1
|
Juliani do Amaral M, Soares de Oliveira L, Cordeiro Y. Zinc ions trigger the prion protein liquid-liquid phase separation. Biochem Biophys Res Commun 2025; 753:151489. [PMID: 39983547 DOI: 10.1016/j.bbrc.2025.151489] [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: 10/21/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Abstract
Prion diseases are characterized by the misfolding and conversion of the monomeric prion protein (PrP) to a multimeric aggregated pathogenic form, known as PrPSc. We and others have recently shown that biomolecular condensates formed via liquid-liquid phase separation of PrP can undergo maturation to solid-like species that resemble pathological aggregates, and this process is modulated by DNA, RNA, and oxidative conditions. Conversely, the most well-studied ligand of PrP, copper ions, induce liquid-like condensates of PrP that accumulate Cu2+in vitro, and live PrPC-expressing cells show condensation at the cell surface as triggered by physiologically relevant conditions of Cu2+ and protein concentrations. Since PrP can also bind to Zn2+ through its intrinsically disordered N-terminal domain, though with different affinities and binding modes than Cu2+, we hypothesized that Zn2+ could modulate PrP phase separation differently from copper ions. Using an appropriate buffer with negligible metal ion binding, as well as relevant pH, ionic strength, molecular crowding, and Zn2+ concentrations, we show that recombinant PrP undergoes phase separation with Zn2+. Furthermore, we show that metal ion-induced condensation of PrP is dependent on the N-terminal domain (residues 23-90). In vitro Fluorescence Recovery After Photobleaching (FRAP) experiments and thioflavin T aggregation kinetics support key differences in the molecular properties of PrP:Zn2+versus PrP:Cu2+ phase separated states. FRAP analysis indicated that both Cu2+ and Zn2+ promote liquid-like PrP condensates; however, PrP:Zn2+condensates exhibit a faster recovery. Cu2+ pronouncedly inhibits seed-induced PrP misfolding, whereas Zn2+ provides a milder delay in PrP aggregation. Our findings provide insights on Zn2+-induced phase separation of PrP, supporting a variety of previously proposed functions of PrP in metal sequestering and uptake, processes that could be effectively regulated through biomolecular condensation.
Collapse
Affiliation(s)
| | | | - Yraima Cordeiro
- Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
López-Guerrero V, Posadas Y, Sánchez-López C, Smart A, Miranda J, Singewald K, Bandala Y, Juaristi E, Den Auwer C, Perez-Cruz C, González-Mariscal L, Millhauser G, Segovia J, Quintanar L. A Copper-Binding Peptide with Therapeutic Potential against Alzheimer's Disease: From the Blood-Brain Barrier to Metal Competition. ACS Chem Neurosci 2025; 16:241-261. [PMID: 39723808 PMCID: PMC11741003 DOI: 10.1021/acschemneuro.4c00796] [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: 11/25/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia worldwide. AD brains are characterized by the accumulation of amyloid-β peptides (Aβ) that bind Cu2+ and have been associated with several neurotoxic mechanisms. Although the use of copper chelators to prevent the formation of Cu2+-Aβ complexes has been proposed as a therapeutic strategy, recent studies show that copper is an important neuromodulator that is essential for a neuroprotective mechanism mediated by Cu2+ binding to the cellular prion protein (PrPC). Therefore, in addition to metal selectivity and blood-brain barrier (BBB) permeability, an emerging challenge for copper chelators is to prevent the formation of neurotoxic Cu2+-Aβ species without perturbing the neuroprotective Cu2+-PrPC interaction. Previously, we reported the design of a tetrapeptide (TP) that withdraws Cu2+ from Aβ(1-16) and impacts the Cu2+-induced aggregation of Aβ(1-40). In this study, we improved the drug-like properties of TP in a BBB model, evaluated the metal selectivity of the optimized peptide (TP*), and tested its effect on Cu2+ coordination to PrPC and proteins involved in copper trafficking, such as copper transporter 1 and albumin. Our results show that changing the stereochemistry of the first residue prevents TP degradation in the BBB model and coadministration of TP with a peptide that increases BBB permeability allows its passage through the BBB model. TP* is highly selective toward Cu2+ in the presence of Zn2+ ions, transfers Cu2+ to copper-trafficking proteins, and forms a ternary TP*-Cu2+-PrP species that does not perturb the physiological conformation of PrP and displays only a minor impact in the neuroprotective Cu2+-dependent interaction of PrPC with the N-methyl-d-aspartate receptor. Overall, these results show that TP* displays desirable features for a copper chelator with therapeutic potential against AD. Moreover, this is the first study that explores the effect of a Cu2+ chelator with therapeutic potential for AD on Cu2+ coordination to PrPC (an emerging key player in AD pathology), integrating recent knowledge about metalloproteins involved in AD with the design of copper chelators against AD.
Collapse
Affiliation(s)
- Victor
E. López-Guerrero
- Department
of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
- Department
of Chemistry, Center for Research and Advanced
Studies (Cinvestav), Mexico City 07360, Mexico
| | - Yanahi Posadas
- Department
of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
- Department
of Pharmacology, Center for Research and
Advanced Studies (Cinvestav), Mexico
City 07360, Mexico
| | - Carolina Sánchez-López
- Center
for Research in Aging, Center for Research
and Advanced Studies (Cinvestav), Mexico City 14330, Mexico
| | - Amanda Smart
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, 1156, Santa Cruz 95064, United States
| | - Jael Miranda
- Department
of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Kevin Singewald
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, 1156, Santa Cruz 95064, United States
| | - Yamir Bandala
- Department
of Chemistry, Center for Research and Advanced
Studies (Cinvestav), Mexico City 07360, Mexico
| | - Eusebio Juaristi
- Department
of Chemistry, Center for Research and Advanced
Studies (Cinvestav), Mexico City 07360, Mexico
- El Colegio
Nacional, Mexico City 06020, Mexico
| | | | - Claudia Perez-Cruz
- Department
of Pharmacology, Center for Research and
Advanced Studies (Cinvestav), Mexico
City 07360, Mexico
| | - Lorenza González-Mariscal
- Department
of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Glenn Millhauser
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, 1156, Santa Cruz 95064, United States
| | - Jose Segovia
- Department
of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Liliana Quintanar
- Department
of Chemistry, Center for Research and Advanced
Studies (Cinvestav), Mexico City 07360, Mexico
- Center
for Research in Aging, Center for Research
and Advanced Studies (Cinvestav), Mexico City 14330, Mexico
| |
Collapse
|
3
|
Casey C, Sleator RD. Prions: structure, function, evolution, and disease. Arch Microbiol 2024; 207:1. [PMID: 39572454 DOI: 10.1007/s00203-024-04200-3] [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: 09/02/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024]
Abstract
Prions are proteinaceous infectious particles implicated in fatal neurodegenerative disorders known as prion diseases. Herein, we provide an overview of prion biology, emphasizing the structural, functional, and evolutionary aspects of prions, along with their potential applications in protein engineering. Understanding the structure-function relationships of both healthy and disease-associated prion proteins enables a deeper understanding of the mechanisms of prion-induced neurotoxicity. Furthermore, we describe how insights into prion evolution have begun to shed light on their ancient origins and evolutionary resilience, offering deeper insights into the potential roles of prions in primordial chemical processes.
Collapse
Affiliation(s)
- Clara Casey
- Department of Biological Sciences, Munster Technological University, Bishopstown, Cork, T12 P928, Ireland
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Roy D Sleator
- Department of Biological Sciences, Munster Technological University, Bishopstown, Cork, T12 P928, Ireland.
| |
Collapse
|
4
|
Pehar M, Hewitt M, Wagner A, Sandhu JK, Khalili A, Wang X, Cho JY, Sim VL, Kulka M. Histamine stimulates human microglia to alter cellular prion protein expression via the HRH2 histamine receptor. Sci Rep 2024; 14:25519. [PMID: 39462031 PMCID: PMC11513956 DOI: 10.1038/s41598-024-75982-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Although the cellular prion protein (PrPC) has been evolutionarily conserved, the role of this protein remains elusive. Recent evidence indicates that PrPC may be involved in neuroinflammation and the immune response in the brain, and its expression may be modified via various mechanisms. Histamine is a proinflammatory mediator and neurotransmitter that stimulates numerous cells via interactions with histamine receptors 1-4 (HRH1-4). Since microglia are the innate immune cells of the central nervous system, we hypothesized that histamine-induced stimulation regulates the expression of PrPC in human-derived microglia. The human microglial clone 3 (HMC3) cell line was treated with histamine, and intracellular calcium levels were measured via a calcium flux assay. Cytokine production was monitored by enzyme-linked immunosorbent assay (ELISA). Western blotting and quantitative reverse transcription-polymerase chain reaction were used to determine protein and gene expression of HRH1-4. Flow cytometry and western blotting were used to measure PrPC expression levels. Fluorescence microscopy was used to examine Iba-1 and PrPC localization. HMC3 cells stimulated by histamine exhibited increased intracellular calcium levels and increased release of IL-6 and IL-8, while also modifying PrPC localization. HMC3 stimulated with histamine for 6 and 24 hours exhibited increased surface PrPC expression. Specifically, we found that stimulation of the HRH2 receptor was responsible for changes in surface PrPC. Histamine-induced increases in surface PrPC were attenuated following inhibition of the HRH2 receptor via the HRH2 antagonist ranitidine. These changes were unique to HRH2 activation, as stimulation of HRH1, HRH3, or HRH4 did not alter surface PrPC. Prolonged stimulation of HMC3 decreased PrPC expression following 48 and 72 hours of histamine stimulation. HMC3 cells can be stimulated by histamine to undergo intracellular calcium influx. Surface expression levels of PrPC on HMC3 cells are altered by histamine exposure, primarily mediated by HRH2. While histamine exposure also increases release of IL-6 and IL-8 in these cells, this cytokine release is not fully dependent on PrPC levels, as IL-6 release is only partially reduced and IL-8 release is unchanged under the conditions of HRH2 blockade that prevent PrPC changes. Overall, this suggests that PrPC may play a role in modulating microglial responses.
Collapse
Affiliation(s)
- Marcus Pehar
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - Melissa Hewitt
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Ashley Wagner
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
| | - Jagdeep K Sandhu
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Aria Khalili
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Xinyu Wang
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Jae-Young Cho
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Valerie L Sim
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Marianna Kulka
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada.
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
5
|
Striebel JF, Carroll JA, Race B, Leung JM, Schwartz C, Reese ED, Bowes Rickman C, Chesebro B, Klingeborn M. The prion protein is required for normal responses to light stimuli by photoreceptors and bipolar cells. iScience 2024; 27:110954. [PMID: 39381753 PMCID: PMC11460503 DOI: 10.1016/j.isci.2024.110954] [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: 05/02/2024] [Revised: 07/15/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
The prion protein, PrPC, is well known as an essential susceptibility factor for neurodegenerative prion diseases, yet its function in normal, healthy cells remains uncertain. A role in synaptic function has been proposed for PrPC, supported by its cell surface expression in neurons and glia. Here, in mouse retina, we localized PrPC to the junctions between photoreceptors and bipolar cells using synaptic proteins EAAT5, CtBP2, and PSD-95. PrPC localized most densely with bipolar cell dendrites synapsing with cone photoreceptors. In two coisogenic mouse strains, deletion of the gene encoding PrPC, Prnp, significantly altered the scotopic and/or photopic electroretinographic (ERG) responses of photoreceptors and bipolar cells. Cone-dominant pathways showed the most significant ERG changes. Retinal thickness, quantitated by high-resolution optical coherence tomography (OCT), and ribbon synapse morphology were not altered upon deletion of PrPC, suggesting that the ERG changes were driven by functional rather than structural alterations.
