101
|
Petit-Paitel A, Ménard B, Guyon A, Béringue V, Nahon JL, Zsürger N, Chabry J. Prion protein is a key determinant of alcohol sensitivity through the modulation of N-methyl-D-aspartate receptor (NMDAR) activity. PLoS One 2012; 7:e34691. [PMID: 22536327 PMCID: PMC3335038 DOI: 10.1371/journal.pone.0034691] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 03/06/2012] [Indexed: 11/21/2022] Open
Abstract
The prion protein (PrP) is absolutely required for the development of prion diseases; nevertheless, its physiological functions in the central nervous system remain elusive. Using a combination of behavioral, electrophysiological and biochemical approaches in transgenic mouse models, we provide strong evidence for a crucial role of PrP in alcohol sensitivity. Indeed, PrP knock out (PrP−/−) mice presented a greater sensitivity to the sedative effects of EtOH compared to wild-type (wt) control mice. Conversely, compared to wt mice, those over-expressing mouse, human or hamster PrP genes presented a relative insensitivity to ethanol-induced sedation. An acute tolerance (i.e. reversion) to ethanol inhibition of N-methyl-D-aspartate (NMDA) receptor-mediated excitatory post-synaptic potentials in hippocampal slices developed slower in PrP−/− mice than in wt mice. We show that PrP is required to induce acute tolerance to ethanol by activating a Src-protein tyrosine kinase-dependent intracellular signaling pathway. In an attempt to decipher the molecular mechanisms underlying PrP-dependent ethanol effect, we looked for changes in lipid raft features in hippocampus of ethanol-treated wt mice compared to PrP−/− mice. Ethanol induced rapid and transient changes of buoyancy of lipid raft-associated proteins in hippocampus of wt but not PrP−/− mice suggesting a possible mechanistic link for PrP-dependent signal transduction. Together, our results reveal a hitherto unknown physiological role of PrP on the regulation of NMDAR activity and highlight its crucial role in synaptic functions.
Collapse
Affiliation(s)
- Agnès Petit-Paitel
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Baptiste Ménard
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Alice Guyon
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Vincent Béringue
- Institut National de la Recherche Agronomique, UR892, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Jean-Louis Nahon
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Nicole Zsürger
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Joëlle Chabry
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- * E-mail:
| |
Collapse
|
102
|
Abstract
Prion protein (PrP) can be considered a pivotal molecule because it interacts with several partners to perform a diverse range of critical biological functions that might differ in embryonic and adult cells. In recent years, there have been major advances in elucidating the putative role of PrP in the basic biology of stem cells in many different systems. Here, we review the evidence indicating that PrP is a key molecule involved in driving different aspects of the potency of embryonic and tissue-specific stem cells in self-perpetuation and differentiation in many cell types. It has been shown that PrP is involved in stem cell self-renewal, controlling pluripotency gene expression, proliferation, and neural and cardiomyocyte differentiation. PrP also has essential roles in distinct processes that regulate tissue-specific stem cell biology in nervous and hematopoietic systems and during muscle regeneration. Results from our own investigations have shown that PrP is able to modulate self-renewal and proliferation in neural stem cells, processes that are enhanced by PrP interactions with stress inducible protein 1 (STI1). Thus, the available data reveal the influence of PrP in acting upon the maintenance of pluripotent status or the differentiation of stem cells from the early embryogenesis through adulthood.
Collapse
Affiliation(s)
- Marilene H Lopes
- Department of Cell and Developmental Biology, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil.
| | | |
Collapse
|
103
|
Portaluppi F, Tiseo R, Smolensky MH, Hermida RC, Ayala DE, Fabbian F. Circadian rhythms and cardiovascular health. Sleep Med Rev 2012; 16:151-166. [PMID: 21641838 DOI: 10.1016/j.smrv.2011.04.003] [Citation(s) in RCA: 208] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 04/27/2011] [Indexed: 11/30/2022]
Abstract
The functional organization of the cardiovascular system shows clear circadian rhythmicity. These and other circadian rhythms at all levels of organization are orchestrated by a central biological clock, the suprachiasmatic nuclei of the hypothalamus. Preservation of the normal circadian time structure from the level of the cardiomyocyte to the organ system appears to be essential for cardiovascular health and cardiovascular disease prevention. Myocardial ischemia, acute myocardial infarct, and sudden cardiac death are much greater in incidence than expected in the morning. Moreover, supraventricular and ventricular cardiac arrhythmias of various types show specific day-night patterns, with atrial arrhythmias--premature beats, tachycardias, atrial fibrillation, and flutter - generally being of higher frequency during the day than night--and ventricular fibrillation and ventricular premature beats more common, respectively, in the morning and during the daytime activity than sleep span. Furthermore, different circadian patterns of blood pressure are found in arterial hypertension, in relation to different cardiovascular morbidity and mortality risk. Such temporal patterns result from circadian periodicity in pathophysiological mechanisms that give rise to predictable-in-time differences in susceptibility-resistance to cyclic environmental stressors that trigger these clinical events. Circadian rhythms also may affect the pharmacokinetics and pharmacodynamics of cardiovascular and other medications. Knowledge of 24-h patterns in the risk of cardiac arrhythmias and cardiovascular disease morbidity and mortality plus circadian rhythm-dependencies of underlying pathophysiologic mechanisms suggests the requirement for preventive and therapeutic interventions is not the same throughout the day and night, and should be tailored accordingly to improve outcomes.
Collapse
|
104
|
Hedlin P, Taschuk R, Potter A, Griebel P, Napper S. Detection and control of prion diseases in food animals. ISRN VETERINARY SCIENCE 2012; 2012:254739. [PMID: 23738120 PMCID: PMC3658581 DOI: 10.5402/2012/254739] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/22/2011] [Indexed: 12/14/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs), or prion diseases, represent a unique form of infectious disease based on misfolding of a self-protein (PrPC) into a pathological, infectious conformation (PrPSc). Prion diseases of food animals gained notoriety during the bovine spongiform encephalopathy (BSE) outbreak of the 1980s. In particular, disease transmission to humans, to the generation of a fatal, untreatable disease, elevated the perspective on livestock prion diseases from food production to food safety. While the immediate threat posed by BSE has been successfully addressed through surveillance and improved management practices, another prion disease is rapidly spreading. Chronic wasting disease (CWD), a prion disease of cervids, has been confirmed in wild and captive populations with devastating impact on the farmed cervid industries. Furthermore, the unabated spread of this disease through wild populations threatens a natural resource that is a source of considerable economic benefit and national pride. In a worst-case scenario, CWD may represent a zoonotic threat either through direct transmission via consumption of infected cervids or through a secondary food animal, such as cattle. This has energized efforts to understand prion diseases as well as to develop tools for disease detection, prevention, and management. Progress in each of these areas is discussed.
Collapse
Affiliation(s)
- Peter Hedlin
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK, Canada S7N 5E3 ; Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada S7N 5E3
| | | | | | | | | |
Collapse
|
105
|
Fisher SP, Godinho SI, Pothecary CA, Hankins MW, Foster RG, Peirson SN. Rapid assessment of sleep-wake behavior in mice. J Biol Rhythms 2012; 27:48-58. [PMID: 22306973 PMCID: PMC4650254 DOI: 10.1177/0748730411431550] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Sleep is a fundamental biological rhythm involving the interaction of numerous brain structures and diverse neurotransmitter systems. The primary measures used to define sleep are the electroencephalogram (EEG) and electromyogram (EMG). However, EEG-based methods are often unsuitable for use in high-throughput screens as they are time-intensive and involve invasive surgery. As such, the dissection of sleep mechanisms and the discovery of novel drugs that modulate sleep would benefit greatly from further development of rapid behavioral assays to assess sleep in animal models. Here is described an automated noninvasive approach to evaluate sleep duration, latency, and fragmentation using video tracking of mice in their home cage. This approach provides a high correlation with EEG/EMG measures under both baseline conditions and following administration of pharmacological agents. Moreover, the dose-dependent effects of sedatives, stimulants, and light can be readily detected. This approach is robust yet relatively inexpensive to implement and can be easily incorporated into ongoing screening programs to provide a powerful first-pass screen for assessing sleep and allied behaviors.
Collapse
Affiliation(s)
| | - Sofia I.H. Godinho
- Nuffield Laboratory of Ophthalmology, University of Oxford, Levels 5-6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, United Kingdom
| | - Carina A. Pothecary
- Nuffield Laboratory of Ophthalmology, University of Oxford, Levels 5-6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, United Kingdom
| | - Mark W. Hankins
- Nuffield Laboratory of Ophthalmology, University of Oxford, Levels 5-6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, United Kingdom
| | - Russell G. Foster
- Nuffield Laboratory of Ophthalmology, University of Oxford, Levels 5-6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, United Kingdom
| | - Stuart N. Peirson
- Nuffield Laboratory of Ophthalmology, University of Oxford, Levels 5-6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
106
|
Gourdain P, Ballerini C, Nicot AB, Carnaud C. Exacerbation of experimental autoimmune encephalomyelitis in prion protein (PrPc)-null mice: evidence for a critical role of the central nervous system. J Neuroinflammation 2012; 9:25. [PMID: 22281016 PMCID: PMC3305405 DOI: 10.1186/1742-2094-9-25] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 01/26/2012] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The cellular prion protein (PrPc) is a host-encoded glycoprotein whose transconformation into PrP scrapie (PrPSc) initiates prion diseases. The role of PrPc in health is still obscure, but many candidate functions have been attributed to the protein, both in the immune and the nervous systems. Recent data show that experimental autoimmune encephalomyelitis (EAE) is worsened in mice lacking PrPc. Disease exacerbation has been attributed to T cells that would differentiate into more aggressive effectors when deprived of PrPc. However, alternative interpretations such as reduced resistance of neurons to autoimmune insult and exacerbated gliosis leading to neuronal deficits were not considered. METHOD To better discriminate the contribution of immune cells versus neural cells, reciprocal bone marrow chimeras with differential expression of PrPc in the lymphoid or in the central nervous system (CNS) were generated. Mice were subsequently challenged with MOG35-55 peptide and clinical disease as well as histopathology were compared in both groups. Furthermore, to test directly the T cell hypothesis, we compared the encephalitogenicity of adoptively transferred PrPc-deficient versus PrPc-sufficient, anti-MOG T cells. RESULTS First, EAE exacerbation in PrPc-deficient mice was confirmed. Irradiation exacerbated EAE in all the chimeras and controls, but disease was more severe in mice with a PrPc-deleted CNS and a normal immune system than in the reciprocal construction. Moreover, there was no indication that anti-MOG responses were different in PrPc-sufficient and PrPc-deficient mice. Paradoxically, PrPc-deficient anti-MOG 2D2 T cells were less pathogenic than PrPc-expressing 2D2 T cells. CONCLUSIONS In view of the present data, it can be concluded that the origin of EAE exacerbation in PrPc-ablated mice resides in the absence of the prion protein in the CNS. Furthermore, the absence of PrPc on both neural and immune cells does not synergize for disease worsening. These conclusions highlight the critical role of PrPc in maintaining the integrity of the CNS in situations of stress, especially during a neuroinflammatory insult.
Collapse
Affiliation(s)
- Pauline Gourdain
- INSERM, UMR S 938, Centre de Recherche Hôpital Saint-Antoine, Paris, France
| | | | | | | |
Collapse
|
107
|
Fulda S, Romanowski CPN, Becker A, Wetter TC, Kimura M, Fenzel T. Rapid eye movements during sleep in mice: high trait-like stability qualifies rapid eye movement density for characterization of phenotypic variation in sleep patterns of rodents. BMC Neurosci 2011; 12:110. [PMID: 22047102 PMCID: PMC3228710 DOI: 10.1186/1471-2202-12-110] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 11/02/2011] [Indexed: 01/13/2023] Open
Abstract
Background In humans, rapid eye movements (REM) density during REM sleep plays a prominent role in psychiatric diseases. Especially in depression, an increased REM density is a vulnerability marker for depression. In clinical practice and research measurement of REM density is highly standardized. In basic animal research, almost no tools are available to obtain and systematically evaluate eye movement data, although, this would create increased comparability between human and animal sleep studies. Methods We obtained standardized electroencephalographic (EEG), electromyographic (EMG) and electrooculographic (EOG) signals from freely behaving mice. EOG electrodes were bilaterally and chronically implanted with placement of the electrodes directly between the musculus rectus superior and musculus rectus lateralis. After recovery, EEG, EMG and EOG signals were obtained for four days. Subsequent to the implantation process, we developed and validated an Eye Movement scoring in Mice Algorithm (EMMA) to detect REM as singularities of the EOG signal, based on wavelet methodology. Results The distribution of wakefulness, non-REM (NREM) sleep and rapid eye movement (REM) sleep was typical of nocturnal rodents with small amounts of wakefulness and large amounts of NREM sleep during the light period and reversed proportions during the dark period. REM sleep was distributed correspondingly. REM density was significantly higher during REM sleep than NREM sleep. REM bursts were detected more often at the end of the dark period than the beginning of the light period. During REM sleep REM density showed an ultradian course, and during NREM sleep REM density peaked at the beginning of the dark period. Concerning individual eye movements, REM duration was longer and amplitude was lower during REM sleep than NREM sleep. The majority of single REM and REM bursts were associated with micro-arousals during NREM sleep, but not during REM sleep. Conclusions Sleep-stage specific distributions of REM in mice correspond to human REM density during sleep. REM density, now also assessable in animal models through our approach, is increased in humans after acute stress, during PTSD and in depression. This relationship can now be exploited to match animal models more closely to clinical situations, especially in animal models of depression.
Collapse
Affiliation(s)
- Stephany Fulda
- Max Planck Institute of Psychiatry, Kraepelinstrasse 2, 80804 Munich, Germany
| | | | | | | | | | | |
Collapse
|
108
|
Graham JF, Kurian D, Agarwal S, Toovey L, Hunt L, Kirby L, Pinheiro TJT, Banner SJ, Gill AC. Na+/K+-ATPase is present in scrapie-associated fibrils, modulates PrP misfolding in vitro and links PrP function and dysfunction. PLoS One 2011; 6:e26813. [PMID: 22073199 PMCID: PMC3206849 DOI: 10.1371/journal.pone.0026813] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 10/04/2011] [Indexed: 12/21/2022] Open
Abstract
Transmissible spongiform encephalopathies are characterised by widespread deposition of fibrillar and/or plaque-like forms of the prion protein. These aggregated forms are produced by misfolding of the normal prion protein, PrPC, to the disease-associated form, PrPSc, through mechanisms that remain elusive but which require either direct or indirect interaction between PrPC and PrPSc isoforms. A wealth of evidence implicates other non-PrP molecules as active participants in the misfolding process, to catalyse and direct the conformational conversion of PrPC or to provide a scaffold ensuring correct alignment of PrPC and PrPSc during conversion. Such molecules may be specific to different scrapie strains to facilitate differential prion protein misfolding. Since molecular cofactors may become integrated into the growing protein fibril during prion conversion, we have investigated the proteins contained in prion disease-specific deposits by shotgun proteomics of scrapie-associated fibrils (SAF) from mice infected with 3 different strains of mouse-passaged scrapie. Concomitant use of negative control preparations allowed us to identify and discount proteins that are enriched non-specifically by the SAF isolation protocol. We found several proteins that co-purified specifically with SAF from infected brains but none of these were reproducibly and demonstrably specific for particular scrapie strains. The α-chain of Na+/K+-ATPase was common to SAF from all 3 strains and we tested the ability of this protein to modulate in vitro misfolding of recombinant PrP. Na+/K+-ATPase enhanced the efficiency of disease-specific conversion of recombinant PrP suggesting that it may act as a molecular cofactor. Consistent with previous results, the same protein inhibited fibrillisation kinetics of recombinant PrP. Since functional interactions between PrPC and Na+/K+-ATPase have previously been reported in astrocytes, our data highlight this molecule as a key link between PrP function, dysfunction and misfolding.
Collapse
Affiliation(s)
- James F. Graham
- The Roslin Institute and R(D)SVS, Neuropathogenesis Division, University of Edinburgh, Easter Bush, Roslin, Edinburgh, Midlothian, United Kingdom
| | - Dominic Kurian
- Institute for Animal Health, Compton, Newbury, Berkshire, United Kingdom
| | - Sonya Agarwal
- The Roslin Institute and R(D)SVS, Neuropathogenesis Division, University of Edinburgh, Easter Bush, Roslin, Edinburgh, Midlothian, United Kingdom
| | - Lorna Toovey
- The Roslin Institute and R(D)SVS, Neuropathogenesis Division, University of Edinburgh, Easter Bush, Roslin, Edinburgh, Midlothian, United Kingdom
| | - Lawrence Hunt
- Institute for Animal Health, Compton, Newbury, Berkshire, United Kingdom
| | - Louise Kirby
- The Roslin Institute and R(D)SVS, Neuropathogenesis Division, University of Edinburgh, Easter Bush, Roslin, Edinburgh, Midlothian, United Kingdom
| | | | - Steven J. Banner
- The Roslin Institute and R(D)SVS, Neuropathogenesis Division, University of Edinburgh, Easter Bush, Roslin, Edinburgh, Midlothian, United Kingdom
| | - Andrew C. Gill
- The Roslin Institute and R(D)SVS, Neuropathogenesis Division, University of Edinburgh, Easter Bush, Roslin, Edinburgh, Midlothian, United Kingdom
- * E-mail:
| |
Collapse
|
109
|
Abstract
Sleep is a fundamental behavior in higher animals that has been firmly established to be under substantial genetic control. However, the identification of individual genes responsible for primary sleep-wake traits has largely eluded researchers. Genetic studies in animal models have uncovered a variety of genomic loci associated with specific traits, validated the role of key neurotransmitter systems (i.e., monoamines) in sleep-wake regulation, identified novel and unexpected genes responsible for controlling sleep-wake traits, and demonstrated substantial genetic overlap in the regulation of sleep and circadian rhythms. Future studies are expected to reveal additional genes and gene networks underlying certain sleep-wake traits, thereby advancing our understanding of the molecular basis of sleep, which may suggest answers to the ultimate question of why we sleep as well as provide unique insight into the relationship between sleep and chronic diseases.
Collapse
Affiliation(s)
- Keith C. Summa
- Center for Sleep and Circadian Biology, Northwestern University
| | - Fred W. Turek
- Center for Sleep and Circadian Biology, Northwestern University
- Charles E. and Emma H. Morrison Professor of Biology, Department of Neurobiology and Physiology, Northwestern University
| |
Collapse
|
110
|
Miranda A, Pericuesta E, Ramírez MÁ, Gutierrez-Adan A. Prion protein expression regulates embryonic stem cell pluripotency and differentiation. PLoS One 2011; 6:e18422. [PMID: 21483752 PMCID: PMC3070729 DOI: 10.1371/journal.pone.0018422] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 03/06/2011] [Indexed: 01/06/2023] Open
Abstract
Cellular prion protein (PRNP) is a glycoprotein involved in the pathogenesis of transmissible spongiform encephalopathies (TSEs). Although the physiological function of PRNP is largely unknown, its key role in prion infection has been extensively documented. This study examines the functionality of PRNP during the course of embryoid body (EB) differentiation in mouse Prnp-null (KO) and WT embryonic stem cell (ESC) lines. The first feature observed was a new population of EBs that only appeared in the KO line after 5 days of differentiation. These EBs were characterized by their expression of several primordial germ cell (PGC) markers until Day 13. In a comparative mRNA expression analysis of genes playing an important developmental role during ESC differentiation to EBs, Prnp was found to participate in the transcription of a key pluripotency marker such as Nanog. A clear switching off of this gene on Day 5 was observed in the KO line as opposed to the WT line, in which maximum Prnp and Nanog mRNA levels appeared at this time. Using a specific antibody against PRNP to block PRNP pathways, reduced Nanog expression was confirmed in the WT line. In addition, antibody-mediated inhibition of ITGB5 (integrin αvβ5) in the KO line rescued the low expression of Nanog on Day 5, suggesting the regulation of Nanog transcription by Prnp via this Itgb5. mRNA expression analysis of the PRNP-related proteins PRND (Doppel) and SPRN (Shadoo), whose PRNP function is known to be redundant, revealed their incapacity to compensate for the absence of PRNP during early ESC differentiation. Our findings provide strong evidence for a relationship between Prnp and several key pluripotency genes and attribute Prnp a crucial role in regulating self-renewal/differentiation status of ESC, confirming the participation of PRNP during early embryogenesis.
Collapse
Affiliation(s)
- Alberto Miranda
- Departamento de Reproducción Animal y Conservación de Recursos Zoogenéticos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain.
| | | | | | | |
Collapse
|
111
|
Benvegnù S, Roncaglia P, Agostini F, Casalone C, Corona C, Gustincich S, Legname G. Developmental influence of the cellular prion protein on the gene expression profile in mouse hippocampus. Physiol Genomics 2011; 43:711-25. [PMID: 21406608 DOI: 10.1152/physiolgenomics.00205.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The conversion of the cellular prion protein (PrP(C)) to an abnormal and protease-resistant isoform is the key event in prion diseases. Mice lacking PrP(C) are resistant to prion infection, and downregulation of PrP(C) during prion infection prevents neuronal loss and the progression to clinical disease. These results are suggestive of the potential beneficial effect of silencing PrP(C) during prion diseases. However, the silencing of a protein that is widely expressed throughout the central nervous system could be detrimental to brain homeostasis. The physiological role of PrP(C) remains still unclear, but several putative functions (e.g., neuronal development and maintenance) have been proposed. To assess the influence of PrP(C) on gene expression profile in the mouse brain, we undertook a microarray analysis by using RNA isolated from the hippocampus at two different developmental stages: newborn (4.5-day-old) and adult (3-mo-old) mice, both from wild-type and Prnp(0/0) animals. Comparing the different datasets allowed us to identify "commonly" co-regulated genes and "uniquely" deregulated genes during postnatal development. The absence of PrP(C) affected several biological pathways, the most representative being cell signaling, cell-cell communication and transduction processes, calcium homeostasis, nervous system development, synaptic transmission, and cell adhesion. However, there was only a moderate alteration of the gene expression profile in our animal models. PrP(C) deficiency did not lead to a dramatic alteration of gene expression profile and produced moderately altered gene expression levels from young to adult animals. Thus, our results may provide additional support to silencing endogenous PrP(C) levels as therapeutic approach to prion diseases.
Collapse
Affiliation(s)
- Stefano Benvegnù
- Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste
| | | | | | | | | | | | | |
Collapse
|
112
|
Wisniewski T, Goñi F. Immunomodulation for prion and prion-related diseases. Expert Rev Vaccines 2011; 9:1441-52. [PMID: 21105779 DOI: 10.1586/erv.10.131] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Prion diseases are a unique category of illness, affecting both animals and humans, where the underlying pathogenesis is related to a conformational change of a normal self protein called cellular prion protein to a pathological and infectious conformer known as scrapie prion protein (PrP(Sc)). Currently, all prion diseases lack effective treatment and are universally fatal. Past experiences with bovine spongiform encephalopathy and variant Creutzfeldt-Jakob disease mainly in Europe, as well as the current epidemic of chronic wasting disease in North America, have highlighted the need to develop prophylactic and/or therapeutic approaches. In Alzheimer's disease that, like prion disease, is a conformational neurodegenerative disorder, both passive and active immunization has been shown to be highly effective in model animals at preventing disease and cognitive deficits, with emerging data from human trials suggesting that this approach is able to reduce amyloid-related pathology. However, any immunomodulatory approach aimed at a self-antigen has to finely balance an effective humoral immune response with potential autoimmune toxicity. The prion diseases most commonly acquired by infection typically have the alimentary tract as a portal of infectious agent entry. This makes mucosal immunization a potentially attractive method to produce a local immune response that partially or completely prevents prion entry across the gut barrier, while at the same time producing modulated systemic immunity that is unlikely to be associated with toxicity. Our results using an attenuated Salmonella vaccine strain expressing the prion protein showed that mucosal vaccination can protect against prion infection from a peripheral source, suggesting the feasibility of this approach. It is also possible to develop active and/or passive immunomodulatory approaches that more specifically target PrP(Sc) or target the shared pathological conformer found in numerous conformational disorders. Such approaches could have a significant impact on many of the common age-associated dementias.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Department of Psychiatry, Millhauser Laboratories, Room HN419, New York University School of Medicine, 560 First Avenue, New York, NY 10016, USA.
| | | |
Collapse
|
113
|
Abstract
OBJECTIVES We previously demonstrated that the expression of cellular prion protein (PrPC) in islet [beta]-cells is suppressed in hyperglycemic rats suggesting a major role for PrPC in blood glucose regulation. To further characterize the function of PrPC in glucose homeostasis, we studied glucoregulation in PrPC knockout (PrPC KO) mice. METHODS Glucose tolerance, insulin secretion, and insulin sensitivity were analyzed to assess glucoregulation in Zrch I PrPC KO and the C57BL/6 (control) mice. Immunohistochemistry and morphometry were used to measure [beta]-cell mass. RESULTS Male PrPC KO mice had significantly increased blood glucose concentration 60, 120, and 180 minutes after intraperitoneal injection of glucose compared with C57BL/6 mice. Female PrPC KO mice showed a less pronounced phenotype of glucose intolerance. Evaluation of [beta]-cell mass, insulin and proinsulin deficiency, and insulin resistance in male mice revealed essentially no difference between PrPC KO and control mice. The only exception was an increase in serum insulin concentration in male PrPC KO mice 5 minutes after glucose injection. CONCLUSIONS This report is the first to show that PrPC in [beta]-cells is involved in glucoregulation. A further understanding of the role of PrPC in regulating [beta]-cell function will provide valuable insight into the mechanisms of blood glucose regulation.
Collapse
|
114
|
|
115
|
Pushie MJ, Pickering IJ, Martin GR, Tsutsui S, Jirik FR, George GN. Prion protein expression level alters regional copper, iron and zinc content in the mouse brain. Metallomics 2011; 3:206-14. [PMID: 21264406 DOI: 10.1039/c0mt00037j] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The central role of the prion protein (PrP) in a family of fatal neurodegenerate diseases has garnered considerable research interest over the past two decades. Moreover, the role of PrP in neuronal development, as well as its apparent role in metal homeostasis, is increasingly of interest. The host-encoded form of the prion protein (PrP(C)) binds multiple copper atoms via its N-terminal domain and can influence brain copper and iron levels. The importance of PrP(C) to the regulation of brain metal homeostasis and metal distribution, however, is not fully understood. We therefore employed synchrotron-based X-ray fluorescence imaging to map the level and distributions of several key metals in the brains of mice that express different levels of PrP(C). Brain sections from wild-type, prion gene knockout (Prnp(-/-)) and PrP(C) over-expressing mice revealed striking variation in the levels of iron, copper, and even zinc in specific brain regions as a function of PrP(C) expression. Our results indicate that one important function of PrP(C) may be to regulate the amount and distribution of specific metals within the central nervous system. This raises the possibility that PrP(C) levels, or its activity, might regulate the progression of diseases in which altered metal homeostasis is thought to play a pathogenic role such as Alzheimer's, Parkinson's and Wilson's diseases and disorders such as hemochromatosis.
Collapse
Affiliation(s)
- M Jake Pushie
- Molecular and Environmental Science Research Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan, S7N 5E2, Canada.
| | | | | | | | | | | |
Collapse
|
116
|
YAN MM, XU XH, HUANG ZL, YAO MH, URADE Y, QU WM. Selection of optimal epoch duration in assessment of rodent sleep-wake profiles. Sleep Biol Rhythms 2011. [DOI: 10.1111/j.1479-8425.2010.00484.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
117
|
Affiliation(s)
- Chiara Cirelli
- Department of Psychiatry, University of Wisconsin, Madison, WI 53719, USA.
| | | |
Collapse
|
118
|
Abstract
Prion diseases in humans and animals are characterized by progressive neurodegeneration and the formation of infectious particles called prions. Both features are intimately linked to a conformational transition of the cellular prion protein (PrP(C)) into aberrantly folded conformers with neurotoxic and self-replicating activities. Interestingly, there is increasing evidence that the infectious and neurotoxic properties of PrP conformers are not necessarily coupled. Transgenic mouse models revealed that some PrP mutants interfere with neuronal function in the absence of infectious prions. Vice versa, propagation of prions can occur without causing neurotoxicity. Consequently, it appears plausible that two partially independent pathways exist, one pathway leading to the propagation of infectious prions and another one that mediates neurotoxic signaling. In this review we will summarize current knowledge of neurotoxic PrP conformers and discuss the role of PrP(C) as a mediator of both stress-protective and neurotoxic signaling cascades.
Collapse
|
119
|
Montagna P. Fatal familial insomnia and the role of the thalamus in sleep regulation. HANDBOOK OF CLINICAL NEUROLOGY 2011; 99:981-996. [PMID: 21056239 DOI: 10.1016/b978-0-444-52007-4.00018-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Affiliation(s)
- Pasquale Montagna
- Department of Neurological Sciences, University of Bologna Medical School, Bologna, Italy.
| |
Collapse
|
120
|
Borbély AA, Tobler I. Manifestations and functional implications of sleep homeostasis. HANDBOOK OF CLINICAL NEUROLOGY 2011; 98:205-213. [PMID: 21056188 DOI: 10.1016/b978-0-444-52006-7.00013-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Affiliation(s)
- Alexander A Borbély
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
| | | |
Collapse
|
121
|
Park Y, Kim W, Kim AY, Choi HJ, Choi JK, Park N, Koh EK, Seo J, Koh YH. Normal prion protein in Drosophila enhances the toxicity of pathogenic polyglutamine proteins and alters susceptibility to oxidative and autophagy signaling modulators. Biochem Biophys Res Commun 2010; 404:638-45. [PMID: 21146501 DOI: 10.1016/j.bbrc.2010.12.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 12/05/2010] [Indexed: 12/18/2022]
Abstract
To investigate the in vivo functions of normal prion protein (PrP) in Drosophila, we utilized characterized transgenic flies expressing ³(F)⁴-tagged mouse PrP (Mo-PrP³(F)⁴). The neurotoxicity of pathogenic Machado-Joseph Disease (MJD) glutamine (Q) 78 and 127Q proteins were enhanced by the co-expression of Mo-PrP³(F)⁴in the fly eyes, while the eyes of controls flies and flies expressing Mo-PrP³(F)⁴) alone or together with MJD-Q27 or 20Q proteins did not show any defect. Susceptibilities to H₂O₂, paraquat, and Dithiothreitol (DTT) were altered in Mo-PrP³(F)⁴ flies. In addition, Mo-PrP³(F)⁴ flies were significantly more susceptible to the perturbation of autophagy signaling by an autophagy inhibitor, 3-methyladenine (3-MA), and inducer, LiCl. Taken together, our data suggest that Mo-PrP³(F)⁴ may enhance the neurotoxicity of pathogenic Poly-Q proteins by perturbing oxidative and autophagy signaling.
Collapse
Affiliation(s)
- Yunwoong Park
- ILSONG Institute of Life Science, Hallym University, Anyang, Gyeonggi-do 431-060, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Wadsworth JDF, Asante EA, Collinge J. Review: contribution of transgenic models to understanding human prion disease. Neuropathol Appl Neurobiol 2010; 36:576-97. [PMID: 20880036 PMCID: PMC3017745 DOI: 10.1111/j.1365-2990.2010.01129.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 09/16/2010] [Indexed: 01/28/2023]
Abstract
Transgenic mice expressing human prion protein in the absence of endogenous mouse prion protein faithfully replicate human prions. These models reproduce all of the key features of human disease, including long clinically silent incubation periods prior to fatal neurodegeneration with neuropathological phenotypes that mirror human prion strain diversity. Critical contributions to our understanding of human prion disease pathogenesis and aetiology have only been possible through the use of transgenic mice. These models have provided the basis for the conformational selection model of prion transmission barriers and have causally linked bovine spongiform encephalopathy with variant Creutzfeldt-Jakob disease. In the future these models will be essential for evaluating newly identified potentially zoonotic prion strains, for validating effective methods of prion decontamination and for developing effective therapeutic treatments for human prion disease.
Collapse
Affiliation(s)
- J D F Wadsworth
- MRC Prion Unit and Department of Neurodegenerative Disease, Institute of Neurology, University College London, National Hospital for Neurology and Neurosurgery, London, UK.
| | | | | |
Collapse
|
123
|
Abstract
The prion diseases are a family of rare neurodegenerative disorders that result from the accumulation of a misfolded isoform of the prion protein (PrP), a normal constituent of the neuronal membrane. Five subtypes constitute the known human prion diseases; kuru, Creutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker syndrome (GSS), fatal insomnia (FI), and variant CJD (vCJD). These subtypes are distinguished, in part, by their clinical phenotype, but primarily by their associated brain histopathology. Evidence suggests these phenotypes are defined by differences in the pathogenic conformation of misfolded PrP. Although the vast majority of cases are sporadic, 10% to 15% result from an autosomal dominant mutation of the PrP gene (PRNP). General phenotype-genotype correlations can be made for the major subtypes of CJD, GSS, and FI. This paper will review some of the general background related to prion biology and detail the clinical and pathologic features of the major prion diseases, with a particular focus on the genetic aspects that result in prion disease or modification of its risk or phenotype.
Collapse
Affiliation(s)
- Khalilah Brown
- Center for Comprehensive Care and Research on Memory Disorders, Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
124
|
Benvegnù S, Poggiolini I, Legname G. Neurodevelopmental expression and localization of the cellular prion protein in the central nervous system of the mouse. J Comp Neurol 2010; 518:1879-91. [PMID: 20394048 DOI: 10.1002/cne.22357] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs) are neurodegenerative disorders caused by PrP(Sc), or prion, an abnormally folded form of the cellular prion protein (PrP(C)). The abundant expression of PrP(C) in the central nervous system (CNS) is a requirement for prion replication, yet despite years of intensive research the physiological function of PrP(C) still remains unclear. Several routes of investigation point out a potential role for PrP(C) in axon growth and neuronal development. Thus, we undertook a detailed analysis of the spatial and temporal expression of PrP(C) during mouse CNS development. Our findings show regional differences of the expression of PrP, with some specific white matter structures showing the earliest and highest expression of PrP(C). Indeed, all these regions are part of the thalamolimbic neurocircuitry, suggesting a potential role of PrP(C) in the development and functioning of this specific brain system.
Collapse
Affiliation(s)
- Stefano Benvegnù
- Scuola Internazionale Superiore di Studi Avanzati-International School for Advanced Studies (SISSA-ISAS), Neurobiology Sector, I-34151 Trieste, Italy
| | | | | |
Collapse
|
125
|
Yasenkov R, Deboer T. Circadian regulation of sleep and the sleep EEG under constant sleep pressure in the rat. Sleep 2010; 33:631-41. [PMID: 20469805 DOI: 10.1093/sleep/33.5.631] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
STUDY OBJECTIVE Sleep is regulated by homeostatic and circadian processes. Slow wave activity (SWA; 1-4 Hz) in the NREM sleep electroencephalogram (EEG) reflects sleep homeostasis. Activity of faster EEG frequencies (10-25 Hz) is thought to be under influence of circadian factors. The relative contribution of both processes to the distribution of sleep and wakefulness and EEG activity in rodents remains uncertain. DESIGN Continuous EEG recording in rats in constant dark conditions (DD) were performed and a sleep deprivation protocol consisting of 2 h sleep deprivation followed by 2 h of rest (2h/2h) was applied for 48 h to obtain a constant sleep pressure. SETTINGS Basic sleep research laboratory. PATIENTS OR PARTICIPANTS Adult male Wistar rats. INTERVENTION Sleep deprivation. MEASUREMENTS AND RESULTS Under the 2h/2h protocol, the circadian modulation of waking, NREM and REM sleep was markedly reduced compared to the baseline, affecting the frequency of vigilance state episodes and the duration of REM sleep and waking episodes. In contrast, NREM sleep episode duration still showed a daily modulation. Consecutive 2h values of SWA in NREM sleep were stabile during the 2h\2h protocol, while NREM sleep EEG activity within the higher frequencies (7-25 Hz) still demonstrated strong circadian modulation, which did not differ from baseline. CONCLUSIONS In rats, the daily modulation of REM sleep is less pronounced compared to NREM sleep and waking. In contrast to SWA, activity in higher frequencies (7-25 Hz) in the NREM sleep EEG have an endogenous circadian origin and are not influenced by sleep homeostatic mechanisms.
Collapse
Affiliation(s)
- Roman Yasenkov
- Laboratory for Neurophysiology, Department of Molecular Cell Biology Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
126
|
Singh N, Singh A, Das D, Mohan ML. Redox control of prion and disease pathogenesis. Antioxid Redox Signal 2010; 12:1271-94. [PMID: 19803746 PMCID: PMC2864664 DOI: 10.1089/ars.2009.2628] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 09/22/2009] [Accepted: 10/03/2009] [Indexed: 11/12/2022]
Abstract
Imbalance of brain metal homeostasis and associated oxidative stress by redox-active metals like iron and copper is an important trigger of neurotoxicity in several neurodegenerative conditions, including prion disorders. Whereas some reports attribute this to end-stage disease, others provide evidence for specific mechanisms leading to brain metal dyshomeostasis during disease progression. In prion disorders, imbalance of brain-iron homeostasis is observed before end-stage disease and worsens with disease progression, implicating iron-induced oxidative stress in disease pathogenesis. This is an unexpected observation, because the underlying cause of brain pathology in all prion disorders is PrP-scrapie (PrP(Sc)), a beta-sheet-rich conformation of a normal glycoprotein, the prion protein (PrP(C)). Whether brain-iron dyshomeostasis occurs because of gain of toxic function by PrP(Sc) or loss of normal function of PrP(C) remains unclear. In this review, we summarize available evidence suggesting the involvement of oxidative stress in prion-disease pathogenesis. Subsequently, we review the biology of PrP(C) to highlight its possible role in maintaining brain metal homeostasis during health and the contribution of PrP(Sc) in inducing brain metal imbalance with disease progression. Finally, we discuss possible therapeutic avenues directed at restoring brain metal homeostasis and alleviating metal-induced oxidative stress in prion disorders.
Collapse
Affiliation(s)
- Neena Singh
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
127
|
Lee YJ, Baskakov IV. Treatment with normal prion protein delays differentiation and helps to maintain high proliferation activity in human embryonic stem cells. J Neurochem 2010; 114:362-73. [PMID: 20089130 DOI: 10.1111/j.1471-4159.2010.06601.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The normal cellular form of prion protein (PrP(C)) has been shown to exhibit a diverse range of biological activities. Several recent studies highlighted potential involvement of PrP(C) in embryogenesis or in regulating stem cell self-renewal and proliferation. In the current study, we employed human embryonic stem cells (hESCs) for assessing the potential role of prion protein in early human development. Here, we showed that treatment of hESCs with full-length recombinant PrP folded into an alpha-helical conformation similar to that of PrP(C) delayed the spontaneous differentiation of hESCs and helped to maintain their high proliferation activity during spontaneous differentiation. Considering that administration of alpha-rPrP was also found to down-regulate the expression of endogenous PrP(C), the effects of alpha-rPrP were likely to be indirect, i.e. executed by endogenous PrP(C). Together with previous observations, these work support the hypothesis that PrP(C) is involved in regulating self-renewal/differentiation status of stem cells including hESCs.
Collapse
Affiliation(s)
- Young Jin Lee
- Medical Biotechnology Center, University of Maryland Biotechnology Institute, Baltimore, MD 21201, USA
| | | |
Collapse
|
128
|
Benetti F, Gasperini L, Zampieri M, Legname G. Gene expression profiling to identify druggable targets in prion diseases. Expert Opin Drug Discov 2010; 5:177-202. [PMID: 22822917 DOI: 10.1517/17460440903544449] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
IMPORTANCE OF THE FIELD Despite many recent advances in prion research, the molecular mechanisms by which prions cause neurodegeneration have not been established. In fact, the complexity and the novelty characterizing this class of disorders pose a huge challenge to drug discovery. Pharmacogenomics has recently adopted high-throughput transcriptome analyses to predict potential drug target candidates, with promising results in various fields of medicine. AREAS COVERED IN THIS REVIEW The present work offers an overview of the transcriptional alterations induced by prion infection in different biological systems. Hereafter, therapeutic approaches are discussed in light of the identified altered processes. WHAT THE READER WILL GAIN This review offers readers a detailed overview on microarray analyses, taking into account their advantages and limitations. Our work can help readers, from many research areas, to design a suitable microarray experiment. TAKE HOME MESSAGE So far, drugs acting on the pathways identified by microarray analysis have not been found to be effective in prion diseases therapy. An integration of gene expression profiling, proteomics and physiology should be applied to pursue this aim.
Collapse
Affiliation(s)
- Federico Benetti
- Laboratory of Prion Biology, Neurobiology Sector, Scuola Internazionale Superiore di Studi Avanzati-International School of Advanced Studies (SISSA-ISAS), Edificio Q1, Basovizza, Trieste, Italy
| | | | | | | |
Collapse
|
129
|
Deboer T. PERsonalizing sleep. Sleep Med Rev 2009; 14:147-9. [PMID: 19962333 DOI: 10.1016/j.smrv.2009.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Accepted: 10/03/2009] [Indexed: 10/20/2022]
|
130
|
Rial D, Duarte F, Xikota J, Schmitz A, Dafré A, Figueiredo C, Walz R, Prediger R. Cellular prion protein modulates age-related behavioral and neurochemical alterations in mice. Neuroscience 2009; 164:896-907. [DOI: 10.1016/j.neuroscience.2009.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 08/10/2009] [Accepted: 09/01/2009] [Indexed: 02/04/2023]
|
131
|
Rangel A, Madroñal N, Massó AGI, Gavín R, Llorens F, Sumoy L, Torres JM, Delgado-García JM, Río JAD. Regulation of GABA(A) and glutamate receptor expression, synaptic facilitation and long-term potentiation in the hippocampus of prion mutant mice. PLoS One 2009; 4:e7592. [PMID: 19855845 PMCID: PMC2763346 DOI: 10.1371/journal.pone.0007592] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2009] [Accepted: 09/30/2009] [Indexed: 11/18/2022] Open
Abstract
Background Prionopathies are characterized by spongiform brain degeneration, myoclonia, dementia, and periodic electroencephalographic (EEG) disturbances. The hallmark of prioniopathies is the presence of an abnormal conformational isoform (PrPsc) of the natural cellular prion protein (PrPc) encoded by the Prnp gene. Although several roles have been attributed to PrPc, its putative functions in neuronal excitability are unknown. Although early studies of the behavior of Prnp knockout mice described minor changes, later studies report altered behavior. To date, most functional PrPc studies on synaptic plasticity have been performed in vitro. To our knowledge, only one electrophysiological study has been performed in vivo in anesthetized mice, by Curtis and coworkers. They reported no significant differences in paired-pulse facilitation or LTP in the CA1 region after Schaffer collateral/commissural pathway stimulation. Methodology/Principal Findings Here we explore the role of PrPc expression in neurotransmission and neural excitability using wild-type, Prnp −/− and PrPc-overexpressing mice (Tg20 strain). By correlating histopathology with electrophysiology in living behaving mice, we demonstrate that both Prnp −/− mice but, more relevantly Tg20 mice show increased susceptibility to KA, leading to significant cell death in the hippocampus. This finding correlates with enhanced synaptic facilitation in paired-pulse experiments and hippocampal LTP in living behaving mutant mice. Gene expression profiling using Illumina™ microarrays and Ingenuity pathways analysis showed that 129 genes involved in canonical pathways such as Ubiquitination or Neurotransmission were co-regulated in Prnp −/− and Tg20 mice. Lastly, RT-qPCR of neurotransmission-related genes indicated that subunits of GABAA and AMPA-kainate receptors are co-regulated in both Prnp −/− and Tg20 mice. Conclusions/Significance Present results demonstrate that PrPc is necessary for the proper homeostatic functioning of hippocampal circuits, because of its relationships with GABAA and AMPA-Kainate neurotransmission. New PrPc functions have recently been described, which point to PrPc as a target for putative therapies in Alzheimer's disease. However, our results indicate that a “gain of function” strategy in Alzheimer's disease, or a “loss of function” in prionopathies, may impair PrPc function, with devastating effects. In conclusion, we believe that present data should be taken into account in the development of future therapies.
Collapse
Affiliation(s)
- Alejandra Rangel
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, and Department of Cell Biology, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Noelia Madroñal
- Division de Neurociencias. Universidad Pablo de Olavide, Sevilla, Spain
| | | | - Rosalina Gavín
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, and Department of Cell Biology, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Franc Llorens
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, and Department of Cell Biology, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Lauro Sumoy
- Institute of Predictive and Personalized Medicine of Cancer, Badalona, Spain
| | - Juan María Torres
- Centro de Investigación en Sanidad Animal (CISA), INIA, Valdeolmos, Madrid, Spain
| | | | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, and Department of Cell Biology, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- * E-mail: (JADR); (JMDG)
| |
Collapse
|
132
|
Peñarrubia L, Andrés-Colás N, Moreno J, Puig S. Regulation of copper transport in Arabidopsis thaliana: a biochemical oscillator? J Biol Inorg Chem 2009; 15:29-36. [PMID: 19798519 DOI: 10.1007/s00775-009-0591-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 09/08/2009] [Indexed: 01/08/2023]
Abstract
Plants are among the most versatile higher eukaryotes in accommodating environmental copper availability to largely variable demands. In particular, copper deficiency in soils is a threat for plant survival since it mostly affects reproductive structures. One of the strategies that plant cells use to overcome this situation is to increase copper levels by expressing high-affinity copper transporters delivering the metal to the cytosol. In this minireview, we discuss recent advances in the structure, function, and regulation of the CTR/COPT family of copper transporters, and pay special attention to the Arabidopsis thaliana counterparts. These are constituted by transmembrane polypeptides, containing several copper-binding sequences of functional and/or regulatory value, and assembling as trimers. Copper deficiency activates the expression of some members of the COPT family via the interaction of the SPL7 transcription factor with reiterative GTAC motifs present in their promoters. Interestingly, the regulation of the synthesis of these transporters by copper itself constitutes a negative-feedback loop that could cause a sustained oscillation in the cytosolic copper levels. We analyze the theoretical conditions required for this hypothetical copper oscillation and the potential advantages of synchronization with other cycles. Diverse data in other organisms point to the relationship between copper homeostasis and circadian cycles.
Collapse
Affiliation(s)
- Lola Peñarrubia
- Departament de Bioquímica i Biologia Molecular, Universitat de València, Spain.
| | | | | | | |
Collapse
|
133
|
Abstract
Transmissible spongiform encephalopathies (TSEs) are inevitably lethal neurodegenerative diseases that affect humans and a large variety of animals. The infectious agent responsible for TSEs is the prion, an abnormally folded and aggregated protein that propagates itself by imposing its conformation onto the cellular prion protein (PrPC) of the host. PrPCis necessary for prion replication and for prion-induced neurodegeneration, yet the proximal causes of neuronal injury and death are still poorly understood. Prion toxicity may arise from the interference with the normal function of PrPC, and therefore, understanding the physiological role of PrPCmay help to clarify the mechanism underlying prion diseases. Here we discuss the evolution of the prion concept and how prion-like mechanisms may apply to other protein aggregation diseases. We describe the clinical and the pathological features of the prion diseases in human and animals, the events occurring during neuroinvasion, and the possible scenarios underlying brain damage. Finally, we discuss potential antiprion therapies and current developments in the realm of prion diagnostics.
Collapse
|
134
|
Palchykova S, Winsky-Sommerer R, Tobler I. Sleep deprivation in the dark period does not impair memory in OF1 mice. Chronobiol Int 2009; 26:682-96. [PMID: 19444749 DOI: 10.1080/07420520902926025] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
There is increasing evidence that sleep facilitates memory acquisition and consolidation. Moreover, the sleep-wake history preceding memory acquisition and retention as well as circadian timing may be important. We showed previously that sleep deprivation (SD) following learning in OF1 mice impaired their performance on an object recognition task. The learning task was scheduled at the end of the 12 h dark period and the test 24 h later. To investigate the influence of the prominent circadian sleep-wake distribution typical for rodents, we now scheduled the learning task at the beginning of the dark period. Wakefulness following immediately after the learning task was attained either by gentle interference (SD; n = 20) or by spontaneous wheel running (RW; n = 20). Two control groups were used: one had no RW throughout the experiment (n = 23), while the other group's wheel was blocked immediately after acquisition (n = 16), thereby preventing its use until testing. Recognition memory, defined as the difference in exploration of a novel and of familiar objects, was assessed 24 h later during the test phase. Motor activity and RW use were continuously recorded. Remarkably, performance on the object recognition task was not influenced by the protocols; the waking period following acquisition did not impair memory, independent of the method inducing wakefulness (i.e., sleep deprivation or spontaneous running). Thus, all groups explored the novel object significantly longer than the familiar ones during the test phase. Interestingly, neither the amount of rest lost during the SD interventions nor the amount of rest preceding acquisition influenced performance. However, the total amount of rest obtained by the control and SD mice subjected to acquisition at "dark offset" correlated positively (r = 0.66) with memory at test, while no such relationship occurred in the corresponding groups tested at dark onset. Neither the amount of running nor intermediate rest correlated with performance at test in the RW group. We conclude that interfering with sleep during the dark period does not affect object recognition memory consolidation.
Collapse
|
135
|
Sakaguchi S, Ishibashi D, Matsuda H. Antibody-based immunotherapeutic attempts in experimental animal models of prion diseases. Expert Opin Ther Pat 2009; 19:907-17. [PMID: 19514955 DOI: 10.1517/13543770902988530] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND There has been a dramatic decrease in the risk of transmission of bovine spongiform encephalopathy to humans. In contrast, the risk of human-to-human transmission of variant Creutzfeldt-Jakob disease (vCJD) via medical treatments became potentially high since 4 vCJD cases were reported to be possibly transmitted through blood transfusion in the UK. However, no treatments are yet available for curing prion diseases. OBJECTIVE Conversion of the normal prion protein, PrP(C), to the amyloidogenic PrP, PrP(Sc), plays a pivotal role in the pathogenesis. Recently, certain anti-PrP or anti-37/67-kDa laminin receptor (LRP/LR) antibodies were shown to have the potential to cure chronically infected cells, clearing PrP(Sc) from the cells. This has raised the possibility of antibody based-immunotherapy for prion diseases. This article aims to introduce and discuss the recently published attempts of immunotherapy in prion diseases. METHODS Bibliographic research was carried out using the PubMed database. Patent literature was searched using the UK Intellectual Property Office website. RESULTS/CONCLUSION No satisfying consequences in animals could be detected with anti-PrP antibodies directly infused into the brains of animals by the intraventricular route or by anti-PrP or anti-LRP/LR single chain fragment antibodies directly delivered into the brain by virus vector-mediated gene transfer. This is probably because such delivery systems failed to deliver the antibodies to the neurons relevant for the treatments.
Collapse
Affiliation(s)
- Suehiro Sakaguchi
- The University of Tokushima, The Institute for Enzyme Research, Division of Molecular Neurobiology, 3-18-15 Kuramoto-cho, Tokushima, Japan.
| | | | | |
Collapse
|
136
|
Abstract
It has been known for a long time that genetic factors affect sleep quantity and quality. Genetic screens have identified several mutations that affect sleep across species, pointing to an evolutionary conserved regulation of sleep. Moreover, it has also been recognized that sleep affects gene expression. These findings have given valuable insights into the molecular underpinnings of sleep regulation and function that might lead the way to more efficient treatments for sleep disorders.
Collapse
Affiliation(s)
- Chiara Cirelli
- Department of Psychiatry, University of Wisconsin, Madison, Wisconsin 53719, USA.
| |
Collapse
|
137
|
Abstract
The brain uses a variety of mechanisms to survey the immune system constantly. Responses of the immune system to invading pathogens are detected by the central nervous system, which responds by orchestrating complex changes in behavior and physiology. Sleep is one of the behaviors altered in response to immune challenge. The role of cytokines as mediators of responses to infectious challenge and regulators and modulators of sleep is the focus of this article.
Collapse
Affiliation(s)
- Mark R Opp
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48109-0615, USA.
| |
Collapse
|
138
|
Abstract
Human prion diseases (PrD) like Creutzfeldt-Jakob disease (CJD) include sporadic, acquired and familial neurodegenerative disorders. The central events in the neuropathological process of PrDs are severe neuronal loss, spongiform change and accumulation of abnormal prion protein (PrPSc). The latter is a conformational variant of the host-encoded cellular PrP (PrPC), a copper-binding protein. The physiological role of PrPC is debated. Definitive diagnosis of PrD is based on post mortem demonstration of PrPSc by immunohistochemistry or Western blot. Mutations in the PrP gene (PRNP), the polymorphic site at codon 129, and the molecular characteristic of protease resistant PrP influence the phenotype. Clinical symptoms, cranial MRI scan, EEG and investigation of 14-3-3 protein in cerebrospinal fluid (CSF) suggest a diagnosis of probable CJD. Variant CJD, related to bovine spongiform encephalopathy, shows a different clinical course, symmetrical high intensity MRI signal in the pulvinar, presence of PrPSc in tonsil biopsy tissue, and a lower sensitivity of CSF 14-3-3 protein compared to sporadic CJD. Future possibilities in diagnosis of PrDs include either the demonstration of PrPSc in body fluids or disease associated changes in laboratory variables or gene expression.
Collapse
Affiliation(s)
- Gábor G Kovács
- Institute of Neurology, Medical University of Vienna, and Austrian Reference Centre for Human Prion Diseases, Vienna, Austria
| | | | | | | |
Collapse
|
139
|
Kernek KL, Trofatter JA, Mayeda AR, Hofstetter JR. A Locus for Circadian Period of Locomotor Activity on Mouse Proximal Chromosome 3. Chronobiol Int 2009; 21:343-52. [PMID: 15332441 DOI: 10.1081/cbi-120038596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Lengthened circadian period of locomotor activity is a characteristic of a congenic strain of mice carrying a nonsense mutation in exon 5 of the carbonic anhydrase II gene, car2. The null mutation in car2 is located on a DBA/2J inbred strain insert on proximal chromosome 3, on an otherwise C57BL/6J genomic background. Since reducing the size of the congenic region would narrow the possible candidate genes for period, two recombinant congenic strains (R1 and R2) were developed from the original congenic strain. These new congenic strains were assessed for period, genetic composition, and the presence of immunoreactive carbonic anhydrase II. R1 mice were homozygous DBA/2J for the distal portion of the original DBA/2J insert, while R2 mice were homozygous DBA/2J for the proximal portion. R1 mice had a significantly lengthened period compared to R2 mice and wild-type C57BL/6J mice, indicating that the gene(s) affecting period is likely found within the reduced DBA/2J insert (approximately 1 cM) in the R1 mice. The R1 mice also possessed the null mutation in car2. This study confirmed the presence of a gene(s) affecting period on proximal chromosome 3 and significantly reduced the size of the congenic region and the number of candidate genes. Future studies will focus on identifying the gene influencing period.
Collapse
Affiliation(s)
- Kari L Kernek
- Program in Medical Neurobiology, Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | | | | | |
Collapse
|
140
|
White MD, Mallucci GR. Therapy for prion diseases: Insights from the use of RNA interference. Prion 2009; 3:121-8. [PMID: 19597349 PMCID: PMC2802775 DOI: 10.4161/pri.3.3.9289] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Accepted: 06/16/2009] [Indexed: 12/11/2022] Open
Abstract
Insights into the molecular basis and the temporal evolution of neurotoxicity in prion disease are increasing, and recent work in mice leads to new avenues for targeting treatment of these disorders. Using lentivirally mediated RNA interference (RNAi) against native prion protein (PrP), White et al. report the first therapeutic intervention that results in neuronal rescue, prevents symptoms and increases survival in mice with established prion disease.(1) Both the target and the timing of treatment here are crucial to the effectiveness of this strategy: the formation of the neurotoxic prion agent is prevented at a point when diseased neurons can still be saved from death. But the data also give new insights into the timing of treatment in the context of the pattern of spread of prion infection throughout the brain, with implications for developing the most effective treatments.
Collapse
Affiliation(s)
- Melanie D White
- Centre for Neuroscience Research, University of Edinburgh, UK
| | | |
Collapse
|
141
|
New insights into cellular prion protein (PrPc) functions: the "ying and yang" of a relevant protein. ACTA ACUST UNITED AC 2009; 61:170-84. [PMID: 19523487 DOI: 10.1016/j.brainresrev.2009.06.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 05/26/2009] [Accepted: 06/03/2009] [Indexed: 12/19/2022]
Abstract
The conversion of cellular prion protein (PrP(c)), a GPI-anchored protein, into a protease-K-resistant and infective form (generally termed PrP(sc)) is mainly responsible for Transmissible Spongiform Encephalopathies (TSEs), characterized by neuronal degeneration and progressive loss of basic brain functions. Although PrP(c) is expressed by a wide range of tissues throughout the body, the complete repertoire of its functions has not been fully determined. Recent studies have confirmed its participation in basic physiological processes such as cell proliferation and the regulation of cellular homeostasis. Other studies indicate that PrP(c) interacts with several molecules to activate signaling cascades with a high number of cellular effects. To determine PrP(c) functions, transgenic mouse models have been generated in the last decade. In particular, mice lacking specific domains of the PrP(c) protein have revealed the contribution of these domains to neurodegenerative processes. A dual role of PrP(c) has been shown, since most authors report protective roles for this protein while others describe pro-apoptotic functions. In this review, we summarize new findings on PrP(c) functions, especially those related to neural degeneration and cell signaling.
