California San Diego, La Jolla, CA;
| |
Collapse
|
18
|
Zhong T, Jiang N, Li C, Wang G. A highly selective fluorescence and absorption sensor for rapid recognition and detection of Cu 2+ ion in aqueous solution and film. LUMINESCENCE 2021; 37:391-398. [PMID: 34931444 DOI: 10.1002/bio.4180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/06/2022]
Abstract
A fluorescence and absorption chemosensor (SAAT) based on 5-(hydroxymethyl)-salicylaldehyde (SA) and o-aminothiophenol (AT) was designed and synthesized. SAAT in DMSO-HEPES (20.0 mM, v/v, 1:99, pH=7.0) solution shows a highly selective and sensitive absorption and "on-off" fluorescence response to Cu2+ ions in aqueous solutions over all other competitive metal ions including Na+ , Ag+ , Ba2+ , Ca2+ , Cd2+ , Mg2+ , Zn2+ , Cr3+ , Al3+ , Hg2+ , K+ , Mn2+ , Ni2+ , Sr2+ , Tb3+ and Co2+ . SAAT exhibits ratiometric absorption sensing ability for Cu2+ ions. Importantly, SAAT also can sense Cu2+ ions by fluorescence quenching, the fluorescence intensity of SAAT showed a good linear relationship with Cu2+ concentration, and the detection limit of Cu2+ was 0.34 μM. The results of Job's plot, Benesi-Hildebrand plot, mass spectra, and DFT calculations confirmed that the selective absorption and fluorescence response were attributed to the formation of 1:1 complex between SAAT and Cu2+ . SAAT in test film can identify Cu2+ in water samples by the intuitive fluorescence color change under UV lamp. SAAT has great application value as a selective and sensitive chemosensor to discrimination and detection of Cu2+ ions.
Collapse
Affiliation(s)
- Tianyuan Zhong
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Faculty of Chemistry, Northeast Normal University, Changchun, P. R. China
| | - Nan Jiang
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Faculty of Chemistry, Northeast Normal University, Changchun, P. R. China
| | - Chen Li
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Faculty of Chemistry, Northeast Normal University, Changchun, P. R. China
| | - Guang Wang
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Faculty of Chemistry, Northeast Normal University, Changchun, P. R. China
| |
Collapse
|
19
|
Sánchez-López C, Quintanar L. β-cleavage of the human prion protein impacts Cu(II) coordination at its non-octarepeat region. J Inorg Biochem 2021; 228:111686. [PMID: 34929540 DOI: 10.1016/j.jinorgbio.2021.111686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 11/26/2022]
Abstract
The cellular prion protein (PrPC) is a membrane-anchored copper binding protein that undergoes proteolytic processing. β-cleavage of PrPC is associated with a pathogenic condition and it yields two fragments: N2 with residues 23-89, and C2 including residues 90-231. The membrane-bound C2 fragment retains the Cu binding sites at His96 and His111, but it also has a free N-terminal NH2 group. In this study, the impact of β-cleavage of PrPC in its Cu(II) binding properties was evaluated, using the peptide of the human prion protein hPrP(90-115) as a model for the C2 fragment. The Cu(II) coordination properties of hPrP(90-115) were studied using circular dichroism (CD) and electron paramagnetic resonance (EPR); while the H96A and H111A substitutions and its acetylated variants were also studied. Cu binding to hPrP(90-115) is dependent on metal ion concentration: At low copper concentrations the participation of His96 and free NH2-terminus is evident, while at high copper concentrations the His111 site is populated without participation of the N-terminal NH2 group. The presence of a free NH2-terminal group in the C2 fragment significantly impacts the Cu(II) coordination properties of the His96 site, where the NH2 group also anchors the metal ion. This study provides further insights into the impact of proteolytic processing of PrPC in the Cu binding properties of this important neuronal protein.
Collapse
Affiliation(s)
- Carolina Sánchez-López
- Department of Chemistry, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Liliana Quintanar
- Department of Chemistry, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico.
| |
Collapse
|
20
|
Pollock NM, Leighton P, Neil G, Allison WT. Transcriptomic analysis of zebrafish prion protein mutants supports conserved cross-species function of the cellular prion protein. Prion 2021; 15:70-81. [PMID: 34139950 PMCID: PMC8216189 DOI: 10.1080/19336896.2021.1924557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 10/31/2022] Open
Abstract
Cellular Prion Protein (PrPC) is a well-studied protein as the substrate for various progressive untreatable neurodegenerative diseases. Normal functions of PrPC are poorly understood, though recent proteomic and transcriptomic approaches have begun to reveal common themes. We use our compound prp1 and prp2 knockout mutant zebrafish at three days post fertilization to take a transcriptomic approach to investigating potentially conserved PrPC functions during development. Gene ontology analysis shows the biological processes with the largest changes in gene expression include redox processing, transport and cell adhesion. Within these categories several different gene families were prevalent including the solute carrier proteins, cytochrome p450 enzymes and protocadherins. Continuing from previous studies identifying cell adhesion as an important function of PrPC we found that in addition to the protocadherins there was a significant reduction in transcript abundance of both ncam1a and st8sia2. These two genes are involved in the early development of vertebrates. The alterations in cell adhesion transcripts were consistent with past findings in zebrafish and mouse prion protein mutants; however E-cadherin processing after prion protein knockdown failed to reveal any differences compared with wild type in either our double prp1/prp2 mutant fish or after prp1 morpholino knockdown. Our data supports a cross species conserved role for PrPC in the development and maintenance of the central nervous system, particularly by regulating various and important cell adhesion processes.
Collapse
Affiliation(s)
- Niall Mungo Pollock
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, Canada
| | - Patricia Leighton
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, Canada
| | - Gavin Neil
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - W. Ted Allison
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, Canada
| |
Collapse
|
21
|
Posadas Y, López-Guerrero VE, Segovia J, Perez-Cruz C, Quintanar L. Dissecting the copper bioinorganic chemistry of the functional and pathological roles of the prion protein: Relevance in Alzheimer's disease and cancer. Curr Opin Chem Biol 2021; 66:102098. [PMID: 34768088 DOI: 10.1016/j.cbpa.2021.102098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 01/11/2023]
Abstract
The cellular prion protein (PrPC) is a metal-binding biomolecule that can interact with different protein partners involved in pivotal physiological processes, such as neurogenesis and neuronal plasticity. Recent studies profile copper and PrPC as important players in the pathological mechanisms of Alzheimer's disease and cancer. Although the copper-PrPC interaction has been characterized extensively, the role of the metal ion in the physiological and pathological roles of PrPC has been barely explored. In this article, we discuss how copper binding and proteolytic processing may impact the ability of PrPC to recruit protein partners for its functional roles. The importance to dissect the role of copper-PrPC interactions in health and disease is also underscored.
