首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
《朊病毒》2013,7(6):420-428
ABSTRACT

Converging observations from disparate lines of inquiry are beginning to clarify the cause of brain iron dyshomeostasis in sporadic Creutzfeldt-Jakob disease (sCJD), a neurodegenerative condition associated with the conversion of prion protein (PrPC), a plasma membrane glycoprotein, from α-helical to a β-sheet rich PrP-scrapie (PrPSc) isoform. Biochemical evidence indicates that PrPC facilitates cellular iron uptake by functioning as a membrane-bound ferrireductase (FR), an activity necessary for the transport of iron across biological membranes through metal transporters. An entirely different experimental approach reveals an evolutionary link between PrPC and the Zrt, Irt-like protein (ZIP) family, a group of proteins involved in the transport of zinc, iron, and manganese across the plasma membrane. Close physical proximity of PrPC with certain members of the ZIP family on the plasma membrane and increased uptake of extracellular iron by cells that co-express PrPC and ZIP14 suggest that PrPC functions as a FR partner for certain members of this family. The connection between PrPC and ZIP proteins therefore extends beyond common ancestry to that of functional cooperation. Here, we summarize evidence supporting the facilitative role of PrPC in cellular iron uptake, and implications of this activity on iron metabolism in sCJD brains.  相似文献   

2.
Dysregulated body copper homeostasis can negatively impact neuronal functions, but full knowledge of the mechanisms underlying the cell metal distribution has not been achieved yet. The high-affinity copper transporter 1 (Ctr1) is considered the main route for cell copper entry, while the cellular prion protein (PrPC) is presumed to be involved in the same process. Anchored to the outer side of the plasma membrane, this protein has the ability to bind copper ions and undergo internalization. To provide indications about the contribution of Ctr1 and PrPC proteins in cell copper transport, we used a fluorimetric method to characterize the kinetic properties of ion internalization in a neuroblastoma cell model, overexpressing prion protein (B104). Biochemical characteristics of intake delineated in the presence of other metal ions and an excess of extracellular potassium were compatible with PrPC-mediated endocytotic transport. Accordingly, inhibition of clathrin-dependent endocytosis by hypertonic shock and enzymatic removal of surface prion protein reduced copper influx by the same extent. On the whole, experimental evidence collected in a neuron-like cell model sustains a role for PrPC in mediating copper uptake by clathrin-dependent endocytosis.  相似文献   

3.
PrP overdrive     
Knockout of the cellular prion protein (PrPC) in mice is tolerated, as is complete elimination of the protein’s N-terminal domain. However, deletion of select short segments between the N- and C-terminal domains is lethal. How can one reconcile this apparent paradox? Research over the last few years demonstrates that PrPC undergoes α-cleavage in the vicinity of residue 109 (mouse sequence) to release the bioactive N1 and C1 fragments. In biophysical studies, we recently characterized the action of relevant members of the ADAM (A Disintegrin And Metalloproteinase) enzyme family (ADAM8, 10, and 17) and found that they all produce α-cleavage, but at 3 distinct cleavage sites, with proteolytic efficiency modulated by the physiologic metals copper and zinc. Remarkably, the shortest lethal deletion segment in PrPC fully encompasses the three α-cleavage sites. Analysis of all reported PrPC deletion mutants suggests that elimination of α-cleavage, coupled with retention of the protein’s N-terminal residues, segments 23–31 and longer, confers the lethal phenotype. Interestingly, these N-terminal residues are implicated in the activation of several membrane proteins, including synaptic glutamate receptors. We propose that α-cleavage is a general mechanism essential for downregulating PrPC’s intrinsic activity, and that blockage of proteolysis leads to constitutively active PrPC and consequent dyshomeostasis.  相似文献   

4.
AMPA receptors are tetrameric glutamate-gated ion channels that mediate fast synaptic neurotransmission in mammalian brain. Their subunits contain a two-lobed N-terminal domain (NTD) that comprises over 40% of the mature polypeptide. The NTD is not obligatory for the assembly of tetrameric receptors, and its functional role is still unclear. By analyzing full-length and NTD-deleted GluA1–4 AMPA receptors expressed in HEK 293 cells, we found that the removal of the NTD leads to a significant reduction in receptor transport to the plasma membrane, a higher steady state-to-peak current ratio of glutamate responses, and strongly increased sensitivity to glutamate toxicity in cell culture. Further analyses showed that NTD-deleted receptors display both a slower onset of desensitization and a faster recovery from desensitization of agonist responses. Our results indicate that the NTD promotes the biosynthetic maturation of AMPA receptors and, for membrane-expressed channels, enhances the stability of the desensitized state. Moreover, these findings suggest that interactions of the NTD with extracellular/synaptic ligands may be able to fine-tune AMPA receptor-mediated responses, in analogy with the allosteric regulatory role demonstrated for the NTD of NMDA receptors.  相似文献   

5.
Increasing evidence suggests that proteins exhibiting “prion-like” behavior cause distinct neurodegenerative diseases, including inherited, sporadic and acquired types. The conversion of cellular prion protein (PrPC) to its infectious protease resistant counterpart (PrPRes) is the essential feature of prion diseases. However, PrPC also performs important functions in transmembrane signaling, especially in neurodegenerative processes. Beta-amyloid (Aβ) synaptotoxicity and cognitive dysfunction in mouse models of Alzheimer disease are mediated by a PrPC-dependent pathway. Here we review how this pathway converges with proinflammatory cytokine signaling to activate membrane NADPH oxidase (NOX) and generate reactive oxygen species (ROS) leading to dynamic remodeling of the actin cytoskeleton. The NOX signaling pathway may also be integrated with those of other transmembrane receptors clustered in PrPC-enriched membrane domains. Such a signal convergence along the PrPC-NOX axis could explain the relevance of PrPC in a broad spectrum of neurodegenerative disorders, including neuroinflammatory-mediated alterations in synaptic function following traumatic brain injury. PrPC overexpression alone activates NOX and generates a local increase in ROS that initiates cofilin activation and formation of cofilin-saturated actin bundles (rods). Rods sequester cofilin from synaptic regions where it is required for plasticity associated with learning and memory. Rods can also interrupt vesicular transport by occluding the neurite within which they form. Through either or both mechanisms, rods may directly mediate the synaptic dysfunction that accompanies various neurodegenerative disorders.  相似文献   

