1
|
Dar KB, Bhat AH, Amin S, Reshi BA, Zargar MA, Masood A, Ganie SA. Elucidating Critical Proteinopathic Mechanisms and Potential Drug Targets in Neurodegeneration. Cell Mol Neurobiol 2020; 40:313-345. [PMID: 31584139 PMCID: PMC11449027 DOI: 10.1007/s10571-019-00741-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/06/2019] [Indexed: 12/18/2022]
Abstract
Neurodegeneration entails progressive loss of neuronal structure as well as function leading to cognitive failure, apathy, anxiety, irregular body movements, mood swing and ageing. Proteomic dysregulation is considered the key factor for neurodegeneration. Mechanisms involving deregulated processing of proteins such as amyloid beta (Aβ) oligomerization; tau hyperphosphorylation, prion misfolding; α-synuclein accumulation/lewy body formation, chaperone deregulation, acetylcholine depletion, adenosine 2A (A2A) receptor hyperactivation, secretase deregulation, leucine-rich repeat kinase 2 (LRRK2) mutation and mitochondrial proteinopathies have deeper implications in neurodegenerative disorders. Better understanding of such pathological mechanisms is pivotal for exploring crucial drug targets. Herein, we provide a comprehensive outlook about the diverse proteomic irregularities in Alzheimer's, Parkinson's and Creutzfeldt Jakob disease (CJD). We explicate the role of key neuroproteomic drug targets notably Aβ, tau, alpha synuclein, prions, secretases, acetylcholinesterase (AchE), LRRK2, molecular chaperones, A2A receptors, muscarinic acetylcholine receptors (mAchR), N-methyl-D-aspartate receptor (NMDAR), glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) and mitochondrial/oxidative stress-related proteins for combating neurodegeneration and associated cognitive and motor impairment. Cross talk between amyloidopathy, synucleinopathy, tauopathy and several other proteinopathies pinpoints the need to develop safe therapeutics with ability to strike multiple targets in the aetiology of the neurodegenerative disorders. Therapeutics like microtubule stabilisers, chaperones, kinase inhibitors, anti-aggregation agents and antibodies could serve promising regimens for treating neurodegeneration. However, drugs should be target specific, safe and able to penetrate blood-brain barrier.
Collapse
Affiliation(s)
- Khalid Bashir Dar
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Aashiq Hussain Bhat
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Shajrul Amin
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Bilal Ahmad Reshi
- Department of Biotechnology, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Mohammad Afzal Zargar
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Akbar Masood
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Showkat Ahmad Ganie
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India.
| |
Collapse
|
2
|
Leighton PLA, Nadolski NJ, Morrill A, Hamilton TJ, Allison WT. An ancient conserved role for prion protein in learning and memory. Biol Open 2018; 7:bio.025734. [PMID: 29358166 PMCID: PMC5829491 DOI: 10.1242/bio.025734] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The misfolding of cellular prion protein (PrPC) to form PrP Scrapie (PrPSc) is an exemplar of toxic gain-of-function mechanisms inducing propagated protein misfolding and progressive devastating neurodegeneration. Despite this, PrPC function in the brain is also reduced and subverted during prion disease progression; thus understanding the normal function of PrPC in healthy brains is key. Disrupting PrPC in mice has led to a myriad of controversial functions that sometimes map onto disease symptoms, including a proposed role in memory or learning. Intriguingly, PrPC interaction with amyloid beta (Aβ) oligomers at synapses has also linked its function to Alzheimer's disease and dementia in recent years. We set out to test the involvement of PrPC in memory using a disparate animal model, the zebrafish. Here we document an age-dependent memory decline in prp2−/− zebrafish, pointing to a conserved and ancient role of PrPC in memory. Specifically, we found that aged (3-year-old) prp2−/− fish performed poorly in an object recognition task relative to age-matched prp2+/+ fish or 1-year-old prp2−/− fish. Further, using a novel object approach (NOA) test, we found that aged (3-year-old) prp2−/− fish approached the novel object more than either age-matched prp2+/+ fish or 1-year-old prp2−/− fish, but did not have decreased anxiety when we tested them in a novel tank diving test. Taken together, the results of the NOA and novel tank diving tests suggest an altered cognitive appraisal of the novel object in the 3-year-old prp2−/− fish. The learning paradigm established here enables a path forward to study PrPC interactions of relevance to Alzheimer's disease and prion diseases, and to screen for candidate therapeutics for these diseases. The findings underpin a need to consider the relative contributions of loss- versus gain-of-function of PrPC during Alzheimer's and prion diseases, and have implications upon the prospects of several promising therapeutic strategies. Summary: Prion protein dysfunction at the synapse impacts learning in Alzheimer disease. Here, we demonstrate similar roles for prion protein in zebrafish, revealing ancient constructive roles for this infamously toxic protein.
Collapse
Affiliation(s)
- Patricia L A Leighton
- Centre for Prions and Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Nathan J Nadolski
- Department of Psychology, MacEwan University, Edmonton, AB T5J 4S2, Canada
| | - Adam Morrill
- Department of Psychology, MacEwan University, Edmonton, AB T5J 4S2, Canada
| | - Trevor J Hamilton
- Department of Psychology, MacEwan University, Edmonton, AB T5J 4S2, Canada .,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1
| | - W Ted Allison
- Centre for Prions and Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada .,Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1.,Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
3
|
ONODERA T. Dual role of cellular prion protein in normal host and Alzheimer's disease. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:155-173. [PMID: 28413194 PMCID: PMC5489426 DOI: 10.2183/pjab.93.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/26/2017] [Indexed: 06/07/2023]
Abstract
Using PrPC-knockout cell lines, it has been shown that the inhibition of apoptosis through STI1 is mediated by PrPC-dependent SOD activation. Antioxidant PrPC may contribute to suppression of inflammasome activation. PrPC is functionally involved in copper metabolism, signal transduction, neuroprotection, and cell maturation. Recently several reports have shown that PrPC participates in trans-membrane signaling processes associated with hematopoietic stem cell replication and neuronal differentiation. In another role, PrPC also tends to function as a neurotoxic protein. Aβ oligomer, which is associated with neurodegeneration in Alzheimer's disease (AD), has also been reported to act as a ligand of PrPC. However, the physiological role of PrPC as an Aβ42-binding protein is not clear. Actually, PrPC is critical in Aβ42-mediated autophagy in neurons. PrPC shows a beneficial role in lipid rafts to promote autophagy. Further search for PrPC-interaction molecules using Prnp-/- mice and various types of Prnp-/- cell lines under various conditions may elucidate other important PrPC important functions.
Collapse
Affiliation(s)
- Takashi ONODERA
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Peggion C, Bertoli A, Sorgato MC. Almost a century of prion protein(s): From pathology to physiology, and back to pathology. Biochem Biophys Res Commun 2016; 483:1148-1155. [PMID: 27581199 DOI: 10.1016/j.bbrc.2016.07.118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 07/27/2016] [Indexed: 12/30/2022]
Abstract
Prions are one of the few pathogens whose name is renowned at all population levels, after the dramatic years pervaded by the fear of eating prion-infected food. If now this, somehow irrational, scare of bovine meat inexorably transmitting devastating brain disorders is largely subdued, several prion-related issues are still unsolved, precluding the design of therapeutic approaches that could slow, if not halt, prion diseases. One unsolved issue is, for example, the role of the prion protein (PrPC), whole conformational misfolding originates the prion but whose physiologic reason d'etre in neurons, and in cells at large, remains enigmatic. Preceded by a historical outline, the present review will discuss the functional pleiotropicity ascribed to PrPC, and whether this aspect could fall, at least in part, into a more concise framework. It will also be devoted to radically different perspectives for PrPC, which have been recently brought to the attention of the scientific world with unexpected force. Finally, it will discuss the possible reasons allowing an evolutionary conserved and benign protein, as PrPC is, to turn into a high affinity receptor for pathologic misfolded oligomers, and to transmit their toxic message into neurons.
Collapse
Affiliation(s)
- Caterina Peggion
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy.
| | - Alessandro Bertoli
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy
| | - M Catia Sorgato
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy; C.N.R. Institute of Neuroscience, University of Padova, Via Bassi 58/B, 35131 Padova, Italy.
| |
Collapse
|
5
|
Chen LN, Sun J, Yang XD, Xiao K, Lv Y, Zhang BY, Zhou W, Chen C, Gao C, Shi Q, Dong XP. The Brain NO Levels and NOS Activities Ascended in the Early and Middle Stages and Descended in the Terminal Stage in Scrapie-Infected Animal Models. Mol Neurobiol 2016; 54:1786-1796. [PMID: 26887380 DOI: 10.1007/s12035-016-9755-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/26/2016] [Indexed: 12/22/2022]
Abstract
The infections of prion agents may cause progressive and fatal neurodegenerative diseases in humans and a serial of animal species. Previous studies have proposed that the levels of nitric oxide (NO) and nitric oxide synthase (NOS) in the brains of some neurodegeneration diseases changed, while S-nitrosylation (SNO) of many brain proteins altered in prion diseases. To elucidate the potential changes of brain NO levels during prion infection, the NO levels and NOS activities in the brain tissues of three scrapie experimental rodents were measured, including scrapie agent 263 K-infected hamsters and 139A- and ME7-infected mice. Both NO levels and NOS activities, including total NOS (TNOS) and inducible NOS (iNOS), were increased at the terminal stages of scrapie-infected animals. Assays of the brain samples collected at different time points during scrapie infection showed that the NO levels and NOS activities started to increase at early stage, reached to the peak in the middle stage, and dropped down at late stage. Western blots for brain iNOS revealed increased firstly and decreased late, especially in the brains of 139A- and ME7-infected mice. In line with those alterations, the levels of the SNO forms of several selected brain proteins such as aquaporin-1 (AQP1), calcium/calmodulin-dependent protein kinase II (CaMKII), neurogranin, and opalin, underwent similar changing trends, while their total protein levels did not change obviously during scrapie infection. Our data here for the first time illustrate the changing profile of brain NO and NOS during prion infection. Time-dependent alterations of brain NO level and the associated protein S-nitrosylation process may contribute greatly to the neuropathological damage in prion diseases.