Collapse
Affiliation(s)
- Yanahi Posadas
- Department of Chemistry, Center for Research and Advanced Studies (Cinvestav), Mexico City, 07350, Mexico; Department of Pharmacology, Center for Research and Advanced Studies (Cinvestav), Mexico City, 07350, Mexico
| | - Victor E López-Guerrero
- Department of Chemistry, Center for Research and Advanced Studies (Cinvestav), Mexico City, 07350, Mexico; Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies (Cinvestav), Mexico City, 07350, Mexico
| | - José Segovia
- Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies (Cinvestav), Mexico City, 07350, Mexico
| | - Claudia Perez-Cruz
- Department of Pharmacology, Center for Research and Advanced Studies (Cinvestav), Mexico City, 07350, Mexico
| | - Liliana Quintanar
- Department of Chemistry, Center for Research and Advanced Studies (Cinvestav), Mexico City, 07350, Mexico.
| |
Collapse
|
22
|
Chaudhary S, Ashok A, Wise AS, Rana NA, Kritikos AE, Lindner E, Singh N. β-Cleavage of the prion protein in the human eye: Implications for the spread of infectious prions and human ocular disorders. Exp Eye Res 2021; 212:108787. [PMID: 34624335 DOI: 10.1016/j.exer.2021.108787] [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: 07/01/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 11/26/2022]
Abstract
Recently, we reported β-cleavage of the prion protein (PrPC) in human ocular tissues. Here, we explored whether this is unique to the human eye, and its functional implications. A comparison of the cleavage pattern of PrPC in human ocular tissues with common nocturnal and diurnal animals revealed mainly β-cleavage in humans, and mostly full-length PrPC in animal retinas. Soluble FL PrPC and N-terminal fragment (N2) released from β-cleavage was observed in the aqueous and vitreous humor (AH & VH). Expression of human PrPC in ARPE-19 cells, a human retinal pigmented epithelial cell line, also showed β-cleaved PrPC. Surprisingly, β-cleavage was not altered by a variety of insults, including oxidative stress, suggesting a unique role of this cleavage in the human eye. It is likely that β-cleaved C- or N-terminal fragments of PrPC protect from various insults unique to the human eye. On the contrary, β-cleaved C-terminus of PrPC is susceptible to conversion to the pathological PrP-scrapie form, and includes the binding sites for β1-integrin and amyloid-β, molecules implicated in several ocular disorders. Considering the species and tissue-specific cleavage of PrPC, our data suggest re-evaluation of prion infectivity and other ocular disorders of the human eye conducted in mouse models.
Collapse
Affiliation(s)
- Suman Chaudhary
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ajay Ashok
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Aaron S Wise
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Neil A Rana
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Alexander E Kritikos
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ewald Lindner
- Department of Ophthalmology, Medical University of Graz, Auenbruggerplatz 4, 8036, Graz, Austria
| | - Neena Singh
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
23
|
Dexter E, Kong Q. Neuroprotective effect and potential of cellular prion protein and its cleavage products for treatment of neurodegenerative disorders part II: strategies for therapeutics development. Expert Rev Neurother 2021; 21:983-991. [PMID: 34470554 DOI: 10.1080/14737175.2021.1965882] [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] [Indexed: 12/13/2022]
Abstract
Introduction: The cellular prion protein (PrPC), some of its derivatives (especially PrP N-terminal N1 peptide and shed PrP), and PrPC-containing exosomes have strong neuroprotective activities, which have been reviewed in the companion article (Part I) and are briefly summarized here.Areas covered: We propose that elevating the extracellular levels of a protective PrP form using gene therapy and other approaches is a very promising novel avenue for prophylactic and therapeutic treatments against prion disease, Alzheimer's disease, and several other neurodegenerative diseases. We will dissect the pros and cons of various potential PrP-based treatment options and propose a few strategies that are more likely to succeed. The cited references were obtained from extensive PubMed searches of recent literature, including peer-reviewed original articles and review articles.Expert opinion: Concurrent knockdown of celllular PrP expression and elevation of the extracellular levels of a neuroprotective PrP N-terminal peptide via optimized gene therapy vectors is a highly promising broad-spectrum prophylactic and therapeutic strategy against several neurodegenerative diseases, including prion diseases, Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Emily Dexter
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Qingzhong Kong
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Department of Neurology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
24
|
Dexter E, Kong Q. Neuroprotective effect and potential of cellular prion protein and its cleavage products for treatment of neurodegenerative disorders part I. a literature review. Expert Rev Neurother 2021; 21:969-982. [PMID: 34470561 DOI: 10.1080/14737175.2021.1965881] [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] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The cellular prion protein (PrPC) is well known for its pathogenic roles in prion diseases, several other neurodegenerative diseases (such as Alzheimer's disease), and multiple types of cancer, but the beneficial aspects of PrPC and its cleavage products received much less attention. AREAS COVERED Here the authors will systematically review the literatures on the negative as well as protective aspects of PrPC and its derivatives (especially PrP N-terminal N1 peptide and shed PrP). The authors will dissect the current findings on N1 and shed PrP, including evidence for their neuroprotective effects, the categories of PrPC cleavage, and numerous cleavage enzymes involved. The authors will also discuss the protective effects and therapeutic potentials of PrPC-rich exosomes. The cited articles were obtained from extensive PubMed searches of recent literature, including peer-reviewed original articles and review articles. EXPERT OPINION PrP and its N-terminal fragments have strong neuroprotective activities that should be explored for therapeutics and prophylactics development against prion disease, Alzheimer's disease and a few other neurodegenerative diseases. The strategies to develop PrP-based therapeutics and prophylactics for these neurodegenerative diseases will be discussed in a companion article (Part II).
Collapse
Affiliation(s)
- Emily Dexter
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, USA
| | - Qingzhong Kong
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, USA.,Department of Neurology, School of Medicine, Case Western Reserve University, Cleveland, USA
| |
Collapse
|
25
|
Posadas Y, Parra-Ojeda L, Perez-Cruz C, Quintanar L. Amyloid β Perturbs Cu(II) Binding to the Prion Protein in a Site-Specific Manner: Insights into Its Potential Neurotoxic Mechanisms. Inorg Chem 2021; 60:8958-8972. [PMID: 34043332 DOI: 10.1021/acs.inorgchem.1c00846] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Amyloid β (Aβ) is a Cu-binding peptide that plays a key role in the pathology of Alzheimer's disease. A recent report demonstrated that Aβ disrupts the Cu-dependent interaction between cellular prion protein (PrPC) and N-methyl-d-aspartate receptor (NMDAR), inducing overactivation of NMDAR and neurotoxicity. In this context, it has been proposed that Aβ competes for Cu with PrPC; however, there is no spectroscopic evidence to support this hypothesis. Prion protein (PrP) can bind up to six Cu(II) ions: from one to four at the octarepeat (OR) region, producing low- and high-occupancy modes, and two at the His96 and His111 sites. Additionally, PrPC is cleaved by α-secretases at Lys110/His111, yielding a new Cu(II)-binding site at the α-cleaved His111. In this study, the competition for Cu(II) between Aβ(1-16) and peptide models for each Cu-binding site of PrP was evaluated using circular dichroism and electron paramagnetic resonance. Our results show that the impact of Aβ(1-16) on Cu(II) coordination to PrP is highly site-specific: Aβ(1-16) cannot effectively compete with the low-occupancy mode at the OR region, whereas it partially removes the metal ion from the high-occupancy modes and forms a ternary OR-Cu(II)-Aβ(1-16) complex. In contrast, Aβ(1-16) removes all Cu(II) ions from the His96 and His111 sites without formation of ternary species. Finally, at the α-cleaved His111 site, Aβ(1-16) yields at least two different ternary complexes depending on the ratio of PrP/Cu(II)/Aβ. Altogether, our spectroscopic results indicate that only the low-occupancy mode at the OR region resists the effect of Aβ, while Cu(II) coordination to the high-occupancy modes and all other tested sites of PrP is perturbed, by either removal of the metal ion or formation of ternary complexes. These results provide important insights into the intricate effect of Aβ on Cu(II) binding to PrP and the potential neurotoxic mechanisms through which Aβ might affect Cu-dependent functions of PrPC, such as NMDAR modulation.
Collapse
|
26
|
Senthil Murugan A, Kiruthika M, Abel Noelson E, Yogapandi P, Gnana kumar G, Annaraj J. Fluorescent sensor for in-vivo bio-imaging, precise tracking of Fe3+ ions in Zebrafish embryos and visual measuring of Cu2+ ions in pico-molar level. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2020.11.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
27
|
Xia XL, Zhang DB, Zhang JL, Pu SZ. Highly sensitive ruthenium complex-based fluorescent probe for copper ion detection. Tetrahedron 2020. [DOI: 10.1016/j.tet.2020.131526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
28
|
Singh N, Chaudhary S, Ashok A, Lindner E. Prions and prion diseases: Insights from the eye. Exp Eye Res 2020; 199:108200. [PMID: 32858007 DOI: 10.1016/j.exer.2020.108200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/24/2020] [Accepted: 08/21/2020] [Indexed: 12/30/2022]
Abstract
Prion diseases are invariably fatal neurodegenerative disorders that have gained much publicity due to their transmissible nature. Sporadic Creutzfeldt-Jakob disease (sCJD) is the most common human prion disorder, with an incidence of 1 in a million. Inherited prion disorders are relatively rare, and associated with mutations in the prion protein gene. More than 50 different point mutations, deletions, and insertions have been identified so far. Most are autosomal dominant and fully penetrant. Prion disorders also occur in animals, and are of major concern because of the potential for spreading to humans. The principal pathogenic event underlying all prion disorders is a change in the conformation of prion protein (PrPC) from a mainly α-helical to a β-sheet rich isoform, PrP-scrapie (PrPSc). Accumulation of PrPSc in the brain parenchyma is the major cause of neuronal degeneration. The mechanism by which PrPSc is transmitted, propagates, and causes neurodegenerative changes has been investigated over the years, and several clues have emerged. Efforts are also ongoing for identifying specific and sensitive diagnostic tests for sCJD and animal prion disorders, but success has been limited. The eye is suitable for these evaluations because it shares several anatomical and physiological features with the brain, and can be observed in vivo during disease progression. The retina, considered an extension of the central nervous system, is involved extensively in prion disorders. Accordingly, Optical Coherence Tomography and electroretinogram have shown some promise as pre-mortem diagnostic tests for human and animal prion disorders. However, a complete understanding of the physiology of PrPC and pathobiology of PrPSc in the eye is essential for developing specific and sensitive tests. Below, we summarize recent progress in ocular physiology and pathology in prion disorders, and the eye as an anatomically accessible site to diagnose, monitor disease progression, and test therapeutic options.
Collapse
Affiliation(s)
- Neena Singh
- Departments of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Suman Chaudhary
- Departments of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ajay Ashok
- Departments of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ewald Lindner
- Department of Ophthalmology, Medical University of Graz, Auenbruggerplatz 4, 8036, Graz, Austria
| |
Collapse
|
29
|
Mantuano E, Azmoon P, Banki MA, Lam MS, Sigurdson CJ, Gonias SL. A soluble derivative of PrP C activates cell-signaling and regulates cell physiology through LRP1 and the NMDA receptor. J Biol Chem 2020; 295:14178-14188. [PMID: 32788217 DOI: 10.1074/jbc.ra120.013779] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/04/2020] [Indexed: 11/06/2022] Open
Abstract
Cellular prion protein (PrPC) is a widely expressed glycosylphosphatidylinositol-anchored membrane protein. Scrapie prion protein is a misfolded and aggregated form of PrPC responsible for prion-induced neurodegenerative diseases. Understanding the function of the nonpathogenic PrPC monomer is an important objective. PrPC may be shed from the cell surface to generate soluble derivatives. Herein, we studied a recombinant derivative of PrPC (soluble cellular prion protein, S-PrP) that corresponds closely in sequence to a soluble form of PrPC shed from the cell surface by proteases in the A Disintegrin And Metalloprotease (ADAM) family. S-PrP activated cell-signaling in PC12 and N2a cells. TrkA was transactivated by Src family kinases and extracellular signal-regulated kinase 1/2 was activated downstream of Trk receptors. These cell-signaling events were dependent on the N-methyl-d-aspartate receptor (NMDA-R) and low-density lipoprotein receptor-related protein-1 (LRP1), which functioned as a cell-signaling receptor system in lipid rafts. Membrane-anchored PrPC and neural cell adhesion molecule were not required for S-PrP-initiated cell-signaling. S-PrP promoted PC12 cell neurite outgrowth. This response required the NMDA-R, LRP1, Src family kinases, and Trk receptors. In Schwann cells, S-PrP interacted with the LRP1/NMDA-R system to activate extracellular signal-regulated kinase 1/2 and promote cell migration. The effects of S-PrP on PC12 cell neurite outgrowth and Schwann cell migration were similar to those caused by other proteins that engage the LRP1/NMDA-R system, including activated α2-macroglobulin and tissue-type plasminogen activator. Collectively, these results demonstrate that shed forms of PrPC may exhibit important biological activities in the central nervous system and the peripheral nervous system by serving as ligands for the LRP1/NMDA-R system.
Collapse
Affiliation(s)
- Elisabetta Mantuano
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Pardis Azmoon
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Michael A Banki
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Michael S Lam
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Christina J Sigurdson
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Steven L Gonias
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
30
|
Roseman GP, Wu B, Wadolkowski MA, Harris DA, Millhauser GL. Intrinsic toxicity of the cellular prion protein is regulated by its conserved central region. FASEB J 2020; 34:8734-8748. [PMID: 32385908 DOI: 10.1096/fj.201902749rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/08/2020] [Accepted: 04/20/2020] [Indexed: 11/11/2022]
Abstract
The conserved central region (CR) of PrPC has been hypothesized to serve as a passive linker connecting the protein's toxic N-terminal and globular C-terminal domains. Yet, deletion of the CR causes neonatal fatality in mice, implying the CR possesses a protective function. The CR encompasses the regulatory α-cleavage locus, and additionally facilitates a regulatory metal ion-promoted interaction between the PrPC N- and C-terminal domains. To elucidate the role of the CR and determine why CR deletion generates toxicity, we designed PrPC constructs wherein either the cis-interaction or α-cleavage are selectively prevented. These constructs were interrogated using nuclear magnetic resonance, electrophysiology, and cell viability assays. Our results demonstrate the CR is not a passive linker and the native sequence is crucial for its protective role over the toxic N-terminus, irrespective of α-cleavage or the cis-interaction. Additionally, we find that the CR facilitates homodimerization of PrPC , attenuating the toxicity of the N-terminus.