Collapse
Affiliation(s)
- James F. Striebel
- Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - James A. Carroll
- Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Brent Race
- Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Jacqueline M. Leung
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Cindi Schwartz
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Emily D. Reese
- McLaughlin Research Institute, Great Falls, MT 59405, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bruce Chesebro
- Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Mikael Klingeborn
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27710, USA
- McLaughlin Research Institute, Great Falls, MT 59405, USA
| |
Collapse
|
6
|
Irie KI, Honda H, Tateishi T, Mori S, Yamamoto A, Morimitsu M, Shinsuke K, Moritaka T, Kurata S, Kumazoe H, Shijo M, Sasagasako N, Taniwaki T. Dopaminergic neurodegeneration in Gerstmann-Sträussler-Scheinker (P102L) disease: insights from imaging and pathological examination. Front Neurol 2024; 15:1452709. [PMID: 39376689 PMCID: PMC11456421 DOI: 10.3389/fneur.2024.1452709] [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: 06/21/2024] [Accepted: 09/06/2024] [Indexed: 10/09/2024] Open
Abstract
Gerstmann-Sträussler-Scheinker (GSS) disease is an inherited prion disease characterized by dementia, cerebellar ataxia, and painful sensory disturbances. GSS is pathologically defined by the presence of amyloid plaques comprised of prion protein predominantly localized in the cerebral cortex, cerebellar cortex, and basal ganglia, resulting from mutations in the prion protein gene. This study investigated five cases of GSS P102L [GSS caused by a leucine (L) substitution of proline (P) at position 102 of the prion protein gene] with L-dopa-resistant extrapyramidal symptoms and reduced dopamine transporter single-photon emission computed tomography (DAT-SPECT) uptake. Clinical findings revealed diverse manifestations, with all cases exhibiting parkinsonism, and four patients had a vertical gaze palsy. Notably, all patients showed reduced striatal DAT-SPECT uptake, indicating neurodegeneration of the nigrostriatal system. Autopsy findings in one case confirmed prion protein plaques and dopaminergic neuron loss in the substantia nigra of a patient with GSS P102L. Additionally, reduced DAT immunostaining was observed in the putamen compared with a control. While previous studies have identified reduced DAT-SPECT and positron emission tomography uptake in Creutzfeldt-Jakob disease and fatal familial insomnia owing to nigrostriatal neurodegeneration induced by abnormal prion protein deposition, similar phenomena in GSS P102L have not been reported. This study provides support for a correlation between abnormal prion protein deposition and nigrostriatal system degeneration in GSS P102L. Our results reveal the importance of considering GSS P102L in cases of atypical Parkinsonism and abnormal DAT-SPECT results, which would serve as a valuable indicator for subsequent prion genetic testing.
Collapse
Affiliation(s)
- Ken-Ichi Irie
- Division of Respirology, Neurology and Rheumatology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Neuropathology Center, NHO Omuta Hospital, Fukuoka, Japan
| | - Hiroyuki Honda
- Neuropathology Center, NHO Omuta Hospital, Fukuoka, Japan
| | - Takahisa Tateishi
- Division of Respirology, Neurology and Rheumatology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Shinichiro Mori
- Division of Respirology, Neurology and Rheumatology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Neuropathology Center, NHO Omuta Hospital, Fukuoka, Japan
| | - Akifumi Yamamoto
- Division of Neurology, Department of Neurology, Neuro Muscular Center, NHO Omuta Hospital, Fukuoka, Japan
| | - Makoto Morimitsu
- Division of Respirology, Neurology and Rheumatology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Kikuchi Shinsuke
- Division of Respirology, Neurology and Rheumatology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Taiga Moritaka
- Division of Respirology, Neurology and Rheumatology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Seiji Kurata
- Department of Radiology, Kurume University School of Medicine, Kurume, Japan
| | | | - Masahiro Shijo
- Neuropathology Center, NHO Omuta Hospital, Fukuoka, Japan
- Department of Neurology, Kyushu Central Hospital of the Mutual Aid Association of Public School Teachers, Fukuoka, Japan
| | - Naokazu Sasagasako
- Division of Neurology, Department of Neurology, Neuro Muscular Center, NHO Omuta Hospital, Fukuoka, Japan
| | - Takayuki Taniwaki
- Division of Respirology, Neurology and Rheumatology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
7
|
Ono N, Suzuyama K, Minagawa H, Uwatoko K, Yoshikawa M, Ide T, Mitsuoka M, Honda K, Hirai T, Otsuka T, Kai K, Honda H, Kitamoto T, Irie H, Yukitake M, Koike H. Involvement of the nigrostriatal system in Gerstman-Sträussler-Scheinker disease with the PRNP-P102L mutation. J Neurol Sci 2024; 464:123166. [PMID: 39128159 DOI: 10.1016/j.jns.2024.123166] [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: 05/16/2024] [Revised: 07/07/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Gerstmann-Sträussler-Scheinker disease (GSS) is an autosomal-dominant inherited prion disease most often associated with the human prion protein gene (PRNP)-P102L mutation. Although patients manifest considerable phenotypic heterogeneity, the involvement of the nigrostriatal system has not been well-studied. METHODS We performed dopamine transporter single-photon emission computed tomography (DAT-SPECT) using 123I-ioflupane to investigate the nigrostriatal system function in nine patients with the PRNP-P102L mutation. We also examined the pathological findings in another patient whose predominant feature was ataxia and who died 5 years after disease onset. RESULTS Striatum uptake of 123I-ioflupane indicated by specific binding ratio (SBR) values was significantly reduced in two patients. The DAT-SPECT examination was performed 6 months after disease onset in one of these patients who manifested rapidly developing cognitive decline mimicking Creutzfeldt-Jakob disease. DAT-SPECT was also performed 9 years after disease onset in another patient who manifested the conventional features of GSS involving ataxia and dementia in the initial phase but showed akinetic mutism at the examination time. Another patient examined 2 years after disease onset who predominantly manifested ataxia showed marginally abnormal SBR values. An autopsy case showed moderate neuronal loss in the substantia nigra, and the degree of neuronal loss was similar in most other parts of the brain. CONCLUSION Nigrostriatal system involvement may occur in patients with GSS associated with the PRNP-P102L mutation, even though parkinsonism is not the predominant feature.
Collapse
Affiliation(s)
- Natsuki Ono
- Department of Neurology, Imari Arita Kyoritsu Hospital, Arita, Japan; Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan; Department of Neurology, Kouhoukai Takagi Hospital, Okawa, Japan
| | - Kohei Suzuyama
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Hiromu Minagawa
- Department of Neurology, Kouhoukai Takagi Hospital, Okawa, Japan
| | - Kiku Uwatoko
- Department of Neurology, Kouhoukai Takagi Hospital, Okawa, Japan
| | - Masaaki Yoshikawa
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Toshihiro Ide
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Miyuki Mitsuoka
- Department of Radiology, Saga University Hospital, Saga, Japan
| | - Kazuo Honda
- Department of Radiology, Kouhoukai Takagi Hospital, Okawa, Japan
| | - Tetsuyoshi Hirai
- Department of Radiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Takateru Otsuka
- Department of Radiology, Kouhoukai Takagi Hospital, Okawa, Japan
| | - Keita Kai
- Department of Pathology, Saga University Hospital, Saga, Japan
| | - Hiroyuki Honda
- Neuropathology Center, NHO, Omuta Hospital, Omuta, Japan
| | - Tetsuyuki Kitamoto
- Department of Neurological Science, Tohoku University School of Medicine, Sendai, Japan
| | - Hiroyuki Irie
- Department of Radiology, Faculty of Medicine, Saga University, Saga, Japan
| | | | - Haruki Koike
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan.
| |
Collapse
|
8
|
Ghosh S, Jana R, Jana S, Basu R, Chatterjee M, Ranawat N, Das Sarma J. Differential expression of cellular prion protein (PrP C) in mouse hepatitis virus induced neuroinflammation. J Neurovirol 2024; 30:215-228. [PMID: 38922550 DOI: 10.1007/s13365-024-01215-w] [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: 02/21/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024]
Abstract
The cellular prion protein (PrPC) is an extracellular cell membrane protein. Due to its diversified roles, a definite role of PrPC has been difficult to establish. During viral infection, PrPC has been reported to play a pleiotropic role. Here, we have attempted to envision the function of PrPC in the neurotropic m-CoV-MHV-RSA59-induced model of neuroinflammation in C57BL/6 mice. A significant upregulation of PrPC at protein and mRNA levels was evident in infected mouse brains during the acute phase of neuroinflammation. Furthermore, investigation of the effect of MHV-RSA59 infection on PrPC expression in specific neuronal, microglial, and astrocytoma cell lines, revealed a differential expression of prion protein during neuroinflammation. Additionally, siRNA-mediated downregulation of prnp transcripts reduced the expression of viral antigen and viral infectivity in these cell lines. Cumulatively, our results suggest that PrPC expression significantly increases during acute MHV-RSA59 infection and that PrPC also assists in viral infectivity and viral replication.
Collapse
Affiliation(s)
- Satavisha Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
| | - Rishika Jana
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
| | - Soumen Jana
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
- Optical NeuroImaging Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Rahul Basu
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Madhurima Chatterjee
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
| | - Nishtha Ranawat
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
- Burke Neurological Institute, Weill Cornell Medicine, New York, NY, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India.
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
9
|
Giorgi C, Lombardozzi G, Ammannito F, Scenna MS, Maceroni E, Quintiliani M, d’Angelo M, Cimini A, Castelli V. Brain Organoids: A Game-Changer for Drug Testing. Pharmaceutics 2024; 16:443. [PMID: 38675104 PMCID: PMC11054008 DOI: 10.3390/pharmaceutics16040443] [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: 02/07/2024] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Neurological disorders are the second cause of death and the leading cause of disability worldwide. Unfortunately, no cure exists for these disorders, but the actual therapies are only able to ameliorate people's quality of life. Thus, there is an urgent need to test potential therapeutic approaches. Brain organoids are a possible valuable tool in the study of the brain, due to their ability to reproduce different brain regions and maturation stages; they can be used also as a tool for disease modelling and target identification of neurological disorders. Recently, brain organoids have been used in drug-screening processes, even if there are several limitations to overcome. This review focuses on the description of brain organoid development and drug-screening processes, discussing the advantages, challenges, and limitations of the use of organoids in modeling neurological diseases. We also highlighted the potential of testing novel therapeutic approaches. Finally, we examine the challenges and future directions to improve the drug-screening process.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Annamaria Cimini
- Department of Life, Health and Environmental Science, University of L’Aquila, 67100 L’Aquila, Italy; (C.G.); (G.L.); (F.A.); (M.S.S.); (E.M.); (M.Q.); (M.d.)
| | - Vanessa Castelli
- Department of Life, Health and Environmental Science, University of L’Aquila, 67100 L’Aquila, Italy; (C.G.); (G.L.); (F.A.); (M.S.S.); (E.M.); (M.Q.); (M.d.)
| |
Collapse
|
10
|
Perumal N, Yurugi H, Dahm K, Rajalingam K, Grus FH, Pfeiffer N, Manicam C. Proteome landscape and interactome of voltage-gated potassium channel 1.6 (Kv1.6) of the murine ophthalmic artery and neuroretina. Int J Biol Macromol 2024; 257:128464. [PMID: 38043654 DOI: 10.1016/j.ijbiomac.2023.128464] [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: 08/29/2023] [Revised: 11/14/2023] [Accepted: 11/25/2023] [Indexed: 12/05/2023]
Abstract
The voltage-gated potassium channel 1.6 (Kv1.6) plays a vital role in ocular neurovascular beds and exerts its modulatory functions via interaction with other proteins. However, the interactome and their potential roles remain unknown. Here, the global proteome landscape of the ophthalmic artery (OA) and neuroretina was mapped, followed by the determination of Kv1.6 interactome and validation of its functionality and cellular localization. Microfluorimetric analysis of intracellular [K+] and Western blot validated the native functionality and cellular expression of the recombinant Kv1.6 channel protein. A total of 54, 9 and 28 Kv1.6-interacting proteins were identified in the mouse OA and, retina of mouse and rat, respectively. The Kv1.6-protein partners in the OA, namely actin cytoplasmic 2, alpha-2-macroglobulin and apolipoprotein A-I, were implicated in the maintenance of blood vessel integrity by regulating integrin-mediated adhesion to extracellular matrix and Ca2+ flux. Many retinal protein interactors, particularly the ADP/ATP translocase 2 and cytoskeleton protein tubulin, were involved in endoplasmic reticulum stress response and cell viability. Three common interactors were found in all samples comprising heat shock cognate 71 kDa protein, Ig heavy constant gamma 1 and Kv1.6 channel. This foremost in-depth investigation enriched and identified the elusive Kv1.6 channel and, elucidated its complex interactome.
