Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Neurobiol Dis ; 179: 106050, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36809847

RESUMEN

Effective therapies are urgently needed to safely target TDP-43 pathology as it is closely associated with the onset and development of devastating diseases such as frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP) and amyotrophic lateral sclerosis (ALS). In addition, TDP-43 pathology is present as a co-pathology in other neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Our approach is to develop a TDP-43-specific immunotherapy that exploits Fc gamma-mediated removal mechanisms to limit neuronal damage while maintaining physiological TDP-43 function. Thus, using both in vitro mechanistic studies in conjunction with the rNLS8 and CamKIIa inoculation mouse models of TDP-43 proteinopathy, we identified the key targeting domain in TDP-43 to accomplish these therapeutic objectives. Targeting the C-terminal domain of TDP-43 but not the RNA recognition motifs (RRM) reduces TDP-43 pathology and avoids neuronal loss in vivo. We demonstrate that this rescue is dependent on Fc receptor-mediated immune complex uptake by microglia. Furthermore, monoclonal antibody (mAb) treatment enhances phagocytic capacity of ALS patient-derived microglia, providing a mechanism to restore the compromised phagocytic function in ALS and FTD patients. Importantly, these beneficial effects are achieved while preserving physiological TDP-43 activity. Our findings demonstrate that a mAb targeting the C-terminal domain of TDP-43 limits pathology and neurotoxicity, enabling clearance of misfolded TDP-43 through microglia engagement, and supporting the clinical strategy to target TDP-43 by immunotherapy. SIGNIFICANCE STATEMENT: TDP-43 pathology is associated with various devastating neurodegenerative disorders with high unmet medical needs such as frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS) and Alzheimer's disease. Thus, safely and effectively targeting pathological TDP-43 represents a key paradigm for biotechnical research as currently there is little in clinical development. After years of research, we have determined that targeting the C-terminal domain of TDP-43 rescues multiple patho-mechanisms involved in disease progression in two animal models of FTD/ALS. In parallel, importantly, our studies establish that this approach does not alter the physiological functions of this ubiquitously expressed and indispensable protein. Together, our findings substantially contribute to the understanding of TDP-43 pathobiology and support the prioritization for clinical testing of immunotherapy approaches targeting TDP-43.


Asunto(s)
Enfermedad de Alzheimer , Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Enfermedad de Pick , Ratones , Animales , Esclerosis Amiotrófica Lateral/genética , Demencia Frontotemporal/genética , Enfermedad de Alzheimer/genética , Neuroprotección , Proteínas de Unión al ADN/metabolismo , Inmunoterapia
2.
Proc Natl Acad Sci U S A ; 115(4): E782-E791, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311311

RESUMEN

Point mutations in the amyloid-ß (Aß) coding region produce a combination of mutant and WT Aß isoforms that yield unique clinicopathologies in familial Alzheimer's disease (fAD) and cerebral amyloid angiopathy (fCAA) patients. Here, we report a method to investigate the structural variability of amyloid deposits found in fAD, fCAA, and sporadic AD (sAD). Using this approach, we demonstrate that mutant Aß determines WT Aß conformation through prion template-directed misfolding. Using principal component analysis of multiple structure-sensitive fluorescent amyloid-binding dyes, we assessed the conformational variability of Aß deposits in fAD, fCAA, and sAD patients. Comparing many deposits from a given patient with the overall population, we found that intrapatient variability is much lower than interpatient variability for both disease types. In a given brain, we observed one or two structurally distinct forms. When two forms coexist, they segregate between the parenchyma and cerebrovasculature, particularly in fAD patients. Compared with sAD samples, deposits from fAD patients show less intersubject variability, and little overlap exists between fAD and sAD deposits. Finally, we examined whether E22G (Arctic) or E22Q (Dutch) mutants direct the misfolding of WT Aß, leading to fAD-like plaques in vivo. Intracerebrally injecting mutant Aß40 fibrils into transgenic mice expressing only WT Aß induced the deposition of plaques with many biochemical hallmarks of fAD. Thus, mutant Aß40 prions induce a conformation of WT Aß similar to that found in fAD deposits. These findings indicate that diverse AD phenotypes likely arise from one or more initial Aß prion conformations, which kinetically dominate the spread of prions in the brain.


Asunto(s)
Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/metabolismo , Conformación Proteica , Pliegue de Proteína , Péptidos beta-Amiloides/genética , Animales , Ratones Transgénicos , Mutación Puntual
3.
Proc Natl Acad Sci U S A ; 114(24): 6191-6196, 2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28566494

RESUMEN

Throughout biology, amyloids are key structures in both functional proteins and the end product of pathologic protein misfolding. Amyloids might also represent an early precursor in the evolution of life because of their small molecular size and their ability to self-purify and catalyze chemical reactions. They also provide attractive backbones for advanced materials. When ß-strands of an amyloid are arranged parallel and in register, side chains from the same position of each chain align, facilitating metal chelation when the residues are good ligands such as histidine. High-resolution structures of metalloamyloids are needed to understand the molecular bases of metal-amyloid interactions. Here we combine solid-state NMR and structural bioinformatics to determine the structure of a zinc-bound metalloamyloid that catalyzes ester hydrolysis. The peptide forms amphiphilic parallel ß-sheets that assemble into stacked bilayers with alternating hydrophobic and polar interfaces. The hydrophobic interface is stabilized by apolar side chains from adjacent sheets, whereas the hydrated polar interface houses the Zn2+-binding histidines with binding geometries unusual in proteins. Each Zn2+ has two bis-coordinated histidine ligands, which bridge adjacent strands to form an infinite metal-ligand chain along the fibril axis. A third histidine completes the protein ligand environment, leaving a free site on the Zn2+ for water activation. This structure defines a class of materials, which we call metal-peptide frameworks. The structure reveals a delicate interplay through which metal ions stabilize the amyloid structure, which in turn shapes the ligand geometry and catalytic reactivity of Zn2.


Asunto(s)
Amiloide/química , Espectroscopía de Resonancia Magnética/métodos , Zinc/química , Amiloide/metabolismo , Sitios de Unión , Biología Computacional , Histidina/química , Histidina/metabolismo , Metaloproteínas , Modelos Moleculares , Agua/química , Zinc/metabolismo
4.
Biopolymers ; 109(8): e23096, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29319162

RESUMEN

The hydrophobic Aß peptide is highly aggregation prone; it first forms soluble oligomers, which then convert into the amyloid fibrils found in the cerebral plaques of Alzheimer's disease. It is generally understood that as the peptide concentration of Aß increases, the fibrillization process is accelerated, but we examine the limits on this phenomenon. We found that once a threshold concentration of Aß is exceeded, a stable oligomer is formed at the expense of fibril formation. The suppression of fibril formation was observed by amyloid-binding dye Thioflavin T and solution nuclear magnetic resonance (NMR). Small-angle X-ray scattering, size exclusion chromatography, and analytical ultracentrifugation demonstrated that Aß peptides form a range of compact species, with a dimer being an early highly populated oligomer. Solution NMR allowed us to define the secondary structure of this Aß dimer, which shows interlocking contacts between C-terminal peptide strands. Thus, we present a novel Aß oligomer that resists conversion to fibrils and remains stable for more than one year.


Asunto(s)
Péptidos beta-Amiloides/química , Benzotiazoles/química , Resonancia Magnética Nuclear Biomolecular , Fragmentos de Péptidos/química , Agregado de Proteínas , Humanos , Estabilidad Proteica
5.
Proc Natl Acad Sci U S A ; 112(35): E4949-58, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26286986