Collapse
Affiliation(s)
- Li-Na Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China
| | - Jing Sun
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China
| | - Xiao-Dong Yang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China
| | - Kang Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China
| | - Yan Lv
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China
| | - Bao-Yun Zhang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China
| | - Wei Zhou
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China
| | - Cao Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China
| | - Chen Gao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China.
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, People's Republic of China. .,Chinese Academy of Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
6
|
Onodera T, Sakudo A, Tsubone H, Itohara S. Review of studies that have used knockout mice to assess normal function of prion protein under immunological or pathophysiological stress. Microbiol Immunol 2015; 58:361-74. [PMID: 24866463 DOI: 10.1111/1348-0421.12162] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/22/2014] [Accepted: 05/26/2014] [Indexed: 12/29/2022]
Abstract
Deletion of cellular isoform of prion protein (PrP(C)) increases neuronal predisposition to damage by modulating apoptosis and the negative consequences of oxidative stress. In vivo studies have demonstrated that PrP(C)-deficient mice are more prone to seizure, depression, and induction of epilepsy and experience extensive cerebral damage following ischemic challenge or viral infection. In addition, adenovirus-mediated overexpression of PrP(C) reduces brain damage in rat models of cerebral ischemia. In experimental autoimmune encephalomyelitis, PrP(C)-deficient mice reportedly have a more aggressive disease onset and less clinical improvement during the chronic phase than wild-type mice mice. In mice given oral dextran sulfate, PrP(C) has a potential protective role against inflammatory bowel disease. PrP(C)-deficient mice demonstrate significantly greater increases in blood glucose concentrations after intraperitoneal injection of glucose than wild-type mice. Further in vivo challenges to PrP gene-deficient models and conditional knockout models with siRNA and in vivo administration of PrP-ligating agents may assist in refining knowledge of the lymphoid function of PrP(C) and predicting the effects of anti-PrP treatment on the immune system. Together, these findings indicate that PrP(C) may have multiple neuroprotective and anti-inflammatory roles, which explains why this protein is so widely expressed.
Collapse
Affiliation(s)
- Takashi Onodera
- Research Center for Food Safety, School of Agricultural and Life Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-8657
| | | | | | | |
Collapse
|
7
|
Loss of prion protein leads to age-dependent behavioral abnormalities and changes in cytoskeletal protein expression. Mol Neurobiol 2014; 50:923-36. [PMID: 24604355 DOI: 10.1007/s12035-014-8655-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/29/2014] [Indexed: 12/13/2022]
Abstract
The cellular prion protein (PrPC) is a highly conserved protein whose exact physiological role remains elusive. In the present study, we investigated age-dependent behavioral abnormalities in PrPC-knockout (Prnp0/0) mice and wild-type (WT) controls. Prnp0/0 mice showed age-dependent behavioral deficits in memory performance, associative learning, basal anxiety, and nest building behavior. Using a hypothesis-free quantitative proteomic investigation, we found that loss of PrPC affected the levels of neurofilament proteins in an age-dependent manner. In order to understand the biochemical basis of these observations, we analyzed the phosphorylation status of neurofilament heavy chain (NF-H). We found a reduction in NF-H phosphorylation in both Prnp0/0 mice and in PrPC-deficient cells. The expression of Fyn and phospho-Fyn, a potential regulator for NF phosphorylation, was associated with PrPC ablation. The number of β-tubulin III-positive neurons in the hippocampus was diminished in Prnp0/0 mice relative to WT mice. These data indicate that PrPC plays an important role in cytoskeletal organization, brain function, and age-related neuroprotection. Our work represents the first direct biochemical link between these proteins and the observed behavioral phenotypes.
Collapse
|
8
|
Abstract
The human cellular prion protein (PrP(C)) is a glycosylphosphatidylinositol (GPI) anchored membrane glycoprotein with two N-glycosylation sites at residues 181 and 197. This protein migrates in several bands by Western blot analysis (WB). Interestingly, PNGase F treatment of human brain homogenates prior to the WB, which is known to remove the N-glycosylations, unexpectedly gives rise to two dominant bands, which are now known as C-terminal (C1) and N-terminal (N1) fragments. This resembles the β-amyloid precursor protein (APP) in Alzheimer disease (AD), which can be physiologically processed by α-, β-, and γ-secretases. The processing of APP has been extensively studied, while the identity of the cellular proteases involved in the proteolysis of PrP(C) and their possible role in prion biology has remained limited and controversial. Nevertheless, there is a strong correlation between the neurotoxicity caused by prion proteins and the blockade of their normal proteolysis. For example, expression of non-cleavable PrP(C) mutants in transgenic mice generates neurotoxicity, even in the absence of infectious prions, suggesting that PrP(C) proteolysis is physiologically and pathologically important. As many mouse models of prion diseases have recently been developed and the knowledge about the proteases responsible for the PrP(C) proteolysis is accumulating, we examine the historical experimental evidence and highlight recent studies that shed new light on this issue.
Collapse
|
9
|
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]
|
10
|
Bonomo RP, Di Natale G, Rizzarelli E, Tabbì G, Vagliasindi LI. Copper(ii) complexes of prion protein PEG11-tetraoctarepeat fragment: spectroscopic and voltammetric studies. Dalton Trans 2009:2637-46. [DOI: 10.1039/b821727k] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
11
|
Altered neuron excitability and synaptic plasticity in the cerebellar granular layer of juvenile prion protein knock-out mice with impaired motor control. J Neurosci 2008; 28:7091-103. [PMID: 18614678 DOI: 10.1523/jneurosci.0409-08.2008] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Although the role of abnormal prion protein (PrP) conformation in generating infectious brain diseases (transmissible spongiform encephalopathy) has been recognized, the function of PrP in the normal brain remains mostly unknown. In this investigation, we considered the effect of PrP gene knock-out (PrP(0/0)) on cerebellar neural circuits and in particular on granule cells, which show intense PrP expression during development and selective affinity for PrP. At the third postnatal week, when PrP expression would normally attain mature levels, PrP(0/0) mice showed low performance in the accelerating rotarod and runway tests and the functioning of 40% of granule cells was abnormal. Spikes were slow, nonovershooting, and nonrepetitive in relation with a reduction in transient inward and outward membrane currents, and also the EPSPs and EPSCs had slow kinetics. Overall, these alterations closely resembled an immature phenotype. Moreover, in slow-spiking PrP(0/0) granule cells, theta-burst stimulation was unable to induce any long-term potentiation. This profound impairment in synaptic excitation and plasticity was associated with a protracted proliferation of granule cells and disappeared at P40-P50 along with the recovery of normal motor behavior (Büeler et al., 1992). These results suggest that PrP plays an important role in granule cell development eventually regulating cerebellar network formation and motor control.
Collapse
|
12
|
Buchmann A, Mondadori CR, Hänggi J, Aerni A, Vrticka P, Luechinger R, Boesiger P, Hock C, Nitsch RM, de Quervain DJF, Papassotiropoulos A, Henke K. Prion protein M129V polymorphism affects retrieval-related brain activity. Neuropsychologia 2008; 46:2389-402. [DOI: 10.1016/j.neuropsychologia.2008.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 03/03/2008] [Accepted: 03/06/2008] [Indexed: 01/08/2023]
|
13
|
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland,
| | - Frank Baumann
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland,
| | - Juliane Bremer
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland,
| |
Collapse
|
14
|
Linden R, Martins VR, Prado MAM, Cammarota M, Izquierdo I, Brentani RR. Physiology of the prion protein. Physiol Rev 2008; 88:673-728. [PMID: 18391177 DOI: 10.1152/physrev.00007.2007] [Citation(s) in RCA: 435] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Prion diseases are transmissible spongiform encephalopathies (TSEs), attributed to conformational conversion of the cellular prion protein (PrP(C)) into an abnormal conformer that accumulates in the brain. Understanding the pathogenesis of TSEs requires the identification of functional properties of PrP(C). Here we examine the physiological functions of PrP(C) at the systemic, cellular, and molecular level. Current data show that both the expression and the engagement of PrP(C) with a variety of ligands modulate the following: 1) functions of the nervous and immune systems, including memory and inflammatory reactions; 2) cell proliferation, differentiation, and sensitivity to programmed cell death both in the nervous and immune systems, as well as in various cell lines; 3) the activity of numerous signal transduction pathways, including cAMP/protein kinase A, mitogen-activated protein kinase, phosphatidylinositol 3-kinase/Akt pathways, as well as soluble non-receptor tyrosine kinases; and 4) trafficking of PrP(C) both laterally among distinct plasma membrane domains, and along endocytic pathways, on top of continuous, rapid recycling. A unified view of these functional properties indicates that the prion protein is a dynamic cell surface platform for the assembly of signaling modules, based on which selective interactions with many ligands and transmembrane signaling pathways translate into wide-range consequences upon both physiology and behavior.