Collapse
Affiliation(s)
- Graham P Roseman
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Bei Wu
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Mark A Wadolkowski
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Glenn L Millhauser
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| |
Collapse
|
31
|
Carroll JA, Groveman BR, Williams K, Moore R, Race B, Haigh CL. Prion protein N1 cleavage peptides stimulate microglial interaction with surrounding cells. Sci Rep 2020; 10:6654. [PMID: 32313035 PMCID: PMC7171115 DOI: 10.1038/s41598-020-63472-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/29/2020] [Indexed: 01/06/2023] Open
Abstract
Microglia act as the protective immune cell of the brain. By surveying the tissue to identify and rectify problems, they function to maintain the health of brain cells. The prion protein N-terminal cleavage fragment, N1, has demonstrated neuroprotective activities in vitro and in vivo. This study aimed to elucidate whether N1 could modulate microglial function and, if so, determine the consequences for the surrounding tissue. Using a mixed neuronal lineage and microglia co-culture system, we showed that N1 stimulation changed overall morphology and metabolism, suggesting enhanced cellular viability. Furthermore, N1 induced an increase in Cxcl10 secretion in the co-cultures. Recombinant Cxcl10, administered exogenously, mediated the changes in the mixed neuronal lineage culture morphology and metabolism in the absence of microglia, but no effect of Cxcl10 was observed on microglia cultured on their own. Direct cell-to-cell contact was required for N1 to influence microglia in the co-cultures, and this was linked with restructuring of microglial membrane composition to include a higher GM1 content at interaction sites with surrounding cells. Our findings show that N1 can play a regulatory role in microglial function in the context of an inter-connected network of cells by changing both cellular interaction sites and cytokine secretion.
Collapse
Affiliation(s)
- J A Carroll
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - B R Groveman
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - K Williams
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - R Moore
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - B Race
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - C L Haigh
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA.
| |
Collapse
|
32
|
Bacchella C, Nicolis S, Dell'Acqua S, Rizzarelli E, Monzani E, Casella L. Membrane Binding Strongly Affecting the Dopamine Reactivity Induced by Copper Prion and Copper/Amyloid-β (Aβ) Peptides. A Ternary Copper/Aβ/Prion Peptide Complex Stabilized and Solubilized in Sodium Dodecyl Sulfate Micelles. Inorg Chem 2019; 59:900-912. [PMID: 31869218 DOI: 10.1021/acs.inorgchem.9b03153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The combination between dyshomeostatic levels of catecholamine neurotransmitters and redox-active metals such as copper and iron exacerbates the oxidative stress condition that typically affects neurodegenerative diseases. We report a comparative study of the oxidative reactivity of copper complexes with amyloid-β (Aβ40) and the prion peptide fragment 76-114 (PrP76-114), containing the high-affinity binding site, toward dopamine and 4-methylcatechol, in aqueous buffer and in sodium dodecyl sulfate micelles, as a model membrane environment. The competitive oxidative and covalent modifications undergone by the peptides were also evaluated. The high binding affinity of Cu/peptide to micelles and lipid membranes leads to a strong reduction (Aβ40) and quenching (PrP76-114) of the oxidative efficiency of the binary complexes and to a stabilization and redox silencing of the ternary complex CuII/Aβ40/PrP76-114, which is highly reactive in solution. The results improve our understanding of the pathological and protective effects associated with these complexes, depending on the physiological environment.
Collapse
Affiliation(s)
- Chiara Bacchella
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Stefania Nicolis
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Simone Dell'Acqua
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Enrico Rizzarelli
- Istituto di Biostrutture e Bioimmagini , Consiglio Nazionale delle Ricerche , Via P. Gaifami 18 , 95125 Catania , Italy
| | - Enrico Monzani
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Luigi Casella
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| |
Collapse
|
33
|
Hackl S, Becker CFW. Prion protein-Semisynthetic prion protein (PrP) variants with posttranslational modifications. J Pept Sci 2019; 25:e3216. [PMID: 31713950 PMCID: PMC6899880 DOI: 10.1002/psc.3216] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022]
Abstract
Deciphering the pathophysiologic events in prion diseases is challenging, and the role of posttranslational modifications (PTMs) such as glypidation and glycosylation remains elusive due to the lack of homogeneous protein preparations. So far, experimental studies have been limited in directly analyzing the earliest events of the conformational change of cellular prion protein (PrPC ) into scrapie prion protein (PrPSc ) that further propagates PrPC misfolding and aggregation at the cellular membrane, the initial site of prion infection, and PrP misfolding, by a lack of suitably modified PrP variants. PTMs of PrP, especially attachment of the glycosylphosphatidylinositol (GPI) anchor, have been shown to be crucially involved in the PrPSc formation. To this end, semisynthesis offers a unique possibility to understand PrP behavior invitro and invivo as it provides access to defined site-selectively modified PrP variants. This approach relies on the production and chemoselective linkage of peptide segments, amenable to chemical modifications, with recombinantly produced protein segments. In this article, advances in understanding PrP conversion using semisynthesis as a tool to obtain homogeneous posttranslationally modified PrP will be discussed.
Collapse
Affiliation(s)
- Stefanie Hackl
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Vienna, Austria
| | - Christian F W Becker
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Vienna, Austria
| |
Collapse
|
34
|
Ashok A, Kang MH, Wise AS, Pattabiraman P, Johnson WM, Lonigro M, Ravikumar R, Rhee DJ, Singh N. Prion protein modulates endothelial to mesenchyme-like transition in trabecular meshwork cells: Implications for primary open angle glaucoma. Sci Rep 2019; 9:13090. [PMID: 31511544 PMCID: PMC6739364 DOI: 10.1038/s41598-019-49482-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/15/2019] [Indexed: 12/31/2022] Open
Abstract
Endothelial-to-mesenchyme-like transition (Endo-MT) of trabecular meshwork (TM) cells is known to be associated with primary open angle glaucoma (POAG). Here, we investigated whether the prion protein (PrPC), a neuronal protein known to modulate epithelial-to-mesenchymal transition in a variety of cell types, is expressed in the TM, and plays a similar role at this site. Using a combination of primary human TM cells and human, bovine, and PrP-knock-out (PrP−/−) mouse models, we demonstrate that PrPC is expressed in the TM of all three species, including endothelial cells lining the Schlemm’s canal. Silencing of PrPC in primary human TM cells induces aggregation of β1-integrin and upregulation of α-smooth muscle actin, fibronectin, collagen 1A, vimentin, and laminin, suggestive of transition to a mesenchyme-like phenotype. Remarkably, intraocular pressure is significantly elevated in PrP−/− mice relative to wild-type controls, suggesting reduced pliability of the extracellular matrix and increased resistance to aqueous outflow in the absence of PrPC. Since PrPC is cleaved by members of the disintegrin and matrix-metalloprotease family that are increased in the aqueous humor of POAG arising from a variety of conditions, it is likely that concomitant cleavage of PrPC exaggerates and confounds the pathology by inducing Endo-MT-like changes in the TM.
Collapse
Affiliation(s)
- Ajay Ashok
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Min H Kang
- Department of Ophthalmology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Aaron S Wise
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - P Pattabiraman
- Department of Ophthalmology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | | | - Michael Lonigro
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Ranjana Ravikumar
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Douglas J Rhee
- Department of Ophthalmology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Neena Singh
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA.
| |
Collapse
|
35
|
Structural Consequences of Copper Binding to the Prion Protein. Cells 2019; 8:cells8080770. [PMID: 31349611 PMCID: PMC6721516 DOI: 10.3390/cells8080770] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/24/2022] Open
Abstract
Prion, or PrPSc, is the pathological isoform of the cellular prion protein (PrPC) and it is the etiological agent of transmissible spongiform encephalopathies (TSE) affecting humans and animal species. The most relevant function of PrPC is its ability to bind copper ions through its flexible N-terminal moiety. This review includes an overview of the structure and function of PrPC with a focus on its ability to bind copper ions. The state-of-the-art of the role of copper in both PrPC physiology and in prion pathogenesis is also discussed. Finally, we describe the structural consequences of copper binding to the PrPC structure.
