fbpx
Wikipedia

PRNP

Major prion protein (PrP), is encoded in the human by the PRNP gene also known as CD230 (cluster of differentiation 230).[5][6][7][8] Expression of the protein is most predominant in the nervous system but occurs in many other tissues throughout the body.[9][10][11]

PRNP
Available structures
PDBOrtholog search: PDBe RCSB
Identifiers
AliasesPRNP, ASCR, AltPrP, CD230, CJD, GSS, KURU, PRIP, PrP, PrP27-30, PrP33-35C, PrPc, p27-30, prion protein
External IDsOMIM: 176640 MGI: 97769 HomoloGene: 7904 GeneCards: PRNP
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_001278256
NM_011170

RefSeq (protein)

NP_001265185
NP_035300

Location (UCSC)Chr 20: 4.69 – 4.7 MbChr 2: 131.75 – 131.78 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

The protein can exist in multiple isoforms: the normal PrPC form, and the protease-resistant form designated PrPRes such as the disease-causing PrPSc(scrapie) and an isoform located in mitochondria. The misfolded version PrPSc is associated with a variety of cognitive disorders and neurodegenerative diseases such as in animals: ovine scrapie, bovine spongiform encephalopathy (BSE, mad cow disease), feline spongiform encephalopathy, transmissible mink encephalopathy (TME), exotic ungulate encephalopathy, chronic wasting disease (CWD) which affects cervids; and in humans: Creutzfeldt–Jakob disease (CJD), fatal familial insomnia (FFI), Gerstmann–Sträussler–Scheinker syndrome (GSS), kuru, and variant Creutzfeldt–Jakob disease (vCJD). Similarities exist between kuru, thought to be due to human ingestion of diseased individuals, and vCJD, thought to be due to human ingestion of BSE-tainted cattle products.

Gene

The human PRNP gene is located on the short (p) arm of chromosome 20 between the end (terminus) of the arm and position 13, from base pair 4,615,068 to base pair 4,630,233.

Structure

PrP is highly conserved through mammals, lending credence to application of conclusions from test animals such as mice.[12] Comparison between primates is especially similar, ranging from 92.9-99.6% similarity in amino acid sequences. The human protein structure consists of a globular domain with three α-helices and a two-strand antiparallel β-sheet, an NH2-terminal tail, and a short COOH-terminal tail.[13] A glycophosphatidylinositol (GPI) membrane anchor at the COOH-terminal tethers PrP to cell membranes, and this proves to be integral to the transmission of conformational change; secreted PrP lacking the anchor component is unaffected by the infectious isoform.[14]

The primary sequence of PrP is 253 amino acids long before post-translational modification. Signal sequences in the amino- and carboxy- terminal ends are removed posttranslationally, resulting in a mature length of 208 amino acids. For human and golden hamster PrP, two glycosylated sites exist on helices 2 and 3 at Asn181 and Asn197. Murine PrP has glycosylation sites as Asn180 and Asn196. A disulfide bond exists between Cys179 of the second helix and Cys214 of the third helix (human PrPC numbering).

PrP messenger RNA contains a pseudoknot structure (prion pseudoknot), which is thought to be involved in regulation of PrP protein translation.[15]

Ligand-binding

The mechanism for conformational conversion to the scrapie isoform is speculated to be an elusive ligand-protein, but, so far, no such compound has been identified. However, a large body of research has developed on candidates and their interaction with the PrPC.[16]

Copper, zinc, manganese, and nickel are confirmed PrP ligands that bind to its octarepeat region.[17] Ligand binding causes a conformational change with unknown effect. Heavy metal binding at PrP has been linked to resistance to oxidative stress arising from heavy metal toxicity.[17][18]

PrPC (normal cellular) isoform

Although the precise function of PrP is not yet known, it is possibly involved in the transport of ionic copper to cells from the surrounding environment. Researchers have also proposed roles for PrP in cell signaling or in the formation of synapses.[19] PrPC attaches to the outer surface of the cell membrane by a glycosylphosphatidylinositol anchor at its C-terminal Ser231.

Prion protein contains five octapeptide repeats with sequence PHGGGWGQ (though the first repeat has the slightly-modified, histidine-deficient sequence PQGGGGWGQ). This is thought to generate a copper-binding domain via nitrogen atoms in the histidine imidazole side-chains and deprotonated amide nitrogens from the 2nd and 3rd glycines in the repeat. The ability to bind copper is, therefore, pH-dependent. NMR shows copper binding results in a conformational change at the N-terminus.

PrPSc (scrapie) isoform

PrPSc is a conformational isoform of PrPC, but this orientation tends to accumulate in compact, protease-resistant aggregates within neural tissue.[20] The abnormal PrPSc isoform has a different secondary and tertiary structure from PrPC, but identical primary sequence. Circular dichroism shows that normal PrPC has 42% alpha helical and 3% beta sheet content, whereas PrPSc is only 30% alpha helix and 43% beta sheet.[21] However, the presence of alpha helices in infectious PrPSc has come into question, with current models proposing a lack of alpha helices altogether, replaced instead with a total beta sheet composition.[22] This refolding renders the PrPSc isoform extremely resistant to proteolysis.

The propagation of PrPSc is a topic of great interest, as its accumulation is a pathological cause of neurodegeneration. Based on the progressive nature of spongiform encephalopathies, the predominant hypothesis posits that the change from normal PrPC is caused by the presence and interaction with PrPSc.[23] Strong support for this is taken from studies in which PRNP-knockout mice are resistant to the introduction of PrPSc.[24] Despite widespread acceptance of the conformation conversion hypothesis, some studies mitigate claims for a direct link between PrPSc and cytotoxicity.[25]

Polymorphisms at sites 136, 154, and 171 are associated with varying susceptibility to ovine scrapie. (These ovine sites correspond to human sites 133, 151, and 168.) Polymorphisms of the PrP-VRQ form and PrP-ARQ form are associated with increased susceptibility, whereas PrP-ARR is associated with resistance. The National Scrapie Plan of the UK aims to breed out these scrapie polymorphisms by increasing the frequency of the resistant allele.[26] However, PrP-ARR polymorphisms are susceptible to atypical scrapie, so this may prove unfruitful.

Function

Nervous system

The strong association to neurodegenerative diseases raises many questions of the function of PrP in the brain. A common approach is using PrP-knockout and transgenic mice to investigate deficiencies and differences.[27] Initial attempts produced two strains of PrP-null mice that show no physiological or developmental differences when subjected to an array of tests. However, more recent strains have shown significant cognitive abnormalities.[16]

As the null mice age, a marked loss of Purkinje cells in the cerebellum results in decreased motor coordination. However, this effect is not a direct result of PrP's absence, and rather arises from increased Doppel gene expression.[28] Other observed differences include reduced stress response and increased exploration of novel environments.[29][30]

Circadian rhythm is altered in null mice.[11] Fatal familial insomnia is thought to be the result of a point mutation in PRNP at codon 178, which corroborates PrP's involvement in sleep-wake cycles.[31] In addition, circadian regulation has been demonstrated in PrP mRNA, which cycles regularly with day-night.[32]

Memory

While null mice exhibit normal learning ability and short-term memory, long-term memory consolidation deficits have been demonstrated. As with ataxia, this is attributable to Doppel gene expression. However, spatial learning, a predominantly hippocampal-function, is decreased in the null mice and can be recovered with the reinstatement of PrP in neurons; this indicates that loss of PrP function is the cause.[33][34] The interaction of hippocampal PrP with laminin (LN) is pivotal in memory processing and is likely modulated by the kinases PKA and ERK1/2.[35][36]

Further support for PrP's role in memory formation is derived from several population studies. A test of healthy young humans showed increased long-term memory ability associated with an MM or MV genotype when compared to VV.[37] Down syndrome patients with a single valine substitution have been linked to earlier cognitive decline.[38] Several polymorphisms in PRNP have been linked with cognitive impairment in the elderly as well as earlier cognitive decline.[39][40][41] All of these studies investigated differences in codon 129, indicating its importance in the overall functionality of PrP, in particular with regard to memory.

Neurons and synapses

PrP is present in both the pre- and post-synaptic compartments, with the greatest concentration in the pre-synaptic portion.[42] Considering this and PrP's suite of behavioral influences, the neural cell functions and interactions are of particular interest. Based on the copper ligand, one proposed function casts PrP as a copper buffer for the synaptic cleft. In this role, the protein could serve as either a copper homeostasis mechanism, a calcium modulator, or a sensor for copper or oxidative stress.[43] Loss of PrP function has been linked to long-term potentiation (LTP). This effect can be positive or negative and is due to changes in neuronal excitability and synaptic transmission in the hippocampus.[44][45]

Some research indicates PrP involvement in neuronal development, differentiation, and neurite outgrowth. The PrP-activated signal transduction pathway is associated with axon and dendritic outgrowth with a series of kinases.[25][46]

Immune system

Though most attention is focused on PrP's presence in the nervous system, it is also abundant in immune system tissue. PrP immune cells include hematopoietic stem cells, mature lymphoid and myeloid compartments, and certain lymphocytes; also, it has been detected in natural killer cells, platelets, and monocytes. T cell activation is accompanied by a strong up-regulation of PrP, though it is not requisite. The lack of immunoresponse to transmissible spongiform encephalopathies (TSE), neurodegenerative diseases caused by prions, could stem from the tolerance for PrPSc.[47]

Muscles, liver, and pituitary

PrP-null mice provide clues to a role in muscular physiology when subjected to a forced swimming test, which showed reduced locomotor activity. Aging mice with an overexpression of PRNP showed significant degradation of muscle tissue.