RESUMEN

Increasingly, evidence argues that many neurodegenerative diseases, including progressive supranuclear palsy (PSP), are caused by prions, which are alternatively folded proteins undergoing self-propagation. In earlier studies, PSP prions were detected by infecting human embryonic kidney (HEK) cells expressing a tau fragment [TauRD(LM)] fused to yellow fluorescent protein (YFP). Here, we report on an improved bioassay using selective precipitation of tau prions from human PSP brain homogenates before infection of the HEK cells. Tau prions were measured by counting the number of cells with TauRD(LM)-YFP aggregates using confocal fluorescence microscopy. In parallel studies, we fused α-synuclein to YFP to bioassay α-synuclein prions in the brains of patients who died of multiple system atrophy (MSA). Previously, MSA prion detection required ∼120 d for transmission into transgenic mice, whereas our cultured cell assay needed only 4 d. Variation in MSA prion levels in four different brain regions from three patients provided evidence for three different MSA prion strains. Attempts to demonstrate α-synuclein prions in brain homogenates from Parkinson's disease patients were unsuccessful, identifying an important biological difference between the two synucleinopathies. Partial purification of tau and α-synuclein prions facilitated measuring the levels of these protein pathogens in human brains. Our studies should facilitate investigations of the pathogenesis of both tau and α-synuclein prion disorders as well as help decipher the basic biology of those prions that attack the CNS.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Priones/metabolismo , alfa-Sinucleína/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Enfermedades Neurodegenerativas/patología
6.
Proc Natl Acad Sci U S A ; 111(28): 10329-34, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24982137

RESUMEN

An increasing number of studies continue to show that the amyloid ß (Aß) peptide adopts an alternative conformation and acquires transmissibility; hence, it becomes a prion. Here, we report on the attributes of two strains of Aß prions formed from synthetic Aß peptides composed of either 40 or 42 residues. Modifying the conditions for Aß polymerization increased both the protease resistance and prion infectivity compared with an earlier study. Approximately 150 d after intracerebral inoculation, both synthetic Aß40 and Aß42 prions produced a sustained rise in the bioluminescence imaging signal in the brains of bigenic Tg(APP23:Gfap-luc) mice, indicative of astrocytic gliosis. Pathological investigations showed that synthetic Aß40 prions produced amyloid plaques containing both Aß40 and Aß42 in the brains of inoculated bigenic mice, whereas synthetic Aß42 prions stimulated the formation of smaller, more numerous plaques composed predominantly of Aß42. Synthetic Aß40 preparations consisted of long straight fibrils; in contrast, the Aß42 fibrils were much shorter. Addition of 3.47 mM (0.1%) SDS to the polymerization reaction produced Aß42 fibrils that were indistinguishable from Aß40 fibrils produced in the absence or presence of SDS. Moreover, the Aß amyloid plaques in the brains of bigenic mice inoculated with Aß42 prions prepared in the presence of SDS were similar to those found in mice that received Aß40 prions. From these results, we conclude that the composition of Aß plaques depends on the conformation of the inoculated Aß polymers, and thus, these inocula represent distinct synthetic Aß prion strains.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacología , Encéfalo/metabolismo , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Priones , Animales , Humanos , Ratones Transgénicos , Factores de Tiempo
7.
Proc Natl Acad Sci U S A ; 111(28): 10323-8, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24982139

RESUMEN

An increasing number of studies argues that self-propagating protein conformations (i.e., prions) feature in the pathogenesis of several common neurodegenerative diseases. Mounting evidence contends that aggregates of the amyloid-ß (Aß) peptide become self-propagating in Alzheimer's disease (AD) patients. An important characteristic of prions is their ability to replicate distinct strains, the biological information for which is enciphered within different conformations of protein aggregates. To investigate whether distinct strains of Aß prions can be discerned in AD patients, we performed transmission studies in susceptible transgenic mice using brain homogenates from sporadic or heritable (Arctic and Swedish) AD cases. Mice inoculated with the Arctic AD sample exhibited a pathology that could be distinguished from mice inoculated with the Swedish or sporadic AD samples, which was judged by differential accumulation of Aß isoforms and the morphology of cerebrovascular Aß deposition. Unlike Swedish AD- or sporadic AD-inoculated animals, Arctic AD-inoculated mice, like Arctic AD patients, displayed a prominent Aß38-containing cerebral amyloid angiopathy. The divergent transmission behavior of the Arctic AD sample compared with the Swedish and sporadic AD samples was maintained during second passage in mice, showing that Aß strains are serially transmissible. We conclude that at least two distinct strains of Aß prions can be discerned in the brains of AD patients and that strain fidelity was preserved on serial passage in mice. Our results provide a potential explanation for the clinical and pathological heterogeneity observed in AD patients.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacología , Encéfalo/metabolismo , Priones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Humanos , Ratones , Ratones Transgénicos
8.
J Am Chem Soc ; 138(31): 9840-52, 2016 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-27414264

RESUMEN

The amyloid-ß (Aß) peptide of Alzheimer's disease (AD) forms polymorphic fibrils on the micrometer and molecular scales. Various fibril growth conditions have been identified to cause polymorphism, but the intrinsic amino acid sequence basis for this polymorphism has been unclear. Several single-site mutations in the center of the Aß sequence cause different disease phenotypes and fibrillization properties. The E22G (Arctic) mutant is found in familial AD and forms protofibrils more rapidly than wild-type Aß. Here, we use solid-state NMR spectroscopy to investigate the structure, dynamics, hydration and morphology of Arctic E22G Aß40 fibrils. (13)C, (15)N-labeled synthetic E22G Aß40 peptides are studied and compared with wild-type and Osaka E22Δ Aß40 fibrils. Under the same fibrillization conditions, Arctic Aß40 exhibits a high degree of polymorphism, showing at least four sets of NMR chemical shifts for various residues, while the Osaka and wild-type Aß40 fibrils show a single or a predominant set of chemical shifts. Thus, structural polymorphism is intrinsic to the Arctic E22G Aß40 sequence. Chemical shifts and inter-residue contacts obtained from 2D correlation spectra indicate that one of the major Arctic conformers has surprisingly high structural similarity with wild-type Aß42. (13)C-(1)H dipolar order parameters, (1)H rotating-frame spin-lattice relaxation times and water-to-protein spin diffusion experiments reveal substantial differences in the dynamics and hydration of Arctic, Osaka and wild-type Aß40 fibrils. Together, these results strongly suggest that electrostatic interactions in the center of the Aß peptide sequence play a crucial role in the three-dimensional fold of the fibrils, and by inference, fibril-induced neuronal toxicity and AD pathogenesis.


Asunto(s)
Péptidos beta-Amiloides/química , Espectroscopía de Resonancia Magnética , Benzotiazoles , Sitios de Unión , Guanidina/química , Humanos , Concentración de Iones de Hidrógeno , Cinética , Mutación , Péptidos/química , Fenotipo , Conformación Proteica , Temperatura , Tiazoles/química
9.
Biophys J ; 109(4): 793-805, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26287631

RESUMEN

Aberrant self-assembly, induced by structural misfolding of the prion proteins, leads to a number of neurodegenerative disorders. In particular, misfolding of the mostly α-helical cellular prion protein (PrP(C)) into a ß-sheet-rich disease-causing isoform (PrP(Sc)) is the key molecular event in the formation of PrP(Sc) aggregates. The molecular mechanisms underlying the PrP(C)-to-PrP(Sc) conversion and subsequent aggregation remain to be elucidated. However, in persistently prion-infected cell-culture models, it was shown that treatment with monoclonal antibodies against defined regions of the prion protein (PrP) led to the clearing of PrP(Sc) in cultured cells. To gain more insight into this process, we characterized PrP-antibody complexes in solution using a fast protein liquid chromatography coupled with small-angle x-ray scattering (FPLC-SAXS) procedure. High-quality SAXS data were collected for full-length recombinant mouse PrP [denoted recPrP(23-230)] and N-terminally truncated recPrP(89-230), as well as their complexes with each of two Fab fragments (HuM-P and HuM-R1), which recognize N- and C-terminal epitopes of PrP, respectively. In-line measurements by fast protein liquid chromatography coupled with SAXS minimized data artifacts caused by a non-monodispersed sample, allowing structural analysis of PrP alone and in complex with Fab antibodies. The resulting structural models suggest two mechanisms for how these Fabs may prevent the conversion of PrP(C) into PrP(Sc).