Collapse
|
36
|
Pariente Cohen N, Lo Presti E, Dell'Acqua S, Jantz T, Shimon LJW, Levy N, Nassir M, Elbaz L, Casella L, Fischer B. Aminomethylene-Phosphonate Analogue as a Cu(II) Chelator: Characterization and Application as an Inhibitor of Oxidation Induced by the Cu(II)-Prion Peptide Complex. Inorg Chem 2019; 58:8995-9003. [PMID: 31247811 DOI: 10.1021/acs.inorgchem.9b00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recently, we reported on a series of aminomethylene-phosphonate (AMP) analogues, bearing one or two heterocyclic groups on the aminomethylene moiety, as promising Zn(II) chelators. Given the strong Zn(II) binding properties of these compounds, they may find useful applications in metal chelation therapy. With a goal of inhibiting the devastating oxidative damage caused by prion protein in prion diseases, we explored the most promising ligand, {bis[(1H-imidazol-4-yl)methyl]amino}methylphosphonic acid, AMP-(Im)2, 4, as an inhibitor of the oxidative reactivity associated with the Cu(II) complex of prion peptide fragment 84-114. Specifically, we first characterized the Cu(II) complex with AMP-(Im)2 by ultraviolet-visible spectroscopy and electrochemical measurements that indicated the high chemical and electrochemical stability of the complex. Potentiometric pH titration provided evidence of the formation of a stable 1:1 [Cu(II)-AMP-(Im)2]+ complex (ML), with successive binding of a second AMP-(Im)2 molecule yielding ML2 complex [Cu(II)-(AMP-(Im)2)2]+ (log K' = 15.55), and log β' = 19.84 for ML2 complex. The CuN3O1 ML complex was demonstrated by X-ray crystallography, indicating the thermodynamically stable square pyramidal complex. Chelation of Cu(II) by 4 significantly reduced the oxidation potential of the former. CuCl2 and the 1:2 Cu:AMP-(Im)2 complex showed one-electron redox of Cu(II)/Cu(I) at 0.13 and -0.35 V, respectively. Indeed, 4 was found to be a potent antioxidant that at a 1:1:1 AMP-(Im)2:Cu(II)-PrP84-114 molar ratio almost totally inhibited the oxidation reaction of 4-methylcatechol. Circular dichroism data suggest that this antioxidant activity is due to formation of a ternary, redox inactive Cu(II)-Prp84-114-[AMP-(Im)2] complex. Future studies in prion disease animal models are warranted to assess the potential of 4 to inhibit the devastating oxidative damage caused by PrP.
Collapse
Affiliation(s)
| | - Eliana Lo Presti
- Department of Chemistry , University of Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Simone Dell'Acqua
- Department of Chemistry , University of Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Thomas Jantz
- Department of Chemistry , Bar-Ilan University , Ramat Gan 5290002 , Israel
| | - Linda J W Shimon
- Faculty of Chemistry, Crystallography Unit , Weizmann Institute , Rehovot 76100 , Israel
| | - Naomi Levy
- Department of Chemistry , Bar-Ilan University , Ramat Gan 5290002 , Israel
| | - Molhm Nassir
- Department of Chemistry , Bar-Ilan University , Ramat Gan 5290002 , Israel
| | - Lior Elbaz
- Department of Chemistry , Bar-Ilan University , Ramat Gan 5290002 , Israel
| | - Luigi Casella
- Department of Chemistry , University of Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Bilha Fischer
- Department of Chemistry , Bar-Ilan University , Ramat Gan 5290002 , Israel
| |
Collapse
|
37
|
Ding L, Zhao Z, Li D, Wang X, Chen J. An "off-on" fluorescent sensor for copper ion using graphene quantum dots based on oxidation of l-cysteine. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 214:320-325. [PMID: 30798213 DOI: 10.1016/j.saa.2019.02.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/25/2018] [Accepted: 02/16/2019] [Indexed: 06/09/2023]
Abstract
A simple and highly efficient "off-on" fluorescent sensor based on grapheme quantum dots (GQDs) for Cu2+ was developed. In this sensing platform, the fluorescence of GQDs was quenched in the presence of 2,4-dinitrophenylcysteine (DNPC), which is the reaction product of 1-chloro-2,4-dinitrobenzene (CDNB) and l-cysteine, owing to the spectral overlap between the absorption of DNPC and the excitation of GQDs. In the presence of Cu2+, l-cysteine was catalytically oxidized to l-cystine by O2, resulting in the reduction of DNPC. Thus, the fluorescence of GQDs was recovery. Based on this, the fluorescent detection of Cu2+ could be achieved. The proposed sensing strategy offered a selective identification of Cu2+ with a detection limit of 4.5 nM. Additionally, the practical application of this assay for Cu2+ determination in real water samples was also demonstrated.
Collapse
Affiliation(s)
- Longhua Ding
- Institute for Advanced Interdisciplinary Research, University of Jinan, Jinan 250022, PR China.
| | - Zhongyao Zhao
- Department of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, PR China
| | - Dongjun Li
- Department of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, PR China
| | - Xue Wang
- Department of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, PR China
| | - Jialin Chen
- Department of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, PR China
| |
Collapse
|
38
|
Mengel D, Hong W, Corbett GT, Liu W, DeSousa A, Solforosi L, Fang C, Frosch MP, Collinge J, Harris DA, Walsh DM. PrP-grafted antibodies bind certain amyloid β-protein aggregates, but do not prevent toxicity. Brain Res 2019; 1710:125-135. [PMID: 30593771 PMCID: PMC6431553 DOI: 10.1016/j.brainres.2018.12.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/26/2018] [Accepted: 12/23/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND The prion protein (PrP) is known to bind certain soluble aggregates of the amyloid β-protein (Aβ), and two regions of PrP, one centered around residues 19-33, and the other around 87-112, are thought to be particularly important for this interaction. When either of these sequences are grafted into a human IgG the resulting antibodies react with disease-associated PrP conformers, whereas the parental b12 IgG does not. METHODS Human antibodies containing grafts of PrP 19-33 or 87-112 were prepared as before (Solforosi et al., 2007) and tested for their ability to recognize synthetic and Alzheimer's disease (AD) brain-derived Aβ. Since aqueous extracts of AD brain contain a complex mixture of active and inactive Aβ species, we also assessed whether PrP-grafted antibodies could protect against neuritotoxicity mediated by AD brain-derived Aβ. For these experiments, human iPSC-derived neurons were grown in 96-well plates at 5000 cells per well and on post-induction day 21, AD brain extracts were added +/- test antibodies. Neurons were imaged for 3 days using an IncuCyte live-cell imaging system, and neurite number and density quantified. RESULTS Grafted antibodies bound a significant portion of aggregated Aβ in aqueous AD extracts, but when these antibodies were co-incubated with neurons treated with brain extracts they did not reduce toxicity. By contrast, the PrP fragment N1 did protect against Aβ. CONCLUSIONS These results further demonstrate that not all Aβ oligomers are toxic and suggest that PrP derivatives may allow development of agents that differentially recognize toxic and innocuous Aβ aggregates.
Collapse
Affiliation(s)
- David Mengel
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Wei Hong
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Grant T Corbett
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Wen Liu
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alexandra DeSousa
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Laura Solforosi
- Laboratory of Microbiology and Virology, University Vita-Salute San Raffaele, Milan, Italy
| | - Cheng Fang
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Matthew P Frosch
- Massachusetts General Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - John Collinge
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; MRC Prion Unit at UCL, UCL Institute of Prion Diseases and NHS National Prion Clinic, UCL Hospitals NHS Foundation Trust, London, United Kingdom
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Martellucci S, Santacroce C, Santilli F, Piccoli L, Delle Monache S, Angelucci A, Misasi R, Sorice M, Mattei V. Cellular and Molecular Mechanisms Mediated by recPrP C Involved in the Neuronal Differentiation Process of Mesenchymal Stem Cells. Int J Mol Sci 2019; 20:E345. [PMID: 30654447 PMCID: PMC6358746 DOI: 10.3390/ijms20020345] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/11/2019] [Accepted: 01/13/2019] [Indexed: 12/21/2022] Open
Abstract
Human Dental Pulp Stem Cells (hDPSCs) represent a type of adult mesenchymal stem cells that have the ability to differentiate in vitro in several lineages such as odontoblasts, osteoblasts, chondrocytes, adipocytes and neurons. In the current work, we used hDPSCs as the experimental model to study the role of recombinant prion protein 23⁻231 (recPrPC) in the neuronal differentiation process, and in the signal pathway activation of ERK 1/2 and Akt. We demonstrated that recPrPC was able to activate an intracellular signal pathway mediated by extracellular-signal-regulated kinase 1 and 2 (ERK 1/2) and protein kinase B (Akt). Moreover, in order to understand whether endogenous prion protein (PrPC) was necessary to mediate the signaling induced by recPrPC, we silenced PrPC, demonstrating that the presence of endogenous PrPC was essential for ERK 1/2 and Akt phosphorylation. Since endogenous PrPC is a well-known lipid rafts component, we evaluated the role of these structures in the signal pathway induced by recPrPC. Our results suggest that lipid rafts integrity play a key role in recPrPC activity. In fact, lipid rafts inhibitors, such as fumonisin B1 and MβCD, significantly prevented ERK 1/2 and Akt phosphorylation induced by recPrPC. In addition, we investigated the capacity of recPrPC to induce hDPSCs neuronal differentiation process after long-term stimulation through the evaluation of typical neuronal markers expression such as B3-Tubulin, neurofilament-H (NFH) and growth associated protein 43 (GAP43). Accordingly, when we silenced endogenous PrPC, we observed the inhibition of neuronal differentiation induced by recPrPC. The combined data suggest that recPrPC plays a key role in the neuronal differentiation process and in the activation of specific intracellular signal pathways in hDPSCs.
Collapse
Affiliation(s)
- Stefano Martellucci
- Laboratory of Experimental Medicine and Environmental Pathology, Rieti University Hub "Sabina Universitas", 02100 Rieti, Italy.
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy.
| | - Costantino Santacroce
- Laboratory of Experimental Medicine and Environmental Pathology, Rieti University Hub "Sabina Universitas", 02100 Rieti, Italy.
| | - Francesca Santilli
- Laboratory of Experimental Medicine and Environmental Pathology, Rieti University Hub "Sabina Universitas", 02100 Rieti, Italy.
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy.
| | - Luca Piccoli
- Department of Science Dentistry and Maxillofacial, "Sapienza" University, 00161 Rome, Italy.
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Roberta Misasi
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy.
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy.
| | - Vincenzo Mattei
- Laboratory of Experimental Medicine and Environmental Pathology, Rieti University Hub "Sabina Universitas", 02100 Rieti, Italy.
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy.
| |
Collapse
|
40
|
Castle AR, Daude N, Gilch S, Westaway D. Application of high-throughput, capillary-based Western analysis to modulated cleavage of the cellular prion protein. J Biol Chem 2018; 294:2642-2650. [PMID: 30578300 DOI: 10.1074/jbc.ra118.006367] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/20/2018] [Indexed: 12/21/2022] Open
Abstract
The cellular prion protein (PrPC) is a glycoprotein that is processed through several proteolytic pathways. Modulators of PrPC proteolysis are of interest because full-length PrPC and its cleavage fragments differ in their propensity to misfold, a process that plays a key role in the pathogenesis of prion diseases. PrPC may also act as a receptor for neurotoxic, oligomeric species of other proteins that are linked to neurodegeneration. Importantly, the PrPC C-terminal fragment C1 does not contain the reported binding sites for these oligomers. Western blotting would be a simple end point detection method for cell-based screening of compound libraries for effects on PrPC proteolysis or overall expression level. However, traditional Western blotting methods provide unreliable quantification and have only low throughput. Consequently, we explored capillary-based Western technology as a potential alternative; we believe that this study is the first to report analysis of PrPC using such an approach. We successfully optimized the detection and quantification of the deglycosylated forms of full-length PrPC and its C-terminal cleavage fragments C1 and C2, including simultaneous quantification of β-tubulin levels to control for loading error. We also developed and tested a method for performing all cell culture, lysis, and deglycosylation steps in 96-well microplates prior to capillary Western analysis. These advances represent steps along the way to the development of an automated, high-throughput screening pipeline to identify modulators of PrPC expression levels or proteolysis.
Collapse
Affiliation(s)
- Andrew R Castle
- From the Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2M8, Canada.,Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Nathalie Daude
- From the Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2M8, Canada
| | - Sabine Gilch
- Department of Ecosystem and Public Health, Calgary Prion Research Unit, Faculty of Veterinary Medicine and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada, and
| | - David Westaway
- From the Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2M8, Canada .,Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2G3, Canada.,Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
41
|
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
|
42
|
Paciotti R, Storchi L, Marrone A. An insight of early PrP-E200K aggregation by combined molecular dynamics/fragment molecular orbital approaches. Proteins 2018; 87:51-61. [PMID: 30367504 DOI: 10.1002/prot.25621] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/01/2018] [Accepted: 10/16/2018] [Indexed: 01/30/2023]
Abstract
Unveiling the events leading to the formation of prion particles is a nowadays challenge in the field of neurochemistry. Pathogenic mutants of prion protein (PrP) are characterized by both an intrinsic tendency to aggregation and scrapie conversion propensity. However, the question about a possible correlation between these two events lasts still unanswered. Here, a multilayered computational workflow was employed to investigate structure, stability, and molecular interaction properties of a dimer of PrPC -E200K, a well-known mutant of the PrP that represents a reduced model of early aggregates of this protein. Based on the combination of molecular dynamics and quantum mechanical approaches, this study provided for an in depth insight of PrPC -E200K dimer in terms of residue-residue interactions. Assembly hypotheses for the early aggregation of PrPC -E200K are paved and compared with PrPSc models reported in the literature to find a structural link between early and late (scrapie) aggregates of this protein.
Collapse
Affiliation(s)
- Roberto Paciotti
- Department of Pharmacy, Università "G d'Annunzio" di Chieti-Pescara, Chieti, Italy
| | - Loriano Storchi
- Department of Pharmacy, Università "G d'Annunzio" di Chieti-Pescara, Chieti, Italy.,Molecular Discovery Limited, Middlesex, London, United Kingdom.,ISTM - CNR, Perugia, Italy
| | - Alessandro Marrone
- Department of Pharmacy, Università "G d'Annunzio" di Chieti-Pescara, Chieti, Italy
| |
Collapse
|
43
|
Prasher P, Sharma M. Medicinal chemistry of acridine and its analogues. MEDCHEMCOMM 2018; 9:1589-1618. [PMID: 30429967 PMCID: PMC6195008 DOI: 10.1039/c8md00384j] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 08/14/2018] [Indexed: 02/01/2023]
Abstract
'Acridine' along with its functional analogue 'Acridone' is the most privileged pharmacophore in medicinal chemistry with diverse applications ranging from DNA intercalators, endonuclease mimics, ratiometric selective ion sensors, and P-glycoprotein inhibitors in countering the multi-drug resistance, enzyme inhibitors, and reversals of neurodegenerative disorders. Their interaction with DNA and ability of selectively identifying numerous biologically useful ions has cemented exploitability of the acridone nucleus in modern day therapeutics. Additionally, most derivatives and salts of acridine are planar, crystalline, and stable displaying a strong fluorescence which, when coupled with their marked bio selectivity and low cytotoxicity, enables the studying and monitoring of several biochemical, metabolic, and pharmacological processes. In this review, a detailed picture covering the important therapeutic aspects of the acridone nucleus and its functional analogues is discussed.
Collapse
Affiliation(s)
- Parteek Prasher
- UGC Sponsored Centre for Advanced Studies , Department of Chemistry , Guru Nanak Dev University , Amritsar 143005 , India
- Department of Chemistry , University of Petroleum & Energy Studies , Dehradun 248007 , India . ;
| | - Mousmee Sharma
- UGC Sponsored Centre for Advanced Studies , Department of Chemistry , Guru Nanak Dev University , Amritsar 143005 , India
| |
Collapse
|
44
|
Abstract
The cellular prion protein, PrPC, is a small, cell surface glycoprotein with a function that is currently somewhat ill defined. It is also the key molecule involved in the family of neurodegenerative disorders called transmissible spongiform encephalopathies, which are also known as prion diseases. The misfolding of PrPC to a conformationally altered isoform, designated PrPTSE, is the main molecular process involved in pathogenesis and appears to precede many other pathologic and clinical manifestations of disease, including neuronal loss, astrogliosis, and cognitive loss. PrPTSE is also believed to be the major component of the infectious "prion," the agent responsible for disease transmission, and preparations of this protein can cause prion disease when inoculated into a naïve host. Thus, understanding the biochemical and biophysical properties of both PrPC and PrPTSE, and ultimately the mechanisms of their interconversion, is critical if we are to understand prion disease biology. Although entire books could be devoted to research pertaining to the protein, herein we briefly review the state of knowledge of prion biochemistry, including consideration of prion protein structure, function, misfolding, and dysfunction.
Collapse
Affiliation(s)
- Andrew C Gill
- School of Chemistry, Joseph Banks Laboratories, University of Lincoln, Lincoln, United Kingdom; Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, United Kingdom.
| | - Andrew R Castle
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
45
|
Sánchez-López C, Rivillas-Acevedo L, Cruz-Vásquez O, Quintanar L. Methionine 109 plays a key role in Cu(II) binding to His111 in the 92–115 fragment of the human prion protein. Inorganica Chim Acta 2018. [DOI: 10.1016/j.ica.2017.09.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
46
|
Ashok A, Karmakar S, Chandel R, Ravikumar R, Dalal S, Kong Q, Singh N. Prion protein modulates iron transport in the anterior segment: Implications for ocular iron homeostasis and prion transmission. Exp Eye Res 2018; 175:1-13. [PMID: 29859760 PMCID: PMC6167182 DOI: 10.1016/j.exer.2018.05.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/17/2018] [Accepted: 05/30/2018] [Indexed: 12/14/2022]
Abstract
Iron is an essential biometal in the aqueous humor, the principal source of nutrients for the avascular cornea and the lens. Here, we explored whether the ciliary body (CB), the source of aqueous humor, transports iron, and if the prion protein (PrPC) facilitates this process as in the outer retina. Using a combination of human, bovine, and mouse eyes as models, we report the expression of iron export proteins ferroportin and ceruloplasmin, and major iron uptake and storage proteins transferrin, transferrin receptor, and ferritin in the ciliary epithelium, indicating active exchange of iron at this site. Ferroportin and transferrin receptor are also expressed in the corneal endothelium. However, the relative expression of iron export and uptake proteins suggests export from the ciliary epithelium and import by corneal endothelium. In addition, abundant expression of PrPC, a ferrireductase that facilitates iron transport, is noted in pigmented and non-pigmented epithelium of the CB, posterior pigmented epithelium of the iris, corneal endothelium and epithelium, and lens epithelium. Notably, majority of PrPC in the ciliary epithelium is cleaved at the β-site as in retinal pigment epithelial cells, suggesting a role in iron transport. Most of the PrPC in the cornea, however, is full-length, and susceptible to aggregation by intracerebrally inoculated PrP-scrapie, an infectious conformation of PrPC responsible for human and animal prion disorders. Soluble PrPC is present in the aqueous and vitreous humor, a provocative observation with significant implications. Together, these observations suggest independent cycling of iron in the anterior segment, and a prominent role of PrPC in this process. Aggregation of PrPC in the cornea of PrP-scrapie-infected animals raises the alarming possibility of transmission of animal prions through corneal abrasions.
Collapse
Affiliation(s)
- Ajay Ashok
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Shilpita Karmakar
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Rajeev Chandel
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ranjana Ravikumar
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Stuti Dalal
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Qingzhong Kong
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Neena Singh
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
47
|
Linsenmeier L, Mohammadi B, Wetzel S, Puig B, Jackson WS, Hartmann A, Uchiyama K, Sakaguchi S, Endres K, Tatzelt J, Saftig P, Glatzel M, Altmeppen HC. Structural and mechanistic aspects influencing the ADAM10-mediated shedding of the prion protein. Mol Neurodegener 2018; 13:18. [PMID: 29625583 PMCID: PMC5889536 DOI: 10.1186/s13024-018-0248-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/21/2018] [Indexed: 11/10/2022] Open
Abstract
Background Proteolytic processing of the prion protein (PrPC) by endogenous proteases generates bioactive membrane-bound and soluble fragments which may help to explain the pleiotropic roles of this protein in the nervous system and in brain diseases. Shedding of almost full-length PrPC into the extracellular space by the metalloprotease ADAM10 is of peculiar relevance since soluble PrP stimulates axonal outgrowth and is protective in neurodegenerative conditions such as Alzheimer’s and prion disease. However, molecular determinates and mechanisms regulating the shedding of PrP are entirely unknown. Methods We produced an antibody recognizing the neo-epitope of shed PrP generated by ADAM10 in biological samples and used it to study structural and mechanistic aspects affecting the shedding. For this, we investigated genetically modified cellular and murine models by biochemical and morphological approaches. Results We show that the novel antibody specifically detects shed PrP in cell culture supernatants and murine brain. We demonstrate that ADAM10 is the exclusive sheddase of PrPC in the nervous system and reveal that the glycosylation state and type of membrane-anchorage of PrPC severely affect its shedding. Furthermore, we provide evidence that PrP shedding can be modulated by pharmacological inhibition and stimulation and present data suggesting that shedding is a relevant part of a compensatory network ensuring PrPC homeostasis of the cell. Conclusions With the new antibody, our study introduces a new tool to reliably investigate PrP-shedding. In addition, this study provides novel and important insight into the regulation of this cleavage event, which is likely to be relevant for diagnostic and therapeutic approaches even beyond neurodegeneration.
