Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cell Tissue Res ; 392(1): 81-96, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36070155

RESUMO

Prion diseases are devastating neurodegenerative diseases caused by the structural conversion of the normally benign prion protein (PrPC) to an infectious, disease-associated, conformer, PrPSc. After decades of intense research, much is known about the self-templated prion conversion process, a phenomenon which is now understood to be operative in other more common neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. In this review, we provide the current state of knowledge concerning a relatively poorly understood aspect of prion diseases: mechanisms of neurotoxicity. We provide an overview of proposed functions of PrPC and its interactions with other extracellular proteins in the central nervous system, in vivo and in vitro models used to delineate signaling events downstream of prion propagation, the application of omics technologies, and the emerging appreciation of the role played by non-neuronal cell types in pathogenesis.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Doenças Priônicas , Príons , Humanos , Príons/metabolismo , Doenças Priônicas/metabolismo , Doenças Priônicas/patologia , Proteínas Priônicas
2.
PLoS Pathog ; 14(9): e1007283, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30235355

RESUMO

Synaptic degeneration is one of the earliest pathological correlates of prion disease, and it is a major determinant of the progression of clinical symptoms. However, the cellular and molecular mechanisms underlying prion synaptotoxicity are poorly understood. Previously, we described an experimental system in which treatment of cultured hippocampal neurons with purified PrPSc, the infectious form of the prion protein, induces rapid retraction of dendritic spines, an effect that is entirely dependent on expression of endogenous PrPC by the target neurons. Here, we use this system to dissect pharmacologically the underlying cellular and molecular mechanisms. We show that PrPSc initiates a stepwise synaptotoxic signaling cascade that includes activation of NMDA receptors, calcium influx, stimulation of p38 MAPK and several downstream kinases, and collapse of the actin cytoskeleton within dendritic spines. Synaptic degeneration is restricted to excitatory synapses, spares presynaptic structures, and results in decrements in functional synaptic transmission. Pharmacological inhibition of any one of the steps in the signaling cascade, as well as expression of a dominant-negative form of p38 MAPK, block PrPSc-induced spine degeneration. Moreover, p38 MAPK inhibitors actually reverse the degenerative process after it has already begun. We also show that, while PrPC mediates the synaptotoxic effects of both PrPSc and the Alzheimer's Aß peptide in this system, the two species activate distinct signaling pathways. Taken together, our results provide powerful insights into the biology of prion neurotoxicity, they identify new, druggable therapeutic targets, and they allow comparison of prion synaptotoxic pathways with those involved in other neurodegenerative diseases.


Assuntos
Príons/metabolismo , Príons/patogenicidade , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Sinalização do Cálcio , Células Cultivadas , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Potenciais Pós-Sinápticos Excitadores , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/metabolismo , Neurônios/patologia , Proteínas PrPSc/genética , Proteínas PrPSc/metabolismo , Proteínas PrPSc/patogenicidade , Doenças Priônicas/metabolismo , Doenças Priônicas/patologia , Príons/genética , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
3.
PLoS Pathog ; 14(1): e1006826, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29338055

RESUMO

To explore pathogenesis in a young Gerstmann-Sträussler-Scheinker Disease (GSS) patient, the corresponding mutation, an eight-residue duplication in the hydrophobic region (HR), was inserted into the wild type mouse PrP gene. Transgenic (Tg) mouse lines expressing this mutation (Tg.HRdup) developed spontaneous neurologic syndromes and brain extracts hastened disease in low-expressor Tg.HRdup mice, suggesting de novo formation of prions. While Tg.HRdup mice exhibited spongiform change, PrP aggregates and the anticipated GSS hallmark of a proteinase K (PK)-resistant 8 kDa fragment deriving from the center of PrP, the LGGLGGYV insertion also imparted alterations in PrP's unstructured N-terminus, resulting in a 16 kDa species following thermolysin exposure. This species comprises a plausible precursor to the 8 kDa PK-resistant fragment and its detection in adolescent Tg.HRdup mice suggests that an early start to accumulation could account for early disease of the index case. A 16 kDa thermolysin-resistant signature was also found in GSS patients with P102L, A117V, H187R and F198S alleles and has coordinates similar to GSS stop codon mutations. Our data suggest a novel shared pathway of GSS pathogenesis that is fundamentally distinct from that producing structural alterations in the C-terminus of PrP, as observed in other prion diseases such as Creutzfeldt-Jakob Disease and scrapie.