Asunto(s)
Anticuerpos Monoclonales Humanizados/metabolismo , Proteínas PrPC/química , Proteínas PrPC/inmunología , Proteínas PrPSc/química , Animales , Cromatografía Liquida , Ratones , Modelos Moleculares , Proteínas PrPC/genética , Proteínas PrPSc/genética , Proteínas Recombinantes/química , Dispersión del Ángulo Pequeño , Soluciones , Difracción de Rayos X
10.
Biophys J ; 108(6): 1548-1554, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25809267

RESUMEN

Prions are proteins that adopt self-propagating aberrant folds. The self-propagating properties of prions are a direct consequence of their distinct structures, making the understanding of these structures and their biophysical interactions fundamental to understanding prions and their related diseases. The insolubility and inherent disorder of prions have made their structures difficult to study, particularly in the case of the infectious form of the mammalian prion protein PrP. Many investigators have therefore preferred to work with peptide fragments of PrP, suggesting that these peptides might serve as structural and functional models for biologically active prions. We have used x-ray fiber diffraction to compare a series of different-sized fragments of PrP, to determine the structural commonalities among the fragments and the biologically active, self-propagating prions. Although all of the peptides studied adopted amyloid conformations, only the larger fragments demonstrated a degree of structural complexity approaching that of PrP. Even these larger fragments did not adopt the prion structure itself with detailed fidelity, and in some cases their structures were radically different from that of pathogenic PrP(Sc).


Asunto(s)
Priones/química , Amiloide/química , Animales , Encéfalo/metabolismo , Escherichia coli , Proteínas Ligadas a GPI/química , Humanos , Ratones , Ratones Transgénicos , Microscopía Electrónica , Proteínas del Tejido Nervioso/química , Conformación Proteica , Proteínas Recombinantes/química , Difracción de Rayos X
11.
Proc Natl Acad Sci U S A ; 109(9): 3498-503, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22331873

RESUMEN

Currently, there are no animal models of the most common human prion disorder, sporadic Creutzfeldt-Jakob disease (CJD), in which prions are formed spontaneously from wild-type (WT) prion protein (PrP). Interestingly, bank voles (BV) exhibit an unprecedented promiscuity for diverse prion isolates, arguing that bank vole PrP (BVPrP) may be inherently prone to adopting misfolded conformations. Therefore, we constructed transgenic (Tg) mice expressing WT BVPrP. Tg(BVPrP) mice developed spontaneous CNS dysfunction between 108 and 340 d of age and recapitulated the hallmarks of prion disease, including spongiform degeneration, pronounced astrogliosis, and deposition of alternatively folded PrP in the brain. Brain homogenates of ill Tg(BVPrP) mice transmitted disease to Tg(BVPrP) mice in ∼35 d, to Tg mice overexpressing mouse PrP in under 100 d, and to WT mice in ∼185 d. Our studies demonstrate experimentally that WT PrP can spontaneously form infectious prions in vivo. Thus, Tg(BVPrP) mice may be useful for studying the spontaneous formation of prions, and thus may provide insight into the etiology of sporadic CJD.


Asunto(s)
Arvicolinae/genética , Modelos Animales de Enfermedad , Enfermedades por Prión/genética , Priones/genética , Secuencia de Aminoácidos , Animales , Encéfalo/patología , Química Encefálica , Codón/genética , Genes Reporteros , Proteína Ácida Fibrilar de la Glía , Especificidad del Huésped , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Enfermedades por Prión/patología , Enfermedades por Prión/transmisión , Priones/química , Regiones Promotoras Genéticas , Pliegue de Proteína , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Extractos de Tejidos/toxicidad
12.
Proc Natl Acad Sci U S A ; 109(27): 11025-30, 2012 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-22711819