Though present, very low levels of PrP exist in the liver and could be associated with liver fibrosis. Presence in the pituitary has been shown to affect neuroendocrine function in amphibians, but little is known concerning mammalian pituitary PrP.[16]

Cellular

Varying expression of PrP through the cell cycle has led to speculation on involvement in development. A wide range of studies has been conducted investigating the role in cell proliferation, differentiation, death, and survival.[16] Engagement of PrP has been linked to activation of signal transduction.

Modulation of signal transduction pathways has been demonstrated in cross-linking with antibodies and ligand-binding (hop/STI1 or copper).[16] Given the diversity of interactions, effects, and distribution, PrP has been proposed as dynamic surface protein functioning in signaling pathways. Specific sites along the protein bind other proteins, biomolecules, and metals. These interfaces allow specific sets of cells to communicate based on level of expression and the surrounding microenvironment. The anchoring on a GPI raft in the lipid bilayer supports claims of an extracellular scaffolding function.[16]

Diseases caused by PrP misfolding

More than 20 mutations in the PRNP gene have been identified in people with inherited prion diseases, which include the following:[48][49]

The conversion of PrPC to PrPSc conformation is the mechanism of transmission of fatal, neurodegenerative transmissible spongiform encephalopathies (TSE). This can arise from genetic factors, infection from external source, or spontaneously for reasons unknown. Accumulation of PrPSc corresponds with progression of neurodegeneration and is the proposed cause. Some PRNP mutations lead to a change in single amino acids (the building-blocks of proteins) in the prion protein. Others insert additional amino acids into the protein or cause an abnormally short protein to be made. These mutations cause the cell to make prion proteins with an abnormal structure. The abnormal protein PrPSc accumulates in the brain and destroys nerve cells, which leads to the mental and behavioral features of prion diseases.

Several other changes in the PRNP gene (called polymorphisms) do not cause prion diseases but may affect a person's risk of developing these diseases or alter the course of the disorders. An allele that codes for a PRNP variant, G127V, provides resistance to kuru.[52]

In addition, some prion diseases can be transmitted from external sources of PrPSc.[53]

  • Scrapie – fatal neurodegenerative disease in sheep, not transmissible to humans
  • Bovine spongiform encephalopathy (mad-cow disease) – fatal neurodegenerative disease in cows, which can be transmitted to humans by ingestion of brain, spinal, or digestive tract tissue of an infected cow
  • Kuru – TSE in humans, transmitted via funerary cannibalism. Generally, affected family members were given, by tradition, parts of the central nervous system according to ritual when consuming deceased family members.

Alzheimer's disease

PrPC protein is one of several cellular receptors of soluble amyloid beta (Aβ) oligomers, which are canonically implicated in causing Alzheimer's disease.[54] These oligomers are composed smaller Aβ plaques, and are the most damaging to the integrity of a neuron.[54] The precise mechanism of soluble Aβ oligomers directly inducing neurotoxicity is unknown, and experimental deletion of PRNP in animals has yielded several conflicting findings. When Aβ oligomers were injected into the cerebral ventricles of a mouse model of Alzheimer's, PRNP deletion did not offer protection, only anti-PrPC antibodies prevented long-term memory and spatial learning deficits.[55][56] This would suggest either an unequal relation between PRNP and Aβ oligomer-mediated neurodegeneration or a site-specific relational significance. In the case of direct injection of Aβ oligomers into the hippocampus, PRNP-knockout mice were found to be indistinguishable from control with respect to both neuronal death rates and measurements of synaptic plasticity.[54][56] It was further found that Aβ-oligomers bind to PrPC at the postsynaptic density, indirectly overactivating the NMDA receptor via the Fyn enzyme, resulting in excitotoxicity.[55] Soluble Aβ oligomers also bind to PrPC at the dendritic spines, forming a complex with Fyn and excessively activating tau, another protein implicated in Alzheimer's.[55] As the gene FYN codes for the enzyme Fyn, FYN-knockout mice display neither excitotoxic events nor dendritic spine shrinkage when injected with Aβ oligomers.[55] In mammals, the full functional significance of PRNP remains unclear, as PRNP deletion has been prophylactically implemented by the cattle industry without apparent harm.[54] In mice, this same deletion phenotypically varies between Alzheimer's mouse lines, as hAPPJ20 mice and TgCRND8 mice show a slight increase in epileptic activity, contributing to conflicting results when examining Alzheimer's survival rates.[54] Of note, the deletion of PRNP in both APPswe and SEN1dE9, two other transgenic models of Alzheimer's, attenuated the epilepsy-induced death phenotype seen in a subset of these animals.[54] Taken collectively, recent evidence suggests PRNP may be important for conducing the neurotoxic effects of soluble Aβ-oligomers and the emergent disease state of Alzheimer's.[54][55][56]

In humans, the methionine/valine polymorphism at codon 129 of PRNP (rs1799990) is most closely associated with Alzheimer's disease.[57] Variant V allele carriers (VV and MV) show a 13% decreased risk with respect to developing Alzheimer's compared to the methionine homozygote (MM). However, the protective effects of variant V carriers have been found exclusively in Caucasians. The decreased risk in V allele carriers is further limited to late-onset Alzheimer's disease only (≥ 65 years).[57] PRNP can also functionally interact with polymorphisms in two other genes implicated in Alzheimer's, PSEN1 and APOE, to compound risk for both Alzheimer's and sporadic Creutzfeldt–Jakob disease.[54] A point mutation on codon 102 of PRNP at least in part contributed to three separate patients' atypical frontotemporal dementia within the same family, suggesting a new phenotype for Gerstmann–Sträussler–Scheinker syndrome.[54][58] The same study proposed sequencing PRNP in cases of ambiguously diagnosed dementia, as the various forms of dementia can prove challenging to differentially diagnose.[58]

Interactions

A strong interaction exists between PrP and the cochaperone Hop (Hsp70/Hsp90 organizing protein; also called STI1 (Stress-induced protein 1)).[59][60]