Assuntos
Doença de Gerstmann-Straussler-Scheinker/genética , Mutação , Proteínas PrPSc/química , Proteínas PrPSc/genética , Doenças Priônicas/genética , Adulto , Alelos , Sequência de Aminoácidos , Animais , Humanos , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fragmentos de Peptídeos/genética , Proteínas PrPSc/metabolismo , Domínios Proteicos/genética , Precursores de Proteínas/química , Precursores de Proteínas/genética
4.
J Biol Chem ; 288(52): 37241-55, 2013 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-24225951

RESUMO

Widely expressed in the adult central nervous system, the cellular prion protein (PrP(C)) is implicated in a variety of processes, including neuronal excitability. Dipeptidyl aminopeptidase-like protein 6 (DPP6) was first identified as a PrP(C) interactor using in vivo formaldehyde cross-linking of wild type (WT) mouse brain. This finding was confirmed in three cell lines and, because DPP6 directs the functional assembly of K(+) channels, we assessed the impact of WT and mutant PrP(C) upon Kv4.2-based cell surface macromolecular complexes. Whereas a Gerstmann-Sträussler-Scheinker disease version of PrP with eight extra octarepeats was a loss of function both for complex formation and for modulation of Kv4.2 channels, WT PrP(C), in a DPP6-dependent manner, modulated Kv4.2 channel properties, causing an increase in peak amplitude, a rightward shift of the voltage-dependent steady-state inactivation curve, a slower inactivation, and a faster recovery from steady-state inactivation. Thus, the net impact of wt PrP(C) was one of enhancement, which plays a critical role in the down-regulation of neuronal membrane excitability and is associated with a decreased susceptibility to seizures. Insofar as previous work has established a requirement for WT PrP(C) in the Aß-dependent modulation of excitability in cholinergic basal forebrain neurons, our findings implicate PrP(C) regulation of Kv4.2 channels as a mechanism contributing to the effects of oligomeric Aß upon neuronal excitability and viability.


Assuntos
Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Canais de Potássio/metabolismo , Proteínas PrPC/metabolismo , Prosencéfalo/metabolismo , Canais de Potássio Shal/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Membrana Celular/genética , Membrana Celular/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Células HEK293 , Humanos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Mutantes , Mutação , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Canais de Potássio/genética , Proteínas PrPC/genética , Prosencéfalo/citologia , Canais de Potássio Shal/genética
5.
ACS Chem Neurosci ; 15(11): 2265-2282, 2024 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-38743607

RESUMO

Prion diseases are invariably fatal neurodegenerative diseases of humans and other animals for which there are no effective treatment options. Previous work from our laboratory identified phenethylpiperidines as a novel class of anti-prion compounds. While working to identify the molecular target(s) of these molecules, we unexpectedly discovered ten novel antiprion compounds based on their known ability to bind to the sigma receptors, σ1R and σ2R, which are currently being tested as therapeutic or diagnostic targets for cancer and neuropsychiatric disorders. Surprisingly, however, knockout of the respective genes encoding σ1R and σ2R (Sigmar1 and Tmem97) in prion-infected N2a cells did not alter the antiprion activity of these compounds, demonstrating that these receptors are not the direct targets responsible for the antiprion effects of their ligands. Further investigation of the most potent molecules established that they are efficacious against multiple prion strains and protect against downstream prion-mediated synaptotoxicity. While the precise details of the mechanism of action of these molecules remain to be determined, the present work forms the basis for further investigation of these compounds in preclinical studies. Given the therapeutic utility of several of the tested compounds, including rimcazole and haloperidol for neuropsychiatric conditions, (+)-pentazocine for neuropathic pain, and the ongoing clinical trials of SA 4503 and ANAVEX2-73 for ischemic stroke and Alzheimer's disease, respectively, this work has immediate implications for the treatment of human prion disease.