Collapse
Affiliation(s)
- Rafael Linden
- Instituto de Biofísica da Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | | |
Collapse
|
15
|
Bonomo RP, Pappalardo G, Rizzarelli E, Tabbì G, Vagliasindi LI. Studies of nitric oxide interaction with mono- and dinuclear copper(II) complexes of prion protein bis-octarepeat fragments. Dalton Trans 2008:3805-16. [PMID: 18629402 DOI: 10.1039/b719930a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The interaction of nitric oxide with copper(ii) complexes of two octarepeat sequences belonging to the prion protein was studied, considering both mononuclear and dinuclear systems, i.e. Cu-Ac-(PHGGGWGQ)(2)-NH(2) and Cu(2)-Ac-(PHGGGWGQ)(2)-NH(2), respectively. The NO interaction with both systems was followed in aqueous solutions at physiological pH value, by using UV-Vis and EPR spectroscopic techniques as well as cyclic voltammetry. The mechanism of NO interaction with the mononuclear copper complex can be considered similar to that previously observed for the analogous copper systems with Ac-HGGG-NH(2) and Ac-PHGGGWGQ-NH(2). A more complicated behaviour was found with the copper dinuclear system, in which the involvement of two different intermediate complex species was evidenced. A positive cooperativity between the two copper ions, in the reduction process was inferred. When working with a large excess of the Ac-(PHGGGWGQ)(2)-NH(2) ligand, the frozen-solution EPR parameters pertain to the well characterized [Cu(N(im))(4)](2+) unit, which did not exhibit any interaction with NO. The presence of a free coordination site is the necessary requirement for the NO interaction to occur, as found only in the square-pyramidal geometry of [Cu(L)H(-2)] or [Cu(2)(L)H(-4)] complex species, which form when copper and ligand concentrations are similar.
Collapse
Affiliation(s)
- Raffaele P Bonomo
- Dipartimento di Scienze Chimiche, Università di Catania, Viale A. Doria 6, 95125, Catania, Italy.
| | | | | | | | | |
Collapse
|
16
|
Cellular prion protein prevents brain damage after encephalomyocarditis virus infection in mice. Arch Virol 2008; 153:1007-12. [DOI: 10.1007/s00705-008-0086-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 04/01/2008] [Indexed: 12/26/2022]
|
17
|
Jiang L, Fernandes D, Mehta N, Bean JL, Michaelis ML, Zaidi A. Partitioning of the plasma membrane Ca2+-ATPase into lipid rafts in primary neurons: effects of cholesterol depletion. J Neurochem 2007; 102:378-88. [PMID: 17596212 DOI: 10.1111/j.1471-4159.2007.04480.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Spatial and temporal alterations in intracellular calcium [Ca(2+)](i) play a pivotal role in a wide array of neuronal functions. Disruption in Ca(2+) homeostasis has been implicated in the decline in neuronal function in brain aging and in neurodegenerative disorders. The plasma membrane Ca(2+)-ATPase (PMCA) is a high affinity Ca(2+) transporter that plays a crucial role in the termination of [Ca(2+)](i) signals and in the maintenance of low [Ca(2+)](i) essential for signaling. Recent evidence indicates that PMCA is uniquely sensitive to its lipid environment and is stimulated by lipids with ordered acyl chains. Here we show that both PMCA and its activator calmodulin (CaM) are partitioned into liquid-ordered, cholesterol-rich plasma membrane microdomains or 'lipid rafts' in primary cultured neurons. Association of PMCA with rafts was demonstrated in preparations isolated by sucrose density gradient centrifugation and in intact neurons by confocal microscopy. Total raft-associated PMCA activity was much higher than the PMCA activity excluded from these microdomains. Depletion of cellular cholesterol dramatically inhibited the activity of the raft-associated PMCA with no effect on the activity of the non-raft pool. We propose that association of PMCA with rafts represents a novel mechanism for its regulation and, consequently, of Ca(2+) signaling in the central nervous system.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, Kansas 66045, USA
| | | | | | | | | | | |
Collapse
|
18
|
Lee KJ, Panzera A, Rogawski D, Greene LE, Eisenberg E. Cellular prion protein (PrPC) protects neuronal cells from the effect of huntingtin aggregation. J Cell Sci 2007; 120:2663-71. [PMID: 17635996 DOI: 10.1242/jcs.004598] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The effect of normal cellular prion protein (PrP(C)) on abnormal protein aggregation was examined by transfecting huntingtin fragments (Htt) into SN56 neuronal-derived cells depleted of PrP(C) by RNA interference. PrP(C) depletion caused an increase in both the number of cells containing granules and the number of apoptotic cells. Consistent with the increase in Htt aggregation, PrP(C) depletion caused an decrease in proteasome activity and a decrease in the activities of cellular defense enzymes compared with control cells whereas reactive oxygen species (ROS) increased more than threefold. Therefore, PrP(C) may protect against Htt toxicity in neuronal cells by increasing cellular defense proteins, decreasing ROS and increasing proteasome activity thereby increasing Htt degradation. Depletion of endogenous PrP(C) in non-neuronal Caco-2 and HT-29 cells did not affect ROS levels or proteasome activity suggesting that only in neuronal cells does PrP(C) confer protection against Htt toxicity. The protective effect of PrP(C) was further evident in that overexpression of mouse PrP(C) in SN56 cells transfected with Htt caused a decrease in both the number of cells with Htt granules and the number of apoptotic cells, whereas there was no effect of PrP(C) expression in non-neuronal NIH3T3 or CHO cells. Finally, in chronically scrapie (PrP(Sc))-infected cells, ROS increased more than twofold while proteasome activity was decreased compared to control cells. Although this could be a direct effect of PrP(Sc), it is also possible that, since PrP(C) specifically prevents pathological protein aggregation in neuronal cells, partial loss of PrP(C) itself increases PrP(Sc) aggregation.
Collapse
Affiliation(s)
- Kyung-Jin Lee
- Laboratory of Cell Biology, NHBLI, NIH, Bethesda, MD 20892-0301, USA
| | | | | | | | | |
Collapse
|
19
|
Diez M, Groth D, DeArmond SJ, Prusiner SB, Hökfelt T. Changes in neuropeptide expression in mice infected with prions. Neurobiol Aging 2007; 28:748-65. [PMID: 16621165 DOI: 10.1016/j.neurobiolaging.2006.02.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Revised: 02/24/2006] [Accepted: 02/28/2006] [Indexed: 10/24/2022]
Abstract
Prion diseases are neurodegenerative disorders characterized by accumulation of an aberrantly folded isoform (PrP(Sc)) of the normal prion protein (PrP(C)). Using in situ hybridization and immunohistochemistry, we have studied changes in the expression of neuropeptides, acetylcholinesterase and tyrosine hydroxylase in CD1 and FVB wild-type mouse strains, as well as in PrP(C) null mice and in mice overexpressing PrP(C) following intracerebral inoculation with RML or Me7 prions. In the immunohistochemical analysis, neuropeptide Y (NPY), enkephalin and dynorphin-like immunoreactivities increased in mossy fibers of CD1 and FVB mice inoculated with either RML- or Me7 prions, whereas cholecystokinin-like immunoreactivity was decreased. These changes in peptide levels were paralleled by an increase in the transcripts in granule cells for neuropeptide Y, enkephalin, and cholecystokinin. However, the dynorphin transcript was decreased in the granule cells. The changes occurred more rapidly in PrP(C)-overexpressing compared to wild-type mice, and could not be found at all in PrP(C)-knockout mice. These changes in peptide expression, which mostly occur before appearance of symptoms of disease, may reflect attempts to initiate protective and/or regenerative processes.
Collapse
Affiliation(s)
- Margarita Diez
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
20
|
Fernández AP, Serrano J, Rodrigo J, Monleón E, Monzón M, Vargas A, Badiola JJ, Martínez-Murillo R, Martínez A. Changes in the expression pattern of the nitrergic system of ovine cerebellum affected by scrapie. J Neuropathol Exp Neurol 2007; 66:196-207. [PMID: 17356381 DOI: 10.1097/01.jnen.0000248557.37832.b4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The constitutive and inducible isoforms of nitric oxide synthase (NOS) and the end-product of nitration, nitrotyrosine, were analyzed by immunohistochemistry, Western blotting, and enzymatic activity in sheep at different stages of the prion disease, scrapie. Four groups were studied: 1) nonaffected (control), 2) preclinical, 3) clinical, and 4) terminal. Constitutive neuronal NOS (nNOS) was the most abundant isoform present in cerebellar neurons of the sheep. Expression of nNOS increased in preclinical animals but diminished in the late stages of the disease. The Purkinje cells that usually are not immunoreactive for this protein became immunopositive in the clinical phase. In unaffected sheep, the inducible isoform (iNOS) was slightly positive in the Purkinje cells. As the disease progressed, the immunoreactivity of Purkinje neurons for iNOS increased. At the final stages, numerous iNOS-positive microglial cells were found in the molecular layer. There was a basal level of protein nitration in the cerebellum of unaffected sheep, especially in the molecular layer. As the disease progressed, the distal prolongations of the Purkinje cells and the astroglia became immunoreactive for nitrotyrosine. Our results suggest that the nitrergic system reacts to the progression of spongiform diseases and may be part of their pathogenesis mechanism.
Collapse
Affiliation(s)
- Ana Patricia Fernández
- Department of Neuroanatomy and Cell Biology, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lalatta-Costerbosa G, Mazzoni M, Clavenzani P, Di Guardo G, Mazzuoli G, Marruchella G, De Grossi L, Agrimi U, Chiocchetti R. Nitric oxide synthase immunoreactivity and NADPH-d histochemistry in the enteric nervous system of Sarda breed sheep with different PrP genotypes in whole-mount and cryostat preparations. J Histochem Cytochem 2007; 55:387-401. [PMID: 17210925 DOI: 10.1369/jhc.6a7052.2007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Until now, significant differences in the neurochemical pattern of enteric neurons have been demonstrated in all species studied; however, some strong similarities also occur across species, such as the occurrence of nitric oxide synthase immunoreactivity (NOS-IR) in inhibitory motor neurons to muscle. In consideration of the insufficient data regarding the enteric nervous system (ENS) of sheep, we investigated the myenteric plexus and submucosal plexus of the ovine ileum. Since the pivotal role of the ENS in the early pathogenesis of sheep scrapie, the "prototype" of prion diseases, has been suggested, we have focused our observations also on the host's PrP genotype. We have studied the morphology and distribution of NOS-IR neurons and their relationships with the enteric glia in whole-mount preparations and in cryostat sections. NOS-IR neurons, always encircled by glial processes, were located in both plexuses. Many NOS-IR fibers were seen in the circular muscle layer, in the submucosa, and in the mucosa. In the submucosa they were close to the lymphoid tissue. No differences in the distribution and percentage of NOS-IR fibers and neurons were observed among sheep carrying different PrP genotype, thus making unlikely their contribution in the determinism of susceptibility/resistance to scrapie infection.
Collapse
|
22
|
Bonomo RP, Pappalardo G, Rizzarelli E, Santoro AM, Tabbì G, Vagliasindi LI. Nitrogen oxide interaction with copper complexes formed by small peptides belonging to the prion protein octa-repeat region. Dalton Trans 2007:1400-8. [PMID: 17387400 DOI: 10.1039/b617408f] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The interaction between NO and copper(II) complexes formed by peptides coming from the N-terminal prion protein octa-repeat region was studied. Aqueous solutions of the Cu-Ac-HGGG-NH(2) and the Cu-Ac-PHGGGWGQ-NH(2) systems around pH 7.5 were tested after the addition of NONOates as a source of NO. UV-Vis, room temperature and frozen solution EPR spectra showed the occurrence of copper(ii) reduction in all these complexes. The reduction of these complexes is probably mediated by the formation of a labile NO adduct, which, after re-oxidation, leads to a relatively stable NO(2)(-) adduct through the apical coordination along the void site of their square pyramidal structure. In fact, the most significant shifts in EPR magnetic parameters (g(||) and A(||) or g(iso) and A(iso)) as well as in the optical parameters (lambda(max) and epsilon(max)) gave a reason for geometrical changes of the copper coordination polyhedron from a distorted square pyramid to a pseudo-octahedron. The presence of oxygen in the aqueous solution hindered the reduction ability of NO towards copper, but it made it easier to return to the original species. In order to elucidate the possible mechanism of this interaction, the reduction of copper complexed by these ligands was followed by means of zinc powder addition. The further addition of nitrite to the solution containing reduced copper led to the conclusion that nitrite could easily form an adduct, which after re-oxidation presented the same spectral features of the species obtained when the NO interaction was followed. The complexity of this interaction could involve both an inner or an outer-sphere electron transfer mechanism.
Collapse
Affiliation(s)
- Raffaele P Bonomo
- Dipartimento di Scienze Chimiche, Università di Catania, Viale A. Doria 6, 95125, Catania, Italy.
| | | | | | | | | | | |
Collapse
|
23
|
Cheng F, Lindqvist J, Haigh CL, Brown DR, Mani K. Copper-dependent co-internalization of the prion protein and glypican-1. J Neurochem 2006; 98:1445-57. [PMID: 16923158 DOI: 10.1111/j.1471-4159.2006.03981.x] [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] [Indexed: 01/08/2023]
Abstract
Heparan sulfate chains have been found to be associated with amyloid deposits in a number of diseases including transmissible spongiform encephalopathies. Diverse lines of evidence have linked proteoglycans and their glycosaminoglycan chains, and especially heparan sulfate, to the metabolism of the prion protein isoforms. Glypicans are a family of glycosylphosphatidylinositol-anchored, heparan sulfate-containing, cell-associated proteoglycans. Cysteines in glypican-1 can become nitrosylated by endogenously produced nitric oxide. When glypican-1 is exposed to a reducing agent, such as ascorbate, nitric oxide is released and autocatalyses deaminative cleavage of heparan sulfate chains. These processes take place while glypican-1 recycles via a non-classical, caveolin-associated pathway. We have previously demonstrated that prion protein provides the Cu2+ ions required to nitrosylate thiol groups in the core protein of glypican-1. By using confocal immunofluorescence microscopy and immunomagnetic techniques, we now show that copper induces co-internalization of prion protein and glypican-1 from the cell surface to perinuclear compartments. We find that prion protein is controlling both the internalization of glypican-1 and its nitric oxide-dependent autoprocessing. Silencing glypican-1 expression has no effect on copper-stimulated prion protein endocytosis, but in cells expressing a prion protein construct lacking the copper binding domain internalization of glypican-1 is much reduced and autoprocessing is abrogated. We also demonstrate that heparan sulfate chains of glypican-1 are poorly degraded in prion null fibroblasts. The addition of either Cu2+ ions, nitric oxide donors, ascorbate or ectopic expression of prion protein restores heparan sulfate degradation. These results indicate that the interaction between glypican-1 and Cu2+-loaded prion protein is required both for co-internalization and glypican-1 self-pruning.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Experimental Medical Science, Division of Neuroscience, Glycobiology Group, Lund University, Lund, Sweden
| | | | | | | | | |
Collapse
|
24
|
Freixes M, Rodríguez A, Dalfó E, Ferrer I. Oxidation, glycoxidation, lipoxidation, nitration, and responses to oxidative stress in the cerebral cortex in Creutzfeldt-Jakob disease. Neurobiol Aging 2005; 27:1807-15. [PMID: 16310893 DOI: 10.1016/j.neurobiolaging.2005.10.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2005] [Revised: 10/08/2005] [Accepted: 10/18/2005] [Indexed: 11/21/2022]
Abstract
Gel electrophoresis and Western blotting of frontal cortex homogenates have been carried out in sporadic Creutzfeldt-Jakob disease (CJD) cases and age-matched controls to gain understanding of the expression of glycation-end products (AGEs). N-Carboxymethyl-lysine (CML) and N-carboxyethyl-lysine (CEL) were used as markers of glycoxidation; 4-hydroxynonenal (4-HNE) and malondialdehyde-lysine (MDAL) as markers of lipoxidation; and nitrotyrosine (N-tyr) and neuronal, endothelial and inducible nitric oxide synthase (nNOS, eNos and iNos) as markers of protein nitration and as sources of NO production, respectively. Age receptor (RAGE) and Cu/Zn superoxide dismutase (SOD1) and Mn superoxide dismutase (SOD2) expression levels were also examined. The results showed a significant increase in the expression levels of AGE (p<0.05), CEL (p<0.001), RAGE (p<0.05), HNE-modified proteins (p<0.01), nNOS, iNOS and eNOS (p<0.01 and p<0.05, respectively), N-tyr (p<0.05), and SOD1 (p<0.05) and SOD2 (p<0.05). No relationship was observed between PrP genotype, PrP type, PrP burden, and expression levels of oxidative stress markers. The present findings demonstrate oxidative, glycoxidative, lipoxidative and nitrative protein damage, accompanied by increased oxidative responses, in the cerebral cortex in sporadic CJD. These results provide support for the concept that oxidative stress may have important implications in the pathogenesis of prion diseases.
Collapse
Affiliation(s)
- M Freixes
- Institut de Neuropatologia, Servei Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | | | | | | |
Collapse
|
25
|
Galvan C, Camoletto PG, Dotti CG, Aguzzi A, Ledesma MD. Proper axonal distribution of PrPC depends on cholesterol–sphingomyelin-enriched membrane domains and is developmentally regulated in hippocampal neurons. Mol Cell Neurosci 2005; 30:304-15. [PMID: 16139509 DOI: 10.1016/j.mcn.2005.07.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Revised: 06/13/2005] [Accepted: 07/07/2005] [Indexed: 11/25/2022] Open
Abstract
Defects in cellular localization and trafficking seem to facilitate the conversion of PrP(C) into the disease-associated form, PrP(Sc). Still, it is not clear to which membrane compartments PrP(C) localizes in hippocampal neurons a population most affected in the prion disease. We here show that in developing hippocampal neurons in culture PrP(C) is equally distributed to all neurites yet enriched in growth cones. By contrast, in fully mature neurons PrP(C) is restricted to axons. The axonal distribution in mature stages is paralleled by the increased partitioning of PrP(C) into detergent-resistant cholesterol-sphingolipid-rich domains (DRMs). Consistent with a cause-effect mechanism, disruption of DRMs by sphingolipid or cholesterol depletion leads to the non-polarized distribution and impaired endocytosis of PrP(C). These results indicate that DRMs are essential for proper trafficking and distribution of PrP(C) at late stages of neuronal differentiation and that its function, at least in hippocampus, is restricted to the axonal domain.