Collapse
|
142
|
Revel FG, Gottowik J, Gatti S, Wettstein JG, Moreau JL. Rodent models of insomnia: A review of experimental procedures that induce sleep disturbances. Neurosci Biobehav Rev 2009; 33:874-99. [DOI: 10.1016/j.neubiorev.2009.03.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Revised: 03/04/2009] [Accepted: 03/04/2009] [Indexed: 12/21/2022]
|
143
|
Vidal C, Herzog C, Haeberle A, Bombarde C, Miquel M, Carimalo J, Launay J, Mouillet-Richard S, Lasmézas C, Dormont D, Kellermann O, Bailly Y. Early dysfunction of central 5-HT system in a murine model of bovine spongiform encephalopathy. Neuroscience 2009; 160:731-43. [DOI: 10.1016/j.neuroscience.2009.02.072] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Revised: 02/18/2009] [Accepted: 02/19/2009] [Indexed: 12/16/2022]
|
144
|
Abstract
The prion protein is infamous for its role in devastating neurological diseases, but its normal, physiological function has remained mysterious. A new study uses the experimentally tractable zebrafish model to obtain fresh clues to this puzzle.
Collapse
Affiliation(s)
- Roberto Chiesa
- * To whom correspondence should be addressed. E-mail: (RC); (DAH)
| | - David A Harris
- * To whom correspondence should be addressed. E-mail: (RC); (DAH)
| |
Collapse
|
145
|
Burgess STG, Shen C, Ferguson LA, O'Neill GT, Docherty K, Hunter N, Goldmann W. Identification of adjacent binding sites for the YY1 and E4BP4 transcription factors in the ovine PrP (Prion) gene promoter. J Biol Chem 2009; 284:6716-24. [PMID: 19129193 PMCID: PMC2652306 DOI: 10.1074/jbc.m807065200] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Revised: 12/17/2008] [Indexed: 11/06/2022] Open
Abstract
The PrP gene encodes the cellular isoform of the prion protein (PrP(c)) which has been shown to be crucial to the development of transmissible spongiform encephalopathies (TSEs). PrP knock-out mice, which do not express endogenous PrP(c), exhibit resistance to TSE disease. The regulation of PrP gene expression represents, therefore, a crucial factor in the development of TSEs. Two sequence motifs in the PrP promoter (positions -287 to -263 from transcriptional start) were previously reported as being highly conserved, and it was suggested that they represent binding sites for as yet unidentified transcription factors. To test this hypothesis, binding of nuclear proteins was analyzed by electrophoretic mobility shift assays using ovine or murine cells and tissues with radiolabeled DNA probes containing the conserved motif sequences. Specific binding was observed to both motifs, and polymorphic variants of these motifs exhibited differential binding. Two proteins bound to these motifs were identified as the Yin Yang 1 (YY1) (motif 1) and E4BP4 (motif 2) transcription factors. Functional promoter analysis of four different promoter variants revealed that motif 1 (YY1) was associated with inhibitory activity in the context of the PrP promoter, whereas motif 2 (E4BP4) was linked to a slight enhancing activity. This represents the first demonstration of binding of nuclear factors to two highly conserved DNA sequence motifs within mammalian PrP promoters. The action of these factors on the PrP promoter is haplotype-specific, leading us to propose that the prion protein expression pattern and, with it, the distribution of TSE infectivity may be associated with PrP promoter genotype.
Collapse
Affiliation(s)
- Stewart T G Burgess
- Roslin Institute and R(D)SVS, Neuropathogenesis Division, University of Edinburgh, Roslin, Midlothian EH25 9PS, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
146
|
Enhancing livestock through genetic engineering—Recent advances and future prospects. Comp Immunol Microbiol Infect Dis 2009; 32:123-37. [DOI: 10.1016/j.cimid.2007.11.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2007] [Indexed: 11/23/2022]
|
147
|
Le Pichon CE, Valley MT, Polymenidou M, Chesler AT, Sagdullaev BT, Aguzzi A, Firestein S. Olfactory behavior and physiology are disrupted in prion protein knockout mice. Nat Neurosci 2009; 12:60-9. [PMID: 19098904 PMCID: PMC2704296 DOI: 10.1038/nn.2238] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Accepted: 11/06/2008] [Indexed: 11/25/2022]
Abstract
The prion protein PrP(C) is infamous for its role in disease, but its normal physiological function remains unknown. Here we found a previously unknown behavioral phenotype of Prnp(-/-) mice in an odor-guided task. This phenotype was manifest in three Prnp knockout lines on different genetic backgrounds, which provides strong evidence that the phenotype is caused by a lack of PrP(C) rather than by other genetic factors. Prnp(-/-) mice also showed altered behavior in a second olfactory task, suggesting that the phenotype is olfactory specific. Furthermore, PrP(C) deficiency affected oscillatory activity in the deep layers of the main olfactory bulb, as well as dendrodendritic synaptic transmission between olfactory bulb granule and mitral cells. Notably, both the behavioral and electrophysiological alterations found in Prnp(-/-) mice were rescued by transgenic neuronal-specific expression of PrP(C). These data suggest that PrP(C) is important in the normal processing of sensory information by the olfactory system.
Collapse
Affiliation(s)
- Claire E. Le Pichon
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Matthew T. Valley
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | | | - Alexander T. Chesler
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Botir T. Sagdullaev
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zürich, Zürich, Switzerland
| | - Stuart Firestein
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| |
Collapse
|
148
|
Cellular prion protein modulates defensive attention and innate fear-induced behaviour evoked in transgenic mice submitted to an agonistic encounter with the tropical coral snake Oxyrhopus guibei. Behav Brain Res 2008; 194:129-37. [DOI: 10.1016/j.bbr.2008.06.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 06/03/2008] [Accepted: 06/05/2008] [Indexed: 11/24/2022]
|
149
|
Deriziotis P, Tabrizi SJ. Prions and the proteasome. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1782:713-22. [PMID: 18644436 DOI: 10.1016/j.bbadis.2008.06.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Revised: 06/23/2008] [Accepted: 06/24/2008] [Indexed: 10/21/2022]
Abstract
Prion diseases are fatal neurodegenerative disorders that include Creutzfeldt-Jakob disease in humans and bovine spongiform encephalopathy in animals. They are unique in terms of their biology because they are caused by the conformational re-arrangement of a normal host-encoded prion protein, PrPC, to an abnormal infectious isoform, PrPSc. Currently the precise mechanism behind prion-mediated neurodegeneration remains unclear. It is hypothesised than an unknown toxic gain of function of PrPSc, or an intermediate oligomeric form, underlies neuronal death. Increasing evidence suggests a role for the ubiquitin proteasome system (UPS) in prion disease. Both wild-type PrPC and disease-associated PrP isoforms accumulate in cells after proteasome inhibition leading to increased cell death, and abnormal beta-sheet-rich PrP isoforms have been shown to inhibit the catalytic activity of the proteasome. Here we review potential interactions between prions and the proteasome outlining how the UPS may be implicated in prion-mediated neurodegeneration.
Collapse
Affiliation(s)
- Pelagia Deriziotis
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | | |
Collapse
|
150
|
Dossena S, Imeri L, Mangieri M, Garofoli A, Ferrari L, Senatore A, Restelli E, Balducci C, Fiordaliso F, Salio M, Bianchi S, Fioriti L, Morbin M, Pincherle A, Marcon G, Villani F, Carli M, Tagliavini F, Forloni G, Chiesa R. Mutant prion protein expression causes motor and memory deficits and abnormal sleep patterns in a transgenic mouse model. Neuron 2008; 60:598-609. [PMID: 19038218 DOI: 10.1016/j.neuron.2008.09.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Revised: 07/22/2008] [Accepted: 09/04/2008] [Indexed: 11/18/2022]
Abstract
A familial form of Creutzfeldt-Jakob disease (CJD) is linked to the D178N/V129 prion protein (PrP) mutation. Tg(CJD) mice expressing the mouse homolog of this mutant PrP synthesize a misfolded form of the mutant protein, which is aggregated and protease resistant. These mice develop clinical and pathological features reminiscent of CJD, including motor dysfunction, memory impairment, cerebral PrP deposition, and gliosis. Tg(CJD) mice also display electroencephalographic abnormalities and severe alterations of sleep-wake patterns strikingly similar to those seen in a human patient carrying the D178N/V129 mutation. Neurons in these mice show swelling of the endoplasmic reticulum (ER) with intracellular retention of mutant PrP, suggesting that ER dysfunction could contribute to the pathology. These results establish a transgenic animal model of a genetic prion disease recapitulating cognitive, motor, and neurophysiological abnormalities of the human disorder. Tg(CJD) mice have the potential for giving greater insight into the spectrum of neuronal dysfunction in prion diseases.
Collapse
Affiliation(s)
- Sara Dossena
- Dulbecco Telethon Institute, 20156 Milan, Italy; Department of Neuroscience, "Mario Negri" Institute for Pharmacological Research, 20156 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|