References

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000171867 - Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000079037 - Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ Kretzschmar HA, Stowring LE, Westaway D, Stubblebine WH, Prusiner SB, Dearmond SJ (August 1986). "Molecular cloning of a human prion protein cDNA". DNA. 5 (4): 315–24. doi:10.1089/dna.1986.5.315. PMID 3755672.
  6. ^ Sparkes RS, Simon M, Cohn VH, Fournier RE, Lem J, Klisak I, Heinzmann C, Blatt C, Lucero M, Mohandas T (October 1986). "Assignment of the human and mouse prion protein genes to homologous chromosomes". Proc. Natl. Acad. Sci. U.S.A. 83 (19): 7358–62. Bibcode:1986PNAS...83.7358S. doi:10.1073/pnas.83.19.7358. PMC 386716. PMID 3094007.
  7. ^ Liao YC, Lebo RV, Clawson GA, Smuckler EA (July 1986). "Human prion protein cDNA: molecular cloning, chromosomal mapping, and biological implications". Science. 233 (4761): 364–7. Bibcode:1986Sci...233..364L. doi:10.1126/science.3014653. PMID 3014653.
  8. ^ Robakis NK, Devine-Gage EA, Jenkins EC, Kascsak RJ, Brown WT, Krawczun MS, Silverman WP (October 1986). "Localization of a human gene homologous to the PrP gene on the p arm of chromosome 20 and detection of PrP-related antigens in normal human brain". Biochem. Biophys. Res. Commun. 140 (2): 758–65. doi:10.1016/0006-291X(86)90796-5. PMID 2877664.
  9. ^ Prusiner SB (2001). "Shattuck lecture--neurodegenerative diseases and prions". N Engl J Med. 344 (20): 1516–26. doi:10.1056/NEJM200105173442006. PMID 11357156.
  10. ^ Weissmann C (2004). "The state of the prion". Nat Rev Microbiol. 2 (11): 861–71. doi:10.1038/nrmicro1025. PMID 15494743. S2CID 20992257.
  11. ^ a b Zomosa-Signoret V, Arnaud JD, Fontes P, Alvarez-Martinez MT, Liautard JP (2008). "Physiological role of the cellular prion protein" (PDF). Vet. Res. 39 (4): 9. doi:10.1051/vetres:2007048. PMID 18073096.
  12. ^ Damberger FF, Christen B, Pérez DR, Hornemann S, Wüthrich K (October 2011). "Cellular prion protein conformation and function". Proc. Natl. Acad. Sci. U.S.A. 108 (42): 17308–13. Bibcode:2011PNAS..10817308D. doi:10.1073/pnas.1106325108. PMC 3198368. PMID 21987789.
  13. ^ Schätzl HM, Da Costa M, Taylor L, Cohen FE, Prusiner SB (January 1995). "Prion protein gene variation among primates". J. Mol. Biol. 245 (4): 362–74. doi:10.1006/jmbi.1994.0030. PMID 7837269.
  14. ^ Chesebro B, Trifilo M, Race R, Meade-White K, Teng C, LaCasse R, Raymond L, Favara C, Baron G, Priola S, Caughey B, Masliah E, Oldstone M (June 2005). "Anchorless prion protein results in infectious amyloid disease without clinical scrapie". Science. 308 (5727): 1435–9. Bibcode:2005Sci...308.1435C. CiteSeerX 10.1.1.401.781. doi:10.1126/science.1110837. PMID 15933194. S2CID 10064966.
  15. ^ Barrette I, Poisson G, Gendron P, Major F (2001). "Pseudoknots in prion protein mRNAs confirmed by comparative sequence analysis and pattern searching". Nucleic Acids Res. 29 (3): 753–758. doi:10.1093/nar/29.3.753. PMC 30388. PMID 11160898.
  16. ^ a b c d e f Linden R, Martins VR, Prado MA, Cammarota M, Izquierdo I, Brentani RR (April 2008). "Physiology of the prion protein". Physiol. Rev. 88 (2): 673–728. doi:10.1152/physrev.00007.2007. PMID 18391177.
  17. ^ a b Prčina M, Kontseková E, Novák M (2015). "Prion protein prevents heavy metals overloading of cells and thus protects them against their toxicity". Acta Virol. 59 (2): 179–84. doi:10.4149/av_2015_02_179. PMID 26104335.
  18. ^ Brown DR, Clive C, Haswell SJ (January 2001). "Antioxidant activity related to copper binding of native prion protein". J. Neurochem. 76 (1): 69–76. doi:10.1046/j.1471-4159.2001.00009.x. PMID 11145979. S2CID 45647133.
  19. ^ Kanaani J, Prusiner SB, Diacovo J, Baekkeskov S, Legname G (December 2005). "Recombinant prion protein induces rapid polarization and development of synapses in embryonic rat hippocampal neurons in vitro". Journal of Neurochemistry. 95 (5): 1373–86. doi:10.1111/j.1471-4159.2005.03469.x. PMID 16313516. S2CID 24329326.
  20. ^ Ross CA, Poirier MA (July 2004). "Protein aggregation and neurodegenerative disease". Nat. Med. 10 Suppl (7): S10–7. doi:10.1038/nm1066. PMID 15272267. S2CID 205383483.
  21. ^ Pan KM, Baldwin M, Nguyen J, Gasset M, Serban A, Groth D, Mehlhorn I, Huang Z, Fletterick RJ, Cohen FE (December 1993). "Conversion of alpha-helices into beta-sheets features in the formation of the scrapie prion proteins". Proc. Natl. Acad. Sci. U.S.A. 90 (23): 10962–6. Bibcode:1993PNAS...9010962P. doi:10.1073/pnas.90.23.10962. PMC 47901. PMID 7902575.
  22. ^ Baskakov, Ilia V.; Caughey, Byron; Requena, Jesús R.; Sevillano, Alejandro M.; Surewicz, Witold K.; Wille, Holger (2019-01-01). "The prion 2018 round tables (I): the structure of PrPSc". Prion. 13 (1): 46–52. doi:10.1080/19336896.2019.1569450. ISSN 1933-6896. PMC 6422368. PMID 30646817.
  23. ^ Sandberg MK, Al-Doujaily H, Sharps B, Clarke AR, Collinge J (February 2011). "Prion propagation and toxicity in vivo occur in two distinct mechanistic phases". Nature. 470 (7335): 540–2. Bibcode:2011Natur.470..540S. doi:10.1038/nature09768. PMID 21350487. S2CID 4399936.
  24. ^ Büeler H, Aguzzi A, Sailer A, Greiner RA, Autenried P, Aguet M, Weissmann C (July 1993). "Mice devoid of PrP are resistant to scrapie". Cell. 73 (7): 1339–47. doi:10.1016/0092-8674(93)90360-3. PMID 8100741.
  25. ^ a b Aguzzi A, Baumann F, Bremer J (2008). "The prion's elusive reason for being". Annu. Rev. Neurosci. 31: 439–77. doi:10.1146/annurev.neuro.31.060407.125620. PMID 18558863.
  26. ^ Atkinson M (October 2001). "National scrapie plan". The Veterinary Record. 149 (15): 462. PMID 11688751.
  27. ^ Weissmann C, Flechsig E (2003). "PrP knock-out and PrP transgenic mice in prion research". Br. Med. Bull. 66: 43–60. doi:10.1093/bmb/66.1.43. PMID 14522848.
  28. ^ Katamine S, Nishida N, Sugimoto T, Noda T, Sakaguchi S, Shigematsu K, Kataoka Y, Nakatani A, Hasegawa S, Moriuchi R, Miyamoto T (December 1998). "Impaired motor coordination in mice lacking prion protein". Cell. Mol. Neurobiol. 18 (6): 731–42. doi:10.1023/A:1020234321879. PMID 9876879. S2CID 23409873.
  29. ^ Nico PB, 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 (July 2005). "Altered behavioural response to acute stress in mice lacking cellular prion protein". Behav. Brain Res. 162 (2): 173–81. doi:10.1016/j.bbr.2005.02.003. PMID 15970215. S2CID 37511702.
  30. ^ Roesler R, Walz R, Quevedo J, de-Paris F, Zanata SM, Graner E, Izquierdo I, Martins VR, Brentani RR (August 1999). "Normal inhibitory avoidance learning and anxiety, but increased locomotor activity in mice devoid of PrP(C)". Brain Res. Mol. Brain Res. 71 (2): 349–53. doi:10.1016/S0169-328X(99)00193-X. PMID 10521590.
  31. ^ Medori R, Tritschler HJ, LeBlanc A, Villare F, Manetto V, Chen HY, Xue R, Leal S, Montagna P, Cortelli P (February 1992). "Fatal familial insomnia, a prion disease with a mutation at codon 178 of the prion protein gene". N. Engl. J. Med. 326 (7): 444–9. doi:10.1056/NEJM199202133260704. PMC 6151859. PMID 1346338.
  32. ^ Cagampang FR, Whatley SA, Mitchell AL, Powell JF, Campbell IC, Coen CW (1999). "Circadian regulation of prion protein messenger RNA in the rat forebrain: a widespread and synchronous rhythm". Neuroscience. 91 (4): 1201–4. doi:10.1016/S0306-4522(99)00092-5. PMID 10391428. S2CID 42892475.
  33. ^ Criado JR, Sánchez-Alavez M, Conti B, Giacchino JL, Wills DN, Henriksen SJ, Race R, Manson JC, Chesebro B, Oldstone MB (2005). "Mice devoid of prion protein have cognitive deficits that are rescued by reconstitution of PrP in neurons". Neurobiol. Dis. 19 (1–2): 255–65. doi:10.1016/j.nbd.2005.01.001. PMID 15837581. S2CID 2618712.
  34. ^ Balducci C, Beeg M, Stravalaci M, Bastone A, Sclip A, Biasini E, Tapella L, Colombo L, Manzoni C, Borsello T, Chiesa R, Gobbi M, Salmona M, Forloni G (February 2010). "Synthetic amyloid-beta oligomers impair long-term memory independently of cellular prion protein". Proc. Natl. Acad. Sci. U.S.A. 107 (5): 2295–300. Bibcode:2010PNAS..107.2295B. doi:10.1073/pnas.0911829107. PMC 2836680. PMID 20133875.
  35. ^ Coitinho AS, Freitas AR, Lopes MH, Hajj GN, Roesler R, Walz R, Rossato JI, Cammarota M, Izquierdo I, Martins VR, Brentani RR (December 2006). "The interaction between prion protein and laminin modulates memory consolidation". Eur. J. Neurosci. 24 (11): 3255–64. doi:10.1111/j.1460-9568.2006.05156.x. PMID 17156386. S2CID 17164351.
  36. ^ Shorter J, Lindquist S (June 2005). "Prions as adaptive conduits of memory and inheritance". Nat. Rev. Genet. 6 (6): 435–50. doi:10.1038/nrg1616. PMID 15931169. S2CID 5575951.
  37. ^ Papassotiropoulos A, Wollmer MA, Aguzzi A, Hock C, Nitsch RM, de Quervain DJ (August 2005). "The prion gene is associated with human long-term memory" (PDF). Hum. Mol. Genet. 14 (15): 2241–6. doi:10.1093/hmg/ddi228. PMID 15987701.
  38. ^ Del Bo R, Comi GP, Giorda R, Crimi M, Locatelli F, Martinelli-Boneschi F, Pozzoli U, Castelli E, Bresolin N, Scarlato G (June 2003). "The 129 codon polymorphism of the prion protein gene influences earlier cognitive performance in Down syndrome subjects". J. Neurol. 250 (6): 688–92. doi:10.1007/s00415-003-1057-5. PMID 12796830. S2CID 21049364.
  39. ^ Berr C, Richard F, Dufouil C, Amant C, Alperovitch A, Amouyel P (September 1998). "Polymorphism of the prion protein is associated with cognitive impairment in the elderly: the EVA study". Neurology. 51 (3): 734–7. doi:10.1212/wnl.51.3.734. PMID 9748018. S2CID 11352163.
  40. ^ Croes EA, Dermaut B, Houwing-Duistermaat JJ, Van den Broeck M, Cruts M, Breteler MM, Hofman A, van Broeckhoven C, van Duijn CM (August 2003). "Early cognitive decline is associated with prion protein codon 129 polymorphism". Ann. Neurol. 54 (2): 275–6. doi:10.1002/ana.10658. PMID 12891686. S2CID 31538672.
  41. ^ Kachiwala SJ, Harris SE, Wright AF, Hayward C, Starr JM, Whalley LJ, Deary IJ (September 2005). "Genetic influences on oxidative stress and their association with normal cognitive ageing". Neurosci. Lett. 386 (2): 116–20. doi:10.1016/j.neulet.2005.05.067. PMID 16023289. S2CID 23642220.
  42. ^ Herms J, Tings T, Gall S, Madlung A, Giese A, Siebert H, Schürmann P, Windl O, Brose N, Kretzschmar H (October 1999). "Evidence of presynaptic location and function of the prion protein". J. Neurosci. 19 (20): 8866–75. doi:10.1523/JNEUROSCI.19-20-08866.1999. PMC 6782778. PMID 10516306.
  43. ^ Kardos J, Kovács I, Hajós F, Kálmán M, Simonyi M (August 1989). "Nerve endings from rat brain tissue release copper upon depolarization. A possible role in regulating neuronal excitability". Neurosci. Lett. 103 (2): 139–44. doi:10.1016/0304-3940(89)90565-X. PMID 2549468. S2CID 24917999.
  44. ^ Bailey CH, Kandel ER, Si K (September 2004). "The persistence of long-term memory: a molecular approach to self-sustaining changes in learning-induced synaptic growth". Neuron. 44 (1): 49–57. doi:10.1016/j.neuron.2004.09.017. PMID 15450159. S2CID 2637074.
  45. ^ Barco A, Bailey CH, Kandel ER (June 2006). "Common molecular mechanisms in explicit and implicit memory". J. Neurochem. 97 (6): 1520–33. doi:10.1111/j.1471-4159.2006.03870.x. PMID 16805766. S2CID 26307975.
  46. ^ Laurén J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM (February 2009). "Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers". Nature. 457 (7233): 1128–32. Bibcode:2009Natur.457.1128L. doi:10.1038/nature07761. PMC 2748841. PMID 19242475.
  47. ^ Isaacs JD, Jackson GS, Altmann DM (October 2006). "The role of the cellular prion protein in the immune system". Clin. Exp. Immunol. 146 (1): 1–8. doi:10.1111/j.1365-2249.2006.03194.x. PMC 1809729. PMID 16968391.
  48. ^ Castilla J, Hetz C, Soto C (2004). "Molecular mechanisms of neurotoxicity of pathological prion protein". Curr Mol Med. 4 (4): 397–403. doi:10.2174/1566524043360654. PMID 15354870.
  49. ^ Kovács GG, Trabattoni G, Hainfellner JA, Ironside JW, Knight RS, Budka H (2002). "Mutations of the prion protein gene phenotypic spectrum". J Neurol. 249 (11): 1567–82. doi:10.1007/s00415-002-0896-9. PMID 12420099. S2CID 22688729.
  50. ^ Collins S, McLean CA, Masters CL (2001). "Gerstmann-Straussler-Scheinker syndrome, fatal familial insomnia, and kuru: a review of these less common human transmissible spongiform encephalopathies". J Clin Neurosci. 8 (5): 387–97. doi:10.1054/jocn.2001.0919. PMID 11535002. S2CID 31976428.
  51. ^ Montagna P, Gambetti P, Cortelli P, Lugaresi E (2003). "Familial and sporadic fatal insomnia". Lancet Neurol. 2 (3): 167–76. doi:10.1016/S1474-4422(03)00323-5. PMID 12849238. S2CID 20822956.
  52. ^ Mead S, Whitfield J, Poulter M, Shah P, Uphill J, Campbell T, Al-Dujaily H, Hummerich H, Beck J, Mein CA, Verzilli C, Whittaker J, Alpers MP, Collinge J (2009). "A Novel Protective Prion Protein Variant that Colocalizes with Kuru Exposure" (PDF). The New England Journal of Medicine. 361 (21): 2056–2065. doi:10.1056/NEJMoa0809716. PMID 19923577.
    • "Brain disease 'resistance gene' evolves in Papua New Guinea community; could offer insights into CJD". ScienceDaily (Press release). November 21, 2009.
  53. ^ Hwang D, Lee IY, Yoo H, Gehlenborg N, Cho JH, Petritis B, Baxter D, Pitstick R, Young R, Spicer D, Price ND, Hohmann JG, Dearmond SJ, Carlson GA, Hood LE (2009). "A systems approach to prion disease". Mol. Syst. Biol. 5 (1): 252. doi:10.1038/msb.2009.10. PMC 2671916. PMID 19308092.
  54. ^ a b c d e f g h i Laurén J (2014). "Cellular prion protein as a therapeutic target in Alzheimer's disease". Journal of Alzheimer's Disease. 38 (2): 227–244. doi:10.3233/JAD-130950. PMID 23948943.
  55. ^ a b c d e Zhou J, Liu B (May 2013). "Alzheimer's disease and prion protein". Intractable & Rare Diseases Research. 2 (2): 35–44. doi:10.5582/irdr.2013.v2.2.35. PMC 4204584. PMID 25343100.
  56. ^ a b c Laurén J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM (Feb 2009). "Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers". Nature. 457 (7233): 1128–1132. Bibcode:2009Natur.457.1128L. doi:10.1038/nature07761. PMC 2748841. PMID 19242475.
  57. ^ a b He J, Li X, Yang J, Huang J, Fu X, Zhang Y, Fan H (Mar 2013). "The association between the methionine/valine (M/V) polymorphism (rs1799990) in the PRNP gene and the risk of Alzheimer disease: an update by meta-analysis". Journal of the Neurological Sciences. 326 (1–2): 89–95. doi:10.1016/j.jns.2013.01.020. PMID 23399523. S2CID 31070331.
  58. ^ a b Giovagnoli AR, Di Fede G, Aresi A, Reati F, Rossi G, Tagliavini F (December 2008). "Atypical frontotemporal dementia as a new clinical phenotype of Gerstmann-Straussler-Scheinker disease with the PrP-P102L mutation. Description of a previously unreported Italian family". Neurological Sciences. 29 (6): 405–10. doi:10.1007/s10072-008-1025-z. PMID 19030774. S2CID 20553167.
  59. ^ Americo TA, Chiarini LB, Linden R (June 2007). "Signaling induced by hop/STI-1 depends on endocytosis". Biochem. Biophys. Res. Commun. 358 (2): 620–5. doi:10.1016/j.bbrc.2007.04.202. PMID 17498662.
  60. ^ Zanata SM, Lopes MH, Mercadante AF, Hajj GN, Chiarini LB, Nomizo R, Freitas AR, Cabral AL, Lee KS, Juliano MA, de Oliveira E, Jachieri SG, Burlingame A, Huang L, Linden R, Brentani RR, Martins VR (Jul 2002). "Stress-inducible protein 1 is a cell surface ligand for cellular prion that triggers neuroprotection". EMBO J. 21 (13): 3307–16. doi:10.1093/emboj/cdf325. PMC 125391. PMID 12093732.