RESUMEN

The aggregation and deposition of amyloid-ß (Aß) peptides are believed to be central events in the pathogenesis of Alzheimer's disease (AD). Inoculation of brain homogenates containing Aß aggregates into susceptible transgenic mice accelerated Aß deposition, suggesting that Aß aggregates are capable of self-propagation and hence might be prions. Recently, we demonstrated that Aß deposition can be monitored in live mice using bioluminescence imaging (BLI). Here, we use BLI to probe the ability of Aß aggregates to self-propagate following inoculation into bigenic mice. We report compelling evidence that Aß aggregates are prions by demonstrating widespread cerebral ß-amyloidosis induced by inoculation of either purified Aß aggregates derived from brain or aggregates composed of synthetic Aß. Although synthetic Aß aggregates were sufficient to induce Aß deposition in vivo, they exhibited lower specific biological activity compared with brain-derived Aß aggregates. Our results create an experimental paradigm that should lead to identification of self-propagating Aß conformations, which could represent novel targets for interrupting the spread of Aß deposition in AD patients.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/síntesis química , Péptidos beta-Amiloides/metabolismo , Amiloidosis/metabolismo , Priones/síntesis química , Priones/metabolismo , Envejecimiento/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/aislamiento & purificación , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Luciferasas/genética , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Degeneración Nerviosa/metabolismo , Priones/genética , Priones/aislamiento & purificación
13.
Proc Natl Acad Sci U S A ; 108(52): 21223-8, 2011 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-22160704

RESUMEN

Some prion protein mutations create anchorless molecules that cause Gerstmann-Sträussler-Scheinker (GSS) disease. To model GSS, we generated transgenic mice expressing cellular prion protein (PrP(C)) lacking the glycosylphosphatidyl inositol (GPI) anchor, denoted PrP(ΔGPI). Mice overexpressing PrP(ΔGPI) developed a late-onset, spontaneous neurologic dysfunction characterized by widespread amyloid deposition in the brain and the presence of a short protease-resistant PrP fragment similar to those found in GSS patients. In Tg(PrP,ΔGPI) mice, disease onset could be accelerated either by inoculation with brain homogenate prepared from spontaneously ill animals or by coexpression of membrane-anchored, full-length PrP(C). In contrast, coexpression of N-terminally truncated PrP(Δ23-88) did not affect disease progression. Remarkably, disease from ill Tg(PrP,ΔGPI) mice transmitted to mice expressing wild-type PrP(C), indicating the spontaneous generation of prions.


Asunto(s)
Amiloide/ultraestructura , Modelos Animales de Enfermedad , Enfermedad de Gerstmann-Straussler-Scheinker/metabolismo , Enfermedad de Gerstmann-Straussler-Scheinker/fisiopatología , Glicosilfosfatidilinositoles/deficiencia , Proteínas PrPC/metabolismo , Animales , Western Blotting , Electroforesis en Gel de Poliacrilamida , Mapeo Epitopo , Enfermedad de Gerstmann-Straussler-Scheinker/genética , Enfermedad de Gerstmann-Straussler-Scheinker/patología , Técnicas Histológicas , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Proteínas PrPC/genética , Pliegue de Proteína
14.
Res Sq ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38765977

RESUMEN

PINK1 loss-of-function mutations and exposure to mitochondrial toxins are causative for Parkinson's disease (PD) and Parkinsonism, respectively. We demonstrate that pathological α-synuclein deposition, the hallmark pathology of idiopathic PD, induces mitochondrial dysfunction, and impairs mitophagy as evidenced by the accumulation of the PINK1 substrate pS65-Ubiquitin (pUb). We discovered MTK458, a brain penetrant small molecule that binds to PINK1 and stabilizes its active complex, resulting in increased rates of mitophagy. Treatment with MTK458 mediates clearance of accumulated pUb and α-synuclein pathology in α-synuclein pathology models in vitro and in vivo. Our findings from preclinical PD models suggest that pharmacological activation of PINK1 warrants further clinical evaluation as a therapeutic strategy for disease modification in PD.

15.
PLoS Pathog ; 7(11): e1002382, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22163178

RESUMEN

The central event in prion diseases is the conformational conversion of the cellular prion protein (PrP(C)) into PrP(Sc), a partially protease-resistant and infectious conformer. However, the mechanism by which PrP(Sc) causes neuronal dysfunction remains poorly understood. Levels of Shadoo (Sho), a protein that resembles the flexibly disordered N-terminal domain of PrP(C), were found to be reduced in the brains of mice infected with the RML strain of prions [1], implying that Sho levels may reflect the presence of PrP(Sc) in the brain. To test this hypothesis, we examined levels of Sho during prion infection using a variety of experimental systems. Sho protein levels were decreased in the brains of mice, hamsters, voles, and sheep infected with different natural and experimental prion strains. