201
|
Grivel J, Cvetkovic V, Bayer L, Machard D, Tobler I, Mühlethaler M, Serafin M. The wake-promoting hypocretin/orexin neurons change their response to noradrenaline after sleep deprivation. J Neurosci 2006; 25:4127-30. [PMID: 15843615 PMCID: PMC6724959 DOI: 10.1523/jneurosci.0666-05.2005] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Sleep deprivation is accompanied by the progressive development of an irresistible need to sleep, a phenomenon whose mechanism has remained elusive. Here, we identified for the first time a reflection of that phenomenon in vitro by showing that, after a short 2 h period of total sleep deprivation, the action of noradrenaline on the wake-promoting hypocretin/orexin neurons changes from an excitation to an inhibition. We propose that such a conspicuous modification of responsiveness should contribute to the growing sleepiness that accompanies sleep deprivation.
Collapse
Affiliation(s)
- Jeremy Grivel
- Département des Neurosciences fondamentales, Centre Médical Universitaire, 1211 Geneva 4, Switzerland
| | | | | | | | | | | | | |
Collapse
|
202
|
Kernek KL, Trofatter JA, Mayeda AR, Lahiri DK, Hofstetter JR. A single copy of carbonic anhydrase 2 restores wild-type circadian period to carbonic anhydrase II-deficient mice. Behav Genet 2006; 36:301-8. [PMID: 16408247 DOI: 10.1007/s10519-005-9032-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Accepted: 07/14/2005] [Indexed: 01/12/2023]
Abstract
Carbonic anhydrase II (CA-II)-deficient mice have long circadian periods compared to their siblings with normal CA-II levels. The CA-II-deficient mice differ genetically from their siblings at proximal chromosome three, where the mutated carbonic anhydrase 2 gene sits on a small insert of DNA from the DBA/2J strain. The rest of the genome is that of the C57BL/6J strain. The goal of this study was to test the hypothesis that the null mutation in carbonic anhydrase 2 and the long circadian period phenotype were linked. In order to separate the effect of the null mutation in carbonic anhydrase 2 from the effect of DBA/2J alleles of other genes on the insert, two new lines of mice were studied. The first line, Kar, was developed from a CA-II-deficient mouse that had a fortuitous recombination restoring functional CA-II without affecting the rest of the DBA/2J insert. The second line was generated by breeding DBA/2J mice and C57BL/6J mice until they had the genomic composition of CA-II-deficient mice without the null mutation. Both lines of mice had circadian periods not different from C57BL/6J mice and shorter than CA-II-deficient mice. The phenotype of the new lines showed that the long circadian period characteristic of the CA-II-deficient mice arises when functional CA-II is absent, not when DBA/2J alleles are present on proximal chromosome three.
Collapse
Affiliation(s)
- Kari L Kernek
- Program in Medical Neurobiology, Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, 46202, USA
| | | | | | | | | |
Collapse
|
203
|
Ludlam CA, Turner ML. Managing the risk of transmission of variant Creutzfeldt Jakob disease by blood products. Br J Haematol 2006; 132:13-24. [PMID: 16371015 DOI: 10.1111/j.1365-2141.2005.05796.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Whereas plasma-derived clotting factor concentrates now have a very good safety record for not being infectious for lipid enveloped viruses, concern has arisen about the possibility that prion diseases might be transmitted by blood products. There is epidemiological evidence that classical sporadic Creutzfeld Jakob disease (CJD) is not transmitted by blood transfusion. There is now good evidence that the abnormal prion associated with variant CJD can be transmitted by transfusion of fresh blood components and infect recipients. To reduce the risk of the pathological prion in the UK infecting recipients of clotting factor concentrates, these are now only manufactured from imported plasma collected from countries where there has not been bovine spongiform encephalopathy (BSE) in cattle and the risk of variant CJD in the population is, therefore, considered negligible. The safety of these concentrates is also enhanced because prion protein is, to an appreciable extent, excluded by the manufacturing process from the final product. To help reduce the chance of prion transmission by fresh blood products, donations are leucodepleted, there is increasing use of imported fresh frozen plasma (especially for treating children) and potential donors, who have been recipients of blood since 1980 (the beginning of the BSE epidemic in cattle) are deferred.
Collapse
|
204
|
McBride SM. Prion protein: a pattern recognition receptor for viral components and uric acid responsible for the induction of innate and adaptive immunity. Med Hypotheses 2005; 65:570-7. [PMID: 15913900 DOI: 10.1016/j.mehy.2005.02.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Accepted: 02/23/2005] [Indexed: 10/25/2022]
Abstract
Prion protein, known as Prp(c), is a GPI-anchored membrane bound glycoprotein ubiquitously expressed in the body. To date, the precise nature of its physiological role remains a mystery. The prion protein's presence on neurons and immune effector cells suggests a dual neurological and immunological function. Some consensus exists regarding the proposed involvement of Prp(c) in neurodevelopment, where it would serve to mediate interactions between the extra-cellular matrix (ECM) and the neuron. There is also evidence that Prp plays a part in immunity, although the exact nature of the role remains unclear. Interestingly, a role in both immunity and development is a functional division seen in other types of receptors, most notably the Toll Receptor. In mammals, toll-like receptors (TLRs) are partly responsible for both innate and adaptive immune activity. However, recently several TLR independent pathways have been identified that initiate such responses. Unfortunately, receptors for such pathways remain unidentified. But based upon its functional homology to Toll Receptors, its known interactions with several viruses, and its possible downstream effector proteins, it is proposed that Prp(c) represents a new type of pattern recognition receptor responsible for TLR-independent induction of myeloid dendritic cell and macrophage maturation and later T-cell activation. From what is known of the ligands for the prion protein, it is proposed that this response would be initiated via the binding of uric acid, viral RNA, or viral structural proteins to Prp(c). It will further be proposed that Prp(c)'s ability to interact with viral components stems from its evolutionary origin as a horizontally transferred gene from an early RNA virus. Finally, Prp(c)'s functional role in immunity will be related to the pathophysiology of TSEs, with observations made concerning immune response to infection and agent composition.
Collapse
Affiliation(s)
- Sean M McBride
- Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.
| |
Collapse
|
205
|
Kopp C, Ressel V, Wigger E, Tobler I. Influence of estrus cycle and ageing on activity patterns in two inbred mouse strains. Behav Brain Res 2005; 167:165-74. [PMID: 16214232 DOI: 10.1016/j.bbr.2005.09.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 08/31/2005] [Accepted: 09/05/2005] [Indexed: 11/30/2022]
Abstract
Despite the widespread use of inbred mice in research, little is known about aging of the circadian system in female mice, although interactions between female gonadal hormones and circadian rhythms have been established. We investigated the influence of the estrus cycle on circadian aspects of running-wheel activity and changes in the course of aging in female C57BL/6 and C3H/He mice recorded continuously between the ages of 3 and 19 months. In the young, cycling mice the second part of the proestrus night was often, but not consistently, characterized by increased motor activity compared to the remaining estrus cycle nights. After estrus cycling had ceased in the course of ageing, the estrus-dependent day-to-day variability in activity was reduced. The amplitude of the daily rest-activity rhythm decreased progressively after the age of 8 months in C3H/He and 10 months in C57BL/6 mice. The capacity for resynchronisation of activity onset to the LD-cycle was compared in young and old mice after an 8-h phase advance of the LD-cycle. Resynchronisation was significantly slower in old C3H/He mice and unaffected by age in C57BL/6 mice. The circadian period in constant darkness did not change with age in either strain. However, the period was shorter in 17-month old C57BL/6 mice compared to an additional group, which was recorded at the same age, after at least 1-month adaptation to the recording conditions. The results show that the reproductive state as well as ageing influence motor activity patterns of female mice in a strain- and cohort-dependent manner.
Collapse
Affiliation(s)
- Caroline Kopp
- Institute of Pharmacology and Toxicology, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | | | | | |
Collapse
|
206
|
Sakurai-Yamashita Y, Sakaguchi S, Yoshikawa D, Okimura N, Masuda Y, Katamine S, Niwa M. Female-specific neuroprotection against transient brain ischemia observed in mice devoid of prion protein is abolished by ectopic expression of prion protein-like protein. Neuroscience 2005; 136:281-7. [PMID: 16198494 DOI: 10.1016/j.neuroscience.2005.06.095] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Revised: 05/22/2005] [Accepted: 06/30/2005] [Indexed: 11/30/2022]
Abstract
This study was designed to examine the function of cellular prion protein and prion protein-like protein/Doppel, in transient ischemia-related neuronal death in the hippocampus. Two different lines of mice devoid of cellular prion protein, Zrch I Prnp(0/0) and Ngsk Prnp(0/0), were used. The former lacks cellular prion protein whereas the latter ectopically expresses prion protein-like protein/Doppel in the brain in the absence of cellular prion protein. Mice were subjected to 10 min-occlusion of the bilateral common carotid arteries with recovery for 14 days. Less than 10% of the pyramidal neurons in the CA1 subfield were degenerated in male and female wild-type mice. In contrast, more than half of the neurons were lost in male Zrch I Prnp(0/0) and Ngsk Prnp(0/0) mice. Such severe neuronal loss was also observed in female Ngsk Prnp(0/0) mice. However, female Zrch I Prnp(0/0) mice showed mild neuronal loss similar to wild-type mice. Flunarizine, a T- and L-type Ca(2+)-channel antagonist, significantly reduced the neuronal loss in female but not in male Ngsk Prnp(0/0) mice. These results indicate that loss of cellular prion protein renders hippocampal neurons susceptible to ischemic insult specifically in male but not female mice and the ectopic expression of prion protein-like protein/Doppel aggravates the ischemic neuronal death in female prion protein-null mice probably via overloading of Ca(2+)-dependent signaling.
Collapse
Affiliation(s)
- Y Sakurai-Yamashita
- Department of Pharmacology 1, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| | | | | | | | | | | | | |
Collapse
|
207
|
Criado JR, Sánchez-Alavez M, Conti B, Giacchino JL, Wills DN, Henriksen SJ, Race R, Manson JC, Chesebro B, Oldstone MBA. Mice devoid of prion protein have cognitive deficits that are rescued by reconstitution of PrP in neurons. Neurobiol Dis 2005; 19:255-65. [PMID: 15837581 DOI: 10.1016/j.nbd.2005.01.001] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2004] [Revised: 12/22/2004] [Accepted: 01/04/2005] [Indexed: 10/25/2022] Open
Abstract
Prion protein (PrP(C)) is a constituent of most normal mammalian cells and plays an essential role in the pathogenesis of transmissible spongiform encephalopathies (TSE). However, the normal cellular function of PrP(C) remains unclear. Here, we document that mice with a selective deletion of PrP(C) exhibited deficits in hippocampal-dependent spatial learning, but non-spatial learning remained intact. mPrP-/- mice also showed reduction in paired-pulse facilitation and long-term potentiation in the dentate gyrus in vivo. These deficits were rescued in transgenic mPrP-/- mice expressing PrP(C) in neurons under control of the neuron-specific enolase (NSE) promoter indicating that they were due to lack of PrP(C) function in neurons. The deficits were seen in mPrP-/- mice with a homogeneous 129/Ola background and in mPrP-/- mice in the mixed (129/Ola x C57BL/10) background indicating that these abnormalities were unlikely due to variability of background genes or alteration of the nearby Prnd (doppel) gene.
Collapse
Affiliation(s)
- José R Criado
- Department of Neuropharmacology (TPC-10), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Vyazovskiy VV, Kopp C, Bösch G, Tobler I. The GABAA receptor agonist THIP alters the EEG in waking and sleep of mice. Neuropharmacology 2005; 48:617-26. [PMID: 15814097 DOI: 10.1016/j.neuropharm.2004.12.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2004] [Revised: 11/30/2004] [Accepted: 12/17/2004] [Indexed: 11/23/2022]
Abstract
THIP is a GABA(A) agonist with hypnotic properties consisting in reducing sleep latency and prolonging and consolidating sleep. THIP has been reported to increase EEG slow-wave activity (SWA; EEG power in the 0.75-4 Hz band) in non-REM (NREM) sleep in both rats and humans. We investigated the effects of THIP on sleep in C57BL/6 mice. EEG recordings were performed after 2, 4 and 6 mg/kg THIP and saline control. The results were compared with analyses of recordings obtained after 6 h of sleep deprivation (SD) in the same strain of mice. The two higher doses of THIP induced an abnormal EEG pattern both in waking and NREM sleep. The EEG was characterized by sporadic asymmetric high-voltage potentials recurring at a low-frequency (<1 Hz) on the background of a low-amplitude EEG pattern. In contrast, after SD the typical regular synchronous high amplitude delta waves predominated. THIP at 4 and 6 mg/kg led to a prominent enhancement of spectral power in the low-frequency range of the waking and sleep EEG which was much higher than the increase attained after 6 h SD. This effect was particularly prominent in the waking EEG. In NREM sleep the increase of spectral power after THIP reflected the frequency of recurrence of the high-voltage potentials, and was restricted to a narrower frequency band than after SD. The EEG changes after 2mg/kg differed little from saline control. Sleep latency was not affected by the two lower doses of THIP, and was prolonged after 6 mg/kg. REM sleep was suppressed after the two higher doses. In contrast to previous results reported in other species, THIP did not have a hypnotic action in mice. The changes induced by THIP in the waking and sleep EEG differed from those caused by enhanced physiological sleep pressure encountered after SD. Considering the abnormal EEG pattern and the similarity of the spectral changes in the sleep and waking EEG, THIP does not seem to exert a specific effect on mechanisms involved in sleep regulation.
Collapse
|
209
|
Rachidi W, Riondel J, McMahon HM, Favier A. [Prion protein and copper: a mysterious relationship]. ACTA ACUST UNITED AC 2005; 53:244-50. [PMID: 15850959 DOI: 10.1016/j.patbio.2003.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2003] [Accepted: 10/30/2003] [Indexed: 11/23/2022]
Abstract
Prion diseases form a group of fatal neurodegenerative disorders including Creutzfeldt-Jakob disease in humans and bovine spongiform encephalopathy in animals. All of which are characterized by the accumulation of abnormally folded isoform of the cellular prion protein (PrP(C)), denoted PrP(Sc), which is the major component of infectious prion diseases. The function of PrP(C) remains elusive. Its amino-terminal region contains a repeated five octapeptide domain that binds copper. The protein is believed to display a superoxide dismutase like activity, and hence a possible protective function against oxidative stress. In this review, relationship between PrP, copper and oxidative stress was analysed. Thus, metal ions and oxidative stress would play an essential role in the pathogenesis of prion diseases and represent important targets for future therapeutic targets or a novel diagnostic marker.
Collapse
Affiliation(s)
- W Rachidi
- Industrial Microbiology, University College Dublin (UCD), Belfield, Ireland.
| | | | | | | |
Collapse
|
210
|
Prion diseases. NEURODEGENER DIS 2005. [DOI: 10.1017/cbo9780511544873.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
211
|
Vyazovskiy VV, Ruijgrok G, Deboer T, Tobler I. Running wheel accessibility affects the regional electroencephalogram during sleep in mice. ACTA ACUST UNITED AC 2005; 16:328-36. [PMID: 15901653 DOI: 10.1093/cercor/bhi110] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Regional aspects of sleep homeostasis were investigated in mice provided with a running wheel for several weeks. Electroencephalogram (EEG) spectra of the primary motor (frontal) and somatosensory cortex (parietal) were recorded for three consecutive days. On a single day (day 2) the wheel was locked to prevent running. Wheel running correlated negatively with the frontal-parietal ratio of slow-wave activity (EEG power between 0.75 and 4.0 Hz) in the first 2 h after sleep onset (r = -0.60; P < 0.01). On day 2 frontal EEG power (2.25-8.0 Hz) in non-rapid eye movement sleep exceeded the level of the previous day, indicating that the diverse behaviors replacing wheel-running elicited more pronounced regional EEG differences. The frontal-parietal power ratio of the lower frequency bin (0.75-1.0 Hz) in the first 2 h of sleep after dark onset correlated positively with the duration of the preceding waking (r = 0.64; P < 0.001), whereas the power ratio in the remaining frequencies of the delta band (1.25-4.0 Hz) was unrelated to waking. The data suggest that in mice EEG power in the lower frequency, corresponding to the slow oscillations described in cats and humans, is related to local sleep homeostasis.
Collapse
Affiliation(s)
- Vladyslav V Vyazovskiy
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | | | | | | |
Collapse
|
212
|
Al Bersaoui R, Robert I, Lutz Y, Blanc F, Sommermeyer-Leroux G, Shibaguchi H, Aunis D, Fuchs JP. Purkinje-cell degeneration in prion protein-deficient mice is associated with a cerebellum-specific Doppel protein species signature. FEBS Lett 2005; 579:2715-21. [PMID: 15862314 DOI: 10.1016/j.febslet.2005.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2005] [Revised: 04/05/2005] [Accepted: 04/06/2005] [Indexed: 11/29/2022]
Abstract
PrP(c) (cellular prion protein) and Doppel are antagonizing proteins, respectively neuroprotective and neurotoxic. Evidence for Doppel neurotoxicity came from PrP(c)-deficient (Prnp(0/0)) mouse lines developing late onset Purkinje-cell degeneration caused by Doppel overexpression in brain. To address the molecular underpinnings of this cell-type specificity, we generated Doppel N-terminal-specific antibodies and started to examine the spatio-temporal expression of Doppel protein species in Ngsk Prnp(0/0) brain. Although Doppel overexpression is ubiquitous, Western analyses of normal and deglycosylated protein extracts revealed cerebellar patterns distinct from the rest of the brain, supporting the idea that neurotoxicity might be linked to a particular Doppel species pattern. Furthermore, our newly raised antibodies allowed the first Doppel immunohistochemical analyses in brain, showing a distribution in Prnp(0/0) cerebellum similar to PrP(c) in wild type.