External links

  • PRNP (PrP) gene at GeneCard
  • PRNP+protein,+human at the US National Library of Medicine Medical Subject Headings (MeSH)
  • Susan Lindquist's Seminar: "The Surprising World of Prion Biology"

prnp, confused, with, prions, infectious, forms, proteins, which, have, been, observed, almost, instances, forms, need, major, prion, protein, encoded, human, gene, also, known, cd230, cluster, differentiation, expression, protein, most, predominant, nervous, . Not to be confused with prions infectious forms of proteins which have so far been observed in almost all instances to be forms of PRNP but need not be Major prion protein PrP is encoded in the human by the PRNP gene also known as CD230 cluster of differentiation 230 5 6 7 8 Expression of the protein is most predominant in the nervous system but occurs in many other tissues throughout the body 9 10 11 PRNPAvailable structuresPDBOrtholog search PDBe RCSBList of PDB id codes1E1G 1E1J 1E1P 1E1S 1E1U 1E1W 1FKC 1FO7 1H0L 1HJM 1HJN 1I4M 1OEH 1OEI 1QLX 1QLZ 1QM0 1QM1 1QM2 1QM3 2IV4 2IV5 2IV6 2K1D 2KUN 2LBG 2LEJ 2LFT 2LSB 2LV1 2OL9 2W9E 3HAF 3HAK 3HEQ 3HER 3HES 3HJ5 3HJX 3MD4 3MD5 3NHC 3NHD 3NVF 4DGI 4E1H 4E1I s1E1G 1E1J 1E1P 1E1S 1E1U 1E1W 1FKC 1FO7 1H0L 1HJM 1HJN 1I4M 1OEH 1OEI 1QLX 1QLZ 1QM0 1QM1 1QM2 1QM3 2IV4 2IV5 2IV6 2K1D 2KUN 2LBG 2LEJ 2LFT 2LSB 2LV1 2M8T 2W9E 3HAF 3HAK 3HEQ 3HER 3HES 3HJ5 3HJX 3MD4 3MD5 3NHC 3NVF 4DGI 4E1H 4E1I 4KML 4N9OIdentifiersAliasesPRNP ASCR AltPrP CD230 CJD GSS KURU PRIP PrP PrP27 30 PrP33 35C PrPc p27 30 prion proteinExternal IDsOMIM 176640 MGI 97769 HomoloGene 7904 GeneCards PRNPGene location Human Chr Chromosome 20 human 1 Band20p13Start4 686 350 bp 1 End4 701 590 bp 1 Gene location Mouse Chr Chromosome 2 mouse 2 Band2 F2 2 64 07 cMStart131 751 848 bp 2 End131 780 349 bp 2 RNA expression patternBgeeHumanMouse ortholog Top expressed inBrodmann area 23Region I of hippocampus properretinal pigment epitheliumorbitofrontal cortexendothelial cellspinal gangliatrigeminal ganglionponsgerminal epitheliumcerebellar vermisTop expressed inentorhinal cortexRegion I of hippocampus properamygdalacingulate gyrussubiculummedial dorsal nucleuslateral geniculate nucleusprimary motor cortexmedial geniculate nucleushippocampus properMore reference expression dataBioGPSMore reference expression dataGene ontologyMolecular functionchaperone binding transmembrane transporter binding microtubule binding ATP dependent protein binding metal ion binding tubulin binding protein binding identical protein binding copper ion binding lamin binding amyloid beta binding protease binding glycosaminoglycan binding type 5 metabotropic glutamate receptor binding type 8 metabotropic glutamate receptor binding signaling receptor activity protein containing complex binding cupric ion binding cuprous ion bindingCellular componentmitochondrial outer membrane membrane mitochondrion integral component of membrane cytoplasm Golgi apparatus plasma membrane cell surface endoplasmic reticulum membrane raft anchored component of membrane extracellular exosome nucleus extrinsic component of membrane cytosol postsynaptic density inclusion body dendrite anchored component of external side of plasma membrane nuclear membrane intracellular membrane bounded organelle postsynapseBiological processnegative regulation of protein phosphorylation negative regulation of interferon gamma production response to cadmium ion negative regulation of calcineurin NFAT signaling cascade response to copper ion negative regulation of apoptotic process response to oxidative stress negative regulation of DNA binding transcription factor activity negative regulation of T cell receptor signaling pathway regulation of protein localization negative regulation of activated T cell proliferation learning or memory cellular copper ion homeostasis cellular response to copper ion cell cycle metabolism negative regulation of interleukin 17 production protein homooligomerization negative regulation of interleukin 2 production regulation of potassium ion transmembrane transport long term memory positive regulation of cell death negative regulation of protein processing protein destabilization activation of protein kinase activity calcium mediated signaling using intracellular calcium source negative regulation of catalytic activity positive regulation of neuron apoptotic process regulation of peptidyl tyrosine phosphorylation positive regulation of peptidyl tyrosine phosphorylation positive regulation of protein tyrosine kinase activity positive regulation of protein targeting to membrane dendritic spine maintenance negative regulation of long term synaptic potentiation regulation of glutamate receptor signaling pathway positive regulation of neuron death negative regulation of amyloid beta formation regulation of intracellular calcium activated chloride channel activity negative regulation of dendritic spine maintenance negative regulation of amyloid precursor protein catabolic process positive regulation of protein localization to plasma membrane response to amyloid beta cellular response to amyloid beta regulation of calcium ion import across plasma membrane neuron projection maintenanceSources Amigo QuickGOOrthologsSpeciesHumanMouseEntrez562119122EnsemblENSG00000171867ENSMUSG00000079037UniProtP04156P04925RefSeq mRNA NM 183079NM 000311NM 001080121NM 001080122NM 001080123NM 001271561NM 001278256NM 011170RefSeq protein NP 000302NP 001073590NP 001073591NP 001073592NP 001258490NP 898902NP 000302 1NP 001073590 1NP 001073591 1NP 001073592 1NP 898902 1NP 001265185NP 035300Location UCSC Chr 20 4 69 4 7 MbChr 2 131 75 131 78 MbPubMed search 3 4 WikidataView Edit HumanView Edit MouseThe protein can exist in multiple isoforms the normal PrPC form and the protease resistant form designated PrPRes such as the disease causing PrPSc scrapie and an isoform located in mitochondria The misfolded version PrPSc is associated with a variety of cognitive disorders and neurodegenerative diseases such as in animals ovine scrapie bovine spongiform encephalopathy BSE mad cow disease feline spongiform encephalopathy transmissible mink encephalopathy TME exotic ungulate encephalopathy chronic wasting disease CWD which affects cervids and in humans Creutzfeldt Jakob disease CJD fatal familial insomnia FFI Gerstmann Straussler Scheinker syndrome GSS kuru and variant Creutzfeldt Jakob disease vCJD Similarities exist between kuru thought to be due to human ingestion of diseased individuals and vCJD thought to be due to human ingestion of BSE tainted cattle products Contents 1 Gene 2 Structure 2 1 Ligand binding 2 2 PrPC normal cellular isoform 2 3 PrPSc scrapie isoform 3 Function 3 1 Nervous system 3 1 1 Memory 3 1 2 Neurons and synapses 3 2 Immune system 3 3 Muscles liver and pituitary 3 4 Cellular 4 Diseases caused by PrP misfolding 4 1 Alzheimer s disease 5 Interactions 6 References 7 External linksGene Edit Chromosome 20 The human PRNP gene is located on the short p arm of chromosome 20 between the end terminus of the arm and position 13 from base pair 4 615 068 to base pair 4 630 233 Structure EditPrP is highly conserved through mammals lending credence to application of conclusions from test animals such as mice 12 Comparison between primates is especially similar ranging from 92 9 99 6 similarity in amino acid sequences The human protein structure consists of a globular domain with three a helices and a two strand antiparallel b sheet an NH2 terminal tail and a short COOH terminal tail 13 A glycophosphatidylinositol GPI membrane anchor at the COOH terminal tethers PrP to cell membranes and this proves to be integral to the transmission of conformational change secreted PrP lacking the anchor component is unaffected by the infectious isoform 14 The primary sequence of PrP is 253 amino acids long before post translational modification Signal sequences in the amino and carboxy terminal ends are removed posttranslationally resulting in a mature length of 208 amino acids For human and golden hamster PrP two glycosylated sites exist on helices 2 and 3 at Asn181 and Asn197 Murine PrP has glycosylation sites as Asn180 and Asn196 A disulfide bond exists between Cys179 of the second helix and Cys214 of the third helix human PrPC numbering PrP messenger RNA contains a pseudoknot structure prion pseudoknot which is thought to be involved in regulation of PrP protein translation 15 Ligand binding Edit The mechanism for conformational conversion to the scrapie isoform is speculated to be an elusive ligand protein but so far no such compound has been identified However a large body of research has developed on candidates and their interaction with the PrPC 16 Copper zinc manganese and nickel are confirmed PrP ligands that bind to its octarepeat region 17 Ligand binding causes a conformational change with unknown effect Heavy metal binding at PrP has been linked to resistance to oxidative stress arising from heavy metal toxicity 17 18 PrPC normal cellular isoform Edit Although the precise function of PrP is not yet known it is possibly involved in the transport of ionic copper to cells from the surrounding environment Researchers have also proposed roles for PrP in cell signaling or in the formation of