6.
Cellular prion protein (PrPC) appears to be involved in numerous physiological processes. We have recently shown a novel modulation of NMDA receptors by PrPC that results in neuroprotection via silencing of NMDA receptors containing NR2D subunits, whereas no effects on AMPA receptor function could be observed (Khosravani, et al. J Cell Biol 2008; 181:551). Here we show that PrP-null mice show a normal response to long-term depression stimuli requiring AMPA receptor activity, thus further supporting our previous findings of a selective action on NMDA receptors among ionotropic glutamate receptors.Key words: AMPA receptor, NMDA receptor, PrP, long term depression, LTDThe role of prion proteins in the pathophysiology of transmissible spongiform encephalopathies is well documented.1 Although there is a growing body of literature associating normal cellular prion protein (PrPC) with functions such as regulation of cell proliferation and survival, cell signalling and immune function,1 the spectrum of physiological roles attributable to PrPC remains to be determined. This may in part be due to the fact that mice lacking PrPC display a relatively mild phenotype, unless subjected to insults such as ischemia or seizures, where increased mortality of the PrP-null mice has been reported.25 Interestingly, the increased neuronal damage in PrP-null mice following excitotoxicity is alleviated upon treatment with the N-Methyl-D-Aspartate (NMDA) receptor (NMDAR) inhibitor MK-801,6 suggesting a neuroprotective role of PrPC via an action on NMDARs, but the mechanism was unclear.We recently described a novel action of PrPC on NMDAR function.7 By examining the neurophysiological properties of hippocampal neurons isolated from PrP-null mice, we were able to show that PrP-null mouse neurons exhibit enhanced and drastically prolonged NMDA evoked currents due to a functional upregulation of NMDARs containing NR2D subunits. Biochemical analyses suggested that NR2D subunits, but not NR2B subunits, co-immunoprecipitated with PrPC, indicating that PrPC and NMDARs form physical signaling complexes in neurons. The increased NMDAR function could be phenocopied by RNA interference and were rescued upon overexpression of exogenous PrPC. The enhanced NMDAR activity resulted in increased neuronal excitability, as well as enhanced glutamatergic-based excitotoxicity in both in vitro and in vivo experiments were neurons were transiently exposed to the selective agonist NMDA. Hence, native PrPC appears to mediate an important neuroprotective role by virtue of its ability to silence NR2D containing NMDARs. In contrast, minor effects on amplitude and rise and decay-time kinetics were observed for both α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and GABAA (miniature and evoked) currents in synaptically mature hippocampal cultures.AMPA and NMDA receptors have been linked to synaptic plasticity, in particular long term potentiation (LTP) and long term depression (LTD). LTP is believed to mostly reflect a strengthening of the postsynaptic response, caused by a brief period of hyperexcitability that releases significant amounts of glutamate such as during a brief tetanic stimulation. This is thought to result in the opening of AMPA receptors, which depolarize the postsynaptic membrane. This in turn increases the activity of postsynaptic NMDARs, because magnesium ions that normally inhibit NMDAR activity, are dislodged by the postsynaptic depolarization, thus allowing NMDARs to become active. This functional activation of NMDARs results in the influx of calcium ions, which in turn initiate a signaling cascade that results in the membrane insertion of additional AMPA receptors, thus strengthening the synapse. This process is thought to involve NMDA receptor isoforms that predominantly contain the NR2A subunits.8A synaptic depotentiation process also can take place that results in the opposite effects of LTP; this process is known as long-term depression or LTD, which also has an NMDAR-dependent component. In contrast to brief tetanic stimulation, as is used in the induction of LTP, establishing LTD requires low frequency stimulation (e.g., 1 Hz for 15 min). Successful and repeatable induction of LTD depends on the parameters used for the conditioning stimulus and more importantly on the age of the animal. In juvenile animals (P12–P21) a low frequency protocol is effective and the mechanism of LTD is believed to depend on the activity of NMDA receptors containing NR2B subunits.8 Although a clear distinction of the roles between NR2A and NR2B containing NMDARs, in LTP and LTD respectively, has remained controversial,9 it is clear that both NR2A and NR2B are key mediators of alterations in synaptic plasticity. In older animals, the conditioning protocol is reported to require modification to include paired-pulses. This is thought to be due to the involvement of predominantly AMPA (and perhaps kainate) receptors in addition to mGluRs responsible for the synaptic depotentiation (reviewed in ref. 10).As mentioned earlier, our data obtained from hippocampal cultures indicated only a minor effect of PrPC knockout on AMPA receptor function. Hence, we hypothesized that AMPA receptor-mediated LTD should be similar in both wild type and PrP-null mice. We therefore examined the effect of PrP on LTD in hippocampal slices obtained from P30–P45 wild type mice and Zurich 1 PrPC knockout mice. Extracellular potentials were recorded using a patch pipette filled with 150 mM NaCl. First, 10 minutes of baseline evoked (every 30 sec) potentials were recorded to ensure stability of the preparation. LTD was then evoked by application of conditioning paired pulses (Δt = 60 ms) delivered at 1 Hz for 15 min. Thereafter, the field response was sampled every 30 sec for 40 min. As shown in Figure 1, this protocol evoked reliable LTD in wild type mice that partially recovered over the time course of about 20 minutes (Fig. 1). In the PrPC-null slices, LTD was indistinguishable from that observed in the wild type slices (Fig. 1).Open in a separate windowFigure 1LTD in the CA1 region of hippocampal slices from adult (P30–P45) WT and PrP-null mice. The conditioning pulse (arrow head) was delivered as paired-pulses (Δt = 60 ms) at 1 Hz for 15 min at the Schaffer collaterals. Analysis of field excitatory postsynaptic potential (fEPSP) slope revealed no statistically significant differences (Student''s t-test, p > 0.05) in the extent of induced LTD or the time course of its recovery to baseline. Numbers in parentheses indicate number of slices.The age of the animals, combined with the paired pulse protocol used in our experiments was designed to isolate AMPA receptor mediated LTD.10 The notion that LTD was unaltered in PrP-null mice is consistent with the observation that AMPA currents were not altered in these mice, and that AMPA receptor-mediated spontaneous synaptic events showed only minute changes compared with wild type animals. These data are also consistent with the notion that PrP-null mice show only mild phenotypes in spatial learning, with no apparent overall short-term memory deficits. Collectively, these data further support a selective action of PrP on NMDA receptors, rather than overall glutamatergic synaptic transmission.  相似文献   

