1
|
Jeong MJ, Kim YC, Jeong BH. The first report of single nucleotide polymorphisms in the open reading frame of the prion-like protein gene in rabbits. Front Vet Sci 2024; 11:1388339. [PMID: 38952802 PMCID: PMC11216025 DOI: 10.3389/fvets.2024.1388339] [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: 03/15/2024] [Accepted: 04/29/2024] [Indexed: 07/03/2024] Open
Abstract
Background Natural cases of prion disease have not been reported in rabbits, and prior attempts to identify a prion conversion agent have been unsuccessful. However, recent applications of prion seed amplifying experimental techniques have sparked renewed interest in the potential susceptibility of rabbits to prion disease infections. Among several factors related to prion disease, polymorphisms within the prion-like protein gene (PRND), a member of the prion protein family, have been reported as significantly associated with disease susceptibility in various species. Therefore, our study aimed to investigate polymorphisms in the PRND gene of rabbits and analyze their genetic characteristics. Methods Genomic DNA was extracted from 207 rabbit samples to investigate leporine PRND polymorphisms. Subsequently, amplicon sequencing targeting the coding region of the leporine PRND gene was conducted. Additionally, linkage disequilibrium (LD) analysis was employed to assess the connection within and between loci. The impact of non-synonymous single nucleotide polymorphisms (SNPs) on the Doppel protein was evaluated using PolyPhen-2. Results We found nine novel SNPs in the leporine PRND gene: c.18A > G, c.76G > C, c.128C > T, c.146C > T, c.315A > G, c.488G > A, c.525G > C, c.544G > A, and c.579A > G. Notably, seven of these PRND SNPs, excluding c.525G > C and c.579A > G, exhibited strong LD values exceeding 0.3. In addition, LD analysis confirmed a robust link between PRNP SNP c.234C > T and PRND SNPs at c.525G > C and c.579A > G. Furthermore, according to PolyPhen-2 and SIFT analyses, the four non-synonymous SNPs were predicted to have deleterious effects on the function or structure of the Doppel protein. However, PANTHER and Missense3D did not indicate such effects. Conclusion In this paper, we have identified novel SNPs in the rabbit PRND gene and predicted their potential detrimental effects on protein function or structure through four non-synonymous SNPs. Additionally, we observed a genetic linkage between SNPs in the PRND and PRNP genes. These findings may provide insights into understanding the characteristics of rabbits as partially resistant species. To the best of our knowledge, this study is the first to genetically characterize PRND SNPs in rabbits.
Collapse
Affiliation(s)
- Min-Ju Jeong
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeonbuk, Republic of Korea
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| | - Yong-Chan Kim
- Department of Biological Sciences, Andong National University, Andong, Republic of Korea
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeonbuk, Republic of Korea
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| |
Collapse
|
2
|
Lan X, Luo M, Li M, Mu L, Li G, Chen G, He Z, Xiao J. Swim bladder-derived biomaterials: structures, compositions, properties, modifications, and biomedical applications. J Nanobiotechnology 2024; 22:186. [PMID: 38632585 PMCID: PMC11022367 DOI: 10.1186/s12951-024-02449-w] [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: 08/10/2023] [Accepted: 04/01/2024] [Indexed: 04/19/2024] Open
Abstract
Animal-derived biomaterials have been extensively employed in clinical practice owing to their compositional and structural similarities with those of human tissues and organs, exhibiting good mechanical properties and biocompatibility, and extensive sources. However, there is an associated risk of infection with pathogenic microorganisms after the implantation of tissues from pigs, cattle, and other mammals in humans. Therefore, researchers have begun to explore the development of non-mammalian regenerative biomaterials. Among these is the swim bladder, a fish-derived biomaterial that is rapidly used in various fields of biomedicine because of its high collagen, elastin, and polysaccharide content. However, relevant reviews on the biomedical applications of swim bladders as effective biomaterials are lacking. Therefore, based on our previous research and in-depth understanding of this field, this review describes the structures and compositions, properties, and modifications of the swim bladder, with their direct (including soft tissue repair, dural repair, cardiovascular repair, and edible and pharmaceutical fish maw) and indirect applications (including extracted collagen peptides with smaller molecular weights, and collagen or gelatin with higher molecular weights used for hydrogels, and biological adhesives or glues) in the field of biomedicine in recent years. This review provides insights into the use of swim bladders as source of biomaterial; hence, it can aid biomedicine scholars by providing directions for advancements in this field.
Collapse
Affiliation(s)
- Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, 646000, China
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Mingdong Luo
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Meiling Li
- Southwest Hospital of Army Military Medical University, Chongqing, 400038, China
| | - Linpeng Mu
- Institute for Advanced Study, Research Center of Composites & Surface and Interface Engineering, Chengdu University, Chengdu, 610106, China
| | - Guangwen Li
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Gong Chen
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| | - Zhoukun He
- Institute for Advanced Study, Research Center of Composites & Surface and Interface Engineering, Chengdu University, Chengdu, 610106, China.
| | - Jingang Xiao
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China.
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
3
|
Lee JH, Park SH, Ryou C, Gye MC. Phthalate plasticizer decreases the prion-like protein doppel essential for structural integrity and function of spermatozoa. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 246:114159. [PMID: 36215882 DOI: 10.1016/j.ecoenv.2022.114159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/02/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
Di-n-butyl phthalate (DBP), a well-known endocrine disruptor, causes male reproductive dysfunction. To understand the underlying mechanisms, we performed histological, endocrinological, and biochemical analyses and assessed the expression of genes involved in spermatogenesis and sperm function according to OECD test guideline 407. Following 28 days of administration of the lowest observed adverse effect level dose of DBP to mice, no significant changes in body weight, testis and epididymis weights and histology, serum testosterone level, or testicular daily sperm production were found. Nonetheless, the motility of the epididymal sperm of the DBP group was significantly decreased together with an increase in the incidence of bent tails and abnormal heads. In the testes of the DBP group, lipid peroxidation (LPO) level was significantly increased and testicular Bcl-2 mRNA level was significantly decreased together with an increase in the Bax/Bcl-2 mRNA ratio. In the testes of the DBP group, levels of Prnd mRNA and protein and Pou4f1 mRNA, an activator of the Prnd promotor, were significantly decreased. Of note, prion-like protein doppel (PRND) was significantly decreased together with decreased PRND immunoreactivity in the head, midpiece, and tail of sperm. In the testes of the DBP group, levels of Sox9, Sgp1, and Sgp2 mRNA, which are functional Sertoli cell markers, were significantly decreased. Level of Amh mRNA, a Sertoli cell immaturity marker, was significantly increased together with that of Inha mRNA, suggesting deregulation of the brain-gonadal axis. Together, our findings suggest that DBP at present dosage may potentiate LPO generation and Sertoli cell immaturity via downregulation of Sox9 and disruption of the Pou4f1-Prnd gene network in post-meiotic germ cells without visible changes in spermatogenesis or testosterone level. This may result in structural and functional abnormalities in spermatozoa. Additionally, our findings suggest that assessment of the male reproductive toxicity of phthalate ester plasticizers based on conventional OECD test guidelines should be reconsidered.
Collapse
Affiliation(s)
- Jae-Hyeon Lee
- Department of Life Science, Institute for Natural Sciences and Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Republic of Korea
| | - Seung Hyun Park
- Department of Life Science, Institute for Natural Sciences and Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Republic of Korea
| | - Chongsuk Ryou
- College of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Republic of Korea
| | - Myung Chan Gye
- Department of Life Science, Institute for Natural Sciences and Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Republic of Korea.
| |
Collapse
|
4
|
Castle AR, Wohlgemuth S, Arce L, Westaway D. Investigating CRISPR/Cas9 gene drive for production of disease-preventing prion gene alleles. PLoS One 2022; 17:e0269342. [PMID: 35671288 PMCID: PMC9173614 DOI: 10.1371/journal.pone.0269342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 05/18/2022] [Indexed: 11/29/2022] Open
Abstract
Prion diseases are a group of fatal neurodegenerative disorders that includes chronic wasting disease, which affects cervids and is highly transmissible. Given that chronic wasting disease prevalence exceeds 30% in some endemic areas of North America, and that eventual transmission to other mammalian species, potentially including humans, cannot be ruled out, novel control strategies beyond population management via hunting and/or culling must be investigated. Prion diseases depend upon post-translational conversion of the cellular prion protein, encoded by the Prnp gene, into a disease-associated conformation; ablation of cellular prion protein expression, which is generally well-tolerated, eliminates prion disease susceptibility entirely. Inspired by demonstrations of gene drive in caged mosquito species, we aimed to test whether a CRISPR/Cas9-based gene drive mechanism could, in principle, promote the spread of a null Prnp allele among mammalian populations. First, we showed that transient co-expression of Cas9 and Prnp-directed guide RNAs in RK13 cells generates indels within the Prnp open-reading frame, indicating that repair of Cas9-induced double-strand breaks by non-homologous end-joining had taken place. Second, we integrated a ~1.2 kb donor DNA sequence into the Prnp open-reading frame in N2a cells by homology-directed repair following Cas9-induced cleavages and confirmed that integration occurred precisely in most cases. Third, we demonstrated that electroporation of Cas9/guide RNA ribonucleoprotein complexes into fertilised mouse oocytes resulted in pups with a variety of disruptions to the Prnp open reading frame, with a new coisogenic line of Prnp-null mice obtained as part of this work. However, a technical challenge in obtaining expression of Cas9 in the male germline prevented implementation of a complete gene drive mechanism in mice.
Collapse
Affiliation(s)
- Andrew R. Castle
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Serene Wohlgemuth
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Luis Arce
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Fan Y, Chen W, Wei R, Qiang W, Pearson JD, Yu T, Bremner R, Chen D. Mapping transgene insertion sites reveals the α-Cre transgene expression in both developing retina and olfactory neurons. Commun Biol 2022; 5:411. [PMID: 35505181 PMCID: PMC9065156 DOI: 10.1038/s42003-022-03379-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 04/18/2022] [Indexed: 02/05/2023] Open
Abstract
The Tg(Pax6-cre,GFP)2Pgr (α-Cre) mouse is a commonly used Cre line thought to be retinal-specific. Using targeted locus amplification (TLA), we mapped the insertion site of the transgene, and defined primers useful to deduce zygosity. Further analyses revealed four tandem copies of the transgene. The insertion site mapped to clusters of vomeronasal and olfactory receptor genes. Using R26R and Ai14 Cre reporter mice, we confirmed retinal Cre activity, but also detected expression in Gα0+ olfactory neurons. Most α-Cre+ olfactory neurons do not express Pax6, implicating the influence of neighboring regulatory elements. RT-PCR and buried food pellet test did not detect any effects of the transgene on flanking genes in the nasal mucosa and retina. Together, these data precisely map α-Cre, show that it does not affect surrounding loci, but reveal previously unanticipated transgene expression in olfactory neurons. The α-Cre mouse can be a valuable tool in both retinal and olfactory research. The Pax6-α-Cre mouse line used in retinal studies actually contains four transgene insertion within gene clusters of olfactory and vomeronasal receptors, leading to expression in not just retinal, but also olfactory and vomeronasal sensory neurons.
Collapse
Affiliation(s)
- Yimeng Fan
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Wenyue Chen
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Ran Wei
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Qiang
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Joel D Pearson
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Departments of Ophthalmology and Visual Science, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tao Yu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Departments of Ophthalmology and Visual Science, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Rod Bremner
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Departments of Ophthalmology and Visual Science, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Danian Chen
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China. .,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China. .,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Departments of Ophthalmology and Visual Science, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
Pimenta JM, Pires VM, Nolasco S, Castelo-Branco P, Marques CC, Apolónio J, Azevedo R, Fernandes MT, Lopes-da-Costa L, Prates J, Pereira RM. Post-transcriptional silencing of Bos taurus prion family genes and its impact on granulosa cell steroidogenesis. Biochem Biophys Res Commun 2022; 598:95-99. [DOI: 10.1016/j.bbrc.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/04/2022] [Indexed: 11/25/2022]
|
7
|
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: 1] [Impact Index Per Article: 0.5] [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
|
8
|
Membrane Domain Localization and Interaction of the Prion-Family Proteins, Prion and Shadoo with Calnexin. MEMBRANES 2021; 11:membranes11120978. [PMID: 34940479 PMCID: PMC8704586 DOI: 10.3390/membranes11120978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 11/30/2022]
Abstract
The cellular prion protein (PrPC) is renowned for its infectious conformational isoform PrPSc, capable of templating subsequent conversions of healthy PrPCs and thus triggering the group of incurable diseases known as transmissible spongiform encephalopathies. Besides this mechanism not being fully uncovered, the protein’s physiological role is also elusive. PrPC and its newest, less understood paralog Shadoo are glycosylphosphatidylinositol-anchored proteins highly expressed in the central nervous system. While they share some attributes and neuroprotective actions, opposing roles have also been reported for the two; however, the amount of data about their exact functions is lacking. Protein–protein interactions and membrane microdomain localizations are key determinants of protein function. Accurate identification of these functions for a membrane protein, however, can become biased due to interactions occurring during sample processing. To avoid such artifacts, we apply a non-detergent-based membrane-fractionation approach to study the prion protein and Shadoo. We show that the two proteins occupy similarly raft and non-raft membrane fractions when expressed in N2a cells and that both proteins pull down the chaperone calnexin in both rafts and non-rafts. These indicate their possible binding to calnexin in both types of membrane domains, which might be a necessary requisite to aid the inherently unstable native conformation during their lifetime.
Collapse
|
9
|
Li M, Zheng C, Wu B, Ding K, Zhang S, Huang X, Lei Y, Wang Y. Glycidyl methacrylate-crosslinked fish swim bladder as a novel cardiovascular biomaterial with improved antithrombotic and anticalcification properties. J Biomater Appl 2021; 36:1188-1200. [PMID: 34719293 DOI: 10.1177/08853282211054205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
At present, commercial artificial biological valves are mostly prepared by crosslinking bovine or porcine pericardia with glutaraldehyde. Swim bladder has similar components and lower immunogenicity compared to bovine or porcine pericardium. In this study, we used a glycidyl methacrylate (GMA)-based radical polymerization method to crosslink decellularized swim bladders. Amino and carboxyl groups in the swim bladder were reacted with epoxy groups on GMA to introduce carbon-carbon double bonds to the swim bladder. The results showed that the platelet adhesion of GMA-crosslinked swim bladders (GMA-SBs) decreased by 35%, as compared to that of glutaraldehyde-crosslinked swim bladders (GLUT-SBs). Moreover, the superior anticoagulant property was further verified by the ex vivo arteriovenous shunt assay. Meanwhile, the subcutaneous implantation in rats showed that GMA-SBs were able to effectively inhibit the calcification compared with GLUT-SBs. In conclusion, GMA-SBs showed improved antithrombotic and anticalcification properties compared to GLUT-SBs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yang Lei
- 12530Sichuan University, Chengdu, China
| | | |
Collapse
|
10
|
Abstract
Prion diseases are neurodegenerative disorders caused by conformational conversion of the cellular prion protein (PrPC) into scrapie prion protein (PrPSc). As the main component of prion, PrPSc acts as an infectious template that recruits and converts normal cellular PrPC into its pathogenic, misfolded isoform. Intriguingly, the phenomenon of prionoid, or prion-like, spread has also been observed in many other disease-associated proteins, such as amyloid β (Aβ), tau and α-synuclein. This Cell Science at a Glance and the accompanying poster highlight recently described physiological roles of prion protein and the advanced understanding of pathogenesis of prion disease they have afforded. Importantly, prion protein may also be involved in the pathogenesis of other neurodegenerative disorders such as Alzheimer's and Parkinson's disease. Therapeutic studies of prion disease have also exploited novel strategies to combat these devastating diseases. Future studies on prion protein and prion disease will deepen our understanding of the pathogenesis of a broad spectrum of neurodegenerative conditions.
Collapse
Affiliation(s)
- Caihong Zhu
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zürich, Zürich, CH-8091, Switzerland
| |
Collapse
|
11
|
Intrinsic disorder and phase transitions: Pieces in the puzzling role of the prion protein in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 183:1-43. [PMID: 34656326 DOI: 10.1016/bs.pmbts.2021.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
After four decades of prion protein research, the pressing questions in the literature remain similar to the common existential dilemmas. Who am I? Some structural characteristics of the cellular prion protein (PrPC) and scrapie PrP (PrPSc) remain unknown: there are no high-resolution atomic structures for either full-length endogenous human PrPC or isolated infectious PrPSc particles. Why am I here? It is not known why PrPC and PrPSc are found in specific cellular compartments such as the nucleus; while the physiological functions of PrPC are still being uncovered, the misfolding site remains obscure. Where am I going? The subcellular distribution of PrPC and PrPSc is wide (reported in 10 different locations in the cell). This complexity is further exacerbated by the eight different PrP fragments yielded from conserved proteolytic cleavages and by reversible post-translational modifications, such as glycosylation, phosphorylation, and ubiquitination. Moreover, about 55 pathological mutations and 16 polymorphisms on the PrP gene (PRNP) have been described. Prion diseases also share unique, challenging features: strain phenomenon (associated with the heterogeneity of PrPSc conformations) and the possible transmissibility between species, factors which contribute to PrP undruggability. However, two recent concepts in biochemistry-intrinsically disordered proteins and phase transitions-may shed light on the molecular basis of PrP's role in physiology and disease.
Collapse
|
12
|
Schmitt-Ulms G, Mehrabian M, Williams D, Ehsani S. The IDIP framework for assessing protein function and its application to the prion protein. Biol Rev Camb Philos Soc 2021; 96:1907-1932. [PMID: 33960099 DOI: 10.1111/brv.12731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 01/06/2023]
Abstract
The quest to determine the function of a protein can represent a profound challenge. Although this task is the mandate of countless research groups, a general framework for how it can be approached is conspicuously lacking. Moreover, even expectations for when the function of a protein can be considered to be 'known' are not well defined. In this review, we begin by introducing concepts pertinent to the challenge of protein function assignments. We then propose a framework for inferring a protein's function from four data categories: 'inheritance', 'distribution', 'interactions' and 'phenotypes' (IDIP). We document that the functions of proteins emerge at the intersection of inferences drawn from these data categories and emphasise the benefit of considering them in an evolutionary context. We then apply this approach to the cellular prion protein (PrPC ), well known for its central role in prion diseases, whose function continues to be considered elusive by many investigators. We document that available data converge on the conclusion that the function of the prion protein is to control a critical post-translational modification of the neural cell adhesion molecule in the context of epithelial-to-mesenchymal transition and related plasticity programmes. Finally, we argue that this proposed function of PrPC has already passed the test of time and is concordant with the IDIP framework in a way that other functions considered for this protein fail to achieve. We anticipate that the IDIP framework and the concepts analysed herein will aid the investigation of other proteins whose primary functional assignments have thus far been intractable.
Collapse
Affiliation(s)
- Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 0S8, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | | | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - Sepehr Ehsani
- Theoretical and Philosophical Biology, Department of Philosophy, University College London, Bloomsbury, London, WC1E 6BT, U.K.,Ronin Institute for Independent Scholarship, Montclair, NJ, 07043, U.S.A
| |
Collapse
|
13
|
Overduin M, Wille H, Westaway D. Multisite interactions of prions with membranes and native nanodiscs. Chem Phys Lipids 2021; 236:105063. [PMID: 33600804 DOI: 10.1016/j.chemphyslip.2021.105063] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/29/2021] [Accepted: 02/12/2021] [Indexed: 02/05/2023]
Abstract
Although prions are known as protein-only infectious particles, they exhibit lipid specificities, cofactor dependencies and membrane-dependent activities. Such membrane interactions play key roles in how prions are processed, presented and regulated, and hence have significant functional consequences. The expansive literature related to prion protein interactions with lipids and native nanodiscs is discussed, and provides a unique opportunity to re-evaluate the molecular composition and mechanisms of its infectious and cellular states. A family of crystal and solution structures of prions are analyzed here for the first time using the membrane optimal docking area (MODA) program, revealling the presence of structured binding elements that could mediate specific lipid recognition. A set of motifs centerred around W99, L125, Y169 and Y226 are consistently predicted as being membrane interactive and form an exposed surface which includes α helical, β strand and loop elements involving the prion protein (PrP) structural domain, while the scrapie form is radically different and doubles the size of the membrane interactive site into an extensible surface. These motifs are highly conserved throughout mammalian evolution, suggesting that prions have long been intrinsically attached to membranes at central and N- and C-terminal points, providing several opportunities for stable and specific bilayer interactions as well as multiple complexed orientations. Resistance or susceptibility to prion disease correlates with increased or decreased membrane binding propensity by mutant forms, respectively, indicating a protective role by lipids. The various prion states found in vivo are increasingly resolvable using native nanodiscs formed by styrene maleic acid (SMA) and stilbene maleic acid (STMA) copolymers rather than classical detergents, allowing the endogenous states to be tackled. These copolymers spontaneously fragment intact membranes into water-soluble discs holding a section of native bilayer, and can accommodate prion multimers and mini-fibrils. Such nanodiscs have also proven useful for understanding how β amyloid and α synuclein proteins contribute to Alzheimer's and Parkinson's diseases, providing further biomedical applications. Structural and functional insights of such proteins in styrene maleic acid lipid particles (SMALPs) can be resolved at high resolution by methods including cryo-electron microscopy (cEM), motivating continued progress in polymer design to resolve biological and pathological mechanisms.