Collapse
Affiliation(s)
- Roméo Al Bersaoui
- Unité 575 INSERM, Physiopathologie du Système Nerveux, Strasbourg, France
| | | | | | | | | | | | | | | |
Collapse
|
213
|
Ji HF, Zhang HY, Shen L. The Role of Electrostatic Interaction in Triggering the Unraveling of Stable Helix 1 in Normal Prion Protein. A Molecular Dynamics Simulation Investigation. J Biomol Struct Dyn 2005; 22:563-70. [PMID: 15702928 DOI: 10.1080/07391102.2005.10507026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The conversion of normal prion protein (PrPC) into scrapie isoform (PrPSc) is a key event in the pathogenesis of prion diseases. However, the conversion mechanism has given rise to much controversy. For instance, there is much debate on the behavior of helix 1 (H1) in the conversion. A series of experiments demonstrated that H1 in isolated state was very stable under a variety of conditions. But, other experiments indicated that helices 2 and 3 rather than H1 were retained in PrPSc. In this paper, molecular dynamics (MD) simulation is employed to investigate the dynamic behavior of H1. It is revealed that although the helix 1 of Human PrPC (HuPrPC) is very stable in the isolated state, it becomes unstable when incorporated into native HuPrPC, which likely results from the long-range electrostatic interaction between Asp147 and Arg208 located in the helices 1 and 3, respectively. This explanation is supported by experimental evaluation and MD simulation on D147N mutant of HuPrPC that the mutant becomes a little more stable than the wild type HuPrPC. This finding not only help to reconcile the existing debate on the role of helix 1 in the PrPC-->PrPSc transition, but also reveals a possible mechanism for triggering the PrPC-->PrPSc conversion.
Collapse
Affiliation(s)
- Hong-Fang Ji
- Laboratory for Computational Biology, Shandong Provincial Research Center for Bioinformatic Engineering and Technique, Shandong University of Technology, Zibo 255049, PR China
| | | | | |
Collapse
|
214
|
Sakudo A, Lee DC, Li S, Nakamura T, Matsumoto Y, Saeki K, Itohara S, Ikuta K, Onodera T. PrP cooperates with STI1 to regulate SOD activity in PrP-deficient neuronal cell line. Biochem Biophys Res Commun 2005; 328:14-9. [PMID: 15670743 DOI: 10.1016/j.bbrc.2004.12.132] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Indexed: 11/26/2022]
Abstract
Cellular prion protein (PrP(C)) plays anti-apoptotic and anti-oxidative roles in apoptosis induced by serum deprivation in an immortalized prion protein gene (Prnp)-deficient neuronal cell line. The octapeptide repeat region (OR) and N-terminal half of the hydrophobic region (HR) of PrP(C) are indispensable for PrP(C) activity, but the mechanisms remain unclear. In the present study, elucidation of the mechanisms by which PrP(C) elicits the anti-oxidative activities was facilitated by evidence of stress-inducible protein 1 (STI1) mediating PrP(C)-dependent superoxide dismutase (SOD) activation. Immunoprecipitation revealed that PrP(C) was associated with STI1. The inhibitory peptides against PrP(C)-STI1 binding [STI1 pep.1 and PrP(113-132)] indicated toxic activity in PrP(C)-expressing cells by inhibiting SOD activity but not in Prnp(-/-) cells. Furthermore, OR and N-terminal half of the HR were required for the inhibitory effect of PrP(113-132) but not STI1 pep.1. These data are consistent with results established with a model where OR and N-terminal half of the HR mediate the action of STI1 upon cell survival and upregulation of SOD activity.
Collapse
Affiliation(s)
- Akikazu Sakudo
- Department of Molecular Immunology, School of Agricultural and Life Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
215
|
Barret A, Forestier L, Deslys JP, Julien R, Gallet PF. Glycosylation-related Gene Expression in Prion Diseases. J Biol Chem 2005; 280:10516-23. [PMID: 15632154 DOI: 10.1074/jbc.m412635200] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Several lines of evidence indicate that some glycoconjugates are efficient effectors of the cellular prion protein (PrP(C)) conversion into its pathogenic (PrP(Sc)) isoform. To assess how glycoconjugate glycan moieties participate in the biogenesis of PrP(Sc), an exhaustive comparative analysis of the expression of about 200 glycosylation-related genes was performed on prion-infected or not, hypothalamus-derived GT1 cells by hybridization of DNA microarrays, semiquantitative RT-PCR, and biochemical assays. A significant up- (30-fold) and down- (17-fold) regulation of the expression of the ChGn1 and Chst8 genes, respectively, was observed in prion-infected cells. ChGn1 and Chst8 are involved in the initiation of the synthesis of chondroitin sulfate and in the 4-O-sulfation of non-reducing N-acetylgalactosamine residues, respectively. A possible role for a hyposulfated chondroitin in PrP(Sc) accumulation was evidenced at the protein level and by determination of chondroitin and heparan sulfate amounts. Treatment of Sc-GT1 cells with a heparan mimetic (HM2602) induced an important reduction of the amount of PrP(Sc), associated with a total reversion of the transcription pattern of the N-acetylgalactosamine-4-O-sulfotransferase 8. It suggests a link between the genetic control of 4-O-sulfation and PrP(Sc) accumulation.
Collapse
Affiliation(s)
- Agnès Barret
- Groupe d'Innovation Diagnostique et Thérapeutique des Infections à Prions, Commissariat à l'Energie Atomique, 18 route du Panorama, 92265, Fontenay-aux-Roses, France
| | | | | | | | | |
Collapse
|
216
|
|
217
|
Purdey M. Elevated silver, barium and strontium in antlers, vegetation and soils sourced from CWD cluster areas: do Ag/Ba/Sr piezoelectric crystals represent the transmissible pathogenic agent in TSEs? Med Hypotheses 2005; 63:211-25. [PMID: 15236778 DOI: 10.1016/j.mehy.2004.02.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Accepted: 02/13/2004] [Indexed: 10/26/2022]
Abstract
High levels of Silver (Ag), Barium (Ba) and Strontium (Sr) and low levels of copper (Cu) have been measured in the antlers, soils and pastures of the deer that are thriving in the chronic wasting disease (CWD) cluster zones in North America in relation to the areas where CWD and other transmissible spongiform encephalopathies (TSEs) have not been reported. The elevations of Ag, Ba and Sr were thought to originate from both natural geochemical and artificial pollutant sources--stemming from the common practise of aerial spraying with 'cloud seeding' Ag or Ba crystal nuclei for rain making in these drought prone areas of North America, the atmospheric spraying with Ba based aerosols for enhancing/refracting radar and radio signal communications as well as the spreading of waste Ba drilling mud from the local oil/gas well industry across pastureland. These metals have subsequently bioconcentrated up the foodchain and into the mammals who are dependent upon the local Cu deficient ecosystems. A dual eco-prerequisite theory is proposed on the aetiology of TSEs which is based upon an Ag, Ba, Sr or Mn replacement binding at the vacant Cu/Zn domains on the cellular prion protein (PrP)/sulphated proteoglycan molecules which impairs the capacities of the brain to protect itself against incoming shockbursts of sound and light energy. Ag/Ba/Sr chelation of free sulphur within the biosystem inhibits the viable synthesis of the sulphur dependent proteoglycans, which results in the overall collapse of the Cu mediated conduction of electric signals along the PrP-proteoglycan signalling pathways; ultimately disrupting GABA type inhibitory currents at the synapses/end plates of the auditory/circadian regulated circuitry, as well as disrupting proteoglycan co-regulation of the growth factor signalling systems which maintain the structural integrity of the nervous system. The resulting Ag, Ba, Sr or Mn based compounds seed piezoelectric crystals which incorporate PrP and ferritin into their structure. These ferrimagnetically ordered crystals multireplicate and choke up the PrP-proteoglycan conduits of electrical conduction throughout the CNS. The second stage of pathogenesis comes into play when the pressure energy from incoming shock bursts of low frequency acoustic waves from low fly jets, explosions, earthquakes, etc. (a key eco-characteristic of TSE cluster environments) are absorbed by the rogue 'piezoelectric' crystals, which duly convert the mechanical pressure energy into an electrical energy which accumulates in the crystal-PrP-ferritin aggregates (the fibrils) until a point of 'saturation polarization' is reached. Magnetic fields are generated on the crystal surface, which initiate chain reactions of deleterious free radical mediated spongiform neurodegeneration in surrounding tissues. Since Ag, Ba, Sr or Mn based piezoelectric crystals are heat resistant and carry a magnetic field inducing pathogenic capacity, it is proposed that these ferroelectric crystal pollutants represent the transmissible, pathogenic agents that initiate TSE.
Collapse
Affiliation(s)
- Mark Purdey
- High Barn Farm, Elworthy, Taunton, Somerset TA4 3PX, UK.
| |
Collapse
|
218
|
Unterberger U, Voigtländer T, Budka H. Pathogenesis of prion diseases. Acta Neuropathol 2005; 109:32-48. [PMID: 15645262 DOI: 10.1007/s00401-004-0953-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2004] [Accepted: 10/18/2004] [Indexed: 11/28/2022]
Abstract
Prion diseases are rare neurological disorders that may be of genetic or infectious origin, but most frequently occur sporadically in humans. Their outcome is invariably fatal. As the responsible pathogen, prions have been implicated. Prions are considered to be infectious particles that represent mainly, if not solely, an abnormal, protease-resistant isoform of a cellular protein, the prion protein or PrP(C). As in other neurodegenerative diseases, aggregates of misfolded protein conformers are deposited in the CNS of affected individuals. Pathogenesis of prion diseases comprises mainly two equally important, albeit essentially distinct, topics: first, the mode, spread, and amplification of infectivity in acquired disease, designated as peripheral pathogenesis. In this field, significant advances have implicated an essential role of lymphoid tissues for peripheral prion replication, before a likely neural spread to the CNS. The second is the central pathogenesis, dealing, in addition to spread and replication of prions within the CNS, with the mechanisms of nerve cell damage and death. Although important roles for microglial neurotoxicity, oxidative stress, and complement activation have been identified, we are far from complete understanding, and therapeutic applications in prion diseases still need to be developed.
Collapse
|
219
|
Moore RA, Vorberg I, Priola SA. Species barriers in prion diseases--brief review. ARCHIVES OF VIROLOGY. SUPPLEMENTUM 2005:187-202. [PMID: 16355873 DOI: 10.1007/3-211-29981-5_15] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Transmissible spongiform encephalopathies (TSEs or prion diseases) are neurological disorders associated with the aggregation of a pathologic isoform of a host-encoded protein, termed prion protein (PrP). The pathologic isoform of PrP, termed PrP(Sc), is a major constituent of the infectious agent. TSE diseases are characterized by neurodegenerative failure and inevitable morbidity. Bovine spongiform encephalopathy (BSE) has been transmitted from cattle to humans to cause a new variant of Creutzfeldt-Jakob syndrome. The potential for chronic wasting disease to similarly cross the species barrier from cervids to humans is considered unlikely but possible. Thus, understanding how TSE agents overcome resistance to transmission between species is crucial if we are to prevent future epidemics. The species barrier usually can be abrogated to varying degrees in laboratory animals. Studies done with transgenic animals, tissue culture, and cell-free assays established PrP as being necessary for TSE pathogenesis and illustrated that certain amino acid residues are more influential than others for conferring resistance to TSE agent transmission. The essence of what constitutes a TSE agent's species compatibility is thought to be orchestrated by a complex interplay of contributions from its primary amino acid sequence, its glycoform patterns, and its three-dimensional structure.
Collapse
Affiliation(s)
- R A Moore
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA
| | | | | |
Collapse
|
220
|
Sakudo A, Lee DC, Nishimura T, Li S, Tsuji S, Nakamura T, Matsumoto Y, Saeki K, Itohara S, Ikuta K, Onodera T. Octapeptide repeat region and N-terminal half of hydrophobic region of prion protein (PrP) mediate PrP-dependent activation of superoxide dismutase. Biochem Biophys Res Commun 2005; 326:600-6. [PMID: 15596141 DOI: 10.1016/j.bbrc.2004.11.092] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2004] [Indexed: 11/28/2022]
Abstract
Cellular prion protein PrP(C) contains two evolutionarily conserved domains among mammals; viz., the octapeptide repeat region (OR; amino acid residue 51-90) and the hydrophobic region (HR; amino acid residue 112-145). Accumulating evidence indicates that PrP(C) acts as an inhibitor of apoptosis and regulator of superoxide dismutase (SOD) activity. To further understand how PrP(C) activates SOD and prevents apoptosis, we provide evidence here that OR and N-terminal half of HR mediate PrP(C)-dependent SOD activation and anti-apoptotic function. Removal of the OR (amino acid residue 53-94) enhances apoptosis and decreases SOD activity. Deletion of the N-terminal half of HR (amino acids residue 95-132) abolishes its ability to activate SOD and to prevent apoptosis, whereas that of the C-terminal half of HR (amino acids residue 124-146) has little if any effect on the anti-apoptotic activity and SOD activation. These data are consistent with a model in which the anti-apoptotic and anti-oxidative function of PrP(C) is regulated by not only OR but also the N-terminal half of HR.
Collapse
Affiliation(s)
- Akikazu Sakudo
- Department of Molecular Immunology, School of Agricultural and Life Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Cuccioloni M, Amici M, Eleuteri AM, Biagetti M, Barocci S, Angeletti M. Binding of recombinant PrPc to human plasminogen: Kinetic and thermodynamic study using a resonant mirror biosensor. Proteins 2004; 58:728-34. [DOI: 10.1002/prot.20346] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
222
|
Drisaldi B, Coomaraswamy J, Mastrangelo P, Strome B, Yang J, Watts JC, Chishti MA, Marvi M, Windl O, Ahrens R, Major F, Sy MS, Kretzschmar H, Fraser PE, Mount HTJ, Westaway D. Genetic Mapping of Activity Determinants within Cellular Prion Proteins. J Biol Chem 2004; 279:55443-54. [PMID: 15459186 DOI: 10.1074/jbc.m404794200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The PrP-like Doppel (Dpl) protein causes apoptotic death of cerebellar neurons in transgenic mice, a process prevented by expression of the wild type (wt) cellular prion protein, PrP(C). Internally deleted forms of PrP(C) resembling Dpl such as PrPDelta32-121 produce a similar PrP(C)-sensitive pro-apoptotic phenotype in transgenic mice. Here we demonstrate that these phenotypic attributes of wt Dpl, wt PrP(C), and PrPDelta132-121 can be accurately recapitulated by transfected mouse cerebellar granule cell cultures. This system was then explored by mutagenesis of the co-expressed prion proteins to reveal functional determinants. By this means, neuroprotective activity of wt PrP(C) was shown to be nullified by a deletion of the N-terminal charged region implicated in endocytosis and retrograde axonal transport (PrPDelta23-28), by deletion of all five octarepeats (PrPDelta51-90), or by glycine replacement of four octarepeat histidine residues required for selective binding of copper ions (Prnp"H/G"). In the case of Dpl, overlapping deletions defined a requirement for the gene interval encoding helices B and B' (DplDelta101-125). These data suggest contributions of copper binding and neuronal trafficking to wt PrP(C) function in vivo and place constraints upon current hypotheses to explain Dpl/PrP(C) antagonism by competitive ligand binding. Further implementation of this assay should provide a fuller understanding of the attributes and subcellular localizations required for activity of these enigmatic proteins.
Collapse
Affiliation(s)
- Bettina Drisaldi
- Centre for Research in Neurodegenerative Diseases, Tanz Neuroscience Building, 6 Queen's Park Crescent West, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Satoh JI, Yamamura T. Gene expression profile following stable expression of the cellular prion protein. Cell Mol Neurobiol 2004; 24:793-814. [PMID: 15672681 PMCID: PMC11529964 DOI: 10.1007/s10571-004-6920-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
1. The cellular prion protein (PrPC) is expressed widely in neural and nonneural tissues at the highest level in neurons in the central nervous system (CNS). 2. Recent studies indicated that transgenic mice with the cytoplasmic accumulation of PrPC exhibited extensive neurodegeneration in the cerebellum, although the underlying mechanism remains unknown. To identify the genes whose expression is controlled by over-expression of PrPC in human cells, we have established a stable PrPC-expressing HEK293 cell line designated P1 by the site-specific recombination technique. 3. Microarray analysis identified 33 genes expressed differentially between P1 and the parent PrPC-non-expressing cell line among 12,814 genes examined. They included 18 genes involved in neuronal and glial functions, 5 related to production of extracellular matrix proteins, and 2 located in the complement cascade. 4. Northern blot analysis verified marked upregulation in P1 of the brain-specific protein phosphatase 2A beta subunit (PPP2R2B), a causative gene of spinocerebellar ataxia 12, and the cerebellar degeneration-related autoantigen (CDR34) gene associated with development of paraneoplastic cerebellar degeneration. 5. These results indicate that accumulation of PrPC in the cell caused aberrant regulation of a battery of the genes important for specific neuronal function. This represents a possible mechanism underlying PrPC-mediated selective neurodegeneration.
Collapse
Affiliation(s)
- Jun-ichi Satoh
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo, Japan.
| | | |
Collapse
|
224
|
Léna C, Popa D, Grailhe R, Escourrou P, Changeux JP, Adrien J. Beta2-containing nicotinic receptors contribute to the organization of sleep and regulate putative micro-arousals in mice. J Neurosci 2004; 24:5711-8. [PMID: 15215293 PMCID: PMC6729220 DOI: 10.1523/jneurosci.3882-03.2004] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The cholinergic system is involved in arousal and in rapid eye movement sleep (REMS). To evaluate the contribution of nicotinic acetylcholine receptors (nAChRs) to these functions, we studied with polygraphic recordings the regulation of sleep in mice lacking the beta2 subunit gene of the nAChRs, a major component of high-affinity nicotine binding sites in the brain. Nicotine (1-2 mg/kg, i.p.) increased wakefulness in wild-type but not knock-out animals, indicating that beta2-containing nAChRs mediate the arousing properties of nicotine. Under normal conditions, the beta2-/- mice displayed the same amounts of waking, non-REM sleep (NREMS) and REMS as their wild-type counterparts. However, they exhibited longer REMS episodes and a reduced fragmentation of NREMS by events characterized notably by a transient drop in EEG power and frequently associated with EMG activation, tentatively referred to as micro-arousals. Respiration monitoring showed that these events were accompanied with, but not caused by, breathing irregularities. Sleep deprivation of beta2-/- mice resulted in a normal increase in REMS episode duration and NREMS delta power but yielded a reduction of the number of micro-arousals in NREMS. In contrast, in beta2-/- mice, a 1 hr immobilization stress failed to produce the normal rebound in REMS in the following 12 hr and, instead, was associated with increased NREMS fragmentation and sustained corticosterone levels. Our results show that the beta2-containing nAChRs contribute to the organization of sleep by regulating the transient phasic activity in NREMS, the REMS onset and duration, and the REMS-promoting effect of stress.
Collapse
Affiliation(s)
- Clément Léna
- Récepteurs et Cognition, Unité de Recherche Associée Centre National de la Recherche Scientifique, Institut Pasteur, 757242 Paris Cedex 15, France
| | | | | | | | | | | |
Collapse
|
225
|
Havsteen BH. Control of the pathogenic conformational change of the prion protein by an attractor of low order. J Theor Biol 2004; 231:39-48. [PMID: 15363928 DOI: 10.1016/j.jtbi.2004.04.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2003] [Revised: 02/14/2004] [Accepted: 04/23/2004] [Indexed: 10/26/2022]
Abstract
The molecular vibrations of the infectious part of the prion protein has been studied by the methods of nonlinear dynamics using NMR-data of the protein in solution to test a new hypothesis for the nature of the pathogenic isomerization. The result was that the conformational change accompanying the conversion of the physiological form (PrP(c)) to the pathological (PrP(Sc)) one displays the characteristic properties of an attractor of the dimension 2.7+/-0.2, whereas the dimensions of the C-terminal, potentially infectious half of the physiological and the pathological forms are 5.3+/-0.3 and 3.9+/-0.3, resp. A plot of the average RMS of the vibrations per atom of the amino acids along the peptide chain suggests a pivotal role of E(167) and D(196). The vibrations of these residues are strongly dampened by the pathogenic conformational change suggesting that the neutralization to the hydrophobic form facilitates the isomerisation. This observation lends credence to the hypothesis that the pathological conformational change is released by the charge neutralization of the abundant, basic side chains lining the cleft in PrP(c) by a pK-shift caused by a transient ion flux from an action potential or by an RNA ligand. The attractor ensures that only the two conformers PrP(c) and PrP(Sc)-monomer, which are switching the polyadenylation of mRNA for synaptic proteins on and off, prevail. Some xenogenic PrP(Sc) seem to have a tendency to polymerize irreversibly, which is likely to inhibit the translation, thus killing neurons. The autocorrelation function is dampened (long correlation length for the 3 cases), the Poincaré plot seems to show the cross-section of a dense attractor for PrP(Sc) and a loose one for PrP(c), the Lyapunov exponent is positive and the power spectrum is broad. The Hurst plots of PrP(Sc) and PcP(c) show monofractality. The attractor hypothesis offers a supplement, or an alternative, to the current, inconclusive ideas of the nature of the PrP isomerization. The identification of the control factors may permit the reversal of the fatal conformational change. The dynamic parameters were checked by the interpoint distance method of Judd and by an analysis of the Fourier-transformed data.
Collapse
Affiliation(s)
- Bent H Havsteen
- Department of Biochemistry, University of Kiel, Olshausenstrasse 40, Kiel D-24098, Germany.
| |
Collapse
|
226
|
Kovács GG, Kalev O, Gelpi E, Haberler C, Wanschitz J, Strohschneider M, Molnár MJ, László L, Budka H. The prion protein in human neuromuscular diseases. J Pathol 2004; 204:241-7. [PMID: 15476279 DOI: 10.1002/path.1633] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The basis of human prion diseases affecting the nervous system is accumulation of a disease-associated conformer (PrPSc) of the normal cellular prion protein (PrPC). Earlier studies demonstrated increased expression of PrPC in inclusion body myositis (IBM), dermato-, and polymyositis, as well as neurogenic muscle atrophy. To define the spectrum and reliability of PrPC immunoreactivity, its expression was examined systematically in a series of pathologically characterized muscular disorders by means of immunohistochemistry, confocal laser microscopy, and immunogold electron microscopy. Anti-PrPC immunolabelling of rimmed vacuoles was observed in IBM, inclusions of myofibrillary myopathy, targets, regenerating, and atrophic fibres, mononuclear cells, in addition to ragged red fibres in mitochondrial myopathies, and focal sarcolemmal immunostaining in non-diseased controls. Quantitative analysis demonstrated that, in neurogenic muscle lesions, anti-PrPC staining detects a significantly broader spectrum of fibres than anti-vimentin or anti-NCAM. In dystrophic muscle, PrPC expression was mainly restricted to regenerating fibres. In IBM, PrPC expression was not confined to rimmed vacuoles or vacuolated fibres and only a small percentage (7.1%) of rimmed vacuoles were PrPC positive. Ultrastructurally, PrPC was observed in the cytoplasm of lymphocytes, in the myofibrillar network of targets, and in rimmed vacuoles. Knowledge of disease circumstances with altered expression of PrPC is important in the setting of a potentially increased chance for extraneural PrPC-PrPSc conversion. In addition, our observations suggest that PrPC may have a general stress-response effect in various neuromuscular disorders.
Collapse
Affiliation(s)
- Gábor G Kovács
- National Institute of Psychiatry and Neurology, Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Tafti M, Franken P, Dauvilliers Y. Genetic Regulation of Sleep. Sleep 2004. [DOI: 10.1201/9780203496732.ch7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
228
|
McLennan NF, Brennan PM, McNeill A, Davies I, Fotheringham A, Rennison KA, Ritchie D, Brannan F, Head MW, Ironside JW, Williams A, Bell JE. Prion protein accumulation and neuroprotection in hypoxic brain damage. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:227-35. [PMID: 15215178 PMCID: PMC1618524 DOI: 10.1016/s0002-9440(10)63291-9] [Citation(s) in RCA: 179] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The function of the normal conformational isoform of prion protein, PrP(C), remains unclear although lines of research have suggested a role in the cellular response to oxidative stress. Here we investigate the expression of PrP(C) in hypoxic brain tissues to examine whether PrP(C) is in part regulated by neuronal stress. Cases of adult cerebral ischemia and perinatal hypoxic-ischemic injury in humans were compared with control tissues. PrP(C) immunoreactivity accumulates within neuronal processes in the penumbra of hypoxic damage in adult brain, and within neuronal soma in cases of perinatal hypoxic-ischemic injury, and in situ hybridization analysis suggests an up-regulation of PrP mRNA during hypoxia. Rodents also showed an accumulation of PrP(C) in neuronal soma within the penumbra of ischemic lesions. Furthermore, the infarct size in PrP-null mice was significantly greater than in the wild type, supporting the proposed role for PrP(C) in the neuroprotective adaptive cellular response to hypoxic injury.
Collapse
Affiliation(s)
- Neil F McLennan
- National Creutzfeldt-Jakob Disease Surveillance Unit and Pathology (Neuropathology), School of Molecular and Clinical Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, Scotland, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Vyazovskiy VV, Welker E, Fritschy JM, Tobler I. Regional pattern of metabolic activation is reflected in the sleep EEG after sleep deprivation combined with unilateral whisker stimulation in mice. Eur J Neurosci 2004; 20:1363-70. [PMID: 15341608 DOI: 10.1111/j.1460-9568.2004.03583.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Regional differences in EEG slow wave activity (SWA) during sleep after sleep deprivation (SD) may be a consequence of differential metabolic activation of cortical areas. We investigated the relationship between the regional EEG dynamics and 2-deoxyglucose (DG) uptake after SD in mice. Six hours' SD were combined with natural unilateral whisker stimulation in an enriched environment to selectively activate the barrel cortex and motor areas. As expected, an interhemispheric asymmetry of 2-DG uptake was found in the barrel cortex immediately after SD. To test whether sleep contributes to recovery of the asymmetry, the stimulation was followed by either undisturbed sleep or by an additional SD. The asymmetry vanished after recovery sleep but also after the additional period of wakefulness without stimulation. In addition, relative 2-DG uptake in the primary motor cortex and retrosplenial area was significantly higher immediately after the SD than after the additional sleep or wakefulness, whereas no other region differed between the groups. Whisker stimulation elicited a greater increase in EEG SWA during non rapid eye movement sleep in the stimulated hemisphere than in the control hemisphere; this increase lasted for 10 h. Within a hemisphere, the initial increase in SWA was higher in the frontal than in the parietal derivation. We conclude that the regional SWA differences during sleep are use-dependent and may be related to the regional pattern of metabolism during the previous waking episode. However, the regional metabolic recovery is not dependent on sleep, and is not directly reflected in changes in SWA during sleep.
Collapse
Affiliation(s)
- Vladyslav V Vyazovskiy
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstr. 190, CH-8057, Switzerland
| | | | | | | |
Collapse
|
230
|
Abstract
Although human prion diseases are rare, they are invariably fatal, and treatments remain elusive. Hundreds of iatrogenic prion transmissions have occurred in the past two decades, and the bovine spongiform encephalopathy epidemic has raised concerns about prion transmission from cattle to humans. Research into therapeutics for prion disease is being pursued in several centres and prominently includes immunological strategies. Currently, the options that are being explored aim either to mobilize the innate and adaptive immune systems towards prion destruction or to suppress or dedifferentiate the lymphoreticular compartments that replicate prions. This article reviews the pathophysiology of prion diseases in mouse models and discusses their relevance to immunotherapeutic and immunoprophylactic antiprion strategies.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zürich, Switzerland.
| | | |
Collapse
|
231
|
Paisley D, Banks S, Selfridge J, McLennan NF, Ritchie AM, McEwan C, Irvine DS, Saunders PTK, Manson JC, Melton DW. Male infertility and DNA damage in Doppel knockout and prion protein/Doppel double-knockout mice. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:2279-88. [PMID: 15161660 PMCID: PMC1615753 DOI: 10.1016/s0002-9440(10)63784-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The prion protein (PrP) and Doppel (Dpl) have many structural and biochemical properties in common, leading to the suggestion that the lack of an obvious phenotype in PrP-deficient mice maybe because of compensation by Dpl. To test this hypothesis and also investigate the function of Dpl we have generated Prnd(-/-) and Prnp(-/-)/Prnd(-/-) mouse lines. Both develop normally and display an identical male sterility phenotype that differs from that reported for another Prnd(-/-) mouse line. Sperm from both our mutant lines were present at normal concentrations, had normal motility, and no morphological abnormalities. Despite only rarely fertilizing oocytes in vivo, because of an inability to perform the acrosome reaction, mutant sperm were capable of fertilization in vitro, albeit at reduced rates compared to wild type. Elevated levels of oxidative DNA damage were found in both types of mutant sperm and resulting embryos failed at an early stage. Therefore we found no evidence that Dpl compensates for the loss of PrP function in mutant mouse lines, but it does have an important anti-oxidant function necessary for sperm integrity and male fertility.
Collapse
Affiliation(s)
- Derek Paisley
- Molecular Medicine Centre, University of Edinburgh, Edinburgh, Scotland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
UCHIYAMA M, KAMEI Y, TAGAYA H, TAKAHASHI K. Poor compensatory function for sleep loss in delayed sleep phase syndrome and non-24-hour sleep-wake syndrome. Sleep Biol Rhythms 2004. [DOI: 10.1111/j.1479-8425.2004.00086.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
233
|
D'Adamo P, Wolfer DP, Kopp C, Tobler I, Toniolo D, Lipp HP. Mice deficient for the synaptic vesicle protein Rab3a show impaired spatial reversal learning and increased explorative activity but none of the behavioral changes shown by mice deficient for the Rab3a regulator Gdi1. Eur J Neurosci 2004; 19:1895-905. [PMID: 15078563 DOI: 10.1111/j.1460-9568.2004.03270.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Rab proteins are small GTPases involved in intracellular trafficking. Among the 60 different Rab proteins described in mammals, Rab3a is the most abundant in brain, where it is involved in synaptic vesicle fusion and neurotransmitter release. Rab3a constitutive knockout mice (Rab3a(-/-)) are characterized by deficient short- and long-term synaptic plasticity in the mossy fiber pathway and altered circadian motor activity, while no effects on spatial learning have been reported so far for these mice. The goals of this study were to analyse possible behavioral consequences of the lack of synaptic plasticity in the mossy fiber pathway using a broad battery of sensitive behavioral measures that has been used previously to analyse the behavior of Gdi1 mice lacking a protein thought to regulate Rab3a. Rab3a(-/-) mice showed normal acquisition but moderately impaired platform reversal learning in the water maze including reference memory and episodic-like memory tasks. A mild deficit in spatial working memory was also observed when tested in the radial maze. Analysis of explorative behavior revealed increased locomotor activity and enhanced exploratory activity in open field, O-maze, dark/light box and novel object tests. Spontaneous activity in normal home cage settings was unaffected but Rab3a(-/-) mice showed increased motor activity when the home cage was equipped with a wheel. No differences were found for delayed and trace fear conditioning or for conditioned taste aversion learning. Congruent with earlier data, these results suggest that Rab3a-dependent synaptic plasticity might play a specific role in the reactivity to novel stimuli and behavioral stability rather than being involved in memory processing. On the other hand, the phenotypic changes in the Rab3a(-/-) mice bore no relation to the behavioral changes as observed in the Gdi1 mice. Such divergence in phenotypes implies that the putative synaptic interaction between Gdi1 and Rab3a should be reconsidered and re-analysed.
Collapse
Affiliation(s)
- Patrizia D'Adamo
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057, Switzerland.
| | | | | | | | | | | |
Collapse
|
234
|
Asante EA, Li YG, Gowland I, Jefferys JGR, Collinge J. Pathogenic human prion protein rescues PrP null phenotype in transgenic mice. Neurosci Lett 2004; 360:33-6. [PMID: 15082172 DOI: 10.1016/j.neulet.2004.01.049] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2003] [Revised: 01/26/2004] [Accepted: 01/26/2004] [Indexed: 10/26/2022]
Abstract
Infectious prion diseases may be acquired, sporadic or inherited in their aetiology. Inherited prion diseases are caused by coding mutations in the prion protein (PrP) gene. We investigated whether one of the commonest of these mutations, E200K, results in a functionally inactive prion protein by expressing human PrP 200K in transgenic mice homozygous for murine PrP null alleles. We examined the intrinsic properties of hippocampal CA1 pyramidal cells in these mice by measuring the resting potential, time constants and amplitude of the slow after-hyperpolarisation (AHP). These mice show rescue of the reduced slow AHP electrophysiological phenotype found in PrP null mice. Using the AHP as a marker for PrP function, we conclude that this pathogenic PrP mutation, does not significantly affect the normal neuronal function of PrP.
Collapse
Affiliation(s)
- Emmanuel A Asante
- MRC Prion Unit and Department of Neurodegenerative Disease, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | | | | | | | | |
Collapse
|
235
|
Abstract
The normal function of prion protein (PrP) is usually disregarded at the expense of the more fascinating role of PrP in transmissible prion diseases. However, the normal PrP may play an important role in cellular function in the central nervous system, since PrP is highly expressed in neurons and motifs in the sequence of PrP are conserved in evolution. The finding that prion null mice do not have a significant overt phenotype suggests that the normal function of PrP is of minor importance. However, the absence of PrP in cells or in vivo contributes to an increased susceptibility to oxidative stress or apoptosis-inducing insults. An alternative explanation is that the PrP normal function is so important that it is redundant. Probing into the characteristics of PrP has revealed a number of features that could mediate important cellular functions. The neuroprotective actions so far identified with PrP are initiated through cell surface signaling, antioxidant activity, or anti-Bax function. Here, we review the characteristics of the PrP and the evidence that PrP protects against neurodegeneration and neuronal cell death.
Collapse
Affiliation(s)
- Xavier Roucou
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| | - Malcolm Gains
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | - Andréa C LeBlanc
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| |
Collapse
|
236
|
Anderson L, Rossi D, Linehan J, Brandner S, Weissmann C. Transgene-driven expression of the Doppel protein in Purkinje cells causes Purkinje cell degeneration and motor impairment. Proc Natl Acad Sci U S A 2004; 101:3644-9. [PMID: 15007176 PMCID: PMC373516 DOI: 10.1073/pnas.0308681101] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Doppel (Dpl) and Prion (PrP) proteins show 25% sequence identity and share several structural features with only minor differences. Dpl shows a PrP-like fold of its C-terminal globular domain and lacks the flexible N-terminal tail. The physiological functions of both proteins are unknown. However, ubiquitous Dpl overexpression in the brain of PrP knockout mice correlated with ataxia and Purkinje cell degeneration in the cerebellum. Interestingly, a similar phenotype was reported in transgenic mice expressing an N-terminally truncated PrP (DeltaPrP) in Purkinje cells by the L7 promoter (TgL7-DeltaPrP). Coexpression of full-length PrP rescued both the neurological syndromes caused by either Dpl or DeltaPrP. To evaluate whether the two proteins caused cerebellar neurodegeneration by the same mechanism, we generated transgenic mice selectively expressing Dpl in Purkinje cells by the same L7 promoter. Such mice showed ataxia and Purkinje cell loss that depended on the level of Dpl expression. Interestingly, the effects of high levels of Dpl were not counterbalanced by the presence of two Prnp alleles. By contrast, PrP coexpression was sufficient to abrogate motor impairment and to delay the neurodegenerative process caused by moderate level of Dpl. A similar situation was reported for the corresponding TgL7-DeltaPrP mice supporting the concept that Dpl and DeltaPrP cause cell death, possibly by interfering with a common signaling cascade essential for cell survival.
Collapse
Affiliation(s)
- Lucy Anderson
- Medical Research Council Prion Unit and Department of Neurodegenerative Disease, and Division of Neuropathology, Institute of Neurology, University College, Queen Square, London WC1N 3BG, United Kingdom
| | | | | | | | | |
Collapse
|
237
|
Kim C, Morré DJ. Prion proteins and ECTO-NOX proteins exhibit similar oscillating redox activities. Biochem Biophys Res Commun 2004; 315:1140-6. [PMID: 14985132 DOI: 10.1016/j.bbrc.2004.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2004] [Indexed: 01/09/2023]
Abstract
Both recombinant full-length mouse prion protein expressed in Escherichia coli and native prion protein (PrPsc) from mouse brain exhibited NADH oxidase and protein disulfide-thiol interchange activities similar to those formerly thought to be properties exclusive to the growth-related, cell surface ECTO-NOX proteins. The two activities exhibited the complex 2+3 pattern of oscillations characteristic of ECTO-NOX proteins where the two activities alternate to generate a period length of 24 min. The oscillations were augmented by copper and diminished by addition of the copper chelator bathocuproene. That the activity might be attributable to a contaminating protein was ruled out by experiments where the purified recombinant prion-containing extracts were resolved by SDS-PAGE and the activity was restricted to a single band corresponding to the predicted Mr of the recombinant prion as verified by Western blot analyses.
Collapse
Affiliation(s)
- Chinpal Kim
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907-2064, USA
| | | |
Collapse
|
238
|
Abstract
Prions have been responsible for an entire century of tragic episodes. Fifty years ago, kuru decimated the population of Papua New Guinea. Then, iatrogenic transmission of prions caused more than 250 cases of Creutzfeldt-Jakob disease. More recently, transmission of bovine spongiform encephalopathy to humans caused a widespread health scare. On the other hand, the biology of prions represents a fascinating and poorly understood phenomenon, which may account for more than just diseases and may represent a fundamental mechanism of crosstalk between proteins. The two decades since Stanley Prusiner's formulation of the protein-only hypothesis have witnessed spectacular advances, and yet some of the most basic questions in prion science have remained unanswered.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zürich, Schmelzbergstr. 12, CH-8091 Zürich, Switzerland.
| | | |
Collapse
|
239
|
Marques MM, Thomson AJ, McWhir J. Challenges and prospects for targeted transgenesis in livestock. Practical applications of gene targeting. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2004; 534:265-78. [PMID: 12903726 DOI: 10.1007/978-1-4615-0063-6_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Affiliation(s)
- Margarita M Marques
- Department of Gene Expression and Development, Roslin Institute, Roslin, Midlothian, Scotland EH 25 9PS, UK
| | | | | |
Collapse
|
240
|
Silveira JR, Caughey B, Baron GS. Prion protein and the molecular features of transmissible spongiform encephalopathy agents. Curr Top Microbiol Immunol 2004; 284:1-50. [PMID: 15148986 DOI: 10.1007/978-3-662-08441-0_1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Transmissible spongiform encephalopathy (TSE) diseases, or prion diseases, are neurodegenerative diseases found in a number of mammals, including man. Although they are generally rare, TSEs are always fatal, and as of yet there are no practical therapeutic avenues to slow the course of disease. The epidemic of bovine spongiform encephalopathy (BSE) in the UK greatly increased the awareness of TSE diseases. Although it appears that BSE has not spread to North America, chronic wasting disease (CWD), a TSE found in cervids, is causing significant concern. Despite decades of investigation, the exact nature of the infectious agent of the TSEs is still controversial. Although many questions remain, substantial efforts have been made to understand the molecular features of TSE agents, with the hope of enhancing diagnosis and treatment of disease, as well as understanding the fundamental nature of the infectious agent itself. This review summarizes the current understanding of these molecular features, focusing on the role of the prion protein (PrP(c)) and its relationship to the disease-associated isoform (PrP(Sc)).
Collapse
Affiliation(s)
- J R Silveira
- Laboratory of Persistent Viral Diseases, NIAID, NIH, Rocky Mountain Laboratories, 903 S. 4th St., Hamilton, MT 59840, USA
| | | | | |
Collapse
|
241
|
Vyazovskiy V, Achermann P, Borbély AA, Tobler I. Interhemispheric coherence of the sleep electroencephalogram in mice with congenital callosal dysgenesis. Neuroscience 2004; 124:481-8. [PMID: 14980397 DOI: 10.1016/j.neuroscience.2003.12.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2003] [Indexed: 10/26/2022]
Abstract
Regional differences in the effect of sleep deprivation on the sleep electroencephalogram (EEG) may be related to interhemispheric synchronization. To investigate the role of the corpus callosum in interhemispheric EEG synchronization, coherence spectra were computed in mice with congenital callosal dysgenesis (B1) under baseline conditions and after 6-h sleep deprivation, and compared with the spectra of a control strain (C57BL/6). In B1 mice coherence was lower than in controls in all vigilance states. The level of coherence in each of the three totally acallosal mice was lower than in the mice with only partial callosal dysgenesis. The difference between B1 and control mice was present over the entire 0.5-25 Hz frequency range in non-rapid eye movement sleep (NREM sleep), and in all frequencies except for the high delta and low theta band (3-7 Hz) in rapid eye movement (REM) sleep and waking. In control mice, sleep deprivation induced a rise of coherence in the Delta band of NREM sleep in the first 2 h of recovery. This effect was absent in B1 mice with total callosal dysgenesis and attenuated in mice with partial callosal dysgenesis. In both strains the effect of sleep deprivation dissipated within 4 h. The results show that EEG synchronization between the hemispheres in sleep and waking is mediated to a large part by the corpus callosum. This applies also to the functional changes induced by sleep deprivation in NREM sleep. In contrast, interhemispheric synchronisation of theta oscillations in waking and REM sleep may be mediated by direct interhippocampal connections.
Collapse
Affiliation(s)
- V Vyazovskiy
- Institute of Pharmacology and Toxicology, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | | | | | |
Collapse
|
242
|
Nunziante M, Gilch S, Schätzl HM. Prion Diseases: From Molecular Biology to Intervention Strategies. Chembiochem 2003; 4:1268-84. [PMID: 14661267 DOI: 10.1002/cbic.200300704] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Prion diseases are fatal neurodegenerative infectious disorders for which no therapeutic or prophylactic regimens exist. Understanding the molecular process of conformational conversion of the cellular prion protein (PrP(c)) into its pathological isoform (PrP(Sc)) will be necessary to devise effective antiprion strategies. In recent years, new findings in the cell biology of PrP(c), in the molecular pathogenesis of PrP(Sc), and in the cellular quality control mechanisms involved in these scenarios have accumulated. A function of the prion protein in signalling, the possible impact of the proteasome, and aggresomes as intracellular waste deposits have been described. Here, important pathogenetic similarities with the more frequent neurodegenerative disorders are evident. The need for therapeutic, postexposure, and prophylactic possibilities was drastically illustrated by the emergence of variant Creutzfeldt-Jakob disease (vCJD), a new human prion disease caused by bovine spongiform encephalopathy (BSE) derived prions. Although prion infectivity in humans is usually restricted to the central nervous system, in vCJD patients prions are present in the lympho-reticular system, posing a theoretical risk of accidental human-to-human transmission. A variety of chemical antiprion substances have been reported in in vitro and cell culture based assays or in animal studies. Occasionally, they have also made their way into the first human trials. In addition, various promising interference strategies have been devised in transgenic models, although they are usually hard to transfer into nontransgenic in vivo situations. New findings in the fields of peripheral prion pathogenesis and immune system involvement fuelled the search for antiprion strategies formerly considered to be entirely impossible. This opened the door towards classical immunological interference techniques. Remarkably, passive and even active vaccination approaches now seem to be realistic goals.
Collapse
Affiliation(s)
- Max Nunziante
- Institute of Virology/Prion Research Group, Technical University of Munich, Biedersteinerstrasse 29, 80802 Munich, Germany
| | | | | |
Collapse
|
243
|
Chacón MA, Barría MI, Lorca R, Huidobro-Toro JP, Inestrosa NC. A human prion protein peptide (PrP(59-91)) protects against copper neurotoxicity. Mol Psychiatry 2003; 8:853-62, 835. [PMID: 14515136 DOI: 10.1038/sj.mp.4001400] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Human cellular prion protein (PrP(C)) is involved in several neurodegenerative disorders; however, its normal function is unknown. We report here that a synthetic peptide corresponding to the four-octarepeat sequence of the PrP(C) (PrP(59-91)) protects hippocampal neurons against copper neurotoxic effects in vivo. Using a rat bilateral intrahippocampal injection model, we found that PrP(59-91) protects against copper-induced neurotoxicity, including a recovery in spatial learning performance and a reduced neuronal cell loss and astrogliosis. Previous studies from our laboratory indicated that a tryptophan (Trp) residue plays a key role in the reduction of copper(II) to copper(I); therefore several PrP(59-91) fragments lacking histidine (His) and Trp residues were tested for their capacity to protect from copper toxicity. A PrP(59-91) peptide lacking His residue shows as much neuroprotection as the native peptide; however, PrP(59-91) without Trp residues only partially protected against copper toxicity. The neuroprotective effect not only occurs with PrP(59-91), in fact a full neuroprotection was also observed using just one octamer of the N-terminal region of prion protein. We conclude that the N-terminal tandem octarepeat of the human PrP(C) protects neurons against copper toxicity by a differential contribution of the binding (His) and reducing (Trp) copper activities of PrP(59-91). Our results are consistent with the idea that PrP(C) function is related to copper homeostasis.
Collapse
Affiliation(s)
- M A Chacón
- Centro de Regulación Celular y Patología 'Dr Joaquín V Luco', MIFAB, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | |
Collapse
|
244
|
Sakudo A, Lee DC, Saeki K, Nakamura Y, Inoue K, Matsumoto Y, Itohara S, Onodera T. Impairment of superoxide dismutase activation by N-terminally truncated prion protein (PrP) in PrP-deficient neuronal cell line. Biochem Biophys Res Commun 2003; 308:660-7. [PMID: 12914801 DOI: 10.1016/s0006-291x(03)01459-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previous studies have reported a neuroprotective role for cellular prion protein (PrP(C)) against apoptosis induced by serum deprivation in an immortalized prion protein gene (Prnp)-deficient neuronal cell line, but the mechanisms remain unclear. In this study, to investigate the mechanisms by which PrP(C) prevents apoptosis, the authors compared apoptosis of Prnp(-/-) cells with that of Prnp(-/-) cells expressing the wild-type PrP(C) or PrP(C) lacking N-terminal octapeptide repeat region under serum-free conditions. Re-introduction of Prnp rescued cells from apoptosis, upregulated superoxide dismutase (SOD) activity, enhanced superoxide anion elimination, and inhibited caspase-3/9 activation. On the other hand, N-terminally truncated PrP(C) enhanced apoptosis accompanied by potentiation of superoxide production and caspase-3/9 activation due to inhibition of SOD. These results suggest that PrP(C) protects Prnp(-/-) cells from apoptosis via superoxide- and caspase-3/9-dependent pathways by upregulating SOD activity. Furthermore, the octapeptide repeat region of PrP(C) plays an essential role in regulating apoptosis and SOD activity.
Collapse
Affiliation(s)
- Akikazu Sakudo
- Department of Molecular Immunology, School of Agricultural and Life Sciences, University of Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
245
|
Ebihara S, Miyazaki S, Sakamaki H, Yoshimura T. Sleep properties of CS mice with spontaneous rhythm splitting in constant darkness. Brain Res 2003; 980:121-7. [PMID: 12865166 DOI: 10.1016/s0006-8993(03)02947-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In mice, genetic differences between inbred strains have been shown for several parameters of sleep and circadian activity rhythms. Our previous studies have demonstrated that CS mice have three remarkable characteristics in the circadian rhythm of locomotor activity: (1) high activity both during the day and night, (2) unstable freerunning period and (3) spontaneous rhythm splitting. In order to characterize sleep properties of CS mice, we compared circadian sleep patterns of CS with those of C57BL/6J and C3H/He mice which have normal circadian activity rhythms. Although C57BL/6J and C3H/He mice exhibited clear daily sleep-wake rhythms in the amount of each sleep parameter (Awake, SWS, PS), CS mice did not show clear rhythms in these parameters. The differences were particularly conspicuous in PS; no apparent day-night differences in the amount of PS, PS counts and PS interval (the interval between successive PS episodes) in CS mice. In addition, the ratio of PS to total sleep time was significantly larger in CS mice than other strains. Of these parameters, the most considerable was PS latency which was extremely short and direct transition from Awake to PS without appearance of SWS frequently occurred in these mice. These results indicate that CS mice may be useful for the understanding of sleep mechanisms and its dysfunction.
Collapse
Affiliation(s)
- Shizufumi Ebihara
- Division of Biomodeling, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Naogya464-8601, Japan.
| | | | | | | |
Collapse
|
246
|
Cui T, Daniels M, Wong BS, Li R, Sy MS, Sassoon J, Brown DR. Mapping the functional domain of the prion protein. EUROPEAN JOURNAL OF BIOCHEMISTRY 2003; 270:3368-76. [PMID: 12899694 DOI: 10.1046/j.1432-1033.2003.03717.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Prion diseases such as Creutzfeldt-Jakob disease are possibly caused by the conversion of a normal cellular glycoprotein, the prion protein (PrPc) into an abnormal isoform (PrPSc). The process that causes this conversion is unknown, but to understand it requires a detailed insight into the normal activity of PrPc. It has become accepted from results of numerous studies that PrPc is a Cu-binding protein and that its normal function requires Cu. Further work has suggested that PrPc is an antioxidant with an activity like that of a superoxide dismutase. We have shown in this investigation that this activity is optimal for the whole protein and that deletion of parts of the protein reduce or abolish this activity. The protein therefore contains an active domain requiring certain regions such as the Cu-binding octameric repeat region and the hydrophobic core. These regions show high evolutionary conservation fitting with the idea that they are important to the active domain of the protein.
Collapse
Affiliation(s)
- Taian Cui
- Department of Biology and Biochemistry, University of Bath, UK
| | | | | | | | | | | | | |
Collapse
|
247
|
Affiliation(s)
- Adriano Aguzzi
- Department of Pathology, University Hospital of Zürich, Schmelzbergstr. 12, CH-8091 Zürich, Switzerland.
| | | |
Collapse
|
248
|
Kim JI, Kuizon S, Rubenstein R. Comparison of PrP transcription and translation in two murine myeloma cell lines. J Neuroimmunol 2003; 140:137-42. [PMID: 12864981 DOI: 10.1016/s0165-5728(03)00181-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The generation of monoclonal antibodies (MAbs) to the prion protein, PrP, is important in order to establish a large repertoire of useful reagents for the diagnosis of transmissible spongiform encephalopathies (TSE), or prion diseases. However, the presence of PrP on the surface of all mammalian cells (PrP(C)) causes self-recognition, thereby restricting the ability of mice to produce an immune response to the PrP immunogen. Although this problem has been alleviated with the generation and use of PrP-knockout mice, the production of MAbs has continued to be severely hampered presumably since the fusion partner for spleen-derived lymphocytes was PrP(C)-containing myeloma cell lines. The availability of a mouse myeloma cell line expressing little or no PrP(C) on the surface would therefore be useful for MAb generation. Our data indicate that cells differ in their levels of PrP(C) expression and suggest that not all murine myeloma cell lines are equally useful for obtaining hybridomas secreting anti-PrP MAbs.
Collapse
Affiliation(s)
- Jae-Il Kim
- New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA.
| | | | | |
Collapse
|
249
|
|
250
|
Flechsig E, Hegyi I, Leimeroth R, Zuniga A, Rossi D, Cozzio A, Schwarz P, Rülicke T, Götz J, Aguzzi A, Weissmann C. Expression of truncated PrP targeted to Purkinje cells of PrP knockout mice causes Purkinje cell death and ataxia. EMBO J 2003; 22:3095-101. [PMID: 12805223 PMCID: PMC162137 DOI: 10.1093/emboj/cdg285] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PrP knockout mice with disruption of only the PrP-encoding region (Zürich I-type) remain healthy, whereas mice with deletions extending upstream of the PrP-encoding exon (Nagasaki-type) suffer Purkinje cell loss and ataxia, associated with ectopic expression of Doppel in brain, particularly in Purkinje cells. The phenotype is abrogated by co-expression of full-length PrP. Doppel is 25% similar to PrP, has the same globular fold, but lacks the flexible N-terminal tail. We now show that in Zürich I-type PrP-null mice, expression of N-terminally truncated PrP targeted to Purkinje cells also leads to Purkinje cell loss and ataxia, which are reversed by PrP. Doppel and truncated PrP probably cause Purkinje cell degeneration by the same mechanism.
Collapse
Affiliation(s)
- Eckhard Flechsig
- Institut für Molekularbiologie, Universität Zürich, CH-8057 Zürich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|