synapses 19 PrPC attaches to the outer surface of the cell membrane by a glycosylphosphatidylinositol anchor at its C terminal Ser231 Prion protein contains five octapeptide repeats with sequence PHGGGWGQ though the first repeat has the slightly modified histidine deficient sequence PQGGGGWGQ This is thought to generate a copper binding domain via nitrogen atoms in the histidine imidazole side chains and deprotonated amide nitrogens from the 2nd and 3rd glycines in the repeat The ability to bind copper is therefore pH dependent NMR shows copper binding results in a conformational change at the N terminus PrPSc scrapie isoform Edit PrPSc is a conformational isoform of PrPC but this orientation tends to accumulate in compact protease resistant aggregates within neural tissue 20 The abnormal PrPSc isoform has a different secondary and tertiary structure from PrPC but identical primary sequence Circular dichroism shows that normal PrPC has 42 alpha helical and 3 beta sheet content whereas PrPSc is only 30 alpha helix and 43 beta sheet 21 However the presence of alpha helices in infectious PrPSc has come into question with current models proposing a lack of alpha helices altogether replaced instead with a total beta sheet composition 22 This refolding renders the PrPSc isoform extremely resistant to proteolysis The propagation of PrPSc is a topic of great interest as its accumulation is a pathological cause of neurodegeneration Based on the progressive nature of spongiform encephalopathies the predominant hypothesis posits that the change from normal PrPC is caused by the presence and interaction with PrPSc 23 Strong support for this is taken from studies in which PRNP knockout mice are resistant to the introduction of PrPSc 24 Despite widespread acceptance of the conformation conversion hypothesis some studies mitigate claims for a direct link between PrPSc and cytotoxicity 25 Polymorphisms at sites 136 154 and 171 are associated with varying susceptibility to ovine scrapie These ovine sites correspond to human sites 133 151 and 168 Polymorphisms of the PrP VRQ form and PrP ARQ form are associated with increased susceptibility whereas PrP ARR is associated with resistance The National Scrapie Plan of the UK aims to breed out these scrapie polymorphisms by increasing the frequency of the resistant allele 26 However PrP ARR polymorphisms are susceptible to atypical scrapie so this may prove unfruitful Function EditNervous system Edit The strong association to neurodegenerative diseases raises many questions of the function of PrP in the brain A common approach is using PrP knockout and transgenic mice to investigate deficiencies and differences 27 Initial attempts produced two strains of PrP null mice that show no physiological or developmental differences when subjected to an array of tests However more recent strains have shown significant cognitive abnormalities 16 As the null mice age a marked loss of Purkinje cells in the cerebellum results in decreased motor coordination However this effect is not a direct result of PrP s absence and rather arises from increased Doppel gene expression 28 Other observed differences include reduced stress response and increased exploration of novel environments 29 30 Circadian rhythm is altered in null mice 11 Fatal familial insomnia is thought to be the result of a point mutation in PRNP at codon 178 which corroborates PrP s involvement in sleep wake cycles 31 In addition circadian regulation has been demonstrated in PrP mRNA which cycles regularly with day night 32 Memory Edit While null mice exhibit normal learning ability and short term memory long term memory consolidation deficits have been demonstrated As with ataxia this is attributable to Doppel gene expression However spatial learning a predominantly hippocampal function is decreased in the null mice and can be recovered with the reinstatement of PrP in neurons this indicates that loss of PrP function is the cause 33 34 The interaction of hippocampal PrP with laminin LN is pivotal in memory processing and is likely modulated by the kinases PKA and ERK1 2 35 36 Further support for PrP s role in memory formation is derived from several population studies A test of healthy young humans showed increased long term memory ability associated with an MM or MV genotype when compared to VV 37 Down syndrome patients with a single valine substitution have been linked to earlier cognitive decline 38 Several polymorphisms in PRNP have been linked with cognitive impairment in the elderly as well as earlier cognitive decline 39 40 41 All of these studies investigated differences in codon 129 indicating its importance in the overall functionality of PrP in particular with regard to memory Neurons and synapses Edit PrP is present in both the pre and post synaptic compartments with the greatest concentration in the pre synaptic portion 42 Considering this and PrP s suite of behavioral influences the neural cell functions and interactions are of particular interest Based on the copper ligand one proposed function casts PrP as a copper buffer for the synaptic cleft In this role the protein could serve as either a copper homeostasis mechanism a calcium modulator or a sensor for copper or oxidative stress 43 Loss of PrP function has been linked to long term potentiation LTP This effect can be positive or negative and is due to changes in neuronal excitability and synaptic transmission in the hippocampus 44 45 Some research indicates PrP involvement in neuronal development differentiation and neurite outgrowth The PrP activated signal transduction pathway is associated with axon and dendritic outgrowth with a series of kinases 25 46 Immune system Edit Though most attention is focused on PrP s presence in the nervous system it is also abundant in immune system tissue PrP immune cells include hematopoietic stem cells mature lymphoid and myeloid compartments and certain lymphocytes also it has been detected in natural killer cells platelets and monocytes T cell activation is accompanied by a strong up regulation of PrP though it is not requisite The lack of immunoresponse to transmissible spongiform encephalopathies TSE neurodegenerative diseases caused by prions could stem from the tolerance for PrPSc 47 Muscles liver and pituitary Edit PrP null mice provide clues to a role in muscular physiology when subjected to a forced swimming test which showed reduced locomotor activity Aging mice with an overexpression of PRNP showed significant degradation of muscle tissue Though present very low levels of PrP exist in the liver and could be associated with liver fibrosis Presence in the pituitary has been shown to affect neuroendocrine function in amphibians but little is known concerning mammalian pituitary PrP 16 Cellular Edit Varying expression of PrP through the cell cycle has led to speculation on involvement in development A wide range of studies has been conducted investigating the role in cell proliferation differentiation death and survival 16 Engagement of PrP has been linked to activation of signal transduction Modulation of signal transduction pathways has been demonstrated in cross linking with antibodies and ligand binding hop STI1 or copper 16 Given the diversity of interactions effects and distribution PrP has been proposed as dynamic surface protein functioning in signaling pathways Specific sites along the protein bind other proteins biomolecules and metals These interfaces allow specific sets of cells to communicate based on level of expression and the surrounding microenvironment The anchoring on a GPI raft in the lipid bilayer supports claims of an extracellular scaffolding function 16 Diseases caused by PrP misfolding EditMain article Transmissible spongiform encephalopathy More than 20 mutations in the PRNP gene have been identified in people with inherited prion diseases which include the following 48 49 Creutzfeldt Jakob disease glutamic acid 200 is replaced by lysine while valine is present at amino acid 129 Gerstmann Straussler Scheinker syndrome usually a change in codon 102 from proline to leucine 50 fatal familial insomnia aspartic acid 178 is replaced by asparagine while methionine is present at amino acid 129 51 The conversion of PrPC to PrPSc conformation is the mechanism of transmission of fatal neurodegenerative transmissible spongiform encephalopathies TSE This can arise from genetic factors infection from external source or spontaneously for reasons unknown Accumulation of PrPSc corresponds with progression of neurodegeneration and is the proposed cause Some PRNP mutations lead to a change in single amino acids the building blocks of proteins in the prion protein Others insert additional amino acids into the protein or cause an abnormally short protein to be made These mutations cause the cell to make prion proteins with an abnormal structure The abnormal protein PrPSc accumulates in the brain and destroys nerve cells which leads to the mental and behavioral features of prion diseases Several other changes in the PRNP gene called polymorphisms do not cause prion diseases but may affect a person s risk of developing these diseases or alter the course of the disorders An allele that codes for a PRNP variant G127V provides resistance to kuru 52 In addition some prion diseases can be transmitted from external sources of PrPSc 53 Scrapie fatal neurodegenerative disease in sheep not transmissible to humans Bovine spongiform encephalopathy mad cow disease fatal neurodegenerative disease in cows which can be transmitted to humans by ingestion of brain spinal or digestive tract tissue of an infected cow Kuru TSE in humans transmitted via funerary cannibalism Generally affected family members were given by tradition parts of the central nervous system according to ritual when consuming deceased family members Alzheimer s disease Edit PrPC protein is one of several cellular receptors of soluble amyloid beta Ab oligomers which are canonically implicated in causing Alzheimer s disease 54 These oligomers are composed smaller Ab plaques and are the most damaging to the integrity of a neuron 54 The precise mechanism of soluble Ab oligomers directly inducing neurotoxicity is unknown and experimental deletion of PRNP in animals has yielded several conflicting findings When Ab oligomers were injected into the cerebral ventricles of a mouse model of Alzheimer s PRNP deletion did not offer protection only anti PrPC antibodies prevented long term memory and spatial learning deficits 55 56 This would suggest either an unequal relation between PRNP and Ab oligomer mediated neurodegeneration or a site specific relational significance In the case of direct injection of Ab oligomers into the hippocampus PRNP knockout mice were found to be indistinguishable from control with respect to both neuronal death rates and measurements of synaptic plasticity 54 56 It was further found that Ab oligomers bind to PrPC at the postsynaptic density indirectly overactivating the NMDA receptor via the Fyn enzyme resulting in excitotoxicity 55 Soluble Ab oligomers also bind to PrPC at the dendritic spines forming a complex with Fyn and excessively activating tau another protein implicated in Alzheimer s 55 As the gene FYN codes for the enzyme Fyn FYN knockout mice display neither excitotoxic events nor dendritic spine shrinkage when injected with Ab oligomers 55 In mammals the full functional significance of PRNP remains unclear as PRNP deletion has been prophylactically implemented by the cattle industry without apparent harm 54 In mice this same deletion phenotypically varies between Alzheimer s mouse lines as hAPPJ20 mice and TgCRND8 mice show a slight increase in epileptic activity contributing to conflicting results when examining Alzheimer s survival rates 54 Of note the deletion of PRNP in both APPswe and SEN1dE9 two other transgenic models of Alzheimer s attenuated the epilepsy induced death phenotype seen in a subset of these animals 54 Taken collectively recent evidence suggests PRNP may be important for conducing the neurotoxic effects of soluble Ab oligomers and the emergent disease state of Alzheimer s 54 55 56 In humans the methionine valine polymorphism at codon 129 of PRNP rs1799990 is most closely associated with Alzheimer s disease 57 Variant V allele carriers VV and MV show a 13 decreased risk with respect to developing Alzheimer s compared to the methionine homozygote MM However the protective effects of variant V carriers have been found exclusively in Caucasians The decreased risk in V allele carriers is further limited to late onset Alzheimer s disease only 65 years 57 PRNP can also functionally interact with polymorphisms in two other genes implicated in Alzheimer s PSEN1 and APOE to compound risk for both Alzheimer s and sporadic Creutzfeldt Jakob disease 54 A point mutation on codon 102 of PRNP at least in part contributed to three separate patients atypical frontotemporal dementia within the same family suggesting a new phenotype for Gerstmann Straussler Scheinker syndrome 54 58 The same study proposed sequencing PRNP in cases of ambiguously diagnosed dementia as the various forms of dementia can prove challenging to differentially diagnose 58 Interactions EditA strong interaction exists between PrP and the cochaperone Hop Hsp70 Hsp90 organizing protein also called STI1 Stress induced protein 1 59 60 References Edit a b c GRCh38 Ensembl release 89 ENSG00000171867 Ensembl May 2017 a b c GRCm38 Ensembl release 89 ENSMUSG00000079037 Ensembl May 2017 Human PubMed Reference National Center for Biotechnology Information U S National Library of Medicine Mouse PubMed Reference National Center for Biotechnology Information U S National Library of Medicine Kretzschmar HA Stowring LE Westaway D Stubblebine WH Prusiner SB Dearmond SJ August 1986 Molecular cloning of a human prion protein cDNA DNA 5 4 315 24 doi 10 1089 dna 1986 5 315 PMID 3755672 Sparkes RS Simon M Cohn VH Fournier RE Lem J Klisak I Heinzmann C Blatt C Lucero M Mohandas T October 1986 Assignment of the human and mouse prion protein genes to homologous chromosomes Proc Natl Acad Sci U S A 83 19 7358 62 Bibcode 1986PNAS 83 7358S doi 10 1073 pnas 83 19 7358 PMC 386716 PMID 3094007 Liao YC Lebo RV Clawson GA Smuckler EA July 1986 Human prion protein cDNA molecular cloning chromosomal mapping and biological implications Science 233 4761 364 7 Bibcode 1986Sci 233 364L doi 10 1126 science 3014653 PMID 3014653 Robakis NK Devine Gage EA Jenkins EC Kascsak RJ Brown WT Krawczun MS Silverman WP October 1986 Localization of a human gene homologous to the PrP gene on the p arm of chromosome 20 and detection of PrP related antigens in normal human brain Biochem Biophys Res Commun 140 2 758 65 doi 10 1016 0006 291X 86 90796 5 PMID 2877664 Prusiner SB 2001 Shattuck lecture neurodegenerative diseases and prions N Engl J Med 344 20 1516 26 doi 10 1056 NEJM200105173442006 PMID 11357156 Weissmann C 2004 The state of the prion Nat Rev Microbiol 2 11 861 71 doi 10 1038 nrmicro1025 PMID 15494743 S2CID 20992257 a b Zomosa Signoret V Arnaud JD Fontes P Alvarez Martinez MT Liautard JP 2008 Physiological role of the cellular prion protein PDF Vet Res 39 4 9 doi 10 1051 vetres 2007048 PMID 18073096 Damberger FF Christen B Perez DR Hornemann S Wuthrich K October 2011 Cellular prion protein conformation and function Proc Natl Acad Sci U S A 108 42 17308 13 Bibcode 2011PNAS 10817308D doi 10 1073 pnas 1106325108 PMC 3198368 PMID 21987789 Schatzl HM Da Costa M Taylor L Cohen FE Prusiner SB January 1995 Prion protein gene variation among primates J Mol Biol 245 4 362 74 doi 10 1006 jmbi 1994 0030 PMID 7837269 Chesebro B Trifilo M Race R Meade White K Teng C LaCasse R Raymond L Favara C Baron G Priola S Caughey B Masliah E Oldstone M June 2005 Anchorless prion protein results in infectious amyloid disease without clinical scrapie Science 308 5727 1435 9 Bibcode 2005Sci 308 1435C CiteSeerX 10 1 1 401 781 doi 10 1126 science 1110837 PMID 15933194 S2CID 10064966 Barrette I Poisson G Gendron P Major F 2001 Pseudoknots in prion protein mRNAs confirmed by comparative sequence analysis and pattern searching Nucleic Acids Res 29 3 753 758 doi 10 1093 nar 29 3 753 PMC 30388 PMID 11160898 a b c d e f Linden R Martins VR Prado MA Cammarota M Izquierdo I Brentani RR April 2008 Physiology of the prion protein Physiol Rev 88 2 673 728 doi 10 1152 physrev 00007 2007 PMID 18391177 a b Prcina M Kontsekova E Novak M 2015 Prion protein prevents heavy metals overloading of cells and thus protects them against their toxicity Acta Virol 59 2 179 84 doi 10 4149 av 2015 02 179 PMID 26104335 Brown DR Clive C Haswell SJ January 2001 Antioxidant activity related to copper binding of native prion protein J Neurochem 76 1 69 76 doi 10 1046 j 1471 4159 2001 00009 x PMID 11145979 S2CID 45647133 Kanaani J Prusiner SB Diacovo J Baekkeskov S Legname G December 2005 Recombinant prion protein induces rapid polarization and development of synapses in embryonic rat hippocampal neurons in vitro Journal of Neurochemistry 95 5 1373 86 doi 10 1111 j 1471 4159 2005 03469 x PMID 16313516 S2CID 24329326 Ross CA Poirier MA July 2004 Protein aggregation and neurodegenerative disease Nat Med 10 Suppl 7 S10 7 doi 10 1038 nm1066 PMID 15272267 S2CID 205383483 Pan KM Baldwin M Nguyen J Gasset M Serban A Groth D Mehlhorn I Huang Z Fletterick RJ Cohen FE December 1993 Conversion of alpha helices into beta sheets features in the formation of the scrapie prion proteins Proc Natl Acad Sci U S A 90 23 10962 6 Bibcode 1993PNAS 9010962P doi 10 1073 pnas 90 23 10962 PMC 47901 PMID 7902575 Baskakov Ilia V Caughey Byron Requena Jesus R Sevillano Alejandro M Surewicz Witold K Wille Holger 2019 01 01 The prion 2018 round tables I the structure of PrPSc Prion 13 1 46 52 doi 10 1080 19336896 2019 1569450 ISSN 1933 6896 PMC 6422368 PMID 30646817 Sandberg MK Al Doujaily H Sharps B Clarke AR Collinge J February 2011 Prion propagation and toxicity in vivo occur in two distinct mechanistic phases Nature 470 7335 540 2 Bibcode 2011Natur 470 540S doi 10 1038 nature09768 PMID 21350487 S2CID 4399936 Bueler H Aguzzi A Sailer A Greiner RA Autenried P Aguet M Weissmann C July 1993 Mice devoid of PrP are resistant to scrapie Cell 73 7 1339 47 doi 10 1016 0092 8674 93 90360 3 PMID 8100741 a b Aguzzi A Baumann F Bremer J 2008 The prion s elusive reason for being Annu Rev Neurosci 31 439 77 doi 10 1146 annurev neuro 31 060407 