Collapse
Affiliation(s)
- Cristian Galvan
- Fondazione Cavalieri Ottolenghi Scientific Institute, Universita degli Studi di Torino, A.O. San Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano (Turin), Italy
| | | | | | | | | |
Collapse
|
26
|
Nico PBC, Lobão-Soares B, Landemberger MC, Marques W, Tasca CI, de Mello CF, Walz R, Carlotti CG, Brentani RR, Sakamoto AC, Bianchin MM. Impaired exercise capacity, but unaltered mitochondrial respiration in skeletal or cardiac muscle of mice lacking cellular prion protein. Neurosci Lett 2005; 388:21-6. [PMID: 16039050 DOI: 10.1016/j.neulet.2005.06.033] [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] [Received: 01/07/2005] [Revised: 05/13/2005] [Accepted: 06/11/2005] [Indexed: 10/25/2022]
Abstract
The studies of physiological roles for cellular prion protein (PrP(c)) have focused on possible functions of this protein in the CNS, where it is largely expressed. However, the observation that PrP(c) is expressed also in muscle tissue suggests that the physiological role of PrP(c) might not be limited to the central nervous system. In the present study, we investigated possible functions of PrP(c) in muscle using PrP(c) gene (Prnp) null mice (Prnp(0/0)). For this purpose, we submitted Prnp(0/0) animals to different protocols of exercise, and compared their performance to that of their respective wild-type controls. Prnp(0/0) mice showed an exercise-dependent impairment of locomotor activity. In searching for possible mechanisms associated with the impairment observed, we evaluated mitochondrial respiration (MR) in skeletal or cardiac muscle from these mice during resting or after different intensities of exercise. Baseline MR (states 3 and 4), respiratory control ratio (RCR) and mitochondrial membrane potential (DeltaPsi) were evaluated and were not different in skeletal or cardiac muscle tissue of Prnp(0/0) mice when compared with wild-type animals. We concluded that Prnp(0/0) mice show impairment of swimming capacity, perhaps reflecting impairment of muscular activity under more extreme exercise conditions. In spite of the mitochondrial abnormalities reported in Prnp(0/0) mice, our observation seems not to be related to MR. Our results indicate that further investigations should be conducted in order to improve our knowledge about the function of PrP(c) in muscle physiology and its possible role in several different neuromuscular pathologies.
Collapse
Affiliation(s)
- Patrícia Barreto Costa Nico
- Departamento de Neurologia, Psiquiatria e Psicologia Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (FMRP-USP), Hospital das Clínicas/CIREP, Campus Universitário-Monte Alegre, Ribeirão Preto, SP 14048 900, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Nico PBC, de-Paris F, Vinadé ER, Amaral OB, Rockenbach I, Soares BL, Guarnieri R, Wichert-Ana L, Calvo F, Walz R, Izquierdo I, Sakamoto AC, Brentani R, Martins VR, Bianchin MM. Altered behavioural response to acute stress in mice lacking cellular prion protein. Behav Brain Res 2005; 162:173-81. [PMID: 15970215 DOI: 10.1016/j.bbr.2005.02.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Revised: 01/31/2005] [Accepted: 02/10/2005] [Indexed: 10/25/2022]
Abstract
Although many studies have investigated the function of cellular prion protein (PrPc), its physiologic role remains elusive. PrPc null mice have been reported to develop normally and to show normal performance in most behavioural tests. In the present study we investigated whether this also holds true after episodes of acute stress. PrPc gene ablated (Prnp0/0) and wild-type mice were subjected to restraint stress, electric foot shock, or swimming and compared with non-stressed animals. Immediately after the stressful situation, the anxiety levels and locomotion of the animals were measured using plus-maze and open-field tests. Among non-stressed animals, there was no significant difference in performance between Prnp0/0 and wild type animals in either test. However, after acute stress provoked by a foot shock or a swimming trial, Prnp0/0 animals showed a significant decrease in anxiety levels when compared with control animals. Moreover, after the swimming test, knockout mice presented decreased locomotion when compared to wild-type mice. Because of this observation, we also assessed both types of mice in a forced swimming test with the objective of better evaluating muscle function and found that Prnp0/0 animals presented reduced forced swimming capacity when compared to controls. As far as we know, this is the first report suggesting that cellular prion protein is involved in modulation of anxiety or muscular activity after acute psychic or physical stress.
Collapse
Affiliation(s)
- Patrícia Barreto Costa Nico
- Departamento de Neurologia, Faculdade de Medicina de Ribeirão Preto, Psiquiatria e Psicologia Médica, Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brasil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Dremina ES, Sharov VS, Schöneich C. Protein tyrosine nitration in rat brain is associated with raft proteins, flotillin-1 and α-tubulin: effect of biological aging. J Neurochem 2005; 93:1262-71. [PMID: 15934946 DOI: 10.1111/j.1471-4159.2005.03115.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein 3-nitrotyrosine (3-NY) immunoreactivity of rat brain homogenate was localized to a ca. 50 kDa protein band by western blot (WB) analysis. The nitrated proteins were localized to the raft fraction obtained by centrifugation of the homogenate in a sucrose density gradient, which contained specific raft markers such as flotillin-1 and caveolin-1. Purification of the nitrated raft proteins either by a combination of reversed-phase high-performance liquid chromatography (HPLC) and sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE) or by immunoprecipitation (IP) with protein- and modification-specific antibodies coupled to WB and HPLC-electrospray ionization-tandem mass spectrometry (ESI--MS/MS) analysis allowed us to identify two proteins modified by 3-NY: flotillin-1 and alpha-tubulin. Both alpha- and beta-tubulin were detected in the rat brain raft fraction as abundant proteins, which co-immunoprecipitate with flotillin-1 and caveolin-1. Importantly, some protein-protein interactions in rafts were disrupted in 3-NY-containing proteins, e.g. caveolin-1 was dissociated from a complex with flotillin-1 and alpha-tubulin. The analysis of age dependencies did not show any significant change in protein nitration and expression of flotillin-1 and alpha-tubulin, but a decrease in the brain caveolin-1 level for old (34 months) versus young (6 months) rats. The putative mechanism of nitric oxide synthase (NOS) activity regulation by the level of caveolin expression and raft protein-protein interactions is discussed.
Collapse
Affiliation(s)
- Elena S Dremina
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, USA
| | | | | |
Collapse
|
29
|
Abstract
Prions--pathogens that are lethal to humans and other animals--are thought to be conformational isomers of the cellular prion protein. Their unique biology, and the potential for a wider pathobiological significance of prion-like mechanisms, has motivated much research into understanding prion neurodegeneration. Moreover, concerns that extensive dietary exposure to bovine spongiform encephalopathy (BSE) prions might have infected many individuals--who might eventually develop its human counterpart, variant Creutzfeldt-Jakob disease (vCJD)--has focused much interest on therapeutics. The challenge of interrupting this aggressive, diffuse and uniformly fatal neurodegenerative process is daunting. However, the recent finding that the onset of clinical disease in established neuroinvasive prion infection in a mouse model can be halted and early pathology reversed is a source for considerable optimism. A therapeutic focus on the cellular prion protein, rather than prions themselves, which might not be directly neurotoxic, is suggested.
Collapse
Affiliation(s)
- Giovanna Mallucci
- Medical Research Council Prion Unit and Department of Neurodegenerative Disease, Institute of Neurology, University College London, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| | | |
Collapse
|
30
|
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
|
31
|
Hijazi N, Shaked Y, Rosenmann H, Ben-Hur T, Gabizon R. Copper binding to PrPC may inhibit prion disease propagation. Brain Res 2004; 993:192-200. [PMID: 14642846 DOI: 10.1016/j.brainres.2003.09.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although it has been well established that PrP(C), the normal isoform of PrP(Sc), is a copper-binding protein, the role of this metal in the function of PrP(C) as well as in prion disease pathology remains unclear. Here, we show that when scrapie-infected neuroblastoma cells were cultured in the presence of copper, the accumulation of PrP(Sc) in these cells was markedly reduced. In addition, our results indicate that when normal neuroblastoma cells were cultured in the presence of copper ions, they could no longer bind and internalize PrP(Sc). In another set of experiments, copper was added to the drinking water of normal and scrapie-infected hamsters. Our results show that administration of copper to normal hamsters induced cerebellar PrP(C) accumulation. Most important, a significant delay in prion disease onset was observed when scrapie-infected hamsters were treated with copper. As shown before for neuroblastoma cells, also in vivo most of the copper-induced accumulation of PrP(C) was intracellular. We hypothesized that PrP(C) internalization by copper may hinder PrP(Sc) interaction with this molecule, and thereby affect prion disease propagation.
Collapse
Affiliation(s)
- Nuha Hijazi
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital, Ein Karem, Jerusalem 91120, Israel
| | | | | | | | | |
Collapse
|
32
|
Picanço-Diniz CW, Boche D, Gomes-Leal W, Perry VH, Cunningham C. Neuropil and neuronal changes in hippocampal NADPH-diaphorase histochemistry in the ME7 model of murine prion disease. Neuropathol Appl Neurobiol 2004; 30:292-303. [PMID: 15175082 DOI: 10.1111/j.1365-2990.2004.00537.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nitric oxide (NO) has been implicated in neurotoxicity and cerebral blood flow changes in chronic neurodegeneration, but its activity in the mammalian prion diseases has not been studied in detail. Nicotine adenine dinucleotide phosphate (NADPH)-diaphorase (NADPH-d) histochemistry is a simple and robust histochemical procedure that allows localization of the tissue distribution of NO synthases. The aim of the present study is to assess whether NADPH-d histochemical activity is altered in the hippocampus in the ME7 model of prion disease in C57BL/6J mice. At early and late stages after the initiation of the disease we assessed features of the NADPH-d positive cells and the neuropil histochemical activity in CA1 and dentate gyrus using densitometric analysis. In C57BL/6J mice 13 weeks postinjection of the prion agent ME7, when behavioural changes first become apparent, neuropil NADPH-d histochemical staining increases, whereas at late stages it decreases dramatically. Both type I and type II NADPH-d positive cells were found to survive throughout the hippocampal formation into the late stages of the disease, but diaphorase activity was reduced in dendritic branches and abnormal varicosities were present in both dendritic and axonal processes of NADPH-d positive type I cells. The pathophysiological implications of the results remain to be investigated but both blood flow alteration and NO neurotoxicity may be features of the disease.
Collapse
Affiliation(s)
- C W Picanço-Diniz
- Universidade Federal do Pará, Centro de Ciências Biológicas, Departamento de Morfologia, Laboratório de Neuroanatomia Funcional, Belém, Brazil
| | | | | | | | | |
Collapse
|
33
|
Abstract
3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase inhibitors, or statins, reduce the incidence of strokes and reduce infarct volume after cerebral ischemia in mice. Excitoxicity caused by overstimulation of glutamate receptors is a major cause of neuronal death after an ischemic brain insult. Experiments presented here explored whether statins protect cultured neurons from excitotoxic death caused by the glutamate receptor agonist NMDA. Treatment with statins preserved NMDA receptor-expressing cortical neurons and potently and substantially reduced lactate dehydrogenase release caused by exposure of embryonic mouse neocortical cultures to NMDA. The rank order of neuroprotective potency was rosuvastatin = simvastatin > atorvastatin = mevastatin > pravastatin, which is similar to the known rank order of potency for inhibition of the HMG-CoA reductase enzyme. Resistance of cultures to NMDA excitotoxicity developed after several days of statin exposure. Neuroprotection by rosuvastatin was coincident with a decrease in cell sterols and occurred with a similar potency as inhibition of cholesterol biosynthesis. Neuroprotection was substantially attenuated by cotreatment with either mevalonate or cholesterol and was mimicked by acute treatment with the cholesterol-extracting agent beta-cyclodextrin, suggesting that neuroprotection was mediated by depletion of a cellular pool of cholesterol because of the inhibition of cholesterol biosynthesis. These results suggest the possibility that, in addition to effects on cerebrovascular function, statins have the potential to render cortical neurons more resistant to NMDA-induced excitotoxic death as a result of changes in cell cholesterol homeostasis.
Collapse
|
34
|
Morot-Gaudry-Talarmain Y, Rezaei H, Guermonprez L, Treguer E, Grosclaude J. Selective prion protein binding to synaptic components is modulated by oxidative and nitrosative changes induced by copper(II) and peroxynitrite in cholinergic synaptosomes, unveiling a role for calcineurin B and thioredoxin. J Neurochem 2003; 87:1456-70. [PMID: 14713301 DOI: 10.1046/j.1471-4159.2003.02111.x] [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: 11/20/2022]
Abstract
Choline acetyltransferase (ChAT) and choline transport are decreased after nitrosative stress. ChAT activity is altered in scrapie-infected neurons, where oxidative stress develops. Cellular prion protein (PrPc) may play a neuroprotective function in participating in the redox control of neuronal environment and regulation of copper metabolism, a role impaired when PrPc is transformed into PrPSc in prion pathologies. The complex cross-talk between PrPc and cholinergic neurons was analyzed in vitro using peroxynitrite and Cu2+ treatments on nerve endings isolated from Torpedo marmorata, a model of the motoneuron pre-synaptic element. Specific interactions between solubilized synaptic components and recombinant ovine prion protein (PrPrec) could be demonstrated by Biacore technology. Peroxynitrite abolished this interaction in a concentration-dependent way and induced significant alterations of neuronal targets. Interaction was restored by prior addition of peroxynitrite trapping agents. Cu2+ (in the form of CuSO4) treatment of synaptosomes triggered a milder oxidative effect leading to a bell-shaped increase of PrPrec binding to synaptosomal components, counteracted by the natural thiol agents, glutathione and thioredoxin. Copper(II)-induced modifications of thiols in several neuronal proteins. A positive correlation was observed between PrPrec binding and immunoreactive changes for calcineurin B and its partners, suggesting a synergy between calcineurin complex and PrP for copper regulation.
Collapse
|
35
|
Abstract
Transmissible spongiform encephalopathies (TSE) are fatal neurodegenerative disorders present in various mammals. TSEs have been studies intensively, even more so following the BSE crisis and the subsequent threat of a human nvCJD epidemic. In the 'protein-only' hypothesis, the infectious agent, called prion, is assumed to be a misfolded host protein. Transgenesis has mainly been applied to study the role of this protein, its structure-function relationship with respect to its pathogenic properties and to assess the genetic origin of the well-recognised species barrier effect. This approach has somewhat supplemented the lack of in vitro models. This review will try to summarise the impressive work that has been done in this field. Although many questions remain unanswered, transgenic experiments have and will still improve our knowledge on this disease and might help us to develop critically needed therapeutic approaches.
Collapse
Affiliation(s)
- Jean-Luc Vilotte
- Laboratoire de Génétique Biochimique et Cytogénétique, Institut National de la Recherche Agronomique, 78352, Jouy-en-Josas Cedex, France.
| | | |
Collapse
|
36
|
Weissmann C. Molecular genetics of transmissible spongiform encephalopathies: an introduction. J Toxicol Sci 2002; 27:69-77. [PMID: 12058449 DOI: 10.2131/jts.27.69] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Prnp knockout mice disrupted PrPC-related genes have played an essential role to elucidate the relationship between PrPC, a normal host gene product, and PrPSc, a protease-resistant, infectious PrP; Prnp knockout mice developed by Büeler et al. (1992) were completely protected against scrapie disease when challenged with mouse prions. Further, varying expression levels in PrPC were revisited along with a varying susceptibility of mouse prions, when mouse Prnp genes were introduced into Prnp% mice. How these murine models for human prion-related disease would contribute to the presently ongoing TSE research?
Collapse
Affiliation(s)
- Charles Weissmann
- MRC Prion Unit, Department of Neurodegenerative Disease, Institute of Neurology, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
37
|
Abstract
Recent data demonstrate that transition metal ions such as copper not only bind the prion protein with high affinities, but also modify its biochemical properties. This has important consequences for the potential function of the protein in metal-ion transport or as an anti-oxidant molecule. In addition, this relationship between the prion protein and metal ions is likely to play a critical role in the physiopathology of prion diseases.
Collapse
Affiliation(s)
- Sylvain Lehmann
- Institut de Génétique Humaine, CNRS U.P.R. 1142, 141, rue de la Cardonille, 34396 Cedex 5, Montpellier, France.
| |
Collapse
|
38
|
Mallucci G, Ratté S, Asante E, Linehan J, Gowland I, Jefferys J, Collinge J. Post-natal knockout of prion protein alters hippocampal CA1 properties, but does not result in neurodegeneration. EMBO J 2002; 21:202-10. [PMID: 11823413 PMCID: PMC125833 DOI: 10.1093/emboj/21.3.202] [Citation(s) in RCA: 274] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Prion protein (PrP) plays a crucial role in prion disease, but its physiological function remains unclear. Mice with gene deletions restricted to the coding region of PrP have only minor phenotypic deficits, but are resistant to prion disease. We generated double transgenic mice using the Cre-loxP system to examine the effects of PrP depletion on neuronal survival and function in adult brain. Cre-mediated ablation of PrP in neurons occurred after 9 weeks. We found that the mice remained healthy without evidence of neurodegeneration or other histopathological changes for up to 15 months post-knockout. However, on neurophysiological evaluation, they showed significant reduction of afterhyperpolarization potentials (AHPs) in hippocampal CA1 cells, suggesting a direct role for PrP in the modulation of neuronal excitability. These data provide new insights into PrP function. Furthermore, they show that acute depletion of PrP does not affect neuronal survival in this model, ruling out loss of PrP function as a pathogenic mechanism in prion disease and validating therapeutic approaches targeting PrP.
Collapse
Affiliation(s)
| | - S. Ratté
- MRC Prion Unit and Department of Neurodegenerative Disease, Institute of Neurology, Queen Square, London WC1N 3BG and
Department of Neurophysiology, Division of Neuroscience, The Medical School, University of Birmingham, Birmingham B15 2TT, UK Corresponding author e-mail:
| | | | | | | | - J.G.R. Jefferys
- MRC Prion Unit and Department of Neurodegenerative Disease, Institute of Neurology, Queen Square, London WC1N 3BG and
Department of Neurophysiology, Division of Neuroscience, The Medical School, University of Birmingham, Birmingham B15 2TT, UK Corresponding author e-mail:
| | - J. Collinge
- MRC Prion Unit and Department of Neurodegenerative Disease, Institute of Neurology, Queen Square, London WC1N 3BG and
Department of Neurophysiology, Division of Neuroscience, The Medical School, University of Birmingham, Birmingham B15 2TT, UK Corresponding author e-mail:
| |
Collapse
|
39
|
Brown DR, Nicholas RSJ, Canevari L. Lack of prion protein expression results in a neuronal phenotype sensitive to stress. J Neurosci Res 2002; 67:211-24. [PMID: 11782965 DOI: 10.1002/jnr.10118] [Citation(s) in RCA: 177] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The prion protein is a highly conserved glycoprotein expressed most highly in the synapse. Evidence has recently been put forward to suggest that the prion protein is an antioxidant. However, the functional importance of the prion protein has been disputed; it is claimed that mice genetically ablated to lack prion protein expression are normal and have no specific phenotype. We have reexamined the phenotype of prion protein knockout mice and found that there are multiple biochemical changes in the mice, including increased levels of nuclear factor NF-kappaB and Mn superoxide dismutase, COX-IV decreased levels of Cu/Zn superoxide dismutase activity, decreased p53, and altered melatonin levels. Additionally, cultured cells from these mice are more sensitive to a range of insults, all linked to increased neuronal sensitivity to oxidative stress. These results imply that prion protein knockout mice are more sensitive to oxidative stress and have an altered phenotype that must be taken into account when considering the additional effects of increased levels of proteins such as Doppel. The implication of these results is that the consequence of genetic ablation of genes must include biochemical analysis as well as analyses of possible developmental and behavioral changes.
Collapse
Affiliation(s)
- David R Brown
- Department of Biochemistry, Cambridge University, Cambridge, United Kingdom.
| | | | | |
Collapse
|
40
|
Tagliavini F, Forloni G, D'Ursi P, Bugiani O, Salmona M. Studies on peptide fragments of prion proteins. ADVANCES IN PROTEIN CHEMISTRY 2002; 57:171-201. [PMID: 11447690 DOI: 10.1016/s0065-3233(01)57022-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- F Tagliavini
- Istituto Nazionale Neurologico Carlo Besta, Milano, Italy
| | | | | | | | | |
Collapse
|
41
|
Ostlund P, Lindegren H, Pettersson C, Bedecs K. Altered insulin receptor processing and function in scrapie-infected neuroblastoma cell lines. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 97:161-70. [PMID: 11750072 DOI: 10.1016/s0169-328x(01)00316-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The underlying neurochemical changes contributing to prion-induced neurodegeneration remain largely unknown. This study shows that scrapie infection induced a 2-fold increase of insulin receptor (IR) protein and aberrantly processed IR beta-chain in scrapie-infected N2a neuroblastoma cells (ScN2a) as measured by Western blot of immunoprecipitated IR, in the absence of increased IR mRNA. Elevated IR protein level was further confirmed in an independently scrapie-infected neuroblastoma cell line N1E-115 (ScN1E-115). Proliferation studies showed that the increased IR level in ScN2a did not result in an increased insulin-mediated cell growth compared to normal N2a cells. Binding studies indicated that this apparent paradox was due to a 65% decrease in specific [(125)I]insulin binding sites in ScN2a when compared to the amount of immunoreactive IR, although the IR binding affinity was unchanged. Analysis of insulin stimulated IR tyrosine phosphorylation showed a slight but not significant reduction in ScN2a, when related to the increased level of immunoreactive IR. However, comparing the IR tyrosine phosphorylation to the loss of binding sites in ScN2a, we demonstrated an increased IR tyrosine phosphorylation of the remaining functional IR. In addition to these differences in IR properties, the basal extracellular signal regulated kinase-2 (ERK2) phosphorylation detected by Western blot, was significantly elevated and the insulin stimulated ERK2 phosphorylation was subsequently decreased in ScN2a. Together, these data show that scrapie infection affects the level and processing of the IR and signal transduction mediated by the IR in neuroblastoma cells, as well as induces an elevated basal ERK2 phosphorylation. Aberrant regulation of neuroprotective receptors may contribute to neurodegeneration in prion diseases.
Collapse
Affiliation(s)
- P Ostlund
- Department of Neurochemistry and Neurotoxicology, University of Stockholm, Svante Arrhenius v. 21A, S-10691 Stockholm, Sweden
| | | | | | | |
Collapse
|
42
|
Ostlund P, Lindegren H, Pettersson C, Bedecs K. Up-regulation of functionally impaired insulin-like growth factor-1 receptor in scrapie-infected neuroblastoma cells. J Biol Chem 2001; 276:36110-5. [PMID: 11461928 DOI: 10.1074/jbc.m105710200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A growing body of evidence suggests that an altered level or function of the neurotrophic insulin-like growth factor-1 receptor (IGF-1R), which supports neuronal survival, may underlie neurodegeneration. This study has focused on the expression and function of the IGF-1R in scrapie-infected neuroblastoma cell lines. Our results show that scrapie infection induces a 4-fold increase in the level of IGF-1R in two independently scrapie-infected neuroblastomas, ScN2a and ScN1E-115 cells, and that the increased IGF-1R level was accompanied by increased IGF-1R mRNA levels. In contrast to the elevated IGF-1R expression in ScN2a, receptor binding studies revealed an 80% decrease in specific (125)I-IGF-1-binding sites compared with N2a cells. This decrease in IGF-1R-binding sites was shown to be caused by a 7-fold decrease in IGF-1R affinity. Furthermore, ScN2a showed no significant difference in IGF-1 induced proliferative response, despite the noticeable elevated IGF-1R expression, putatively explained by the reduced IGF-1R binding affinity. Additionally, IGF-1 stimulated IGF-1Rbeta tyrosine phosphorylation showed no major change in the dose-response between the cell types, possibly due to altered tyrosine kinase signaling in scrapie-infected neuroblastoma cells. Altogether these data indicate that scrapie infection affects the expression, binding affinity, and signal transduction mediated by the IGF-1R in neuroblastoma cells. Altered IGF-1R expression and function may weaken the trophic support in scrapie-infected neurons and thereby contribute to neurodegeneration in prion diseases.
Collapse
Affiliation(s)
- P Ostlund
- Department of Neurochemistry and Neurotoxicology, University of Stockholm, Svante Arrhenius v. 21A, S-10691 Stockholm, Sweden
| | | | | | | |
Collapse
|
43
|
Fabrizi C, Silei V, Menegazzi M, Salmona M, Bugiani O, Tagliavini F, Suzuki H, Lauro GM. The stimulation of inducible nitric-oxide synthase by the prion protein fragment 106--126 in human microglia is tumor necrosis factor-alpha-dependent and involves p38 mitogen-activated protein kinase. J Biol Chem 2001; 276:25692-6. [PMID: 11316802 DOI: 10.1074/jbc.m100133200] [Citation(s) in RCA: 56] [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
A synthetic peptide consisting of amino acid residues 106-126 of the human prion protein (PrP-(106--126)) has been previously demonstrated to be neurotoxic and to induce microglial activation. The present study investigated the expression of the inducible form of the nitric-oxide synthase (NOS-II) in human microglial cells treated with PrP-(106--126). Using reverse transcriptase-polymerase chain reaction, we found that PrP-(106--126) induces NOS-II gene expression after 24 h of treatment and that this effect is accompanied by a peak of nuclear factor kappa B (NF-kappa B) binding at 30 min as evaluated by electrophoretic mobility shift assay. Since our previous data demonstrated tumor necrosis factor-alpha (TNF-alpha) to be a potent inducer of NOS-II in these cells, we analyzed the expression of this cytokine in PrP-(106--126)-treated microglia. PrP-(106--126) caused the release of TNF-alpha as detected by enzyme-linked immunosorbent assay, and a blocking antibody, anti-TNF-alpha, abolished NOS-II induction elicited by this peptide. Moreover, PrP-(106-126) activates p38 mitogen-activated protein kinase, and the inhibition of this pathway determines the ablation of NF-kappa B binding induced by this fragment peptide.
Collapse
Affiliation(s)
- C Fabrizi
- Dipartimento di Biologia, Università di Roma Tre, Viale Marconi 446, 00146, Roma, Italy
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Cereghetti GM, Schweiger A, Glockshuber R, Van Doorslaer S. Electron paramagnetic resonance evidence for binding of Cu(2+) to the C-terminal domain of the murine prion protein. Biophys J 2001; 81:516-25. [PMID: 11423433 PMCID: PMC1301530 DOI: 10.1016/s0006-3495(01)75718-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Transmissible spongiform encephalopathies in mammals are believed to be caused by scrapie form of prion protein (PrP(Sc)), an abnormal, oligomeric isoform of the monomeric cellular prion protein (PrP(C)). One of the proposed functions of PrP(C) in vivo is a Cu(II) binding activity. Previous studies revealed that Cu(2+) binds to the unstructured N-terminal PrP(C) segment (residues 23-120) through conserved histidine residues. Here we analyzed the Cu(II) binding properties of full-length murine PrP(C) (mPrP), of its isolated C-terminal domain mPrP(121-231) and of the N-terminal fragment mPrP(58-91) in the range of pH 3-8 with electron paramagnetic resonance spectroscopy. We find that the C-terminal domain, both in its isolated form and in the context of the full-length protein, is capable of interacting with Cu(2+). Three Cu(II) coordination types are observed for the C-terminal domain. The N-terminal segment mPrP(58-91) binds Cu(2+) only at pH values above 5.0, whereas both mPrP(121-231) and mPrP(23-231) already show identical Cu(II) coordination in the pH range 3-5. As the Cu(2+)-binding N-terminal segment 58-91 is not required for prion propagation, our results open the possibility that Cu(2+) ions bound to the C-terminal domain are involved in the replication of prions, and provide the basis for further analytical studies on the specificity of Cu(II) binding by PrP.
Collapse
Affiliation(s)
- G M Cereghetti
- Institute of Molecular Biology and Biophysics, Swiss Federal Institute of Technology, Hönggerberg, CH-8093 Zurich, Switzerland
| | | | | | | |
Collapse
|
45
|
|
46
|
Wong BS, Liu T, Paisley D, Li R, Pan T, Chen SG, Perry G, Petersen RB, Smith MA, Melton DW, Gambetti P, Brown DR, Sy MS. Induction of HO-1 and NOS in doppel-expressing mice devoid of PrP: implications for doppel function. Mol Cell Neurosci 2001; 17:768-75. [PMID: 11312611 DOI: 10.1006/mcne.2001.0963] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ectopic expression of the doppel (Dpl) protein, a homologue of the prion protein (PrP), was recently associated with cerebellar Purkinje cell degeneration observed in two aging prion protein knock-out (Prnp(0/0)) mouse lines. We investigated the possible role of Dpl in oxidative metabolism. Two Prnp(0/0) mouse lines of similar genetic background were studied. One line expresses Dpl in the brain and displays Dpl-associated cerebellar abnormalities. The other has no elevated expression of Dpl and no cerebellar abnormalities. We observed a correlation between Dpl expression and the induction of both heme oxygenase 1 (HO-1) and nitric oxide synthase systems (nNOS and iNOS). These responses are suggestive of increased oxidative stress in the brains of the Dpl-expressing Prnp(0/0) mice. No induction was observed with Hsp-60, indicating a specific response by the HO/NOS system. We proposed that Dpl expression exacerbates oxidative damage that is antagonistic to the protective function of wild-type PrP.
Collapse
Affiliation(s)
- B S Wong
- Institute of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Van Doorslaer S, Cereghetti GM, Glockshuber R, Schweiger A. Unraveling the Cu2+ Binding Sites in the C-Terminal Domain of the Murine Prion Protein: A Pulse EPR and ENDOR Study. J Phys Chem B 2001. [DOI: 10.1021/jp003115y] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Sabine Van Doorslaer
- Laboratory of Physical Chemistry, ETH Zentrum, CH-8092 Zurich, Switzerland, and Institute of Molecular Biology and Biophysics, ETH Zurich Hönggerberg, CH-8093 Zurich, Switzerland
| | - Grazia M. Cereghetti
- Laboratory of Physical Chemistry, ETH Zentrum, CH-8092 Zurich, Switzerland, and Institute of Molecular Biology and Biophysics, ETH Zurich Hönggerberg, CH-8093 Zurich, Switzerland
| | - Rudi Glockshuber
- Laboratory of Physical Chemistry, ETH Zentrum, CH-8092 Zurich, Switzerland, and Institute of Molecular Biology and Biophysics, ETH Zurich Hönggerberg, CH-8093 Zurich, Switzerland
| | - Arthur Schweiger
- Laboratory of Physical Chemistry, ETH Zentrum, CH-8092 Zurich, Switzerland, and Institute of Molecular Biology and Biophysics, ETH Zurich Hönggerberg, CH-8093 Zurich, Switzerland
| |
Collapse
|
48
|
Massmann GA, Zhang J, Sallah J, Figueroa JP. Developmental and regional expression patterns of Type I Nitric Oxide Synthase mRNA and protein in fetal sheep brain during the last third of gestation. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2000; 124:141-52. [PMID: 11113524 DOI: 10.1016/s0165-3806(00)00095-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Type I NOS (nNOS) catalytic activity represents the activity of full-size protein and truncated protein variants originated from many different spliced mRNA variants. Splice mRNA variants are thought to be important in determining the differential organ and subcellular expression of Type I NOS. The present study was directed to increase our understanding of the developmental regulation of Type I NOS in fetal brain. In four discrete areas of the fetal brain, we measured steady-state mRNA levels and catalytic activity and protein mass in the soluble and particulate fractions. Under general anesthesia, we collected sensory-motor cortex, striatum, hippocampus and cerebellum from sheep fetuses at 105, 115, 125 and 135 days gestation (32 fetuses). NOS protein in the soluble and particulate fractions was characterized using Western blot (molecular weight) and arginine to citrulline conversion (enzymatic activity). At the mRNA level, steady state levels were determined using probes directed against exon 2 and exon 21/22 by ribonuclease protection assay (RPA). Our data show that NOS catalytic activity is regulated in a region, subcellular and temporal manner. NOS activity was higher in the soluble fraction in all brain regions and significantly higher levels were found in the soluble fraction of striatum and particulate fraction of hippocampus (P<0.05 by ANOVA). Western blot analysis revealed three distinct molecular weight bands for Type I NOS (155, 144 and 136 kDa). The bands were present in all brain regions and in both cellular compartments with the 155 kDa band being the most abundant molecular form. Truncated protein variants accounted for 25% and 15% of total Type I NOS protein in the soluble fraction and particulate fraction respectively. RPA analysis showed a differential regulation of mRNA variants with exon 2 frame deletions in striatum and hippocampus. A coordinated increase with advancing gestational age of catalytic activity, the full-length protein, the protein variants and steady state mRNA levels was observed in cortex and striatum as demonstrated by higher levels at 125 and 135 days gestation (P<0.05 by ANOVA). NOS enzymatic activity was Ca(2+) and calmodulin dependent. However, in the particulate fraction 20% of the NOS activity was resistant to calmodulin inhibition. In summary, fetal brain Type I NOS mRNA variants are differentially regulated according to brain regions. Our data suggests that exon 2 deleted mRNA variants have low translation efficiency as indicated by the lack of parallel expression of truncated Type I NOS protein variants.
Collapse
Affiliation(s)
- G A Massmann
- Perinatal Research Laboratory, Department of Obstetrics and Gynecology and Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | | | |
Collapse
|
49
|
Keshet GI, Bar-Peled O, Yaffe D, Nudel U, Gabizon R. The cellular prion protein colocalizes with the dystroglycan complex in the brain. J Neurochem 2000; 75:1889-97. [PMID: 11032878 DOI: 10.1046/j.1471-4159.2000.0751889.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The function of PrP(C), the cellular prion protein (PrP), is still unknown. Like other glycophosphatidylinositol-anchored proteins, PrP resides on Triton-insoluble, cholesterol-rich membranous microdomains, termed rafts. We have recently shown that the activity and subcellular localization of the neuronal isoform of nitric oxide synthase (nNOS) are impaired in adult PrP(0/0) mice as well as in scrapie-infected mice. In this study, we sought to determine whether PrP and nNOS are part of the same functional complex and, if so, to identify additional components of such a complex. To this aim, we looked for proteins that coimmunoprecipitated with PrP in the presence of detergents either that completely dissociate rafts, to identify stronger interactions, or that preserve the raft structure, to identify weaker interactions. Using this detergent-dependent immunoprecipitation protocol we found that PrP interacts strongly with dystroglycan, a transmembrane protein that is the core of the dystrophin-glycoprotein complex (DGC). Additional results suggest that PrP also interacts with additional members of the DGC, including nNOS. PrP coprecipitated only with established presynaptic proteins, consistent with recent findings suggesting that PrP is a presynaptic protein.
Collapse
Affiliation(s)
- G I Keshet
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
50
|
El-Husseini AE, Topinka JR, Lehrer-Graiwer JE, Firestein BL, Craven SE, Aoki C, Bredt DS. Ion channel clustering by membrane-associated guanylate kinases. Differential regulation by N-terminal lipid and metal binding motifs. J Biol Chem 2000; 275:23904-10. [PMID: 10779526 DOI: 10.1074/jbc.m909919199] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The postsynaptic density protein PSD-95 and related membrane-associated guanylate kinase (MAGUK) proteins assemble signal transduction complexes at sites of cell-cell contact including synapses. Whereas PSD-95 and PSD-93 occur only at postsynaptic sites in hippocampal neurons, SAP-102 also occurs in axons. In heterologous cells, PSD-95 and PSD-93 mediate cell surface ion channel clustering, but SAP-102 and SAP-97 do not. This selective ion channel clustering activity by MAGUKs is explained by differential palmitoylation, as PSD-93 and PSD-95 are palmitoylated though SAP-97, and SAP-102 are not. Rather than being palmitoylated, we find that N-terminal cysteines from SAP-102 tightly bind to zinc. And, appending the N terminus of SAP-102 to PSD-95 results in localization of the chimera to both axons and dendrites. These data suggest that lipid modifications and heavy metal associations with the N termini of MAGUKs mediate differential functions and subcellular localizations of these synaptic scaffolds.
Collapse
Affiliation(s)
- A E El-Husseini
- Departments of Physiology and University of California, San Francisco, California 94143, USA
| | | | | | | | | | | | | |
Collapse
|