Collapse
Affiliation(s)
- Natarajan Perumal
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Hajime Yurugi
- Cell Biology Unit, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
| | - Katrin Dahm
- Cell Biology Unit, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
| | - Franz H Grus
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
11
|
do Amaral MJ, Mohapatra S, Passos AR, Lopes da Silva TS, Carvalho RS, da Silva Almeida M, Pinheiro AS, Wegmann S, Cordeiro Y. Copper drives prion protein phase separation and modulates aggregation. SCIENCE ADVANCES 2023; 9:eadi7347. [PMID: 37922348 PMCID: PMC10624353 DOI: 10.1126/sciadv.adi7347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/03/2023] [Indexed: 11/05/2023]
Abstract
Prion diseases are characterized by prion protein (PrP) transmissible aggregation and neurodegeneration, which has been linked to oxidative stress. The physiological function of PrP seems related to sequestering of redox-active Cu2+, and Cu2+ dyshomeostasis is observed in prion disease brain. It is unclear whether Cu2+ contributes to PrP aggregation, recently shown to be mediated by PrP condensation. This study indicates that Cu2+ promotes PrP condensation in live cells at the cell surface and in vitro through copartitioning. Molecularly, Cu2+ inhibited PrP β-structure and hydrophobic residues exposure. Oxidation, induced by H2O2, triggered liquid-to-solid transition of PrP:Cu2+ condensates and promoted amyloid-like PrP aggregation. In cells, overexpression of PrPC initially protected against Cu2+ cytotoxicity but led to PrPC aggregation upon extended copper exposure. Our data suggest that PrP condensates function as a buffer for copper that prevents copper toxicity but can transition into PrP aggregation at prolonged oxidative stress.
Collapse
Affiliation(s)
- Mariana Juliani do Amaral
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | | | - Aline Ribeiro Passos
- Brazilian Synchrotron Light Laboratory (LNLS), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | | | | | - Marcius da Silva Almeida
- Plataforma Avançada de Biomoléculas, Centro Nacional de Biologia Estrutural e Bioimagem, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anderson Sá Pinheiro
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susanne Wegmann
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Yraima Cordeiro
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Viles JH. Imaging Amyloid-β Membrane Interactions: Ion-Channel Pores and Lipid-Bilayer Permeability in Alzheimer's Disease. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 135:e202215785. [PMID: 38515735 PMCID: PMC10952214 DOI: 10.1002/ange.202215785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Indexed: 03/08/2023]
Abstract
The accumulation of the amyloid-β peptides (Aβ) is central to the development of Alzheimer's disease. The mechanism by which Aβ triggers a cascade of events that leads to dementia is a topic of intense investigation. Aβ self-associates into a series of complex assemblies with different structural and biophysical properties. It is the interaction of these oligomeric, protofibril and fibrillar assemblies with lipid membranes, or with membrane receptors, that results in membrane permeability and loss of cellular homeostasis, a key event in Alzheimer's disease pathology. Aβ can have an array of impacts on lipid membranes, reports have included: a carpeting effect; a detergent effect; and Aβ ion-channel pore formation. Recent advances imaging these interactions are providing a clearer picture of Aβ induced membrane disruption. Understanding the relationship between different Aβ structures and membrane permeability will inform therapeutics targeting Aβ cytotoxicity.
Collapse
Affiliation(s)
- John H. Viles
- Department of Biochemistry, SBBS, Queen MaryUniversity of LondonUK
| |
Collapse
|
13
|
Viles JH. Imaging Amyloid-β Membrane Interactions: Ion-Channel Pores and Lipid-Bilayer Permeability in Alzheimer's Disease. Angew Chem Int Ed Engl 2023; 62:e202215785. [PMID: 36876912 PMCID: PMC10953358 DOI: 10.1002/anie.202215785] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
The accumulation of the amyloid-β peptides (Aβ) is central to the development of Alzheimer's disease. The mechanism by which Aβ triggers a cascade of events that leads to dementia is a topic of intense investigation. Aβ self-associates into a series of complex assemblies with different structural and biophysical properties. It is the interaction of these oligomeric, protofibril and fibrillar assemblies with lipid membranes, or with membrane receptors, that results in membrane permeability and loss of cellular homeostasis, a key event in Alzheimer's disease pathology. Aβ can have an array of impacts on lipid membranes, reports have included: a carpeting effect; a detergent effect; and Aβ ion-channel pore formation. Recent advances imaging these interactions are providing a clearer picture of Aβ induced membrane disruption. Understanding the relationship between different Aβ structures and membrane permeability will inform therapeutics targeting Aβ cytotoxicity.
Collapse
Affiliation(s)
- John H. Viles
- Department of Biochemistry, SBBS, Queen MaryUniversity of LondonUK
| |
Collapse
|
14
|
Lawrence JA, Aguilar-Calvo P, Ojeda-Juárez D, Khuu H, Soldau K, Pizzo DP, Wang J, Malik A, Shay TF, Sullivan EE, Aulston B, Song SM, Callender JA, Sanchez H, Geschwind MD, Roy S, Rissman RA, Trejo J, Tanaka N, Wu C, Chen X, Patrick GN, Sigurdson CJ. Diminished Neuronal ESCRT-0 Function Exacerbates AMPA Receptor Derangement and Accelerates Prion-Induced Neurodegeneration. J Neurosci 2023; 43:3970-3984. [PMID: 37019623 PMCID: PMC10219035 DOI: 10.1523/jneurosci.1878-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
Endolysosomal defects in neurons are central to the pathogenesis of prion and other neurodegenerative disorders. In prion disease, prion oligomers traffic through the multivesicular body (MVB) and are routed for degradation in lysosomes or for release in exosomes, yet how prions impact proteostatic pathways is unclear. We found that prion-affected human and mouse brain showed a marked reduction in Hrs and STAM1 (ESCRT-0), which route ubiquitinated membrane proteins from early endosomes into MVBs. To determine how the reduction in ESCRT-0 impacts prion conversion and cellular toxicity in vivo, we prion-challenged conditional knockout mice (male and female) having Hrs deleted from neurons, astrocytes, or microglia. The neuronal, but not astrocytic or microglial, Hrs-depleted mice showed a shortened survival and an acceleration in synaptic derangements, including an accumulation of ubiquitinated proteins, deregulation of phosphorylated AMPA and metabotropic glutamate receptors, and profoundly altered synaptic structure, all of which occurred later in the prion-infected control mice. Finally, we found that neuronal Hrs (nHrs) depletion increased surface levels of the cellular prion protein, PrPC, which may contribute to the rapidly advancing disease through neurotoxic signaling. Taken together, the reduced Hrs in the prion-affected brain hampers ubiquitinated protein clearance at the synapse, exacerbates postsynaptic glutamate receptor deregulation, and accelerates neurodegeneration.SIGNIFICANCE STATEMENT Prion diseases are rapidly progressive neurodegenerative disorders characterized by prion aggregate spread through the central nervous system. Early disease features include ubiquitinated protein accumulation and synapse loss. Here, we investigate how prion aggregates alter ubiquitinated protein clearance pathways (ESCRT) in mouse and human prion-infected brain, discovering a marked reduction in Hrs. Using a prion-infection mouse model with neuronal Hrs (nHrs) depleted, we show that low neuronal Hrs is detrimental and markedly shortens survival time while accelerating synaptic derangements, including ubiquitinated protein accumulation, indicating that Hrs loss exacerbates prion disease progression. Additionally, Hrs depletion increases the surface distribution of prion protein (PrPC), linked to aggregate-induced neurotoxic signaling, suggesting that Hrs loss in prion disease accelerates disease through enhancing PrPC-mediated neurotoxic signaling.
Collapse
Affiliation(s)
- Jessica A Lawrence
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
| | - Patricia Aguilar-Calvo
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
| | - Daniel Ojeda-Juárez
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
| | - Helen Khuu
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
| | - Katrin Soldau
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
| | - Donald P Pizzo
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
| | - Jin Wang
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
| | - Adela Malik
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
| | - Timothy F Shay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Erin E Sullivan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Brent Aulston
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Seung Min Song
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
| | - Julia A Callender
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
| | - Henry Sanchez
- Department of Pathology, University of California, San Francisco, San Francisco, California 94143
| | - Michael D Geschwind
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California 94143
| | - Subhojit Roy
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - JoAnn Trejo
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093
| | - Nobuyuki Tanaka
- Division of Tumor Immunobiology, Miyagi Cancer Center Research Institute, Natori 981-1293, Japan
- Division of Tumor Immunobiology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Chengbiao Wu
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Xu Chen
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Gentry N Patrick
- Department of Biology, University of California, San Diego, La Jolla, California 92093
| | - Christina J Sigurdson
- Department of Pathology, University of California, San Diego, La, Jolla, California, 92093
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, Davis, California 95616
- Department of Medicine, University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
15
|
Gielnik M, Szymańska A, Dong X, Jarvet J, Svedružić ŽM, Gräslund A, Kozak M, Wärmländer SKTS. Prion Protein Octarepeat Domain Forms Transient β-Sheet Structures upon Residue-Specific Binding to Cu(II) and Zn(II) Ions. Biochemistry 2023. [PMID: 37163663 DOI: 10.1021/acs.biochem.3c00129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Misfolding of the cellular prion protein (PrPC) is associated with the development of fatal neurodegenerative diseases called transmissible spongiform encephalopathies (TSEs). Metal ions appear to play a crucial role in PrPC misfolding. PrPC is a combined Cu(II) and Zn(II) metal-binding protein, where the main metal-binding site is located in the octarepeat (OR) region. Thus, the biological function of PrPC may involve the transport of divalent metal ions across membranes or buffering concentrations of divalent metal ions in the synaptic cleft. Recent studies have shown that an excess of Cu(II) ions can result in PrPC instability, oligomerization, and/or neuroinflammation. Here, we have used biophysical methods to characterize Cu(II) and Zn(II) binding to the isolated OR region of PrPC. Circular dichroism (CD) spectroscopy data suggest that the OR domain binds up to four Cu(II) ions or two Zn(II) ions. Binding of the first metal ion results in a structural transition from the polyproline II helix to the β-turn structure, while the binding of additional metal ions induces the formation of β-sheet structures. Fluorescence spectroscopy data indicate that the OR region can bind both Cu(II) and Zn(II) ions at neutral pH, but under acidic conditions, it binds only Cu(II) ions. Molecular dynamics simulations suggest that binding of either metal ion to the OR region results in the formation of β-hairpin structures. As the formation of β-sheet structures can be a first step toward amyloid formation, we propose that high concentrations of either Cu(II) or Zn(II) ions may have a pro-amyloid effect in TSE diseases.
Collapse
Affiliation(s)
- Maciej Gielnik
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, PL 61-614 Poznań, Poland
| | - Aneta Szymańska
- Department of Biomedical Chemistry, Faculty of Chemistry, Gdańsk University, PL 80-308 Gdańsk, Poland
| | - Xiaolin Dong
- Chemistry Section, Stockholm University, 10691 Stockholm, Sweden
| | - Jüri Jarvet
- Chemistry Section, Stockholm University, 10691 Stockholm, Sweden
- The National Institute of Chemical Physics and Biophysics, 12618 Tallinn, Estonia
| | - Željko M Svedružić
- Department of Biotechnology, University of Rijeka, HR 51000 Rijeka, Croatia
| | - Astrid Gräslund
- Chemistry Section, Stockholm University, 10691 Stockholm, Sweden
| | - Maciej Kozak
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, PL 61-614 Poznań, Poland
- National Synchrotron Radiation Centre SOLARIS, Jagiellonian University, PL 30-392 Kraków, Poland
| | | |
Collapse
|
16
|
Ojeda-Juárez D, Lawrence JA, Soldau K, Pizzo DP, Wheeler E, Aguilar-Calvo P, Khuu H, Chen J, Malik A, Funk G, Nam P, Sanchez H, Geschwind MD, Wu C, Yeo GW, Chen X, Patrick GN, Sigurdson CJ. Prions induce an early Arc response and a subsequent reduction in mGluR5 in the hippocampus. Neurobiol Dis 2022; 172:105834. [PMID: 35905927 PMCID: PMC10080886 DOI: 10.1016/j.nbd.2022.105834] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 12/01/2022] Open
Abstract
Synapse dysfunction and loss are central features of neurodegenerative diseases, caused in part by the accumulation of protein oligomers. Amyloid-β, tau, prion, and α-synuclein oligomers bind to the cellular prion protein (PrPC), resulting in the activation of macromolecular complexes and signaling at the post-synapse, yet the early signaling events are unclear. Here we sought to determine the early transcript and protein alterations in the hippocampus during the pre-clinical stages of prion disease. We used a transcriptomic approach focused on the early-stage, prion-infected hippocampus of male wild-type mice, and identify immediate early genes, including the synaptic activity response gene, Arc/Arg3.1, as significantly upregulated. In a longitudinal study of male, prion-infected mice, Arc/Arg-3.1 protein was increased early (40% of the incubation period), and by mid-disease (pre-clinical), phosphorylated AMPA receptors (pGluA1-S845) were increased and metabotropic glutamate receptors (mGluR5 dimers) were markedly reduced in the hippocampus. Notably, sporadic Creutzfeldt-Jakob disease (sCJD) post-mortem cortical samples also showed low levels of mGluR5 dimers. Together, these findings suggest that prions trigger an early Arc response, followed by an increase in phosphorylated GluA1 and a reduction in mGluR5 receptors.
Collapse
Affiliation(s)
- Daniel Ojeda-Juárez
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Jessica A Lawrence
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Katrin Soldau
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Donald P Pizzo
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Emily Wheeler
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Helen Khuu
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Joy Chen
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Adela Malik
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Gail Funk
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Percival Nam
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Henry Sanchez
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA
| | - Michael D Geschwind
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Xu Chen
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Gentry N Patrick
- Division of Biological Sciences, Section of Neurobiology, University of California San Diego, La Jolla, CA, USA
| | - Christina J Sigurdson
- Department of Pathology, University of California San Diego, La Jolla, CA, USA; Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Pathology, Microbiology and Immunology, University of California Davis, Davis, CA, USA.
| |
Collapse
|
17
|
Early Changes in Transcriptomic Profiles in Synaptodendrosomes Reveal Aberrant Synaptic Functions in Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms23168888. [PMID: 36012153 PMCID: PMC9408306 DOI: 10.3390/ijms23168888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most prevalent neurodegenerative disorders characterized by the progressive decline of cognitive functions, and is closely associated with the dysfunction of synapses, which comprise the basic structure that mediates the communication between neurons. Although the protein architecture and machinery for protein translation at synapses are extensively studied, the impact that local changes in the mRNA reservoir have on AD progression is largely unknown. Here, we investigated the changes in transcriptomic profiles in the synaptodendrosomes purified from the cortices of AD mice at ages 3 and 6 months, a stage when early signatures of synaptic dysfunction are revealed. The transcriptomic profiles of synaptodendrosomes showed a greater number of localized differentially expressed genes (DEGs) in 6-month-old AD mice compared with mice 3 months of age. Gene Ontology (GO) analysis showed that these DEGs are majorly enriched in mitochondrial biogenesis and metabolic activity. More specifically, we further identified three representative DEGs in mitochondrial and metabolic pathways—Prnp, Cst3, and Cox6c—that regulate the dendritic spine density and morphology in neurons. Taken together, this study provides insights into the transcriptomic changes in synaptodendrosomes during AD progression, which may facilitate the development of intervention strategies targeting local translation to ameliorate the pathological progression of AD.
Collapse
|
18
|
Betancor M, Pérez-Lázaro S, Otero A, Marín B, Martín-Burriel I, Blennow K, Badiola JJ, Zetterberg H, Bolea R. Neurogranin and Neurofilament Light Chain as Preclinical Biomarkers in Scrapie. Int J Mol Sci 2022; 23:7182. [PMID: 35806183 PMCID: PMC9266981 DOI: 10.3390/ijms23137182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/19/2022] [Accepted: 06/27/2022] [Indexed: 01/27/2023] Open
Abstract
Prion diseases are diagnosed in the symptomatic stage, when the neuronal damage is spread throughout the central nervous system (CNS). The assessment of biological features that allow the detection of asymptomatic cases is needed, and, in this context, scrapie, where pre-symptomatic infected animals can be detected through rectal biopsy, becomes a good study model. Neurogranin (Ng) and neurofilament light chain (NfL) are proteins that reflect synaptic and axonal damage and have been studied as cerebrospinal fluid (CSF) biomarkers in different neurodegenerative disorders. In this study, we evaluated Ng and NfL both at the protein and transcript levels in the CNS of preclinical and clinical scrapie-affected sheep compared with healthy controls and assessed their levels in ovine CSF. The correlation between these proteins and the main neuropathological events in prion diseases, PrPSc deposition and spongiosis, was also assessed. The results show a decrease in Ng and NfL at the protein and gene expression levels as the disease progresses, and significant changes between the control and preclinical animals. On the contrary, the CSF levels of NfL increased throughout the progression of the disease. Negative correlations between neuropathological markers of prion disease and the concentration of the studied proteins were also found. Although further research is needed, these results suggest that Ng and NfL could act as biomarkers for neurodegeneration onset and intensity in preclinical cases of scrapie.
Collapse
Affiliation(s)
- Marina Betancor
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragon, 50009 Zaragoza, Spain; (M.B.); (S.P.-L.); (B.M.); (I.M.-B.); (J.J.B.); (R.B.)
| | - Sonia Pérez-Lázaro
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragon, 50009 Zaragoza, Spain; (M.B.); (S.P.-L.); (B.M.); (I.M.-B.); (J.J.B.); (R.B.)
| | - Alicia Otero
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragon, 50009 Zaragoza, Spain; (M.B.); (S.P.-L.); (B.M.); (I.M.-B.); (J.J.B.); (R.B.)
| | - Belén Marín
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragon, 50009 Zaragoza, Spain; (M.B.); (S.P.-L.); (B.M.); (I.M.-B.); (J.J.B.); (R.B.)
| | - Inmaculada Martín-Burriel
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragon, 50009 Zaragoza, Spain; (M.B.); (S.P.-L.); (B.M.); (I.M.-B.); (J.J.B.); (R.B.)
- Laboratory of Biochemical Genetics (LAGENBIO), Faculty of Veterinary, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28220 Madrid, Spain
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Mölndal, Sweden; (K.B.); (H.Z.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
| | - Juan José Badiola
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragon, 50009 Zaragoza, Spain; (M.B.); (S.P.-L.); (B.M.); (I.M.-B.); (J.J.B.); (R.B.)
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Mölndal, Sweden; (K.B.); (H.Z.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Neurodegenerative Disease, University College LondonInstitute of Neurology, Queen Square, London WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Rosa Bolea
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragon, 50009 Zaragoza, Spain; (M.B.); (S.P.-L.); (B.M.); (I.M.-B.); (J.J.B.); (R.B.)
| |
Collapse
|
19
|
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: 23] [Impact Index Per Article: 7.7] [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
|
20
|
Lindner E, Woltsche N, Merle D, Steinwender G, Strohmaier H, Nairz M, Ivastinovic D. Prion Protein on Human Leukocytes Is Reduced in Iron Deficiency - Possible Implications for Age-related Macular Degeneration? Curr Eye Res 2020; 46:1178-1183. [PMID: 33317353 DOI: 10.1080/02713683.2020.1863432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
MATERIALS AND METHODS Patients presenting to the department of ophthalmology of the Medical University of Graz for reasons unrelated to prion diseases were enrolled. Parameters of iron metabolism, including ferritin and soluble transferrin receptor were measured by routine laboratory tests. Serum prion protein was determined by enzyme-linked immunosorbent assay. Surface prion protein on CD14+ monocytes and CD4+ T cells was analyzed by fluorescence activated cell sorting. RESULTS 95 patients were enrolled. Soluble transferrin receptor correlated significantly with prion protein levels on CD14+POM1+ monocytes (P = .001, r = -0.7) and on CD4+POM1+ T cells (P = .01, r = -0.62). CONCLUSION Our findings suggest a connection between the physiological function of the prion protein and iron metabolism in humans.
Collapse
Affiliation(s)
- Ewald Lindner
- Department of Ophthalmology, Medical University Graz, Graz, Austria
| | - Nora Woltsche
- Department of Ophthalmology, Medical University Graz, Graz, Austria
| | - David Merle
- Department of Ophthalmology, Medical University Graz, Graz, Austria
| | | | - Heimo Strohmaier
- Core Facility Imaging, Centre of Medical Research Graz, Graz, Austria
| | - Manfred Nairz
- Department of General Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
21
|
Kishimoto Y, Hirono M, Atarashi R, Sakaguchi S, Yoshioka T, Katamine S, Kirino Y. Impairment of cerebellar long-term depression and GABAergic transmission in prion protein deficient mice ectopically expressing PrPLP/Dpl. Sci Rep 2020; 10:15900. [PMID: 32985542 PMCID: PMC7522223 DOI: 10.1038/s41598-020-72753-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/27/2020] [Indexed: 12/22/2022] Open
Abstract
Prion protein (PrPC) knockout mice, named as the “Ngsk” strain (Ngsk Prnp0/0 mice), show late-onset cerebellar Purkinje cell (PC) degeneration because of ectopic overexpression of PrPC-like protein (PrPLP/Dpl). Our previous study indicated that the mutant mice also exhibited alterations in cerebellum-dependent delay eyeblink conditioning, even at a young age (16 weeks of age) when neurological changes had not occurred. Thus, this electrophysiological study was designed to examine the synaptic function of the cerebellar cortex in juvenile Ngsk Prnp0/0 mice. We showed that Ngsk Prnp0/0 mice exhibited normal paired-pulse facilitation but impaired long-term depression of excitatory synaptic transmission at synapses between parallel fibres and PCs. GABAA-mediated inhibitory postsynaptic currents recorded from PCs were also weakened in Ngsk Prnp0/0 mice. Furthermore, we confirmed that Ngsk Prnp0/0 mice (7–8-week-old) exhibited abnormalities in delay eyeblink conditioning. Our findings suggest that these alterations in both excitatory and inhibitory synaptic transmission to PCs caused deficits in delay eyeblink conditioning of Ngsk Prnp0/0 mice. Therefore, the Ngsk Prnp0/0 mouse model can contribute to study underlying mechanisms for impairments of synaptic transmission and neural plasticity, and cognitive deficits in the central nervous system.