7.
The cellular prion protein (PrPC) is a zinc-binding protein that contributes to the regulation of Zn2+ and other divalent species of the central nervous system. Zn2+ coordinates to the flexible, N-terminal repeat region of PrPC and drives a tertiary contact between this repeat region and a well-defined cleft of the C-terminal domain. The tertiary structure promoted by Zn2+ is thought to regulate inherent PrPC toxicity. Despite the emerging consensus regarding the interaction between Zn2+ and PrPC, there is little direct spectroscopic confirmation of the metal ion’s coordination details. Here, we address this conceptual gap by using Cd2+ as a surrogate for Zn2+. NMR finds that Cd2+ binds exclusively to the His imidazole side chains of the repeat segment, with a dissociation constant of ~1.2 mM, and promotes an N-terminal-C-terminal cis interaction very similar to that observed with Zn2+. Analysis of 113Cd NMR spectra of PrPC, along with relevant control proteins and peptides, suggests that coordination of Cd2+ in the full-length protein is consistent with a three- or four-His geometry. Examination of the mutation E199K in mouse PrPC (E200K in humans), responsible for inherited Creutzfeldt-Jakob disease, finds that the mutation lowers metal ion affinity and weakens the cis interaction. These findings not only provide deeper insight into PrPC metal ion coordination but they also suggest new perspectives on the role of familial mutations in prion disease.  相似文献   

8.
《朊病毒》2013,7(3):245-251
Knowledge of the natural roles of cellular prion protein (PrPC) is essential to an understanding of the molecular basis of prion pathologies. This GPI-anchored protein has been described in synaptic contacts, and loss of its synaptic function in complex systems may contribute to the synaptic loss and neuronal degeneration observed in prionopathy. In addition, Prnp knockout mice show enhanced susceptibility to several excitotoxic insults, GABAA receptor-mediated fast inhibition was weakened, LTP was modified and cellular stress increased. Although little is known about how PrPC exerts its function at the synapse or the downstream events leading to PrPC-mediated neuroprotection against excitotoxic insults, PrPC has recently been reported to interact with two glutamate receptor subunits (NR2D and GluR6/7). In both cases the presence of PrPC blocks the neurotoxicity induced by NMDA and Kainate respectively. Furthermore, signals for seizure and neuronal cell death in response to Kainate in Prnp knockout mouse are associated with JNK3 activity, through enhancing the interaction of GluR6 with PSD-95. In combination with previous data, these results shed light on the molecular mechanisms behind the role of PrPC in excitotoxicity. Future experimental approaches are suggested and discussed.  相似文献   

9.
Knowledge of the natural roles of cellular prion protein (PrPC) is essential to an understanding of the molecular basis of prion pathologies. This GPI-anchored protein has been described in synaptic contacts, and loss of its synaptic function in complex systems may contribute to the synaptic loss and neuronal degeneration observed in prionopathy. In addition, Prnp knockout mice show enhanced susceptibility to several excitotoxic insults, GABAA receptor-mediated fast inhibition was weakened, LTP was modified and cellular stress increased. Although little is known about how PrPC exerts its function at the synapse or the downstream events leading to PrPC-mediated neuroprotection against excitotoxic insults, PrPC has recently been reported to interact with two glutamate receptor subunits (NR2D and GluR6/7). In both cases the presence of PrPC blocks the neurotoxicity induced by NMDA and Kainate respectively. Furthermore, signals for seizure and neuronal cell death in response to Kainate in Prnp knockout mouse are associated with JNK3 activity, through enhancing the interaction of GluR6 with PSD-95. In combination with previous data, these results shed light on the molecular mechanisms behind the role of PrPC in excitotoxicity. Future experimental approaches are suggested and discussed.  相似文献   

10.
11.
Most excitatory transmission in the brain is mediated by alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors (AMPA receptors). Therefore, the presence of these receptors at synapses has to be carefully regulated in order to ensure correct neuronal communication. Interestingly, AMPA receptors are not static components of synapses. On the contrary, they are continuously being delivered and removed in and out of synapses in response to neuronal activity. This dynamic behavior of AMPA receptors is an important mechanism to modify synaptic strength during brain development and also during experience-dependent plasticity. AMPA receptor trafficking involves an intricate network of protein-protein interactions that start with the biosynthesis of the receptors, continues with their transport along dendrites, and ends with their local insertion and removal from synapses. The molecular and cellular mechanisms that regulate each of these processes, and their importance for synaptic plasticity, are now starting to be unraveled.  相似文献   

12.
The cellular prion protein (PrPC) is a metal-binding biomolecule that can interact with different protein partners involved in pivotal physiological processes, such as neurogenesis and neuronal plasticity. Recent studies profile copper and PrPC as important players in the pathological mechanisms of Alzheimer's disease and cancer. Although the copper-PrPC interaction has been characterized extensively, the role of the metal ion in the physiological and pathological roles of PrPC has been barely explored. In this article, we discuss how copper binding and proteolytic processing may impact the ability of PrPC to recruit protein partners for its functional roles. The importance to dissect the role of copper-PrPC interactions in health and disease is also underscored.  相似文献   

13.
In prion diseases, the cellular form of the prion protein, PrPC, undergoes a conformational conversion to the infectious isoform, PrPSc. PrPC associates with lipid rafts through its glycosyl-phosphatidylinositol (GPI) anchor and a region in its N-terminal domain which also binds to heparan sulfate proteoglycans (HSPGs). We show that heparin displaces PrPC from rafts and promotes its endocytosis, suggesting that heparin competes with an endogenous raft-resident HSPG for binding to PrPC. We then utilised a transmembrane-anchored form of PrP (PrP-TM), which is targeted to rafts solely by its N-terminal domain, to show that both heparin and phosphatidylinositol-specific phospholipase C can inhibit its association with detergent-resistant rafts, implying that a GPI-anchored HSPG targets PrPC to rafts. Depletion of the major neuronal GPI-anchored HSPG, glypican-1, significantly reduced the raft association of PrP-TM and displaced PrPC from rafts, promoting its endocytosis. Glypican-1 and PrPC colocalised on the cell surface and both PrPC and PrPSc co-immunoprecipitated with glypican-1. Critically, treatment of scrapie-infected N2a cells with glypican-1 siRNA significantly reduced PrPSc formation. In contrast, depletion of glypican-1 did not alter the inhibitory effect of PrPC on the β-secretase cleavage of the Alzheimer''s amyloid precursor protein. These data indicate that glypican-1 is a novel cellular cofactor for prion conversion and we propose that it acts as a scaffold facilitating the interaction of PrPC and PrPSc in lipid rafts.  相似文献   

14.
Prion protein (PrPC) is a cell surface glycoprotein that is abundantly expressed in nervous system. The elucidation of the PrPC interactome network and its significance on neural physiology is crucial to understanding neurodegenerative events associated with prion and Alzheimer's diseases. PrPC co‐opts stress inducible protein 1/alpha7 nicotinic acetylcholine receptor (STI1/α7nAChR) or laminin/Type I metabotropic glutamate receptors (mGluR1/5) to modulate hippocampal neuronal survival and differentiation. However, potential cross‐talk between these protein complexes and their role in peripheral neurons has never been addressed. To explore this issue, we investigated PrPC‐mediated axonogenesis in peripheral neurons in response to STI1 and laminin‐γ1 chain‐derived peptide (Ln‐γ1). STI1 and Ln‐γ1 promoted robust axonogenesis in wild‐type neurons, whereas no effect was observed in neurons from PrPC‐null mice. PrPC binding to Ln‐γ1 or STI1 led to an increase in intracellular Ca2+ levels via distinct mechanisms: STI1 promoted extracellular Ca2+ influx, and Ln‐γ1 released calcium from intracellular stores. Both effects depend on phospholipase C activation, which is modulated by mGluR1/5 for Ln‐γ1, but depends on, C‐type transient receptor potential (TRPC) channels rather than α7nAChR for STI1. Treatment of neurons with suboptimal concentrations of both ligands led to synergistic actions on PrPC‐mediated calcium response and axonogenesis. This effect was likely mediated by simultaneous binding of the two ligands to PrPC. These results suggest a role for PrPC as an organizer of diverse multiprotein complexes, triggering specific signaling pathways and promoting axonogenesis in the peripheral nervous system.  相似文献   

15.
Ionotropic glutamate receptor (iGluR) subunits contain a approximately 400-residue extracellular N-terminal domain ("X domain"), which is sequence-related to bacterial amino acid-binding proteins and to class C G-protein-coupled receptors. The X domain has been implicated in the assembly, transport to the cell surface, allosteric ligand binding, and desensitization in various members of the iGluR family, but its actual role in these events is poorly characterized. We have studied the properties of homomeric alpha-amino-3-hydroxy-5-methylisoxazolepropionate (AMPA)-selective GluR-D glutamate receptors carrying N-terminal deletions. Our analysis indicates that, surprisingly, transport to the cell surface, ligand binding properties, agonist-triggered channel activation, rapid desensitization, and allosteric potentiation by cyclothiazide can occur normally in the complete absence of the X domain (residues 22-402). The relatively intact ligand-gated channel function of a homomeric AMPA receptor in the absence of the X domain indirectly suggests more subtle roles for this domain in AMPA receptors, e.g. in the assembly of heteromeric receptors and in synaptic protein interactions.  相似文献   

16.
Prion diseases are characterized biochemically by protein aggregation of infectious prion isoforms (PrPSc), which result from the conformational conversion of physiological prion proteins (PrPC). PrPC are variable post-translationally modified glycoproteins, which exist as full length and as aminoterminally truncated glycosylated proteins and which exhibit differential detergent solubility. This implicates the presence of heterogeneous phenotypes, which overlap as protein complexes at the same molecular masses. Although the biological function of PrPC is still enigmatic, evidence reveals that PrPC exhibits metal-binding properties, which result in structural changes and decreased solubility. In this study, we analyzed the yield of PrPC metal binding affiliated with low solubility and changes in protein banding patterns. By implementing a high-speed centrifugation step, the interaction of zinc ions with PrPC was shown to generate large quantities of proteins with low solubility, consisting mainly of full-length glycosylated PrPC; whereas unglycosylated PrPC remained in the supernatants as well as truncated glycosylated proteins which lack of octarepeat sequence necessary for metal binding. This effect was considerably lower when PrPC interacted with copper ions; the presence of other metals tested exhibited no effect under these conditions. The binding of zinc and copper to PrPC demonstrated differentially soluble protein yields within distinct PrPC subtypes. PrPC–Zn2+-interaction may provide a means to differentiate glycosylated and unglycosylated subtypes and offers detailed analysis of metal-bound and metal-free protein conversion assays.  相似文献   

17.
PrPC contains several octapeptide repeats sequences toward the N-terminus which have binding affinity for divalent metals such as copper, zinc, nickel and manganese. However, the link between PrPC expression and zinc metabolism remains elusive. Here we studied the relationship between PrPC and zinc ions intracellular homeostasis using a cell line expressing a doxycycline-inducible PrPC gene. No significant difference in 65Zn2+ uptake was observed in cells expressing PrPC when compared with control cells. However, PrPC-expressing cells were more resistant to zinc-induced toxicity, suggesting an adaptative mechanism induced by PrPC. Using zinquin-ethyl-ester, a specific fluorophore for vesicular free zinc, we observed a significant re-localization of intracellular exchangeable zinc in vesicles after PrPC expression. Finally, we demonstrated that PrPC expression induces metallothionein (MT) expression, a zinc-upregulated zinc-binding protein. Taken together, these results suggest that PrPC modifies the intracellular localization of zinc rather than the cellular content and induces MT upregulation. These findings are of major importance since zinc deregulation is implicated in several neurodegenerative disorders. It is postulated that in prion diseases the conversion of PrPC to PrPSc may deregulate zinc homeostasis mediated by metallothionein.  相似文献   

18.
Synucleinopathies are a group of neurodegenerative diseases characterized by the accumulation of α-synuclein amyloids in several regions of the brain. α-Synuclein fibrils are able to spread via cell-to-cell transfer, and once inside the cells, they can template the misfolding and aggregation of the endogenous α-synuclein. Multiple mechanisms have been shown to participate in the process of propagation: endocytosis, tunneling nanotubes and macropinocytosis. Recently, we published a research showing that the cellular form of the prion protein (PrPC) acts as a receptor for α-synuclein amyloid fibrils, facilitating their internalization through and endocytic pathway. This interaction occurs by a direct interaction between the fibrils and the N-terminal domain of PrPC. In cell lines expressing the pathological form of PrP (PrPSc), the binding between PrPC and α-synuclein fibrils prevents the formation and accumulation of PrPSc, since PrPC is no longer available as a substrate for the pathological conversion templated by PrPSc. On the contrary, PrPSc deposits are cleared over passages, probably due to the increased processing of PrPC into the neuroprotective fragments N1 and C1. Starting from these data, in this work we present new insights into the role of PrPC in the internalization of protein amyloids and the possible therapeutic applications of these findings.  相似文献   

19.
Zinc and copper are indispensable trace metals for life with a recognized role as catalysts in enzyme actions. We now review evidence supporting the role of trace metals as novel allosteric modulators of ionotropic receptors: a new and fundamental physiological role for zinc and copper in neuronal and brain excitability. The review is focussed on ionotropic receptor channels including nucleotide receptors, in particular the P2X receptor family. Since zinc and copper are stored within synaptic vesicles in selected brain regions, and released to the synaptic cleft upon electrical nerve ending depolarization, it is plausible that zinc and copper reach concentrations in the synapse that profoundly affect ligand-gated ionic channels, including the ATP-gated currents of P2X receptors. The identification of key P2X receptor amino acids that act as ligands for trace metal coordination, carves the structural determinants underlying the allosteric nature of the trace metal modulation. The recognition that the identified key residues such as histidines, aspartic and glutamic acids or cysteines in the extracellular domain are different for each P2X receptor subtype and may be different for each metal, highlights the notion that each P2X receptor subtype evolved independent strategies for metal coordination, which form upon the proper three-dimensional folding of the receptor channels. The understanding of the molecular mechanism of allosteric modulation of ligand-operated ionic channels by trace metals is a new contribution to metallo-neurobiology.  相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号