Collapse
Affiliation(s)
- Michael Overduin
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada.
| | - Holger Wille
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada; Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - David Westaway
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada; Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
14
|
The Role of Cellular Prion Protein in Promoting Stemness and Differentiation in Cancer. Cancers (Basel) 2021; 13:cancers13020170. [PMID: 33418999 PMCID: PMC7825291 DOI: 10.3390/cancers13020170] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/31/2020] [Accepted: 01/03/2021] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Aside from its well-established role in prion disorders, in the last decades the significance of cellular prion protein (PrPC) expression in human cancers has attracted great attention. An extensive body of work provided evidence that PrPC contributes to tumorigenesis by regulating tumor growth, differentiation, and resistance to conventional therapies. In particular, PrPC over-expression has been related to the acquisition of a malignant phenotype of cancer stem cells (CSCs) in a variety of solid tumors, encompassing pancreatic ductal adenocarcinoma, osteosarcoma, breast, gastric, and colorectal cancers, and primary brain tumors as well. According to consensus, increased levels of PrPC endow CSCs with self-renewal, proliferative, migratory, and invasive capacities, along with increased resistance to anti-cancer agents. In addition, increasing evidence demonstrates that PrPc also participates in multi-protein complexes to modulate the oncogenic properties of CSCs, thus sustaining tumorigenesis. Therefore, strategies aimed at targeting PrPC and/or PrPC-organized complexes could be a promising approach for anti-cancer therapy. Abstract Cellular prion protein (PrPC) is seminal to modulate a variety of baseline cell functions to grant homeostasis. The classic role of such a protein was defined as a chaperone-like molecule being able to rescue cell survival. Nonetheless, PrPC also represents the precursor of the deleterious misfolded variant known as scrapie prion protein (PrPSc). This variant is detrimental in a variety of prion disorders. This multi-faceted role of PrP is greatly increased by recent findings showing how PrPC in its folded conformation may foster tumor progression by acting at multiple levels. The present review focuses on such a cancer-promoting effect. The manuscript analyzes recent findings on the occurrence of PrPC in various cancers and discusses the multiple effects, which sustain cancer progression. Within this frame, the effects of PrPC on stemness and differentiation are discussed. A special emphasis is provided on the spreading of PrPC and the epigenetic effects, which are induced in neighboring cells to activate cancer-related genes. These detrimental effects are further discussed in relation to the aberrancy of its physiological and beneficial role on cell homeostasis. A specific paragraph is dedicated to the role of PrPC beyond its effects in the biology of cancer to represent a potential biomarker in the follow up of patients following surgical resection.
Collapse
|
15
|
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: 2] [Impact Index Per Article: 0.5] [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
|
16
|
Puig B, Yang D, Brenna S, Altmeppen HC, Magnus T. Show Me Your Friends and I Tell You Who You Are: The Many Facets of Prion Protein in Stroke. Cells 2020; 9:E1609. [PMID: 32630841 PMCID: PMC7407975 DOI: 10.3390/cells9071609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke belongs to the leading causes of mortality and disability worldwide. Although treatments for the acute phase of stroke are available, not all patients are eligible. There is a need to search for therapeutic options to promote neurological recovery after stroke. The cellular prion protein (PrPC) has been consistently linked to a neuroprotective role after ischemic damage: it is upregulated in the penumbra area following stroke in humans, and animal models of stroke have shown that lack of PrPC aggravates the ischemic damage and lessens the functional outcome. Mechanistically, these effects can be linked to numerous functions attributed to PrPC: (1) as a signaling partner of the PI3K/Akt and MAPK pathways, (2) as a regulator of glutamate receptors, and (3) promoting stem cell homing mechanisms, leading to angio- and neurogenesis. PrPC can be cleaved at different sites and the proteolytic fragments can account for the manifold functions. Moreover, PrPC is present on extracellular vesicles (EVs), released membrane particles originating from all types of cells that have drawn attention as potential therapeutic tools in stroke and many other diseases. Thus, identification of the many mechanisms underlying PrPC-induced neuroprotection will not only provide further understanding of the physiological functions of PrPC but also new ideas for possible treatment options after ischemic stroke.
Collapse
Affiliation(s)
- Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | - Denise Yang
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | | | - Tim Magnus
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| |
Collapse
|
17
|
Thellung S, Corsaro A, Bosio AG, Zambito M, Barbieri F, Mazzanti M, Florio T. Emerging Role of Cellular Prion Protein in the Maintenance and Expansion of Glioma Stem Cells. Cells 2019; 8:cells8111458. [PMID: 31752162 PMCID: PMC6912268 DOI: 10.3390/cells8111458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Cellular prion protein (PrPC) is a membrane-anchored glycoprotein representing the physiological counterpart of PrP scrapie (PrPSc), which plays a pathogenetic role in prion diseases. Relatively little information is however available about physiological role of PrPC. Although PrPC ablation in mice does not induce lethal phenotypes, impairment of neuronal and bone marrow plasticity was reported in embryos and adult animals. In neurons, PrPC stimulates neurite growth, prevents oxidative stress-dependent cell death, and favors antiapoptotic signaling. However, PrPC activity is not restricted to post-mitotic neurons, but promotes cell proliferation and migration during embryogenesis and tissue regeneration in adult. PrPC acts as scaffold to stabilize the binding between different membrane receptors, growth factors, and basement proteins, contributing to tumorigenesis. Indeed, ablation of PrPC expression reduces cancer cell proliferation and migration and restores cell sensitivity to chemotherapy. Conversely, PrPC overexpression in cancer stem cells (CSCs) from different tumors, including gliomas—the most malignant brain tumors—is predictive for poor prognosis, and correlates with relapses. The mechanisms of the PrPC role in tumorigenesis and its molecular partners in this activity are the topic of the present review, with a particular focus on PrPC contribution to glioma CSCs multipotency, invasiveness, and tumorigenicity.
Collapse
Affiliation(s)
- Stefano Thellung
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
| | - Alessandro Corsaro
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
| | - Alessia G. Bosio
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
| | - Martina Zambito
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
| | - Federica Barbieri
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
| | - Michele Mazzanti
- Dipartimento di Bioscienze, Università di Milano, 20133 Milano, Italy
- Correspondence: (T.F.); (M.M.); Tel.: +39-01-0353-8806 (T.F.); +39-02-5031-4958 (M.M.)
| | - Tullio Florio
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Correspondence: (T.F.); (M.M.); Tel.: +39-01-0353-8806 (T.F.); +39-02-5031-4958 (M.M.)
| |
Collapse
|
18
|
Ryskalin L, Busceti CL, Biagioni F, Limanaqi F, Familiari P, Frati A, Fornai F. Prion Protein in Glioblastoma Multiforme. Int J Mol Sci 2019; 20:ijms20205107. [PMID: 31618844 PMCID: PMC6834196 DOI: 10.3390/ijms20205107] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/07/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022] Open
Abstract
The cellular prion protein (PrPc) is an evolutionarily conserved cell surface protein encoded by the PRNP gene. PrPc is ubiquitously expressed within nearly all mammalian cells, though most abundantly within the CNS. Besides being implicated in the pathogenesis and transmission of prion diseases, recent studies have demonstrated that PrPc contributes to tumorigenesis by regulating tumor growth, differentiation, and resistance to conventional therapies. In particular, PrPc over-expression has been related to the acquisition of a malignant phenotype of cancer stem cells (CSCs) in a variety of solid tumors, encompassing pancreatic ductal adenocarcinoma (PDAC), osteosarcoma, breast cancer, gastric cancer, and primary brain tumors, mostly glioblastoma multiforme (GBM). Thus, PrPc is emerging as a key in maintaining glioblastoma cancer stem cells’ (GSCs) phenotype, thereby strongly affecting GBM infiltration and relapse. In fact, PrPc contributes to GSCs niche’s maintenance by modulating GSCs’ stem cell-like properties while restraining them from differentiation. This is the first review that discusses the role of PrPc in GBM. The manuscript focuses on how PrPc may act on GSCs to modify their expression and translational profile while making the micro-environment surrounding the GSCs niche more favorable to GBM growth and infiltration.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy.
| | - Carla L Busceti
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli, Italy.
| | | | - Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy.
| | - Pietro Familiari
- Department of Neuroscience, Mental Health and Sense Organs NESMOS, Sapienza University of Rome, 00185 Rome, Italy.
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy.
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli, Italy.
| |
Collapse
|
19
|
Jeong MJ, Jeong BH. No polymorphisms in the coding region of the prion-like protein gene in Thoroughbred racehorses. Acta Vet Hung 2019; 67:174-182. [PMID: 31238729 DOI: 10.1556/004.2019.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Prion diseases are fatal neurodegenerative diseases characterised by the accumulation of an abnormal prion protein isoform (PrPSc), which is converted from the normal prion protein (PrPC). Prion diseases have been reported in an extensive number of species but not in horses up to now; therefore, horses are known to be a species resistant to prion diseases. The prion-like protein gene (PRND) is closely located downstream of the prion protein gene (PRNP) and the prion-like protein (Doppel) is a homologue with PrP. Previous studies have shown that an association between prion diseases and polymorphisms of the PRND gene is reported in the main hosts of prion diseases. Hence, we examined the genetic variations of the PRND gene in Thoroughbred horses. Interestingly, polymorphisms of the PRND gene were not detected. In addition, we conducted a comparative analysis of the amino acid sequences of the PRND gene to identify the differences between horses and other species. The amino acid sequence of the horse PRND gene showed the highest identity to that of sheep (83.7%), followed by that of goats, cattle and humans. To the best of our knowledge, this is the first genetic study of the PRND gene in horses.
Collapse
Affiliation(s)
- Min-Ju Jeong
- 1Korea Zoonosis Research Institute, Chonbuk National University, 820-120 Hana-ro, Iksan, Jeonbuk 54531, Republic of Korea
- 2Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| | - Byung-Hoon Jeong
- 1Korea Zoonosis Research Institute, Chonbuk National University, 820-120 Hana-ro, Iksan, Jeonbuk 54531, Republic of Korea
- 2Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| |
Collapse
|
20
|
Li Q, Zhang F, Wang H, Pan T. Preparation and characterization of a novel acellular swim bladder as dura mater substitute. Neurol Res 2019; 41:242-249. [PMID: 30912483 DOI: 10.1080/01616412.2018.1550139] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVES This paper aimed to develop a novel dura mater substitute made from swim bladders. METHODS The swim bladders were decellularized by diverse methods. The physical structure, residual DNA amount, mechanical properties and hemolysis rate were tested. The in vitro mouse embryonic fibroblast cells (MEFs) co-culture and in vivo dural repair surgery were performed to evaluate the biocompatibility of acellular swim bladder (ASBs). RESULTS The characteristics of different ASBs were evaluated, and the materials prepared via 'freezing-thawing and DNase-I' method showed the most appropriate features as dura mater substitute. The loosen fiber layer structure and three-dimensional porous structure were formed after decellularization. The residual DNA content was low (9.2 ± 2.0 ng/mg) and the mechanical properties could meet the clinical requirement (the maximum tensile strength was 34.77 ± 4.28 N and the maximum stitch tear strength was 7.15 ± 1.84 N). The hemolysis rate was up to 2.8 ± 0.15%. In the MEFs co-culture test, ASBs could support the adhesion, migration and proliferation of cells. The dural repair experiment demonstrated ASBs could prevent the leak of cerebrospinal fluid, and the materials were gradually replaced by autologous connective tissue. The novel dura mater substitute improved dura repair and regeneration without causing adhesion or severe inflammation. DISCUSSION The ASBs prepared via 'freezing-thawing and DNase-I' method had the ideal physical and biological properties as a dura mater substitute for clinical application.
Collapse
Affiliation(s)
- Qing Li
- a Qingdao Chunghao Tissue Engineering Co,LTD , Qingdao , Shandong Province , China
| | - Fenghua Zhang
- a Qingdao Chunghao Tissue Engineering Co,LTD , Qingdao , Shandong Province , China
| | - Hongmei Wang
- a Qingdao Chunghao Tissue Engineering Co,LTD , Qingdao , Shandong Province , China
| | - Tao Pan
- a Qingdao Chunghao Tissue Engineering Co,LTD , Qingdao , Shandong Province , China
| |
Collapse
|
21
|
Jeong MJ, Kim YC, Jeong BH. Prion-like protein gene (PRND) polymorphisms associated with scrapie susceptibility in Korean native black goats. PLoS One 2018; 13:e0206209. [PMID: 30359416 PMCID: PMC6201918 DOI: 10.1371/journal.pone.0206209] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 10/09/2018] [Indexed: 11/19/2022] Open
Abstract
The polymorphisms of the prion protein (PRNP) gene, which encodes normal prion proteins (PrP), are known to be involved in the susceptibility of prion diseases. The prion-like protein (Doppel) gene (PRND) is the paralog of the PRNP gene and is closely located downstream of the PRNP gene. In addition, the polymorphisms of PRND correlate with disease susceptibility in several animals. We analyzed the genotype and allele frequencies of PRND polymorphisms in 246 Korean native black goats and found a total of six single nucleotide polymorphisms (SNPs) with one novel SNP, c.99C>T. We observed linkage disequilibrium (LD) within and between loci. PRND c.28T>C, c.151A>G, and c.385G>C and PRND c.65C>T and c.286G>A were in perfect LD and we have reported for the first time strong LD between PRND and PRNP or prion-related protein gene (PRNT) loci. Specifically, between the PRND c.28T>C, c.151A>G and c.385G>C and the PRNP codon 143, PRND c.99C>T and the PRNP codon 102 or PRND SNPs (c.28T>C, c.151A>G and c.385G>C) and PRNT SNP (c.321T>C). Furthermore, we confirmed that the genotype distribution of the PRNP p.His143Arg was significantly different according to that of the PRND c.28T>C (P < 0.0001). Finally, using PolyPhen-2 and PROVEAN, we predicted that two non-synonymous SNPs, c.65C>T and c.286G>A, in the PRND gene can have a detrimental effect on Doppel. To the best of our knowledge, this is the first report of genetic characteristics of the PRND gene in Korean native black goats.
Collapse
Affiliation(s)
- Min-Ju Jeong
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, Jeonbuk, Republic of Korea
- Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| | - Yong-Chan Kim
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, Jeonbuk, Republic of Korea
- Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, Jeonbuk, Republic of Korea
- Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| |
Collapse
|
22
|
Tosh JL, Rickman M, Rhymes E, Norona FE, Clayton E, Mucke L, Isaacs AM, Fisher EM, Wiseman FK. The integration site of the APP transgene in the J20 mouse model of Alzheimer's disease. Wellcome Open Res 2018; 2:84. [PMID: 29062914 PMCID: PMC5645710 DOI: 10.12688/wellcomeopenres.12237.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2017] [Indexed: 02/02/2023] Open
Abstract
Background: Transgenic animal models are a widely used and powerful tool to investigate human disease and develop therapeutic interventions. Making a transgenic mouse involves random integration of exogenous DNA into the host genome that can have the effect of disrupting endogenous gene expression. The J20 mouse model of Alzheimer's disease (AD) is a transgenic overexpresser of human APP with familial AD mutations and has been extensively utilised in preclinical studies and our aim was to determine the genomic location of the J20 transgene insertion. Methods: We used a combination of breeding strategy and Targeted Locus Amplification with deep sequencing to identify the insertion site of the J20 transgene array. To assess RNA and protein expression of Zbtb20, we used qRT-PCR and Western Blotting. Results: We demonstrate that the J20 transgene construct has inserted within the genetic locus of endogenous mouse gene Zbtb20 on chromosome 16 in an array , disrupting expression of mRNA from this gene in adult hippocampal tissue, while expression of Zbtb20 protein remains unchanged. We note that the endogenous mouse App gene also lies on chromosome 16, although 42 Mb from the Zbtb20 locus. Conclusions: These data will be useful for future studies utilising this popular model of AD, particularly those investigating gene interactions between the J20 APP transgene and other genes present on Mmu16 in the mouse.
Collapse
Affiliation(s)
- Justin L. Tosh
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Matthew Rickman
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Ellie Rhymes
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Frances E. Norona
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Emma Clayton
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease and University of California, San Francisco, CA, 4158, USA
| | - Adrian M. Isaacs
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Elizabeth M.C. Fisher
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,
| | - Frances K. Wiseman
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,
| |
Collapse
|
23
|
Tosh JL, Rickman M, Rhymes E, Norona FE, Clayton E, Mucke L, Isaacs AM, Fisher EM, Wiseman FK. The integration site of the APP transgene in the J20 mouse model of Alzheimer's disease. Wellcome Open Res 2018; 2:84. [PMID: 29062914 PMCID: PMC5645710 DOI: 10.12688/wellcomeopenres.12237.2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2018] [Indexed: 02/02/2023] Open
Abstract
Background: Transgenic animal models are a widely used and powerful tool to investigate human disease and develop therapeutic interventions. Making a transgenic mouse involves random integration of exogenous DNA into the host genome that can have the effect of disrupting endogenous gene expression. The J20 mouse model of Alzheimer's disease (AD) is a transgenic overexpresser of human APP with familial AD mutations and has been extensively utilised in preclinical studies and our aim was to determine the genomic location of the J20 transgene insertion. Methods: We used a combination of breeding strategy and Targeted Locus Amplification with deep sequencing to identify the insertion site of the J20 transgene array. To assess RNA and protein expression of Zbtb20, we used qRT-PCR and Western Blotting. Results: We demonstrate that the J20 transgene construct has inserted within the genetic locus of endogenous mouse gene Zbtb20 on chromosome 16 in an array , disrupting expression of mRNA from this gene in adult hippocampal tissue. Preliminary data suggests that ZBTB20 protein levels remain unchanged in this tissue, however further study is necessary. We note that the endogenous mouse App gene also lies on chromosome 16, although 42 Mb from the Zbtb20 locus. Conclusions: These data will be useful for future studies utilising this popular model of AD, particularly those investigating gene interactions between the J20 APP transgene and other genes present on Mmu16 in the mouse.
Collapse
Affiliation(s)
- Justin L. Tosh
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Matthew Rickman
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Ellie Rhymes
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Frances E. Norona
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Emma Clayton
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease and University of California, San Francisco, CA, 4158, USA
| | - Adrian M. Isaacs
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Elizabeth M.C. Fisher
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,
| | - Frances K. Wiseman
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,
| |
Collapse
|
24
|
A bispecific immunotweezer prevents soluble PrP oligomers and abolishes prion toxicity. PLoS Pathog 2018; 14:e1007335. [PMID: 30273408 PMCID: PMC6181439 DOI: 10.1371/journal.ppat.1007335] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 10/11/2018] [Accepted: 09/13/2018] [Indexed: 11/28/2022] Open
Abstract
Antibodies to the prion protein, PrP, represent a promising therapeutic approach against prion diseases but the neurotoxicity of certain anti-PrP antibodies has caused concern. Here we describe scPOM-bi, a bispecific antibody designed to function as a molecular prion tweezer. scPOM-bi combines the complementarity-determining regions of the neurotoxic antibody POM1 and the neuroprotective POM2, which bind the globular domain (GD) and flexible tail (FT) respectively. We found that scPOM-bi confers protection to prion-infected organotypic cerebellar slices even when prion pathology is already conspicuous. Moreover, scPOM-bi prevents the formation of soluble oligomers that correlate with neurotoxic PrP species. Simultaneous targeting of both GD and FT was more effective than concomitant treatment with the individual molecules or targeting the tail alone, possibly by preventing the GD from entering a toxic-prone state. We conclude that simultaneous binding of the GD and flexible tail of PrP results in strong protection from prion neurotoxicity and may represent a promising strategy for anti-prion immunotherapy. Antibody immunotherapy is considered a viable strategy against prion disease. We previously showed that antibodies against the so-called globular domain of Prion Protein (PrP) can cause PrP dependent neurotoxicity; this does not happen for antibodies against the flexible tail of PrP, which therefore ought to be preferred for therapy. Here we show that simultaneous targeting of both globular domain and flexible tail by a bispecific, combination of a toxic and a non-toxic antibody, results in stronger protection against prion toxicity, even if the bispecific is administered when prion pathology is already conspicuous. We hypothesize that neurotoxicity arises from binding to specific “toxicity triggering sites” in the globular domain. We designed our bispecific with two aims: i) occupying one such site and preventing prion or other factors from docking to it and ii) binding to the flexible tail to engage the region of PrP necessary for neurotoxicity. We also show that neurotoxic antibodies cause the formation of soluble PrP oligomers that cause toxicity on PrP expressing cell lines; these are not formed in the presence of prion protective antibodies. We suggest that these soluble species might play a role in prion toxicity, similarly to what is generally agreed to happen in other neurodegenerative disorders.
Collapse
|
25
|
Prion protein testis specific (PRNT) gene polymorphisms and transcript level in ovine spermatozoa: Implications in freezability, fertilization and embryo production. Theriogenology 2018; 115:124-132. [PMID: 29754043 DOI: 10.1016/j.theriogenology.2018.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/11/2018] [Accepted: 04/09/2018] [Indexed: 11/21/2022]
Abstract
An essential role of prion protein testis specific (PRNT) and prion protein 2 dublet (PRND) genes in the male reproductive function has been highlighted, although a deeper knowledge for the mechanisms involved is still lacking. Our goal was to determine the importance of the PRNT haplotypic variants and mRNA expression levels in ovine spermatozoa freezability and ability for fertilization and embryo developmental processes. Their association with the PRND gene polymorphisms was also analyzed. DNA from rams belonging to three Portuguese sheep breeds (n = 28) was screened by single-strand conformation polymorphism (SSCP) analysis to identify the PRNT and PRND polymorphisms. Semen collected from these rams was cryopreserved and fertility traits evaluated. The SSCP analyses revealed polymorphisms in the codons 6, 38, 43 and 48 of the PRNT coding region - respectively c.17C > T (p.Ser6Phe, which disrupts a consensus arginine-X-X serine/threonine motif); c.112G > C (p.Gly38 > Arg); and synonymous c.129T > C and c.144A > G. The polymorphisms in codons 6, 38 and 48 occur simultaneously while the one in codon 43 occurs independently. Six haplotypes were identified in the PRNT coding region, resulting in three different amino acid polymorphic variants (6S-38G-43C-48V, S6F-G38R-43C-48V and 6F-38R-43C-48V). The PRNT gene mRNA transcript level in spermatozoa was related to the identified haplotypic variants, either considering the codons 6-38-48 (P ≤ 0.0001) or the codon 43 alone (P ≤ 0.0001) or altogether (P ≤ 0.0001). An interaction between PRNT haplotypes and PRND genotypes on PRNT transcript level was also identified (P = 0.0003). Rams carrying the 17C-112G-144A PRNT haplotype had sperm with the highest post-thawed individual motility (P ≤ 0.03). Combined PRNT and PRND polymorphic variation influenced the post-thawed individual motility (P = 0.01). The male PRNT haplotypic, either considering the codons 6-38-48 and 43 altogether or the codon 43 alone, interfered (P ≤ 0.04) in embryo production rates. In conclusion, our data confirm that the PRNT gene is highly polymorphic in sheep and that the PRNT and PRND genotypes are associated. The identified polymorphisms of PRNT coding region seems to interfere on the ram spermatozoa mRNA transcript level and on male fertility, specifically in sperm freezability and ability for embryo development.
Collapse
|
26
|
The function of the cellular prion protein in health and disease. Acta Neuropathol 2018; 135:159-178. [PMID: 29151170 DOI: 10.1007/s00401-017-1790-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/11/2022]
Abstract
The essential role of the cellular prion protein (PrPC) in prion disorders such as Creutzfeldt-Jakob disease is well documented. Moreover, evidence is accumulating that PrPC may act as a receptor for protein aggregates and transduce neurotoxic signals in more common neurodegenerative disorders, such as Alzheimer's disease. Although the pathological roles of PrPC have been thoroughly characterized, a general consensus on its physiological function within the brain has not yet been established. Knockout studies in various organisms, ranging from zebrafish to mice, have implicated PrPC in a diverse range of nervous system-related activities that include a key role in the maintenance of peripheral nerve myelination as well as a general ability to protect against neurotoxic stimuli. Thus, the function of PrPC may be multifaceted, with different cell types taking advantage of unique aspects of its biology. Deciphering the cellular function(s) of PrPC and the consequences of its absence is not simply an academic curiosity, since lowering PrPC levels in the brain is predicted to be a powerful therapeutic strategy for the treatment of prion disease. In this review, we outline the various approaches that have been employed in an effort to uncover the physiological and pathological functions of PrPC. While these studies have revealed important clues about the biology of the prion protein, the precise reason for PrPC's existence remains enigmatic.
Collapse
|
27
|
Leighton PLA, Allison WT. Protein Misfolding in Prion and Prion-Like Diseases: Reconsidering a Required Role for Protein Loss-of-Function. J Alzheimers Dis 2018; 54:3-29. [PMID: 27392869 DOI: 10.3233/jad-160361] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Prion disease research has contributed much toward understanding other neurodegenerative diseases, including recent demonstrations that Alzheimer's disease (AD) and other neurodegenerative diseases are prion-like. Prion-like diseases involve the spread of degeneration between individuals and/or among cells or tissues via template directed misfolding, wherein misfolded protein conformers propagate disease by causing normal proteins to misfold. Here we use the premise that AD, amyotrophic lateral sclerosis, Huntington's disease, and other similar diseases are prion-like and ask: Can we apply knowledge gained from studies of these prion-like diseases to resolve debates about classical prion diseases? We focus on controversies about what role(s) protein loss-of-function might have in prion diseases because this has therapeutic implications, including for AD. We examine which loss-of-function events are recognizable in prion-like diseases by considering the normal functions of the proteins before their misfolding and aggregation. We then delineate scenarios wherein gain-of-function and/or loss-of-function would be necessary or sufficient for neurodegeneration. We consider roles of PrPC loss-of-function in prion diseases and in AD, and conclude that the conventional wisdom that prion diseases are 'toxic gain-of-function diseases' has limitations. While prion diseases certainly have required gain-of-function components, we propose that disease phenotypes are predominantly caused by deficits in the normal physiology of PrPC and its interaction partners as PrPC converts to PrPSc. In this model, gain-of-function serves mainly to spread disease, and loss-of-function directly mediates neuron dysfunction. We propose experiments and predictions to assess our conclusion. Further study on the normal physiological roles of these key proteins is warranted.
Collapse
Affiliation(s)
- Patricia L A Leighton
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - W Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
28
|
|
29
|
What Is Our Current Understanding of PrP Sc-Associated Neurotoxicity and Its Molecular Underpinnings? Pathogens 2017; 6:pathogens6040063. [PMID: 29194372 PMCID: PMC5750587 DOI: 10.3390/pathogens6040063] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/21/2017] [Accepted: 11/27/2017] [Indexed: 01/15/2023] Open
Abstract
The prion diseases are a collection of fatal, transmissible neurodegenerative diseases that cause rapid onset dementia and ultimately death. Uniquely, the infectious agent is a misfolded form of the endogenous cellular prion protein, termed PrPSc. Despite the identity of the molecular agent remaining the same, PrPSc can cause a range of diseases with hereditary, spontaneous or iatrogenic aetiologies. However, the link between PrPSc and toxicity is complex, with subclinical cases of prion disease discovered, and prion neurodegeneration without obvious PrPSc deposition. The toxic mechanisms by which PrPSc causes the extensive neuropathology are still poorly understood, although recent advances are beginning to unravel the molecular underpinnings, including oxidative stress, disruption of proteostasis and induction of the unfolded protein response. This review will discuss the diseases caused by PrPSc toxicity, the nature of the toxicity of PrPSc, and our current understanding of the downstream toxic signaling events triggered by the presence of PrPSc.
Collapse
|
30
|
Ricci A, Allende A, Bolton D, Chemaly M, Davies R, Fernández Escámez PS, Gironés R, Herman L, Koutsoumanis K, Lindqvist R, Nørrung B, Robertson L, Ru G, Sanaa M, Skandamis P, Speybroeck N, Simmons M, Kuile BT, Threlfall J, Wahlström H, Acutis PL, Andreoletti O, Goldmann W, Langeveld J, Windig JJ, Ortiz Pelaez A, Snary E. Genetic resistance to transmissible spongiform encephalopathies (TSE) in goats. EFSA J 2017; 15:e04962. [PMID: 32625625 PMCID: PMC7010077 DOI: 10.2903/j.efsa.2017.4962] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Breeding programmes to promote resistance to classical scrapie, similar to those for sheep in existing transmissible spongiform encephalopathies (TSE) regulations, have not been established in goats. The European Commission requested a scientific opinion from EFSA on the current knowledge of genetic resistance to TSE in goats. An evaluation tool, which considers both the weight of evidence and strength of resistance to classical scrapie of alleles in the goat PRNP gene, was developed and applied to nine selected alleles of interest. Using the tool, the quality and certainty of the field and experimental data are considered robust enough to conclude that the K222, D146 and S146 alleles both confer genetic resistance against classical scrapie strains known to occur naturally in the EU goat population, with which they have been challenged both experimentally and under field conditions. The weight of evidence for K222 is greater than that currently available for the D146 and S146 alleles and for the ARR allele in sheep in 2001. Breeding for resistance can be an effective tool for controlling classical scrapie in goats and it could be an option available to member states, both at herd and population levels. There is insufficient evidence to assess the impact of K222, D146 and S146 alleles on susceptibility to atypical scrapie and bovine spongiform encephalopathy (BSE), or on health and production traits. These alleles are heterogeneously distributed across the EU Member States and goat breeds, but often at low frequencies (< 10%). Given these low frequencies, high selection pressure may have an adverse effect on genetic diversity so any breeding for resistance programmes should be developed at Member States, rather than EU level and their impact monitored, with particular attention to the potential for any negative impact in rare or small population breeds.
Collapse
|
31
|
Carlson GA. Prion Protein and Genetic Susceptibility to Diseases Caused by Its Misfolding. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:123-145. [PMID: 28838658 DOI: 10.1016/bs.pmbts.2017.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Early genetic studies on scrapie, an infectious neurodegenerative disease of sheep that was adapted to mice, provided evidence in support of the hypothesis that the agent was a slow virus with a nucleic acid genome independent of the host. Particularly compelling support for an independent genome came from the existence of strains of scrapie agent, some of which were true breeding, while others appeared to mutate under selective pressure. Kuru, a neurodegenerative disease in the remote highlands of Papua New Guinea, had pathological changes similar to those in scrapie and also proved to be transmissible. Genetic studies with the tools of molecular biology and transgenic mice forced a reevaluation of earlier work and supported the prion hypothesis of a novel pathogen devoid of nucleic acid. In this chapter, I discuss the contributions of classical and molecular genetics to understanding PrP prion diseases and to determining that heritable information is enciphered in protein conformation.
Collapse
|
32
|
Diack AB, Alibhai JD, Manson JC. Gene Targeted Transgenic Mouse Models in Prion Research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:157-179. [PMID: 28838660 DOI: 10.1016/bs.pmbts.2017.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The production of transgenic mice expressing different forms of the prion protein (PrP) or devoid of PrP has enabled researchers to study the role of PrP in the infectious process of a prion disease and its normal function in the healthy individual. A wide range of transgenic models have been produced ranging from PrP null mice, normal expression levels to overexpression models, models expressing different species of the Prnp gene and different mutations and polymorphisms within the gene. Using this range of transgenic models has allowed us to define the influence of PrP expression on disease susceptibility and transmission, assess zoonotic potential, define strains of human prion diseases, elucidate the function of PrP, and start to unravel the mechanisms involved in chronic neurodegeneration. This chapter focuses mainly on the use of the gene targeted transgenic models and summarizes the ways in which they have allowed us to study the role of PrP in prion disease and the insights they have provided into the mechanisms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Abigail B Diack
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, United Kingdom.
| | - James D Alibhai
- The National CJD Research and Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jean C Manson
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, United Kingdom
| |
Collapse
|
33
|
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: 11] [Impact Index Per Article: 1.6] [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
|
34
|
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
|
35
|
Macedo JA, Schrama D, Duarte I, Tavares E, Renaut J, Futschik ME, Rodrigues PM, Melo EP. Membrane-enriched proteome changes and prion protein expression during neural differentiation and in neuroblastoma cells. BMC Genomics 2017; 18:319. [PMID: 28431525 PMCID: PMC5401558 DOI: 10.1186/s12864-017-3694-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/08/2017] [Indexed: 01/12/2023] Open
Abstract
Background The function of the prion protein, involved in the so-called prion diseases, remains a subject of intense debate and the possibility that it works as a pleiotropic protein through the interaction with multiple membrane proteins is somehow supported by recent reports. Therefore, the use of proteomic and bioinformatics combined to uncover cellular processes occurring together with changes in the expression of the prion protein may provide further insight into the putative pleiotropic role of the prion protein. Results This study assessed the membrane-enriched proteome changes accompanying alterations in the expression of the prion protein. A 2D-DIGE approach was applied to two cell lines after prefractionation towards the membrane protein subset: an embryonic stem cell line and the PK1 subline of neuroblastoma cells which efficiently propagates prion infection. Several proteins were differentially abundant with the increased expression of the prion protein during neural differentiation of embryonic stem cells and with the knockdown of the prion protein in PK1 cells. The identity of around 20% of the differentially abundant proteins was obtained by tandem MS. The catalytic subunit A of succinate dehydrogenase, a key enzyme for the aerobic energy metabolism and redox homeostasis, showed a similar abundance trend as the prion protein in both proteomic experiments. A gene ontology analysis revealed “myelin sheath”, “organelle membrane” and “focal adhesion” associated proteins as the main cellular components, and “protein folding” and “ATPase activity” as the biological processes enriched in the first set of differentially abundant proteins. The known interactome of these differentially abundant proteins was customized to reveal four interactors with the prion protein, including two heat shock proteins and a protein disulfide isomerase. Conclusions Overall, our study shows that expression of the prion protein occurs concomitantly with changes in chaperone activity and cell-redox homeostasis, emphasizing the functional link between these cellular processes and the prion protein. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3694-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- J A Macedo
- CBMR, Center for Biomedical Research, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - D Schrama
- CCMAR, Centre of Marine Sciences of Algarve, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - I Duarte
- CBMR, Center for Biomedical Research, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - E Tavares
- CBMR, Center for Biomedical Research, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - J Renaut
- LIST, Luxembourg Institute of Science and Technology, Belvaux, Luxembourg
| | - M E Futschik
- CCMAR, Centre of Marine Sciences of Algarve, University of Algarve, Campus de Gambelas, Faro, Portugal.,School of Biomedical & Healthcare Sciences, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| | - P M Rodrigues
- CCMAR, Centre of Marine Sciences of Algarve, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - E P Melo
- CBMR, Center for Biomedical Research, University of Algarve, Campus de Gambelas, Faro, Portugal.
| |
Collapse
|
36
|
Castle AR, Gill AC. Physiological Functions of the Cellular Prion Protein. Front Mol Biosci 2017; 4:19. [PMID: 28428956 PMCID: PMC5382174 DOI: 10.3389/fmolb.2017.00019] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/22/2017] [Indexed: 01/09/2023] Open
Abstract
The prion protein, PrPC, is a small, cell-surface glycoprotein notable primarily for its critical role in pathogenesis of the neurodegenerative disorders known as prion diseases. A hallmark of prion diseases is the conversion of PrPC into an abnormally folded isoform, which provides a template for further pathogenic conversion of PrPC, allowing disease to spread from cell to cell and, in some circumstances, to transfer to a new host. In addition to the putative neurotoxicity caused by the misfolded form(s), loss of normal PrPC function could be an integral part of the neurodegenerative processes and, consequently, significant research efforts have been directed toward determining the physiological functions of PrPC. In this review, we first summarise important aspects of the biochemistry of PrPC before moving on to address the current understanding of the various proposed functions of the protein, including details of the underlying molecular mechanisms potentially involved in these functions. Over years of study, PrPC has been associated with a wide array of different cellular processes and many interacting partners have been suggested. However, recent studies have cast doubt on the previously well-established links between PrPC and processes such as stress-protection, copper homeostasis and neuronal excitability. Instead, the functions best-supported by the current literature include regulation of myelin maintenance and of processes linked to cellular differentiation, including proliferation, adhesion, and control of cell morphology. Intriguing connections have also been made between PrPC and the modulation of circadian rhythm, glucose homeostasis, immune function and cellular iron uptake, all of which warrant further investigation.
Collapse
|
37
|
Brandner S, Jaunmuktane Z. Prion disease: experimental models and reality. Acta Neuropathol 2017; 133:197-222. [PMID: 28084518 PMCID: PMC5250673 DOI: 10.1007/s00401-017-1670-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 01/04/2023]
Abstract
The understanding of the pathogenesis and mechanisms of diseases requires a multidisciplinary approach, involving clinical observation, correlation to pathological processes, and modelling of disease mechanisms. It is an inherent challenge, and arguably impossible to generate model systems that can faithfully recapitulate all aspects of human disease. It is, therefore, important to be aware of the potentials and also the limitations of specific model systems. Model systems are usually designed to recapitulate only specific aspects of the disease, such as a pathological phenotype, a pathomechanism, or to test a hypothesis. Here, we evaluate and discuss model systems that were generated to understand clinical, pathological, genetic, biochemical, and epidemiological aspects of prion diseases. Whilst clinical research and studies on human tissue are an essential component of prion research, much of the understanding of the mechanisms governing transmission, replication, and toxicity comes from in vitro and in vivo studies. As with other neurodegenerative diseases caused by protein misfolding, the pathogenesis of prion disease is complex, full of conundra and contradictions. We will give here a historical overview of the use of models of prion disease, how they have evolved alongside the scientific questions, and how advancements in technologies have pushed the boundaries of our understanding of prion biology.
Collapse
Affiliation(s)
- Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London, WC1N 3BG UK
| | - Zane Jaunmuktane
- Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London, WC1N 3BG UK
| |
Collapse
|
38
|
Ferreira LM, Garcia-Herreros M, Domingos A, Marques CC, Mesquita P, Barbas JP, Baptista MC, Pimenta J, Horta AEM, Prates JAM, Pereira RMLN. Prion protein 2 (dublet) gene (PRND): role in ovine semen capacitation, cryopreservation and fertility. Reprod Fertil Dev 2017; 29:985-997. [DOI: 10.1071/rd15214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 01/24/2016] [Indexed: 12/20/2022] Open
Abstract
The aim of the present study was to examine the role of Doppel protein in the capacitation process and fertilising ability of both fresh and frozen–thawed (FT) spermatozoa from rams carrying different prion protein 2 (dublet) (PRND) gene polymorphisms. The detection efficacy of new anti-Doppel monoclonal antibodies and PRND mRNA quantification were also explored in ovine spermatozoa. Three different genotypes (AA, GA, GG) were identified for codon 26 of ovine PRND-c.78G>A. Using flow cytometry, a higher fluorescence was detected in fresh compared with FT sperm samples incubated with anti-Doppel primary and fluorescein isothiocyanate-conjugated secondary antibodies (P < 0.05). Capacitation was affected by semen treatment (fresh and FT) and male PRND genotype (P < 0.05). After IVF, the use of fresh semen resulted in a higher cleavage rate than the use of FT spermatozoa (P = 0.004). IVF using spermatozoa from individuals classified as carriers of the AA or GA PRND genotypes resulted in higher cleavage rates than seen using spermatozoa from GG carriers (P ≤ 0.0006). Finally, using semen from rams with the AA PRND genotype resulted in the highest Day 6 and Day 8 embryo rates (P ≤ 0.04). In conclusion, the results of the present study confirm that the identification of different PRND genotypes is important for studying the sperm capacitation process and for improving sperm cryoresistance and embryo production. Furthermore, the detection of Doppel in ejaculated ovine spermatozoa, along with its low expression after cryopreservation, strongly suggests an important physiological function of this protein in male fertility.
Collapse
|
39
|
ONODERA T. Dual role of cellular prion protein in normal host and Alzheimer's disease. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:155-173. [PMID: 28413194 PMCID: PMC5489426 DOI: 10.2183/pjab.93.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/26/2017] [Indexed: 06/07/2023]
Abstract
Using PrPC-knockout cell lines, it has been shown that the inhibition of apoptosis through STI1 is mediated by PrPC-dependent SOD activation. Antioxidant PrPC may contribute to suppression of inflammasome activation. PrPC is functionally involved in copper metabolism, signal transduction, neuroprotection, and cell maturation. Recently several reports have shown that PrPC participates in trans-membrane signaling processes associated with hematopoietic stem cell replication and neuronal differentiation. In another role, PrPC also tends to function as a neurotoxic protein. Aβ oligomer, which is associated with neurodegeneration in Alzheimer's disease (AD), has also been reported to act as a ligand of PrPC. However, the physiological role of PrPC as an Aβ42-binding protein is not clear. Actually, PrPC is critical in Aβ42-mediated autophagy in neurons. PrPC shows a beneficial role in lipid rafts to promote autophagy. Further search for PrPC-interaction molecules using Prnp-/- mice and various types of Prnp-/- cell lines under various conditions may elucidate other important PrPC important functions.
Collapse
Affiliation(s)
- Takashi ONODERA
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
40
|
Nyeste A, Stincardini C, Bencsura P, Cerovic M, Biasini E, Welker E. The prion protein family member Shadoo induces spontaneous ionic currents in cultured cells. Sci Rep 2016; 6:36441. [PMID: 27819308 PMCID: PMC5098206 DOI: 10.1038/srep36441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 10/17/2016] [Indexed: 01/01/2023] Open
Abstract
Some mutant forms of the cellular prion protein (PrPC) carrying artificial deletions or point mutations associated with familial human prion diseases are capable of inducing spontaneous ionic currents across the cell membrane, conferring hypersensitivity to certain antibiotics to a wide range of cultured cells and primary cerebellar granular neurons (CGNs). These effects are abrogated when the wild type (WT) form is co-expressed, suggesting that they might be related to a physiological activity of PrPC. Interestingly, the prion protein family member Shadoo (Sho) makes cells hypersensitive to the same antibiotics as mutant PrP-s, an effect that is diminished by the co-expression of WT-PrP. Here, we report that Sho engages in another mutant PrP-like activity: it spontaneously induces large ionic currents in cultured SH-SY5Y cells, as detected by whole-cell patch clamping. These currents are also decreased by the co-expression of WT-PrP. Furthermore, deletion of the N-terminal (RXXX)8 motif of Sho, mutation of the eight arginine residues of this motif to glutamines, or replacement of the hydrophobic domain by that of PrP, also diminish Sho-induced ionic currents. Our results suggest that the channel activity that is also characteristic to some pathogenic PrP mutants may be linked to a physiological function of Sho.
Collapse
Affiliation(s)
- Antal Nyeste
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary
- Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Claudia Stincardini
- Dulbecco Telethon Laboratory of Prions and Amyloids, Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, ITALY
| | - Petra Bencsura
- Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Milica Cerovic
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri, 20156 Milano, ITALY
| | - Emiliano Biasini
- Dulbecco Telethon Laboratory of Prions and Amyloids, Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, ITALY
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri, 20156 Milano, ITALY
| | - Ervin Welker
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary
- Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
41
|
Daude N, Gapeshina H, Dong B, Winship I, Westaway D. Neuroprotective properties of the PrP-like Shadoo glycoprotein assessed in the middle cerebral artery occlusion model of ischemia. Prion 2016; 9:376-93. [PMID: 26516793 PMCID: PMC4964864 DOI: 10.1080/19336896.2015.1105432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Biochemical similarities have been noted between the natively unstructured region of the cellular prion protein, PrPC, and a GPI-linked glycoprotein called Shadoo (Sho); these proteins are encoded by the Prnp and Sprn genes, respectively. Both proteins are expressed in the adult central nervous system and they share overlapping partners, including each other, in interactome studies. As prior studies have ascribed neuroprotective properties to the N-terminal region of PrPC, specifically the octarepeat region, we investigated Sho's neuroprotective properties. To this end we assessed Sho-null (Sprn0/0) and hemizygous (Sprn0/+) mice in the middle cerebral artery occlusion (MCAO) model versus wild type mice and also vs. transgene-rescued Sprn0/0-TgSprn mice. Sprn0/0 mice had a tendency to greater fragility in reaching endpoint and deficits in parameters including infarct volume and neurogenesis, with a reciprocal trend noted in transgene-rescued mice; however these effects did not reach significance. Loss of both PrPC and Sho immunostaining occurred in parallel to neuronal loss on the ipsilateral side of MCAO-lesioned animals; while focal elevations in immunostaining in the penumbra region were sometimes evident for PrPC, they were not noted for Sho. Our studies argue against discernible neuroprotective action of Sho in the genetic backgrounds used for this MCAO paradigm. Whether or not the positively charged N-terminal regions in Sho and PrPC fulfil different roles in vivo remains to be determined.
Collapse
Affiliation(s)
- Nathalie Daude
- a Center for Prion and Protein Folding Diseases; University of Alberta ; Edmonton , AB , Canada
| | - Hristina Gapeshina
- a Center for Prion and Protein Folding Diseases; University of Alberta ; Edmonton , AB , Canada
| | - Bin Dong
- b Neurochemical Research Unit; University of Alberta ; Edmonton , AB , Canada
| | - Ian Winship
- b Neurochemical Research Unit; University of Alberta ; Edmonton , AB , Canada
| | - David Westaway
- a Center for Prion and Protein Folding Diseases; University of Alberta ; Edmonton , AB , Canada
| |
Collapse
|
42
|
Paltrinieri S, Comazzi S, Spagnolo V, Rondena M, Ponti W, Ceciliani F. Bovine Doppel (Dpl) and Prion Protein (PrP) Expression on Lymphoid Tissue and Circulating Leukocytes. J Histochem Cytochem 2016; 52:1639-45. [PMID: 15557218 DOI: 10.1369/jhc.4a6441.2004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Doppel (Dpl) protein shares some structural features with prion protein (PrP), whose pathologic isoform (PrPsc) is considered to be the causative agent of transmissible spongiform encephalopathies. Dpl is mainly expressed in testes but, when ectopically expressed in the central nervous system, is neurotoxic. We have examined the expression pattern of Dpl and PrP on bovine lymphoid tissues and circulating leukocytes. A polyclonal anti-Dpl antibody along with a panel of monoclonal antibodies specific for leukocyte membrane antigens or PrP were used to examine frozen sections from spleen, lymph nodes, and bone marrow by immunohistochemistry. Blood was analyzed by flow cytometry. Double staining was used to study the possible coexpression of the two proteins and to characterize cells expressing Dpl and/or PrP. Dpl was expressed in B-cells, in dendritic cells within lymphoid follicles, bone marrow, circulating myeloid cells, and circulating B-cells. The distribution of Dpl was quite similar to that of PrP. The only differences in expression observed concerned the low number of Dpl + cells in lymph nodes and the strong Dpl positivity of circulating granulocytes. The two proteins were rarely co-expressed, suggesting an independent expression mechanism in resting cells. The role of Dpl+ leukocytes in the pathogenesis of Dpl- or PrP-induced diseases merits further investigation.
Collapse
Affiliation(s)
- Saverio Paltrinieri
- Department of Veterinary Pathology, Hygiene and Health, University of Milan, Milan, Italy.
| | | | | | | | | | | |
Collapse
|
43
|
Disparate Modes of Evolution Shaped Modern Prion (PRNP) and Prion-Related Doppel (PRND) Variation in Domestic Cattle. PLoS One 2016; 11:e0155924. [PMID: 27224046 PMCID: PMC4880211 DOI: 10.1371/journal.pone.0155924] [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: 03/30/2016] [Accepted: 05/06/2016] [Indexed: 11/19/2022] Open
Abstract
Previous investigations aimed at determining whether the mammalian prion protein actually facilitates tangible molecular aspects of either a discrete or pleiotropic functional niche have been debated, especially given the apparent absence of overt behavioral or physiological phenotypes associated with several mammalian prion gene (PRNP) knockout experiments. Moreover, a previous evaluation of PRNP knockout cattle concluded that they were normal, suggesting that the bovine prion protein is physiologically dispensable. Herein, we examined the frequency and distribution of nucleotide sequence variation within the coding regions of bovine PRNP and the adjacent Doppel (PRND) gene, a proximal paralogue to PRNP on BTA13. Evaluation of PRND variation demonstrated that the gene does not depart from a strictly neutral model of molecular evolution, and would therefore not be expected to influence tests of selection within PRNP. Collectively, our analyses confirm that intense purifying selection is indeed occurring directly on bovine PRNP, which is indicative of a protein with an important role. These results suggest that the lack of observed fitness effects may not manifest in the controlled environmental conditions used to care for and raise PRNP knockout animals.
Collapse
|
44
|
Choi W, Kim E, Yum SY, Lee C, Lee J, Moon J, Ramachandra S, Malaweera BO, Cho J, Kim JS, Kim S, Jang G. Efficient PRNP deletion in bovine genome using gene-editing technologies in bovine cells. Prion 2016. [PMID: 26217959 DOI: 10.1080/19336896.2015.1071459] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Even though prion (encoded by the PRNP gene) diseases like bovine spongiform encephalopathy (BSE) are fatal neurodegenerative diseases in cattle, their study via gene deletion has been limited due to the absence of cell lines or mutant models. In this study, we aim to develop an immortalized fibroblast cell line in which genome-engineering technology can be readily applied to create gene-modified clones for studies. To this end, this study is designed to 1) investigate the induction of primary fibroblasts to immortalization by introducing Bmi-1 and hTert genes; 2) investigate the disruption of the PRNP in those cells; and 3) evaluate the gene expression and embryonic development using knockout (KO) cell lines. Primary cells from a male neonate were immortalized with Bmi-1and hTert. Immortalized cells were cultured for more than 180 days without any changes in their doubling time and morphology. Furthermore, to knockout the PRNP gene, plasmids that encode transcription activator-like effector nuclease (TALEN) pairs were transfected into the cells, and transfected single cells were propagated. Mutated clonal cell lines were confirmed by T7 endonuclease I assay and sequencing. Four knockout cell lines were used for somatic cell nuclear transfer (SCNT), and the resulting embryos were developed to the blastocyst stage. The genes (CSNK2A1, FAM64A, MPG and PRND) were affected after PRNP disruption in immortalized cells. In conclusion, we established immortalized cattle fibroblasts using Bmi-1 and hTert genes, and used TALENs to knockout the PRNP gene in these immortalized cells. The efficient PRNP KO is expected to be a useful technology to develop our understanding of in vitro prion protein functions in cattle.
Collapse
Affiliation(s)
- WooJae Choi
- a Laboratory of Theriogenology and Biotechnology; Department of Veterinary Clinical Science ; College of Veterinary Medicine and the Research Institute of Veterinary Science; Seoul National University ; Seoul , Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Al-Hilal TA, Chung SW, Choi JU, Alam F, Park J, Kim SW, Kim SY, Ahsan F, Kim IS, Byun Y. Targeting prion-like protein doppel selectively suppresses tumor angiogenesis. J Clin Invest 2016; 126:1251-66. [PMID: 26950422 DOI: 10.1172/jci83427] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 01/21/2016] [Indexed: 01/06/2023] Open
Abstract
Controlled and site-specific regulation of growth factor signaling remains a major challenge for current antiangiogenic therapies, as these antiangiogenic agents target normal vasculature as well tumor vasculature. In this article, we identified the prion-like protein doppel as a potential therapeutic target for tumor angiogenesis. We investigated the interactions between doppel and VEGFR2 and evaluated whether blocking the doppel/VEGFR2 axis suppresses the process of angiogenesis. We discovered that tumor endothelial cells (TECs), but not normal ECs, express doppel; tumors from patients and mouse xenografts expressed doppel in their vasculatures. Induced doppel overexpression in ECs enhanced vascularization, whereas doppel constitutively colocalized and complexed with VEGFR2 in TECs. Doppel inhibition depleted VEGFR2 from the cell membrane, subsequently inducing the internalization and degradation of VEGFR2 and thereby attenuating VEGFR2 signaling. We also synthesized an orally active glycosaminoglycan (LHbisD4) that specifically binds with doppel. We determined that LHbisD4 concentrates over the tumor site and that genetic loss of doppel in TECs decreases LHbisD4 binding and targeting both in vitro and in vivo. Moreover, LHbisD4 eliminated VEGFR2 from the cell membrane, prevented VEGF binding in TECs, and suppressed tumor growth. Together, our results demonstrate that blocking doppel can control VEGF signaling in TECs and selectively inhibit tumor angiogenesis.
Collapse
|
46
|
Nuvolone M, Hermann M, Sorce S, Russo G, Tiberi C, Schwarz P, Minikel E, Sanoudou D, Pelczar P, Aguzzi A. Strictly co-isogenic C57BL/6J-Prnp-/- mice: A rigorous resource for prion science. J Exp Med 2016; 213:313-27. [PMID: 26926995 PMCID: PMC4813672 DOI: 10.1084/jem.20151610] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 01/25/2016] [Indexed: 12/18/2022] Open
Abstract
Although its involvement in prion replication and neurotoxicity during transmissible spongiform encephalopathies is undisputed, the physiological role of the cellular prion protein (PrP(C)) remains enigmatic. A plethora of functions have been ascribed to PrP(C) based on phenotypes of Prnp(-/-) mice. However, all currently available Prnp(-/-) lines were generated in embryonic stem cells from the 129 strain of the laboratory mouse and mostly crossed to non-129 strains. Therefore, Prnp-linked loci polymorphic between 129 and the backcrossing strain resulted in systematic genetic confounders and led to erroneous conclusions. We used TALEN-mediated genome editing in fertilized mouse oocytes to create the Zurich-3 (ZH3) Prnp-ablated allele on a pure C57BL/6J genetic background. Genomic, transcriptional, and phenotypic characterization of Prnp(ZH3/ZH3) mice failed to identify phenotypes previously described in non-co-isogenic Prnp(-/-) mice. However, aged Prnp(ZH3/ZH3) mice developed a chronic demyelinating peripheral neuropathy, confirming the crucial involvement of PrP(C) in peripheral myelin maintenance. This new line represents a rigorous genetic resource for studying the role of PrP(C) in physiology and disease.
Collapse
Affiliation(s)
- Mario Nuvolone
- Institute of Neuropathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Mario Hermann
- Institute of Neuropathology, University Hospital of Zurich, 8091 Zurich, Switzerland Institute of Laboratory Animal Science, University of Zurich, 8091 Zurich, Switzerland
| | - Silvia Sorce
- Institute of Neuropathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Giancarlo Russo
- Functional Genomics Center Zurich (FGCZ), 8057 Zurich, Switzerland
| | - Cinzia Tiberi
- Institute of Neuropathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Petra Schwarz
- Institute of Neuropathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Eric Minikel
- Prion Alliance, Cambridge, MA 02139 Broad Institute, Cambridge, MA 02142 Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114
| | - Despina Sanoudou
- Fourth Department of Internal Medicine, Attikon Hospital, Medical School, University of Athens, 115 27 Athens, Greece
| | - Pawel Pelczar
- Institute of Laboratory Animal Science, University of Zurich, 8091 Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
47
|
Nyeste A, Bencsura P, Vida I, Hegyi Z, Homolya L, Fodor E, Welker E. Expression of the Prion Protein Family Member Shadoo Causes Drug Hypersensitivity That Is Diminished by the Coexpression of the Wild Type Prion Protein. J Biol Chem 2016; 291:4473-86. [PMID: 26721882 DOI: 10.1074/jbc.m115.679035] [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] [Received: 07/29/2015] [Indexed: 11/06/2022] Open
Abstract
The prion protein (PrP) seems to exert both neuroprotective and neurotoxic activities. The toxic activities are associated with the C-terminal globular parts in the absence of the flexible N terminus, specifically the hydrophobic domain (HD) or the central region (CR). The wild type prion protein (PrP-WT), having an intact flexible part, exhibits neuroprotective qualities by virtue of diminishing many of the cytotoxic effects of these mutant prion proteins (PrPΔHD and PrPΔCR) when coexpressed. The prion protein family member Doppel, which possesses a three-dimensional fold similar to the C-terminal part of PrP, is also harmful to neuronal and other cells in various models, a phenotype that can also be eliminated by the coexpression of PrP-WT. In contrast, another prion protein family member, Shadoo (Sho), a natively disordered protein possessing structural features similar to the flexible N-terminal tail of PrP, exhibits PrP-WT-like protective properties. Here, we report that, contrary to expectations, Sho expression in SH-SY5Y or HEK293 cells induces the same toxic phenotype of drug hypersensitivity as PrPΔCR. This effect is exhibited in a dose-dependent manner and is also counteracted by the coexpression of PrP-WT. The opposing effects of Shadoo in different model systems revealed here may be explored to help discern the relationship of the various toxic activities of mutant PrPs with each other and the neurotoxic effects seen in neurodegenerative diseases, such as transmissible spongiform encephalopathy and Alzheimer disease.
Collapse
Affiliation(s)
- Antal Nyeste
- From the Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary
| | - Petra Bencsura
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| | - István Vida
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and the Institute of Chemistry, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Zoltán Hegyi
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| | - László Homolya
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| | - Elfrieda Fodor
- From the Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary
| | - Ervin Welker
- From the Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary, the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| |
Collapse
|
48
|
Allais-Bonnet A, Castille J, Pannetier M, Passet B, Elzaïat M, André M, Montazer-Torbati F, Moazami-Goudarzi K, Vilotte JL, Pailhoux E. A specific role for PRND in goat foetal Leydig cells is suggested by prion family gene expression during gonad development in goats and mice. FEBS Open Bio 2016; 6:4-15. [PMID: 27047737 PMCID: PMC4794797 DOI: 10.1002/2211-5463.12002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 10/22/2015] [Accepted: 11/20/2015] [Indexed: 12/17/2022] Open
Abstract
Three genes of the prion protein gene family are expressed in gonads. Comparative analyses of their expression patterns in mice and goats revealed constant expression of PRNP and SPRN in both species and in both male and female gonads, but with a weaker expression of SPRN. By contrast, expression of PRND was found to be sex‐dimorphic, in agreement with its role in spermatogenesis. More importantly, our study revealed that PRND seems to be a key marker of foetal Leydig cells specifically in goats, suggesting a yet unknown role for its encoded protein Doppel during gonadal differentiation in nonrodent mammals.
Collapse
Affiliation(s)
- Aurélie Allais-Bonnet
- Biologie du Développement et Reproduction INRA, UMR 1198 Jouy-en-Josas France; ALLICE Paris France
| | - Johan Castille
- Génétique Animale et Biologie Intégrative INRA, UMR 1313 Jouy-en-Josas France
| | - Maëlle Pannetier
- Biologie du Développement et Reproduction INRA, UMR 1198 Jouy-en-Josas France
| | - Bruno Passet
- Génétique Animale et Biologie Intégrative INRA, UMR 1313 Jouy-en-Josas France
| | - Maëva Elzaïat
- Biologie du Développement et Reproduction INRA, UMR 1198 Jouy-en-Josas France
| | - Marjolaine André
- Biologie du Développement et Reproduction INRA, UMR 1198 Jouy-en-Josas France
| | | | | | - Jean-Luc Vilotte
- Génétique Animale et Biologie Intégrative INRA, UMR 1313 Jouy-en-Josas France
| | - Eric Pailhoux
- Biologie du Développement et Reproduction INRA, UMR 1198 Jouy-en-Josas France
| |
Collapse
|
49
|
Schmitz M, Zafar S, Silva CJ, Zerr I. Behavioral abnormalities in prion protein knockout mice and the potential relevance of PrP(C) for the cytoskeleton. Prion 2015; 8:381-6. [PMID: 25517431 DOI: 10.4161/19336896.2014.983746] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The cellular prion protein (PrP(C)) is a highly conserved protein, which is anchored to the outer surface of the plasma membrane. Even though its physiological function has already been investigated in different cell or mouse models where PrP(C) expression is either upregulated or depleted, its exact physiological role in a mammalian organism remains elusive. Recent studies indicate that PrP(C) has multiple functions and is involved in cognition, learning, anxiety, locomotion, depression, offensive aggression and nest building behavior. While young animals (3 months of age) show only marginal abnormalities, most of the deficits become apparent as the animals age, which might indicate its role in neurodegeneration or neuroprotection. However, the exact biochemical mechanism and signal transduction pathways involving PrP(C) are only gradually becoming clearer. We report the observations made in different studies using different Prnp0/0 mouse models and propose that PrP(C) plays an important role in the regulation of the cytoskeleton and associated proteins. In particular, we showed a nocodazole treatment influenced colocalization of PrP(C) and α tubulin 1. In addition, we confirmed the observed deficits in nest building using a different backcrossed Prnp0/0 mouse line.
Collapse
Affiliation(s)
- Matthias Schmitz
- a Department of Neurology ; University Medical Center Göttingen, and German Center for Neurodegenerative Diseases (DZNE) ; Göttingen , Germany
| | | | | | | |
Collapse
|
50
|
Stroylova YY, Kiselev GG, Schmalhausen EV, Muronetz VI. Prions and chaperones: friends or foes? BIOCHEMISTRY (MOSCOW) 2015; 79:761-75. [PMID: 25365486 DOI: 10.1134/s0006297914080045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This review highlights the modern perception of anomalous folding of the prion protein and the role of chaperones therein. Special attention is paid to prion proteins from mammalian species, which are prone to amyloid-like prion diseases due to a unique aggregation pathway. Despite being a significantly popular current subject of investigations, the etiology, structure, and function of both normal and anomalous prion proteins still hold many mysteries. The most interesting of those are connected to the interaction with chaperone system, which is responsible for stabilizing protein structure and disrupting aggregates. In the case of prion proteins the following question is of the most importance - can chaperones influence different stages of the formation of pathological aggregates (these vary from intermediate oligomers to mature amyloid-like fibrils) and the whole transition from native prion protein to its amyloid-like fibril-enriched form? The existing inconsistencies and ambiguities in the observations made so far can be attributed to the fact that most of the investigations did not take into account the type and functional state of the chaperones. This review discusses in detail our previous works that have demonstrated fundamental differences between eukaryotic and prokaryotic chaperones in the action exerted on the amyloid-like transformation of the prion protein along with the dependence of the observed effects on the functional state of the chaperone.
Collapse
Affiliation(s)
- Y Y Stroylova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.
| | | | | | | |
Collapse
|