Collapse
Affiliation(s)
- Yasushi Kishimoto
- Laboratory of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Kagawa, 769-2193, Japan.
| | - Moritoshi Hirono
- Department of Physiology, Faculty of Medicine, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan.
| | - Ryuichiro Atarashi
- Division of Microbiology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Suehiro Sakaguchi
- Division of Molecular Neurobiology, Institute for Enzyme Research (KOSOKEN), Tokushima University, Tokushima, 770-8501, Japan
| | - Tohru Yoshioka
- Center of Excellence for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Shigeru Katamine
- Center for International Collaborative Research, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Yutaka Kirino
- Laboratory of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Kagawa, 769-2193, Japan
| |
Collapse
|
22
|
Carvalho JF, Viana VS, Leon EP, Bonfa E, Pasoto SG, Martins VR. RETRACTED: Antibodies to cellular prion protein and its cognate ligand stress-inducible protein 1 in systemic lupus erythematosus. Lupus 2020; 29:NP1-NP7. [PMID: 32588733 DOI: 10.1177/0961203320935987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Jozélio F Carvalho
- Rheumatology Division, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Vilma St Viana
- Rheumatology Division, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Elaine P Leon
- Rheumatology Division, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Eloisa Bonfa
- Rheumatology Division, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sandra G Pasoto
- Rheumatology Division, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Vilma R Martins
- Ludwig Institute for Cancer Research, Hospital Alemão Oswaldo Cruz, São Paulo, Brazil
| |
Collapse
|
23
|
Corbett GT, Wang Z, Hong W, Colom-Cadena M, Rose J, Liao M, Asfaw A, Hall TC, Ding L, DeSousa A, Frosch MP, Collinge J, Harris DA, Perkinton MS, Spires-Jones TL, Young-Pearse TL, Billinton A, Walsh DM. PrP is a central player in toxicity mediated by soluble aggregates of neurodegeneration-causing proteins. Acta Neuropathol 2020; 139:503-526. [PMID: 31853635 PMCID: PMC7035229 DOI: 10.1007/s00401-019-02114-9] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 12/30/2022]
Abstract
Neurodegenerative diseases are an enormous public health problem, affecting tens of millions of people worldwide. Nearly all of these diseases are characterized by oligomerization and fibrillization of neuronal proteins, and there is great interest in therapeutic targeting of these aggregates. Here, we show that soluble aggregates of α-synuclein and tau bind to plate-immobilized PrP in vitro and on mouse cortical neurons, and that this binding requires at least one of the same N-terminal sites at which soluble Aβ aggregates bind. Moreover, soluble aggregates of tau, α-synuclein and Aβ cause both functional (impairment of LTP) and structural (neuritic dystrophy) compromise and these deficits are absent when PrP is ablated, knocked-down, or when neurons are pre-treated with anti-PrP blocking antibodies. Using an all-human experimental paradigm involving: (1) isogenic iPSC-derived neurons expressing or lacking PRNP, and (2) aqueous extracts from brains of individuals who died with Alzheimer's disease, dementia with Lewy bodies, and Pick's disease, we demonstrate that Aβ, α-synuclein and tau are toxic to neurons in a manner that requires PrPC. These results indicate that PrP is likely to play an important role in a variety of late-life neurodegenerative diseases and that therapeutic targeting of PrP, rather than individual disease proteins, may have more benefit for conditions which involve the aggregation of more than one protein.
Collapse
Affiliation(s)
- Grant T Corbett
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Zemin Wang
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Wei Hong
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Marti Colom-Cadena
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, University of Edinburgh, Edinburgh, EH89JZ, UK
| | - Jamie Rose
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, University of Edinburgh, Edinburgh, EH89JZ, UK
| | - Meichen Liao
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Adhana Asfaw
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Tia C Hall
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Lai Ding
- Program for Interdisciplinary Neuroscience, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Alexandra DeSousa
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Matthew P Frosch
- Massachusetts General Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - John Collinge
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | | | - Tara L Spires-Jones
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, University of Edinburgh, Edinburgh, EH89JZ, UK
| | - Tracy L Young-Pearse
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Andrew Billinton
- Neuroscience, IMED Biotechnology Unit, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
EFFECT OF ORAL COPPER SUPPLEMENTATION ON SUSCEPTIBILITY IN WHITE-TAILED DEER ( ODOCOILEUS VIRGINIANUS) TO CHRONIC WASTING DISEASE. J Wildl Dis 2020; 56:568-575. [PMID: 32073993 DOI: 10.7589/2019-10-260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Chronic wasting disease (CWD) is an infectious disease, but reported associations suggest several metals-especially copper (Cu) and manganese-potentially play a role in this and other prion diseases. To assess the utility of dietary Cu supplementation in protecting white-tailed deer (Odocoileus virginianus) from CWD, we compared incidence and disease course among individuals naturally exposed to CWD while being maintained on sustained-release Cu boluses or unsupplemented (control). Oral Cu supplementation increased liver tissue Cu concentrations compared to controls but did not affect susceptibility to CWD or survival after natural exposure in the captive white-tailed deer we studied. Over the 27 mo study, 89% (8/9) of the Cu-supplemented deer and 86% (6/7) of control deer became CWD-infected. Survival to 27 mo postexposure did not differ between Cu-supplemented and control deer: model-averaged survival probabilities to 27 mo were 0.45-0.47 for all combinations of Cu treatment and PRNP gene haplotype presence. The PRNP gene haplotype influenced the probability of deer remaining biopsy negative for at least 17 mo but did not affect overall susceptibility.
Collapse
|
25
|
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
|
26
|
De Mario A, Peggion C, Massimino ML, Norante RP, Zulian A, Bertoli A, Sorgato MC. The Link of the Prion Protein with Ca 2+ Metabolism and ROS Production, and the Possible Implication in Aβ Toxicity. Int J Mol Sci 2019; 20:ijms20184640. [PMID: 31546771 PMCID: PMC6770541 DOI: 10.3390/ijms20184640] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 01/05/2023] Open
Abstract
The cellular prion protein (PrPC) is an ubiquitous cell surface protein mostly expressed in neurons, where it localizes to both pre- and post-synaptic membranes. PrPC aberrant conformers are the major components of mammalian prions, the infectious agents responsible for incurable neurodegenerative disorders. PrPC was also proposed to bind aggregated misfolded proteins/peptides, and to mediate their neurotoxic signal. In spite of long-lasting research, a general consensus on the precise pathophysiologic mechanisms of PrPC has not yet been reached. Here we review our recent data, obtained by comparing primary neurons from PrP-expressing and PrP-knockout mice, indicating a central role of PrPC in synaptic transmission and Ca2+ homeostasis. Indeed, by controlling gene expression and signaling cascades, PrPC is able to optimize glutamate secretion and regulate Ca2+ entry via store-operated channels and ionotropic glutamate receptors, thereby protecting neurons from threatening Ca2+ overloads and excitotoxicity. We will also illustrate and discuss past and unpublished results demonstrating that Aβ oligomers perturb Ca2+ homeostasis and cause abnormal mitochondrial accumulation of reactive oxygen species by possibly affecting the PrP-dependent downregulation of Fyn kinase activity.
Collapse
Affiliation(s)
- Agnese De Mario
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| | - Caterina Peggion
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| | - Maria Lina Massimino
- CNR Neuroscience Institute, Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| | - Rosa Pia Norante
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| | - Alessandra Zulian
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| | - Alessandro Bertoli
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
- CNR Neuroscience Institute, Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
- Padova Neuroscience Center, University of Padova, 35131 Padova, Italy.
| | - Maria Catia Sorgato
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
- CNR Neuroscience Institute, Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
27
|
Lin SC, Lin CH, Shih NC, Liu HL, Wang WC, Lin KY, Liu ZY, Tseng YJ, Chang HK, Lin YC, Yeh YC, Minato H, Fujii T, Wu YC, Chen MY, Chou TY. Cellular prion protein transcriptionally regulated by NFIL3 enhances lung cancer cell lamellipodium formation and migration through JNK signaling. Oncogene 2019; 39:385-398. [PMID: 31477838 DOI: 10.1038/s41388-019-0994-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/10/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022]
Abstract
Tumor invasion and metastasis are the major causes of treatment failure and mortality in lung cancer patients. In this study, we identified a group of genes with differential expression in in situ and invasive lung adenocarcinoma tissues by expression profiling; among these genes we further characterized the association of the upregulation of PRNP, the gene encoding cellular Prion protein (PrPc), with lung adenocarcinoma invasiveness. Immunohistochemistry on clinical specimens showed an association of PrPc expression with invasive but not in situ lung adenocarcinoma. Consistently, the expression of PrPc was higher in the highly invasive than in the lowly invasive lung adenocarcinoma cell lines. Knockdown of PrPc expression in cultured lung adenocarcinoma cells decreased their lamellipodium formation, in vitro migration and invasion, and in vivo experimental lung metastasis. Phosphorylation of JNKs was found to correlate with PrPc expression and the inhibition of JNKs suppressed the PrPc-induced up-regulation of lamellipodium formation, cell migration, and invasion. Moreover, we identified the nuclear factor, interleukin 3 regulated (NFIL3) protein as a transcriptional activator of the PRNP promoter. Accordingly, NFIL3 promoted lung cancer cell migration and invasion in a PrPc-dependent manner. High NFIL3 expression in clinical specimens of lung adenocarcinoma was also associated with tumor invasiveness. Overall, our observations suggest that the NFIL3/PrPc axis, through regulating lamellipodium formation and cell mobility via JNK signaling, plays a critical role in lung cancer invasiveness and metastasis.
Collapse
Affiliation(s)
- Shin-Chih Lin
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, 11221, Taiwan.,Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Chia-Hung Lin
- Division of Molecular Pathology, Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, 11221, Taiwan
| | - Nien-Chu Shih
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Hsin-Ling Liu
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Wen-Chao Wang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Kun-Yang Lin
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Zih-Yu Liu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Yu-Jhen Tseng
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Hsueh-Kai Chang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Yi-Cheng Lin
- Division of Molecular Pathology, Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, 11221, Taiwan
| | - Yi-Chen Yeh
- Division of Molecular Pathology, Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, 11221, Taiwan
| | - Hiroshi Minato
- Department of Pathology and Laboratory Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Takeshi Fujii
- Department of Pathology, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Yu-Chung Wu
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, 11221, Taiwan
| | - Mei-Yu Chen
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, 11221, Taiwan. .,Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, 11221, Taiwan.
| | - Teh-Ying Chou
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, 11221, Taiwan. .,Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, 11221, Taiwan. .,Division of Molecular Pathology, Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, 11221, Taiwan. .,Institute of Clinical Medicine, National Yang-Ming University, Taipei, 11221, Taiwan.
| |
Collapse
|
28
|
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
|
29
|
Nguyen XTA, Tran TH, Cojoc D, Legname G. Copper Binding Regulates Cellular Prion Protein Function. Mol Neurobiol 2019; 56:6121-6133. [PMID: 30729399 DOI: 10.1007/s12035-019-1510-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/24/2019] [Indexed: 01/27/2023]
Abstract
The cellular prion protein (PrPC), mainly known for its role in neurodegenerative diseases, is involved in several physiological processes including neuritogenesis. In addition, its ability to bind copper or zinc has been suggested for its role in metal homeostasis. Although PrPC has been known as a copper-binding molecule, little is known about how copper can affect PrPC physiological functions. By combining genomic approaches, cellular assays, and focal stimulation technique, we found that PrPC neuritogenesis function is directly influenced by N-terminal copper-binding amino acids. Several recombinant mouse PrP (recMoPrP) mutants at N-terminal copper-binding sites were produced, and primary hippocampal cultures were treated either in bulk or exposed near the hippocampal growth cones (GC) of single neurons in local stimulation manner. While focal stimulation of GC with wild-type recMoPrP induced neurite outgrowth and rapid GC turning toward the source, N-terminal mutants fail to support this effect. Indeed, disrupting all the copper-binding sites at the N-terminus of PrPC was toxic to neurons indicating that these regions are crucial for the protein function. Mutants at both octarepeat and non-octarepeat region abolished the neuritogenesis effect. Altogether, our findings indicate the crucial role of copper-binding sites in maintaining the neuritogenesis function in PrP, suggesting a potential link between loss-of-function of the protein and disease initiation.
Collapse
Affiliation(s)
- Xuan T A Nguyen
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Thanh Hoa Tran
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Dan Cojoc
- Optical Manipulation (OM)-Lab, Institute of Materials (IOM), Consiglio Nazionale delle Ricerche (CNR), Trieste, Italy.
| | - Giuseppe Legname
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy.
| |
Collapse
|
30
|
Markham KA, Roseman GP, Linsley RB, Lee HW, Millhauser GL. Molecular Features of the Zn 2+ Binding Site in the Prion Protein Probed by 113Cd NMR. Biophys J 2019; 116:610-620. [PMID: 30678993 DOI: 10.1016/j.bpj.2019.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 10/27/2022] Open
Abstract
The cellular prion protein (PrPC) is a zinc-binding protein that contributes to the regulation of Zn2+ and other divalent species of the central nervous system. Zn2+ coordinates to the flexible, N-terminal repeat region of PrPC and drives a tertiary contact between this repeat region and a well-defined cleft of the C-terminal domain. The tertiary structure promoted by Zn2+ is thought to regulate inherent PrPC toxicity. Despite the emerging consensus regarding the interaction between Zn2+ and PrPC, there is little direct spectroscopic confirmation of the metal ion's coordination details. Here, we address this conceptual gap by using Cd2+ as a surrogate for Zn2+. NMR finds that Cd2+ binds exclusively to the His imidazole side chains of the repeat segment, with a dissociation constant of ∼1.2 mM, and promotes an N-terminal-C-terminal cis interaction very similar to that observed with Zn2+. Analysis of 113Cd NMR spectra of PrPC, along with relevant control proteins and peptides, suggests that coordination of Cd2+ in the full-length protein is consistent with a three- or four-His geometry. Examination of the mutation E199K in mouse PrPC (E200K in humans), responsible for inherited Creutzfeldt-Jakob disease, finds that the mutation lowers metal ion affinity and weakens the cis interaction. These findings not only provide deeper insight into PrPC metal ion coordination but they also suggest new perspectives on the role of familial mutations in prion disease.
Collapse
Affiliation(s)
- Kate A Markham
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, California
| | - Graham P Roseman
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, California
| | - Richard B Linsley
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, California
| | - Hsiau-Wei Lee
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, California
| | - Glenn L Millhauser
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, California.
| |
Collapse
|
31
|
Orrù CD, Soldau K, Cordano C, Llibre-Guerra J, Green AJ, Sanchez H, Groveman BR, Edland SD, Safar JG, Lin JH, Caughey B, Geschwind MD, Sigurdson CJ. Prion Seeds Distribute throughout the Eyes of Sporadic Creutzfeldt-Jakob Disease Patients. mBio 2018; 9:e02095-18. [PMID: 30459197 PMCID: PMC6247090 DOI: 10.1128/mbio.02095-18] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022] Open
Abstract
Sporadic Creutzfeldt-Jakob disease (sCJD) is the most common prion disease in humans and has been iatrogenically transmitted through corneal graft transplantation. Approximately 40% of sCJD patients develop visual or oculomotor symptoms and may seek ophthalmological consultation. Here we used the highly sensitive real-time quaking-induced conversion (RT-QuIC) assay to measure postmortem prion seeding activities in cornea, lens, ocular fluid, retina, choroid, sclera, optic nerve, and extraocular muscle in the largest series of sCJD patient eyes studied by any assay to date. We detected prion seeding activity in 100% of sCJD eyes, representing three common sCJD subtypes, with levels varying by up to 4 log-fold among individuals. The retina consistently showed the highest seed levels, which in some cases were only slightly lower than brain. Within the retina, prion deposits were detected by immunohistochemistry (IHC) in the retinal outer plexiform layer in most sCJD cases, and in some eyes the inner plexiform layer, consistent with synaptic prion deposition. Prions were not detected by IHC in any other eye region. With RT-QuIC, prion seed levels generally declined in eye tissues with increased distance from the brain, and yet all corneas had prion seeds detectable. Prion seeds were also present in the optic nerve, extraocular muscle, choroid, lens, vitreous, and sclera. Collectively, these results reveal that sCJD patients accumulate prion seeds throughout the eye, indicating the potential diagnostic utility as well as a possible biohazard.IMPORTANCE Cases of iatrogenic prion disease have been reported from corneal transplants, yet the distribution and levels of prions throughout the eye remain unknown. This study probes the occurrence, level, and distribution of prions in the eyes of patients with sporadic Creutzfeldt-Jakob disease (sCJD). We tested the largest series of prion-infected eyes reported to date using an ultrasensitive technique to establish the prion seed levels in eight regions of the eye. All 11 cases had detectable prion seeds in the eye, and in some cases, the seed levels in the retina approached those in brain. In most cases, prion deposits could also be seen by immunohistochemical staining of retinal tissue; other ocular tissues were negative. Our results have implications for estimating the risk for iatrogenic transmission of sCJD as well as for the development of antemortem diagnostic tests for prion diseases.
Collapse
Affiliation(s)
- Christina D Orrù
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, Montana, USA
| | - Katrin Soldau
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Christian Cordano
- Department of Neurology, Multiple Sclerosis Center, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Jorge Llibre-Guerra
- Cognitive and Behavioral Research Unit, National Institute of Neurology, Havana, Cuba
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Ari J Green
- Department of Neurology, Multiple Sclerosis Center, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Henry Sanchez
- Department of Pathology, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Bradley R Groveman
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, Montana, USA
| | - Steven D Edland
- Department of Family Medicine & Public Health, University of California, San Diego, La Jolla, California, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Jiri G Safar
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neurology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jonathan H Lin
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Byron Caughey
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, Montana, USA
| | - Michael D Geschwind
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Christina J Sigurdson
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
- Department of Pathology, Immunology, and Microbiology, University of California, Davis, Davis, California, USA
| |
Collapse
|
32
|
Brás IC, Lopes LV, Outeiro TF. Sensing α-Synuclein From the Outside via the Prion Protein: Implications for Neurodegeneration. Mov Disord 2018; 33:1675-1684. [DOI: 10.1002/mds.27478] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/30/2022] Open
Affiliation(s)
- Inês Caldeira Brás
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration; University Medical Center Göttingen; Göttingen Germany
| | - Luísa V. Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina; Universidade de Lisboa; Lisboa Portugal
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration; University Medical Center Göttingen; Göttingen Germany
- CEDOC, Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciências Médicas; Universidade Nova de Lisboa, Campo dos Mártires da Pátria; Lisboa Portugal
- Max Planck Institute for Experimental Medicine; Göttingen Germany
- Institute of Neuroscience, The Medical School; Newcastle University; Newcastle Upon Tyne UK
| |
Collapse
|
33
|
Abstract
Sporadic Creutzfeldt-Jakob disease (CJD), the most common human prion disease, is generally regarded as a spontaneous neurodegenerative illness, arising either from a spontaneous PRNP somatic mutation or a stochastic PrP structural change. Alternatively, the possibility of an infection from animals or other source remains to be completely ruled out. Sporadic CJD is clinically characterized by rapidly progressive dementia with ataxia, myoclonus, or other neurologic signs and, neuropathologically, by the presence of aggregates of abnormal prion protein, spongiform change, neuronal loss, and gliosis. Despite these common features the disease shows a wide phenotypic variability which was recognized since its early descriptions. In the late 1990s the identification of key molecular determinants of phenotypic expression and the availability of a large series of neuropathologically verified cases led to the characterization of definite clinicopathologic and molecular disease subtypes and to an internationally recognized disease classification. By showing that these disease subtypes correspond to specific agent strain-host genotype combinations, recent transmission studies have confirmed the biologic basis of this classification. The introduction of brain magnetic resonance imaging techniques such as fluid-attenuated inversion recovery and diffusion-weighted imaging sequences and cerebrospinal fluid biomarker assays for the detection of brain-derived proteins as well as real-time quaking-induced conversion assay, allowing the specific detection of prions in accessible biologic fluids and tissues, has significantly contributed to the improved accuracy of the clinical diagnosis of sporadic CJD in recent years.
Collapse
Affiliation(s)
- Inga Zerr
- Department of Neurology, University Hospital, Georg-August-University, Goettingen, Germany.
| | - Piero Parchi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna and IRCCS Institute of Neurological Sciences, Bologna, Italy
| |
Collapse
|
34
|
The Prion Protein Regulates Synaptic Transmission by Controlling the Expression of Proteins Key to Synaptic Vesicle Recycling and Exocytosis. Mol Neurobiol 2018; 56:3420-3436. [PMID: 30128651 DOI: 10.1007/s12035-018-1293-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/01/2018] [Indexed: 12/16/2022]
Abstract
The cellular prion protein (PrPC), whose misfolded conformers are implicated in prion diseases, localizes to both the presynaptic membrane and postsynaptic density. To explore possible molecular contributions of PrPC to synaptic transmission, we utilized a mass spectrometry approach to quantify the release of glutamate from primary cerebellar granule neurons (CGN) expressing, or deprived of (PrP-KO), PrPC, following a depolarizing stimulus. Under the same conditions, we also tracked recycling of synaptic vesicles (SVs) in the two neuronal populations. We found that in PrP-KO CGN these processes decreased by 40 and 60%, respectively, compared to PrPC-expressing neurons. Unbiased quantitative mass spectrometry was then employed to compare the whole proteome of CGN with the two PrP genotypes. This approach allowed us to assess that, relative to the PrPC-expressing counterpart, the absence of PrPC modified the protein expression profile, including diminution of some components of SV recycling and fusion machinery. Subsequent quantitative RT-PCR closely reproduced proteomic data, indicating that PrPC is committed to ensuring optimal synaptic transmission by regulating genes involved in SV dynamics and neurotransmitter release. These novel molecular and cellular aspects of PrPC add insight into the underlying mechanisms for synaptic dysfunctions occurring in neurodegenerative disorders in which a compromised PrPC is likely to intervene.
Collapse
|
35
|
Prion acute synaptotoxicity is largely driven by protease-resistant PrPSc species. PLoS Pathog 2018; 14:e1007214. [PMID: 30089152 PMCID: PMC6101418 DOI: 10.1371/journal.ppat.1007214] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/20/2018] [Accepted: 07/12/2018] [Indexed: 01/09/2023] Open
Abstract
Although misfolding of normal prion protein (PrPC) into abnormal conformers (PrPSc) is critical for prion disease pathogenesis our current understanding of the underlying molecular pathophysiology is rudimentary. Exploiting an electrophysiology paradigm, herein we report that at least modestly proteinase K (PK)-resistant PrPSc (PrPres) species are acutely synaptotoxic. Brief exposure to ex vivo PrPSc from two mouse-adapted prion strains (M1000 and MU02) prepared as crude brain homogenates (cM1000 and cMU02) and cell lysates from chronically M1000-infected RK13 cells (MoRK13-Inf) caused significant impairment of hippocampal CA1 region long-term potentiation (LTP), with the LTP disruption approximating that reported during the evolution of murine prion disease. Proof of PrPSc (especially PrPres) species as the synaptotoxic agent was demonstrated by: significant rescue of LTP following selective immuno-depletion of total PrP from cM1000 (dM1000); modestly PK-treated cM1000 (PK+M1000) retaining full synaptotoxicity; and restoration of the LTP impairment when employing reconstituted, PK-eluted, immuno-precipitated M1000 preparations (PK+IP-M1000). Additional detailed electrophysiological analyses exemplified by impairment of post-tetanic potentiation (PTP) suggest possible heightened pre-synaptic vulnerability to the acute synaptotoxicity. This dysfunction correlated with cumulative insufficiency of replenishment of the readily releasable pool (RRP) of vesicles during repeated high-frequency stimulation utilised for induction of LTP. Broadly comparable results with LTP and PTP impairment were obtained utilizing hippocampal slices from PrPC knockout (PrPo/o) mice, with cM1000 serial dilution assessments revealing similar sensitivity of PrPo/o and wild type (WT) slices. Size fractionation chromatography demonstrated that synaptotoxic PrP correlated with PK-resistant species >100kDa, consistent with multimeric PrPSc, with levels of these species >6 ng/ml appearing sufficient to induce synaptic dysfunction. Biochemical analyses of hippocampal slices manifesting acute synaptotoxicity demonstrated reduced levels of multiple key synaptic proteins, albeit with noteworthy differences in PrPo/o slices, while such changes were absent in hippocampi demonstrating rescued LTP through treatment with dM1000. Our findings offer important new mechanistic insights into the synaptic impairment underlying prion disease, enhancing prospects for development of targeted effective therapies. Misfolding of the normal prion protein (PrPC) into disease-associated conformations (PrPSc) is the critical initiating step for prion diseases. Similar to other neurodegenerative disorders, progressive failure of brain synapses is considered a primary deleterious event underpinning prion disease evolution. Our current understanding of the underlying mechanisms associated with synaptic failure is rudimentary contributing to difficulties in developing effective treatments. Herein we report the use of an electrophysiology paradigm that allowed us to demonstrate that at least modestly proteinase K (PK)-resistant PrPSc species from two mouse-adapted prion strains (M1000 and MU02) are directly synaptotoxic causing significant acute impairment of hippocampal CA1 region long-term potentiation (LTP). Of note, the LTP disruption approximated that reported in prion animal models. Additional detailed analyses provided novel pathophysiological insights suggesting possible heightened pre-synaptic vulnerability to the acute synaptotoxicity through impairment of replenishment of the readily releasable pool of neurotransmitter vesicles, while biochemical analyses demonstrated reduced levels of multiple key pre-and post-synaptic proteins. Broadly similar acute synaptic dysfunction and dose-response susceptibility were observed in slices from mice not expressing PrPC albeit with minor but noteworthy differences in electrophysiological and biochemical findings. Our study offers important new mechanistic insights into the synaptic impairment underlying prion disease, enhancing prospects for development effective therapies.
Collapse
|
36
|
Younan ND, Chen KF, Rose RS, Crowther DC, Viles JH. Prion protein stabilizes amyloid-β (Aβ) oligomers and enhances Aβ neurotoxicity in a Drosophila model of Alzheimer's disease. J Biol Chem 2018; 293:13090-13099. [PMID: 29887525 DOI: 10.1074/jbc.ra118.003319] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/05/2018] [Indexed: 12/16/2022] Open
Abstract
The cellular prion protein (PrPC) can act as a cell-surface receptor for β-amyloid (Aβ) peptide; however, a role for PrPC in the pathogenesis of Alzheimer's disease (AD) is contested. Here, we expressed a range of Aβ isoforms and PrPC in the Drosophila brain. We found that co-expression of Aβ and PrPC significantly reduces the lifespan, disrupts circadian rhythms, and increases Aβ deposition in the fly brain. In contrast, under the same conditions, expression of Aβ or PrPC individually did not lead to these phenotypic changes. In vitro studies revealed that substoichiometric amounts of PrPC trap Aβ as oligomeric assemblies and fragment-preformed Aβ fibers. The ability of membrane-anchored PrPC to trap Aβ as cytotoxic oligomers at the membrane surface and fragment inert Aβ fibers suggests a mechanism by which PrPC exacerbates Aβ deposition and pathogenic phenotypes in the fly, supporting a role for PrPC in AD. This study provides a second animal model linking PrPC expression with Aβ toxicity and supports a role for PrPC in AD pathogenesis. Blocking the interaction of Aβ and PrPC represents a potential therapeutic strategy.
Collapse
Affiliation(s)
- Nadine D Younan
- From the School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom
| | - Ko-Fan Chen
- the Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London WC1N 3BG, United Kingdom, and
| | - Ruth-Sarah Rose
- From the School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom
| | - Damian C Crowther
- the Neuroscience IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, United Kingdom
| | - John H Viles
- From the School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom,
| |
Collapse
|
37
|
Osborne C, West E, Bate C. The phospholipase A 2 pathway controls a synaptic cholesterol ester cycle and synapse damage. J Cell Sci 2018; 131:jcs.211789. [PMID: 29588394 DOI: 10.1242/jcs.211789] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 03/19/2018] [Indexed: 11/20/2022] Open
Abstract
The cellular prion protein (PrPC) acts as a scaffold protein that organises signalling complexes. In synaptosomes, the aggregation of PrPC by amyloid-β (Aβ) oligomers attracts and activates cytoplasmic phospholipase A2 (cPLA2), leading to synapse degeneration. The signalling platform is dependent on cholesterol released from cholesterol esters by cholesterol ester hydrolases (CEHs). The activation of cPLA2 requires cholesterol released from cholesterol esters by cholesterol ester hydrolases (CEHs), enzymes dependent upon platelet activating factor (PAF) released by activated cPLA2 This demonstrates a positive feedback system in which activated cPLA2 increased cholesterol concentrations, which in turn facilitated cPLA2 activation. PAF was also required for the incorporation of the tyrosine kinase Fyn and cyclooxygenase (COX)-2 into Aβ-PrPC-cPLA2 complexes. As a failure to deactivate signalling complexes can lead to pathology, the mechanisms involved in their dispersal were studied. PAF facilitated the incorporation of acyl-coenzyme A:cholesterol acyltransferase (ACAT)-1 into Aβ-PrPC-cPLA2-COX-2-Fyn complexes. The esterification of cholesterol reduced cholesterol concentrations, causing dispersal of Aβ-PrPC-cPLA2-COX-2-Fyn complexes and the cessation of signalling. This study identifies PAF as a key mediator regulating the cholesterol ester cycle, activation of cPLA2 and COX-2 within synapses, and synapse damage.
Collapse
Affiliation(s)
- Craig Osborne
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, UK AL9 7TA
| | - Ewan West
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, UK AL9 7TA
| | - Clive Bate
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, UK AL9 7TA
| |
Collapse
|
38
|
Abstract
Several studies have indicated that certain misfolded amyloids composed of tau, β-amyloid or α-synuclein can be transferred from cell to cell, suggesting the contribution of mechanisms reminiscent of those by which infective prions spread through the brain. This process of a 'prion-like' spreading between cells is also relevant as a novel putative therapeutic target that could block the spreading of proteinaceous aggregates throughout the brain which may underlie the progressive nature of neurodegenerative diseases. The relevance of β-amyloid oligomers and cellular prion protein (PrPC) binding has been a focus of interest in Alzheimer's disease (AD). At the molecular level, β-amyloid/PrPC interaction takes place in two differently charged clusters of PrPC. In addition to β-amyloid, participation of PrPC in α-synuclein binding and brain spreading also appears to be relevant in α-synucleopathies. This review summarizes current knowledge about PrPC as a putative receptor for amyloid proteins and the physiological consequences of these interactions.
Collapse
Affiliation(s)
- José A Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain; Institute of Neuroscience, University of Barcelona, Barcelona, Spain.
| | - Isidre Ferrer
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain; Institute of Neuroscience, University of Barcelona, Barcelona, Spain; Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain; Senior Consultant Neuropathology, Service of Pathology, Bellvitge University Hospital, Hospitalet de Llobregat, Spain.
| | - Rosalina Gavín
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain; Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| |
Collapse
|
39
|
Kovač V, Čurin Šerbec V. Prion Proteins Without the Glycophosphatidylinositol Anchor: Potential Biomarkers in Neurodegenerative Diseases. Biomark Insights 2018; 13:1177271918756648. [PMID: 29449775 PMCID: PMC5808966 DOI: 10.1177/1177271918756648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/23/2017] [Indexed: 01/17/2023] Open
Abstract
Prion protein (PrP) is a biomolecule that is involved in neuronal signaling, myelinization, and the development of neurodegenerative diseases. In the cell, PrP is shed by the ADAM10 protease. This process generates PrP molecules that lack glycophosphatidylinositol anchor, and these molecules incorporate into toxic aggregates and neutralize toxic oligomers. Due to this dual role, these molecules are important biomarkers for neurodegenerative diseases. In this review, we present shed PrP as a potential biomarker, with a focus on PrP226*, which may be the main biomarker for predicting neurodegenerative diseases in humans.
Collapse
Affiliation(s)
- Valerija Kovač
- Department for the Production of Diagnostic Reagents and Research, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Vladka Čurin Šerbec
- Department for the Production of Diagnostic Reagents and Research, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| |
Collapse
|
40
|
Bate C, Williams A. Monomeric amyloid-β reduced amyloid-β oligomer-induced synapse damage in neuronal cultures. Neurobiol Dis 2017; 111:48-58. [PMID: 29272738 DOI: 10.1016/j.nbd.2017.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 11/12/2017] [Accepted: 12/12/2017] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disease characterized by the accumulation of amyloid-β (Aβ) in the brain. Aβ oligomers are believed to cause synapse damage resulting in the memory deficits that are characteristic of this disease. Since the loss of synaptic proteins in the brain correlates closely with the degree of dementia in Alzheimer's disease, the process of Aβ-induced synapse damage was investigated in cultured neurons by measuring the loss of synaptic proteins. Soluble Aβ oligomers, derived from Alzheimer's-affected brains, caused the loss of cysteine string protein and synaptophysin from neurons. When applied to synaptosomes Aβ oligomers increased cholesterol concentrations and caused aberrant activation of cytoplasmic phospholipase A2 (cPLA2). In contrast, Aβ monomer preparations did not affect cholesterol concentrations or activate synaptic cPLA2, nor did they damage synapses. The Aβ oligomer-induced aggregation of cellular prion proteins (PrPC) at synapses triggered the activation of cPLA2 that leads to synapse degeneration. Critically, Aβ monomer preparations did not cause the aggregation of PrPC; rather they reduced the Aβ oligomer-induced aggregation of PrPC. The presence of Aβ monomer preparations also inhibited the Aβ oligomer-induced increase in cholesterol concentrations and activation of cPLA2 in synaptosomes and protected neurons against the Aβ oligomer-induced synapse damage. These results support the hypothesis that Aβ monomers are neuroprotective. We hypothesise that synapse damage may result from a pathological Aβ monomer:oligomer ratio rather than the total concentrations of Aβ within the brain.
Collapse
Affiliation(s)
- Clive Bate
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Herts AL9 7TA, UK.
| | - Alun Williams
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK.
| |
Collapse
|
41
|
Wheeler LC, Harms MJ. Human S100A5 binds Ca 2+ and Cu 2+ independently. BMC BIOPHYSICS 2017; 10:8. [PMID: 29201357 PMCID: PMC5700546 DOI: 10.1186/s13628-017-0040-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/08/2017] [Indexed: 12/21/2022]
Abstract
Background S100A5 is a calcium binding protein found in a small subset of amniote tissues. Little is known about the biological roles of S100A5, but it may be involved in inflammation and olfactory signaling. Previous work indicated that S100A5 displays antagonism between binding of Ca2+ and Cu2+ ions-one of the most commonly cited features of the protein. We set out to characterize the interplay between Ca2+ and Cu2+ binding by S100A5 using isothermal titration calorimetry (ITC), circular dichroism spectroscopy (CD), and analytical ultracentrifugation (AUC). Results We found that human S100A5 is capable of binding both Cu2+ and Ca2+ ions simultaneously. The wildtype protein was extremely aggregation-prone in the presence of Cu2+ and Ca2+. A Cys-free version of S100A5, however, was not prone to precipitation or oligomerization. Mutation of the cysteines does not disrupt the binding of either Ca2+ or Cu2+ to S100A5. In the Cys-free background, we measured Ca2+ and Cu2+ binding in the presence and absence of the other metal using ITC. Saturating concentrations of Ca2+ or Cu2+ do not disrupt the binding of one another. Ca2+ and Cu2+ binding induce structural changes in S100A5, which are measurable using CD spectroscopy. We show via sedimentation velocity AUC that the wildtype protein is prone to the formation of soluble oligomers, which are not present in Cys-free samples. Conclusions S100A5 can bind Ca2+ and Cu2+ ions simultaneously and independently. This observation is in direct contrast to previously-reported antagonism between binding of Cu2+ and Ca2+ ions. The previous result is likely due to metal-dependent aggregation. Little is known about the biology of S100A5, so an accurate understanding of the biochemistry is necessary to make informed biological hypotheses. Our observations suggest the possibility of independent biological functions for Cu2+ and Ca2+ binding by S100A5.
Collapse
Affiliation(s)
- Lucas C Wheeler
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, 97403 OR USA.,Insitute of Molecular Biology, University of Oregon, Eugene, 97403 OR USA
| | - Michael J Harms
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, 97403 OR USA.,Insitute of Molecular Biology, University of Oregon, Eugene, 97403 OR USA
| |
Collapse
|
42
|
Beckman D, Linden R. A roadmap for investigating the role of the prion protein in depression associated with neurodegenerative disease. Prion 2017; 10:131-42. [PMID: 27057694 DOI: 10.1080/19336896.2016.1152437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The physiological properties of the native, endogenous prion protein (PrP(C)) is a matter of concern, due to its pleiotropic functions and links to neurodegenerative disorders and cancer. In line with our hypothesis that the basic function of PrP(C) is to serve as a cell surface scaffold for the assembly of signaling modules, multiple interactions have been identified of PrP(C) with signaling molecules, including neurotransmitter receptors. We recently reported evidence that PrP(C) may modulate monoaminergic neurotransmission, as well as depressive-like behavior in mice. Here, we discuss how those results, together with a number of other studies, including our previous demonstration that both inflammatory and behavioral stress modulate PrP(C) content in neutrophils, suggest a distributed role of PrP(C) in clinical depression and inflammation associated with neurodegenerative diseases. An overarching understanding of the multiple interventions of PrP(C) upon physiological events may both shed light on the pathogenesis of, as well as help the identification of novel therapeutic targets for clinical depression, Prion and Alzheimer's Diseases.
Collapse
Affiliation(s)
| | - Rafael Linden
- a Instituto de Biofísica da UFRJ, Rio de Janeiro , Brazil
| |
Collapse
|
43
|
Copper- and Zinc-Promoted Interdomain Structure in the Prion Protein: A Mechanism for Autoinhibition of the Neurotoxic N-Terminus. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:35-56. [PMID: 28838668 DOI: 10.1016/bs.pmbts.2017.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The function of the cellular prion protein (PrPC), while still poorly understood, is increasingly linked to its ability to bind physiological metal ions at the cell surface. PrPC binds divalent forms of both copper and zinc through its unstructured N-terminal domain, modulating interactions between PrPC and various receptors at the cell surface and ultimately tuning downstream cellular processes. In this chapter, we briefly discuss the molecular features of copper and zinc uptake by PrPC and summarize evidence implicating these metal ions in PrP-mediated physiology. We then focus our review on recent biophysical evidence revealing a physical interaction between the flexible N-terminal and globular C-terminal domains of PrPC. This interdomain cis interaction is electrostatic in nature and is promoted by the binding of Cu2+ and Zn2+ to the N-terminal octarepeat domain. These findings, along with recent cellular studies, suggest a mechanism whereby NC interactions serve to regulate the activity and/or toxicity of the PrPC N-terminus. We discuss this potential mechanism in relation to familial prion disease mutations, lethal deletions of the PrPC central region, and neurotoxicity induced by certain globular domain ligands, including bona fide prions and toxic amyloid-β oligomers.
Collapse
|
44
|
West E, Osborne C, Bate C. The cholesterol ester cycle regulates signalling complexes and synapse damage caused by amyloid-β. J Cell Sci 2017; 130:3050-3059. [PMID: 28760925 DOI: 10.1242/jcs.205484] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/26/2017] [Indexed: 02/01/2023] Open
Abstract
Cholesterol is required for the formation and function of some signalling platforms. In synaptosomes, amyloid-β (Aβ) oligomers, the causative agent in Alzheimer's disease, bind to cellular prion proteins (PrPC) resulting in increased cholesterol concentrations, translocation of cytoplasmic phospholipase A2 (cPLA2, also known as PLA2G4A) to lipid rafts, and activation of cPLA2 The formation of Aβ-PrPC complexes is controlled by the cholesterol ester cycle. In this study, Aβ activated cholesterol ester hydrolases, which released cholesterol from stores of cholesterol esters and stabilised Aβ-PrPC complexes, resulting in activated cPLA2 Conversely, cholesterol esterification reduced cholesterol concentrations causing the dispersal of Aβ-PrPC complexes. In cultured neurons, the cholesterol ester cycle regulated Aβ-induced synapse damage; cholesterol ester hydrolase inhibitors protected neurons, while inhibition of cholesterol esterification significantly increased Aβ-induced synapse damage. An understanding of the molecular mechanisms involved in the dispersal of signalling complexes is important as failure to deactivate signalling pathways can lead to pathology. This study demonstrates that esterification of cholesterol is a key factor in the dispersal of Aβ-induced signalling platforms involved in the activation of cPLA2 and synapse degeneration.
Collapse
Affiliation(s)
- Ewan West
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Herts, AL9 7TA, UK
| | - Craig Osborne
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Herts, AL9 7TA, UK
| | - Clive Bate
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Herts, AL9 7TA, UK
| |
Collapse
|
45
|
Sarnataro D, Pepe A, Zurzolo C. Cell Biology of Prion Protein. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:57-82. [PMID: 28838675 DOI: 10.1016/bs.pmbts.2017.06.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cellular prion protein (PrPC) is a mammalian glycoprotein which is usually found anchored to the plasma membrane via a glycosylphosphatidylinositol (GPI) anchor. The precise function of PrPC remains elusive but may depend upon its cellular localization. PrPC misfolds to a pathogenic isoform PrPSc, the causative agent of neurodegenerative prion diseases. Nonetheless some forms of prion disease develop in the apparent absence of infectious PrPSc, suggesting that molecular species of PrP distinct from PrPSc may represent the primary neurotoxic culprits. Indeed, in some inherited cases of human prion disease, the predominant form of PrP detectable in the brain is not PrPSc but rather CtmPrP, a transmembrane form of the protein. The relationship between the neurodegeneration occurring in prion diseases involving PrPSc and that associated with CtmPrP remains unclear. However, the different membrane topology of the PrP mutants, as well as the presence of the GPI anchor, could influence both the function and the intracellular localization and trafficking of the protein, all being potentially very important in the pathophysiological mechanism that ultimately causes the disease. Here, we review the latest findings on the fundamental aspects of prions biology, from the PrPC biosynthesis, function, and structure up to its intracellular traffic and analyze the possible roles of the different topological isoforms of the protein, as well as the GPI anchor, in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Daniela Sarnataro
- University of Naples "Federico II", Naples, Italy; Ceinge-Biotecnologie avanzate, s.c.a r.l., Naples, Italy.
| | - Anna Pepe
- University of Naples "Federico II", Naples, Italy; Unité de Trafic Membranaire et Pathogenese, Institut Pasteur, Paris, France
| | - Chiara Zurzolo
- University of Naples "Federico II", Naples, Italy; Unité de Trafic Membranaire et Pathogenese, Institut Pasteur, Paris, France
| |
Collapse
|
46
|
Hirsch TZ, Martin-Lannerée S, Mouillet-Richard S. Functions of the Prion Protein. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:1-34. [PMID: 28838656 DOI: 10.1016/bs.pmbts.2017.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although initially disregarded compared to prion pathogenesis, the functions exerted by the cellular prion protein PrPC have gained much interest over the past two decades. Research aiming at unraveling PrPC functions started to intensify when it became appreciated that it would give clues as to how it is subverted in the context of prion infection and, more recently, in the context of Alzheimer's disease. It must now be admitted that PrPC is implicated in an incredible variety of biological processes, including neuronal homeostasis, stem cell fate, protection against stress, or cell adhesion. It appears that these diverse roles can all be fulfilled through the involvement of PrPC in cell signaling events. Our aim here is to provide an overview of our current understanding of PrPC functions from the animal to the molecular scale and to highlight some of the remaining gaps that should be addressed in future research.
Collapse
Affiliation(s)
- Théo Z Hirsch
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Séverine Martin-Lannerée
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Sophie Mouillet-Richard
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France.
| |
Collapse
|
47
|
Senesi M, Lewis V, Kim JH, Adlard PA, Finkelstein DI, Collins SJ. In vivo prion models and the disconnection between transmissibility and neurotoxicity. Ageing Res Rev 2017; 36:156-164. [PMID: 28450269 DOI: 10.1016/j.arr.2017.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 02/03/2017] [Accepted: 03/17/2017] [Indexed: 02/01/2023]
Abstract
The primary causative event in the development of prion diseases is the misfolding of the normal prion protein (PrPC) into an ensemble of altered conformers (herein collectively denoted as PrPSc) that accumulate in the brain. Prominent amongst currently unresolved key aspects underpinning prion disease pathogenesis is whether transmission and toxicity are sub-served by different molecular species of PrPSc, which may directly impact on the development of effective targeted treatments. The use of murine models of prion disease has been of fundamental importance for probing the relationship between hypothesised "neurotoxic" and "transmissible" PrPSc and the associated kinetic profiles of their production during disease evolution, but unfortunately consensus has not been achieved. Recent in vivo studies have led to formulation of the "two-phase" hypothesis, which postulates that there is first an exponential increase in transmitting PrPSc species followed by an abrupt transition to propagation of neurotoxic PrPSc species. Such observations however, appear inconsistent with previous in vivo murine studies employing detailed time-course behavioural testing, wherein evidence of neurotoxicity could be detected early in disease progression. This review analyses the contributions of in vivo murine models attempting to provide insights into the relationship between transmitting and neurotoxic PrPSc species and explores possible refinements to the "two-phase hypothesis", that better accommodate the available historical and recent evidence.
Collapse
Affiliation(s)
- Matteo Senesi
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville 3010, Australia
| | - Victoria Lewis
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville 3010, Australia
| | - Jee H Kim
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville 3010, Australia
| | - Paul A Adlard
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville 3010, Australia
| | - David I Finkelstein
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville 3010, Australia
| | - Steven J Collins
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville 3010, Australia; The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
48
|
Insights into the mechanisms of copper dyshomeostasis in amyotrophic lateral sclerosis. Expert Rev Mol Med 2017; 19:e7. [PMID: 28597807 DOI: 10.1017/erm.2017.9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a severe neuromuscular disease characterised by a progressive loss of motor neurons that usually results in paralysis and death within 2 to 5 years after disease onset. The pathophysiological mechanisms involved in ALS remain largely unknown and to date there is no effective treatment for this disease. Here, we review clinical and experimental evidence suggesting that dysregulation of copper homeostasis in the central nervous system is a crucial underlying event in motor neuron degeneration and ALS pathophysiology. We also review and discuss novel approaches seeking to target copper delivery to treat ALS. These novel approaches may be clinically relevant not only for ALS but also for other neurological disorders with abnormal copper homeostasis, such as Parkinson's, Huntington's and Prion diseases.
Collapse
|
49
|
Abstract
The misfolding of the cellular prion protein (PrPC) causes fatal neurodegenerative diseases. Yet PrPC is highly conserved in mammals, suggesting that it exerts beneficial functions preventing its evolutionary elimination. Ablation of PrPC in mice results in well-defined structural and functional alterations in the peripheral nervous system. Many additional phenotypes were ascribed to the lack of PrPC, but some of these were found to arise from genetic artifacts of the underlying mouse models. Here, we revisit the proposed physiological roles of PrPC in the central and peripheral nervous systems and highlight the need for their critical reassessment using new, rigorously controlled animal models.
Collapse
Affiliation(s)
- Marie-Angela Wulf
- Institute of Neuropathology, University of Zurich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Assunta Senatore
- Institute of Neuropathology, University of Zurich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Rämistrasse 100, CH-8091, Zürich, Switzerland.
| |
Collapse
|
50
|
Peggion C, Bertoli A, Sorgato MC. Almost a century of prion protein(s): From pathology to physiology, and back to pathology. Biochem Biophys Res Commun 2016; 483:1148-1155. [PMID: 27581199 DOI: 10.1016/j.bbrc.2016.07.118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 07/27/2016] [Indexed: 12/30/2022]
Abstract
Prions are one of the few pathogens whose name is renowned at all population levels, after the dramatic years pervaded by the fear of eating prion-infected food. If now this, somehow irrational, scare of bovine meat inexorably transmitting devastating brain disorders is largely subdued, several prion-related issues are still unsolved, precluding the design of therapeutic approaches that could slow, if not halt, prion diseases. One unsolved issue is, for example, the role of the prion protein (PrPC), whole conformational misfolding originates the prion but whose physiologic reason d'etre in neurons, and in cells at large, remains enigmatic. Preceded by a historical outline, the present review will discuss the functional pleiotropicity ascribed to PrPC, and whether this aspect could fall, at least in part, into a more concise framework. It will also be devoted to radically different perspectives for PrPC, which have been recently brought to the attention of the scientific world with unexpected force. Finally, it will discuss the possible reasons allowing an evolutionary conserved and benign protein, as PrPC is, to turn into a high affinity receptor for pathologic misfolded oligomers, and to transmit their toxic message into neurons.
Collapse
Affiliation(s)
- Caterina Peggion
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy.
| | - Alessandro Bertoli
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy
| | - M Catia Sorgato
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy; C.N.R. Institute of Neuroscience, University of Padova, Via Bassi 58/B, 35131 Padova, Italy.
| |
Collapse
|