Collapse
Affiliation(s)
- Luise Linsenmeier
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Sebastian Wetzel
- Institute of Biochemistry, Christian Albrechts University, Kiel, Germany
| | - Berta Puig
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | | - Alexander Hartmann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Keiji Uchiyama
- Division of Molecular Neurobiology, Institute of Enzyme Research, Tokushima University, Tokushima, Japan
| | - Suehiro Sakaguchi
- Division of Molecular Neurobiology, Institute of Enzyme Research, Tokushima University, Tokushima, Japan
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Jörg Tatzelt
- Institute of Biochemistry and Pathobiochemistry, Ruhr University, Bochum, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian Albrechts University, Kiel, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Hermann C. Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
48
|
Reiten MR, Malachin G, Kommisrud E, Østby GC, Waterhouse KE, Krogenæs AK, Kusnierczyk A, Bjørås M, Jalland CMO, Nekså LH, Røed SS, Stenseth EB, Myromslien FD, Zeremichael TT, Bakkebø MK, Espenes A, Tranulis MA. Stress Resilience of Spermatozoa and Blood Mononuclear Cells without Prion Protein. Front Mol Biosci 2018; 5:1. [PMID: 29417049 PMCID: PMC5787566 DOI: 10.3389/fmolb.2018.00001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/08/2018] [Indexed: 11/19/2022] Open
Abstract
The cellular prion protein PrPC is highly expressed in neurons, but also present in non-neuronal tissues, including the testicles and spermatozoa. Most immune cells and their bone marrow precursors also express PrPC. Clearly, this protein operates in highly diverse cellular contexts. Investigations into putative stress-protective roles for PrPC have resulted in an array of functions, such as inhibition of apoptosis, stimulation of anti-oxidant enzymes, scavenging roles, and a role in nuclear DNA repair. We have studied stress resilience of spermatozoa and peripheral blood mononuclear cells (PBMCs) derived from non-transgenic goats that lack PrPC (PRNPTer/Ter) compared with cells from normal (PRNP+/+) goats. Spermatozoa were analyzed for freeze tolerance, DNA integrity, viability, motility, ATP levels, and acrosome intactness at rest and after acute stress, induced by Cu2+ ions, as well as levels of reactive oxygen species (ROS) after exposure to FeSO4 and H2O2. Surprisingly, PrPC-negative spermatozoa reacted similarly to normal spermatozoa in all read-outs. Moreover, in vitro exposure of PBMCs to Doxorubicin, H2O2 and methyl methanesulfonate (MMS), revealed no effect of PrPC on cellular survival or global accumulation of DNA damage. Similar results were obtained with human neuroblastoma (SH-SY5Y) cell lines stably expressing varying levels of PrPC. RNA sequencing of PBMCs (n = 8 of PRNP+/+ and PRNPTer/Ter) showed that basal level expression of genes encoding DNA repair enzymes, ROS scavenging, and antioxidant enzymes were unaffected by the absence of PrPC. Data presented here questions the in vitro cytoprotective roles previously attributed to PrPC, although not excluding such functions in other cell types or tissues during inflammatory stress.
Collapse
Affiliation(s)
- Malin R Reiten
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Giulia Malachin
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Elisabeth Kommisrud
- Faculty of Education and Natural Sciences, Inland University of Applied Sciences, Hamar, Norway
| | - Gunn C Østby
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Karin E Waterhouse
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway.,Spermvital AS Holsetgata, Hamar, Norway
| | - Anette K Krogenæs
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Anna Kusnierczyk
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Clara M O Jalland
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Liv Heidi Nekså
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Susan S Røed
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Else-Berit Stenseth
- Faculty of Education and Natural Sciences, Inland University of Applied Sciences, Hamar, Norway
| | - Frøydis D Myromslien
- Faculty of Education and Natural Sciences, Inland University of Applied Sciences, Hamar, Norway
| | - Teklu T Zeremichael
- Faculty of Education and Natural Sciences, Inland University of Applied Sciences, Hamar, Norway
| | - Maren K Bakkebø
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Arild Espenes
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Michael A Tranulis
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| |
Collapse
|
49
|
Dai Q, Liu H, Gao C, Li W, Zhu C, Lin C, Tan Y, Yuan Z, Jiang Y. A one-step synthesized acridine-based fluorescent chemosensor for selective detection of copper(ii) ions and living cell imaging. NEW J CHEM 2018. [DOI: 10.1039/c7nj03615a] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A highly selective and sensitive fluorescence quenching chemosensor (ACC) for Cu2+ detection in HEPES buffer and living cell imaging was developed.
Collapse
Affiliation(s)
- Qiuzi Dai
- Department of Chemistry
- Tsinghua University
- Beijing
- P. R. China
- The Ministry-Province Jointly Constructed Base for State Key Lab-Shenzhen Key Laboratory of Chemical Biology
| | - Haiyang Liu
- Department of Chemistry
- Tsinghua University
- Beijing
- P. R. China
- The Ministry-Province Jointly Constructed Base for State Key Lab-Shenzhen Key Laboratory of Chemical Biology
| | - Chunmei Gao
- The Ministry-Province Jointly Constructed Base for State Key Lab-Shenzhen Key Laboratory of Chemical Biology
- the Graduate School at Shenzhen
- Tsinghua University
- Shenzhen
- P. R. China
| | - Wenlu Li
- Department of Chemistry
- Tsinghua University
- Beijing
- P. R. China
- The Ministry-Province Jointly Constructed Base for State Key Lab-Shenzhen Key Laboratory of Chemical Biology
| | - Caizhen Zhu
- School of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- P. R. China
| | - Chengdong Lin
- School of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- P. R. China
| | - Ying Tan
- The Ministry-Province Jointly Constructed Base for State Key Lab-Shenzhen Key Laboratory of Chemical Biology
- the Graduate School at Shenzhen
- Tsinghua University
- Shenzhen
- P. R. China
| | - Zigao Yuan
- Department of Chemistry
- Tsinghua University
- Beijing
- P. R. China
- The Ministry-Province Jointly Constructed Base for State Key Lab-Shenzhen Key Laboratory of Chemical Biology
| | - Yuyang Jiang
- The Ministry-Province Jointly Constructed Base for State Key Lab-Shenzhen Key Laboratory of Chemical Biology
- the Graduate School at Shenzhen
- Tsinghua University
- Shenzhen
- P. R. China
| |
Collapse
|
50
|
Sánchez-López C, Fernández CO, Quintanar L. Neuroprotective alpha-cleavage of the human prion protein significantly impacts Cu(ii) coordination at its His111 site. Dalton Trans 2018; 47:9274-9282. [DOI: 10.1039/c7dt03400h] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alpha-cleavage proteolytic processing of human prion protein significantly impacts its Cu(ii) coordination properties at the His111 site.
Collapse
Affiliation(s)
- Carolina Sánchez-López
- Departamento de Química
- Centro de Investigación y de Estudios Avanzados (Cinvestav)
- Mexico City
- Mexico
| | - Claudio O. Fernández
- Max Planck Laboratory for Structural Biology
- Chemistry and Molecular Biophysics of Rosario (MPLbioR
- UNR-MPIbpC) and Instituto de Investigaciones para el Descubrimiento de Fármacos de Rosario (IIDEFAR
- UNR-CONICET)
- Universidad Nacional de Rosario
| | - Liliana Quintanar
- Departamento de Química
- Centro de Investigación y de Estudios Avanzados (Cinvestav)
- Mexico City
- Mexico
| |
Collapse
|