125620 PMID 18558863 Atkinson M October 2001 National scrapie plan The Veterinary Record 149 15 462 PMID 11688751 Weissmann C Flechsig E 2003 PrP knock out and PrP transgenic mice in prion research Br Med Bull 66 43 60 doi 10 1093 bmb 66 1 43 PMID 14522848 Katamine S Nishida N Sugimoto T Noda T Sakaguchi S Shigematsu K Kataoka Y Nakatani A Hasegawa S Moriuchi R Miyamoto T December 1998 Impaired motor coordination in mice lacking prion protein Cell Mol Neurobiol 18 6 731 42 doi 10 1023 A 1020234321879 PMID 9876879 S2CID 23409873 Nico PB de Paris F Vinade 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 July 2005 Altered behavioural response to acute stress in mice lacking cellular prion protein Behav Brain Res 162 2 173 81 doi 10 1016 j bbr 2005 02 003 PMID 15970215 S2CID 37511702 Roesler R Walz R Quevedo J de Paris F Zanata SM Graner E Izquierdo I Martins VR Brentani RR August 1999 Normal inhibitory avoidance learning and anxiety but increased locomotor activity in mice devoid of PrP C Brain Res Mol Brain Res 71 2 349 53 doi 10 1016 S0169 328X 99 00193 X PMID 10521590 Medori R Tritschler HJ LeBlanc A Villare F Manetto V Chen HY Xue R Leal S Montagna P Cortelli P February 1992 Fatal familial insomnia a prion disease with a mutation at codon 178 of the prion protein gene N Engl J Med 326 7 444 9 doi 10 1056 NEJM199202133260704 PMC 6151859 PMID 1346338 Cagampang FR Whatley SA Mitchell AL Powell JF Campbell IC Coen CW 1999 Circadian regulation of prion protein messenger RNA in the rat forebrain a widespread and synchronous rhythm Neuroscience 91 4 1201 4 doi 10 1016 S0306 4522 99 00092 5 PMID 10391428 S2CID 42892475 Criado JR Sanchez Alavez M Conti B Giacchino JL Wills DN Henriksen SJ Race R Manson JC Chesebro B Oldstone MB 2005 Mice devoid of prion protein have cognitive deficits that are rescued by reconstitution of PrP in neurons Neurobiol Dis 19 1 2 255 65 doi 10 1016 j nbd 2005 01 001 PMID 15837581 S2CID 2618712 Balducci C Beeg M Stravalaci M Bastone A Sclip A Biasini E Tapella L Colombo L Manzoni C Borsello T Chiesa R Gobbi M Salmona M Forloni G February 2010 Synthetic amyloid beta oligomers impair long term memory independently of cellular prion protein Proc Natl Acad Sci U S A 107 5 2295 300 Bibcode 2010PNAS 107 2295B doi 10 1073 pnas 0911829107 PMC 2836680 PMID 20133875 Coitinho AS Freitas AR Lopes MH Hajj GN Roesler R Walz R Rossato JI Cammarota M Izquierdo I Martins VR Brentani RR December 2006 The interaction between prion protein and laminin modulates memory consolidation Eur J Neurosci 24 11 3255 64 doi 10 1111 j 1460 9568 2006 05156 x PMID 17156386 S2CID 17164351 Shorter J Lindquist S June 2005 Prions as adaptive conduits of memory and inheritance Nat Rev Genet 6 6 435 50 doi 10 1038 nrg1616 PMID 15931169 S2CID 5575951 Papassotiropoulos A Wollmer MA Aguzzi A Hock C Nitsch RM de Quervain DJ August 2005 The prion gene is associated with human long term memory PDF Hum Mol Genet 14 15 2241 6 doi 10 1093 hmg ddi228 PMID 15987701 Del Bo R Comi GP Giorda R Crimi M Locatelli F Martinelli Boneschi F Pozzoli U Castelli E Bresolin N Scarlato G June 2003 The 129 codon polymorphism of the prion protein gene influences earlier cognitive performance in Down syndrome subjects J Neurol 250 6 688 92 doi 10 1007 s00415 003 1057 5 PMID 12796830 S2CID 21049364 Berr C Richard F Dufouil C Amant C Alperovitch A Amouyel P September 1998 Polymorphism of the prion protein is associated with cognitive impairment in the elderly the EVA study Neurology 51 3 734 7 doi 10 1212 wnl 51 3 734 PMID 9748018 S2CID 11352163 Croes EA Dermaut B Houwing Duistermaat JJ Van den Broeck M Cruts M Breteler MM Hofman A van Broeckhoven C van Duijn CM August 2003 Early cognitive decline is associated with prion protein codon 129 polymorphism Ann Neurol 54 2 275 6 doi 10 1002 ana 10658 PMID 12891686 S2CID 31538672 Kachiwala SJ Harris SE Wright AF Hayward C Starr JM Whalley LJ Deary IJ September 2005 Genetic influences on oxidative stress and their association with normal cognitive ageing Neurosci Lett 386 2 116 20 doi 10 1016 j neulet 2005 05 067 PMID 16023289 S2CID 23642220 Herms J Tings T Gall S Madlung A Giese A Siebert H Schurmann P Windl O Brose N Kretzschmar H October 1999 Evidence of presynaptic location and function of the prion protein J Neurosci 19 20 8866 75 doi 10 1523 JNEUROSCI 19 20 08866 1999 PMC 6782778 PMID 10516306 Kardos J Kovacs I Hajos F Kalman M Simonyi M August 1989 Nerve endings from rat brain tissue release copper upon depolarization A possible role in regulating neuronal excitability Neurosci Lett 103 2 139 44 doi 10 1016 0304 3940 89 90565 X PMID 2549468 S2CID 24917999 Bailey CH Kandel ER Si K September 2004 The persistence of long term memory a molecular approach to self sustaining changes in learning induced synaptic growth Neuron 44 1 49 57 doi 10 1016 j neuron 2004 09 017 PMID 15450159 S2CID 2637074 Barco A Bailey CH Kandel ER June 2006 Common molecular mechanisms in explicit and implicit memory J Neurochem 97 6 1520 33 doi 10 1111 j 1471 4159 2006 03870 x PMID 16805766 S2CID 26307975 Lauren J Gimbel DA Nygaard HB Gilbert JW Strittmatter SM February 2009 Cellular prion protein mediates impairment of synaptic plasticity by amyloid beta oligomers Nature 457 7233 1128 32 Bibcode 2009Natur 457 1128L doi 10 1038 nature07761 PMC 2748841 PMID 19242475 Isaacs JD Jackson GS Altmann DM October 2006 The role of the cellular prion protein in the immune system Clin Exp Immunol 146 1 1 8 doi 10 1111 j 1365 2249 2006 03194 x PMC 1809729 PMID 16968391 Castilla J Hetz C Soto C 2004 Molecular mechanisms of neurotoxicity of pathological prion protein Curr Mol Med 4 4 397 403 doi 10 2174 1566524043360654 PMID 15354870 Kovacs GG Trabattoni G Hainfellner JA Ironside JW Knight RS Budka H 2002 Mutations of the prion protein gene phenotypic spectrum J Neurol 249 11 1567 82 doi 10 1007 s00415 002 0896 9 PMID 12420099 S2CID 22688729 Collins S McLean CA Masters CL 2001 Gerstmann Straussler Scheinker syndrome fatal familial insomnia and kuru a review of these less common human transmissible spongiform encephalopathies J Clin Neurosci 8 5 387 97 doi 10 1054 jocn 2001 0919 PMID 11535002 S2CID 31976428 Montagna P Gambetti P Cortelli P Lugaresi E 2003 Familial and sporadic fatal insomnia Lancet Neurol 2 3 167 76 doi 10 1016 S1474 4422 03 00323 5 PMID 12849238 S2CID 20822956 Mead S Whitfield J Poulter M Shah P Uphill J Campbell T Al Dujaily H Hummerich H Beck J Mein CA Verzilli C Whittaker J Alpers MP Collinge J 2009 A Novel Protective Prion Protein Variant that Colocalizes with Kuru Exposure PDF The New England Journal of Medicine 361 21 2056 2065 doi 10 1056 NEJMoa0809716 PMID 19923577 Brain disease resistance gene evolves in Papua New Guinea community could offer insights into CJD ScienceDaily Press release November 21 2009 Hwang D Lee IY Yoo H Gehlenborg N Cho JH Petritis B Baxter D Pitstick R Young R Spicer D Price ND Hohmann JG Dearmond SJ Carlson GA Hood LE 2009 A systems approach to prion disease Mol Syst Biol 5 1 252 doi 10 1038 msb 2009 10 PMC 2671916 PMID 19308092 a b c d e f g h i Lauren J 2014 Cellular prion protein as a therapeutic target in Alzheimer s disease Journal of Alzheimer s Disease 38 2 227 244 doi 10 3233 JAD 130950 PMID 23948943 a b c d e Zhou J Liu B May 2013 Alzheimer s disease and prion protein Intractable amp Rare Diseases Research 2 2 35 44 doi 10 5582 irdr 2013 v2 2 35 PMC 4204584 PMID 25343100 a b c Lauren J Gimbel DA Nygaard HB Gilbert JW Strittmatter SM Feb 2009 Cellular prion protein mediates impairment of synaptic plasticity by amyloid beta oligomers Nature 457 7233 1128 1132 Bibcode 2009Natur 457 1128L doi 10 1038 nature07761 PMC 2748841 PMID 19242475 a b He J Li X Yang J Huang J Fu X Zhang Y Fan H Mar 2013 The association between the methionine valine M V polymorphism rs1799990 in the PRNP gene and the risk of Alzheimer disease an update by meta analysis Journal of the Neurological Sciences 326 1 2 89 95 doi 10 1016 j jns 2013 01 020 PMID 23399523 S2CID 31070331 a b Giovagnoli AR Di Fede G Aresi A Reati F Rossi G Tagliavini F December 2008 Atypical frontotemporal dementia as a new clinical phenotype of Gerstmann Straussler Scheinker disease with the PrP P102L mutation Description of a previously unreported Italian family Neurological Sciences 29 6 405 10 doi 10 1007 s10072 008 1025 z PMID 19030774 S2CID 20553167 Americo TA Chiarini LB Linden R June 2007 Signaling induced by hop STI 1 depends on endocytosis Biochem Biophys Res Commun 358 2 620 5 doi 10 1016 j bbrc 2007 04 202 PMID 17498662 Zanata SM Lopes MH Mercadante AF Hajj GN Chiarini LB Nomizo R Freitas AR Cabral AL Lee KS Juliano MA de Oliveira E Jachieri SG Burlingame A Huang L Linden R Brentani RR Martins VR Jul 2002 Stress inducible protein 1 is a cell surface ligand for cellular prion that triggers neuroprotection EMBO J 21 13 3307 16 doi 10 1093 emboj cdf325 PMC 125391 PMID 12093732 External links EditPRNP PrP gene at GeneCard PRNP protein human at the US National Library of Medicine Medical Subject Headings MeSH Susan Lindquist s Seminar The Surprising World of Prion Biology Retrieved from https en wikipedia org w index php title PRNP amp oldid 1132119039, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.