Assuntos
Doenças Priônicas , Receptores sigma , Receptores sigma/metabolismo , Receptores sigma/efeitos dos fármacos , Animais , Ligantes , Doenças Priônicas/tratamento farmacológico , Doenças Priônicas/metabolismo , Camundongos , Humanos , Príons/efeitos dos fármacos , Príons/metabolismo , Receptor Sigma-1 , Linhagem Celular Tumoral
6.
bioRxiv ; 2023 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-38077011

RESUMO

Prion diseases are invariably fatal neurodegenerative diseases of humans and other animals for which there are no treatment options. Previous work from our laboratory identified phenethyl piperidines as novel class of anti-prion compounds. While working to identify the molecular target(s) of these molecules, we unexpectedly discovered ten novel anti-prion compounds based on their known ability to bind to the sigma receptors, σ 1 R and 2 R, which are currently being tested as therapeutic or diagnostic targets for cancer and neuropsychiatric disorders. Surprisingly, however, knockout of the respective genes encoding σ 1 R and σ 2 R ( Sigmar1 and Tmem97 ), in prion infected N2a cells did not alter the anti-prion activity of these compounds, demonstrating that these receptors are not the direct targets responsible the anti-prion effects of their ligands. Further investigation of the most potent molecules established that they are efficacious against multiple prion strains and protect against downstream prion-mediated synaptotoxicity. While the precise details of the mechanism of action of these molecules remains to be determined, the present work forms the basis for further investigations of these compounds in pre-clinical studies. Given the therapeutic utility of several of the tested compounds, including rimcazole and haloperidol for neuropsychiatric conditions, (+)-pentazocine for neuropathic pain, and the ongoing clinical trials of SA 4503 and ANAVEX2-73 for ischemic stroke and Alzheimer's disease, respectively, this work has immediate implications for the treatment of human prion disease.

7.
Biochim Biophys Acta ; 1814(10): 1253-60, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21722757

RESUMO

The existence of a unique sarcomeric actin is demonstrated in teleosts that possess substantial amounts of slow skeletal muscle in the trunk. The slow skeletal isotype is conserved. There is one amino acid substitution between Atlantic herring slow skeletal actin and the equivalent in salmonids. Conversely, the intra-species variation is considerable; 13 substitutions between different herring skeletal isotypes (slow versus fast). The isomorphisms (non-conservative underlined: residues, 2, 3, 103, 155, 160, 165, 278, 281, 310, 329, 358, 360 and 363) are restricted to sub-domains 1 and 3 and include the substitution Asp-360 in 'slow' to Gln in 'fast' which results in an electrophoretic shift at alkaline pH. The musculature of the trunk facilitates the preparation of isoactins for biochemical study. Herring slow skeletal G-actin (Ca.ATP) is more susceptible to thermal, and urea, -induced denaturation and subtilisin cleavage than that in fast skeletal, but more stable than the counterpart in salmonids (one substitution, Gln354Ala) highlighting the critical nature of actin's carboxyl-terminal insert. Fluorescent spectra of G-actin isoforms containing the isomorphism Ser155Ala in complexation with 2'-deoxy 3' O-(N'-Methylanthraniloyl) ATP infer similar polarity of the nucleotide binding cleft. An electrophoretic survey detected two skeletal actins in some (smelt and mackerel) but not all teleosts. One skeletal muscle actin was detected in frog and bird.


Assuntos
Actinas/química , Actinas/metabolismo , Fibras Musculares de Contração Lenta/metabolismo , Vertebrados/metabolismo , Sequência de Aminoácidos , Animais , Sequência Conservada , Peixes/metabolismo , Modelos Moleculares , Músculo Esquelético/metabolismo , Perciformes/metabolismo , Maleabilidade , Conformação Proteica , Dobramento de Proteína , Isoformas de Proteínas/análise , Isoformas de Proteínas/metabolismo , Distribuição Tecidual
8.
EMBO Mol Med ; 7(3): 339-56, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25661904

RESUMO

The cellular prion protein (PrP(C)) comprises a natively unstructured N-terminal domain, including a metal-binding octarepeat region (OR) and a linker, followed by a C-terminal domain that misfolds to form PrP(S) (c) in Creutzfeldt-Jakob disease. PrP(C) ß-endoproteolysis to the C2 fragment allows PrP(S) (c) formation, while α-endoproteolysis blocks production. To examine the OR, we used structure-directed design to make novel alleles, 'S1' and 'S3', locking this region in extended or compact conformations, respectively. S1 and S3 PrP resembled WT PrP in supporting peripheral nerve myelination. Prion-infected S1 and S3 transgenic mice both accumulated similar low levels of PrP(S) (c) and infectious prion particles, but differed in their clinical presentation. Unexpectedly, S3 PrP overproduced C2 fragment in the brain by a mechanism distinct from metal-catalysed hydrolysis reported previously. OR flexibility is concluded to impact diverse biological endpoints; it is a salient variable in infectious disease paradigms and modulates how the levels of PrP(S) (c) and infectivity can either uncouple or engage to drive the onset of clinical disease.


Assuntos
Proteínas PrPC/química , Proteínas PrPC/metabolismo , Doenças Priônicas/patologia , Doenças Priônicas/fisiopatologia , Processamento de Proteína Pós-Traducional , Animais , Linhagem Celular , Análise Mutacional de DNA , Modelos Animais de Doenças , Histocitoquímica , Humanos , Camundongos Transgênicos , Microscopia , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Conformação Proteica , Proteólise
9.
FEBS J ; 281(3): 862-76, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24286250

RESUMO

Cellular prion protein (PrP(C)) misfolds to form infectivity-associated scrapie prion protein and generates C-terminal fragments C1 and C2 in healthy and prion-infected animals. C1 cleavage occurs N-terminally of PrP(C)'s hydrophobic domain, whereas the larger C2 fragment is generated by cleavage at the end of the octarepeat region. As the PrP-like proteins Doppel and Shadoo (Sho) have been reported to inhabit similar membrane environments as PrP(C), we investigated endoproteolysis by using a panel of mutant alleles. Doppel undergoes efficient in vivo cleavage at a C1 site mapped to the start of the globular domain, which is a structurally similar cleavage site to that in PrP(C). Sho is processed to C1 and C2 fragments, and proved refractory to mutagenesis to inactivate C1 cleavage. As a reciprocal product of C1 cleavage, Sho also engenders a metabolically stable N1 fragment with a C-terminus after its hydrophobic domain, an observation that may account for N1's association with membrane and/or cellular fractions in vitro and in vivo. Our data indicate that glycosylation status and yet to be identified proteases modulate internal C1 and C2 proteolysis events within the mammalian prion protein family.


Assuntos
Endopeptidases/metabolismo , Glicoproteínas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas PrPC/metabolismo , Príons/metabolismo , Animais , Encéfalo/enzimologia , Encéfalo/metabolismo , Linhagem Celular , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Glicoproteínas/química , Glicoproteínas/genética , Glicosilação , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Neurônios/enzimologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteínas PrPC/química , Proteínas PrPC/genética , Príons/química , Príons/genética , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteólise , Coelhos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Testículo/enzimologia , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA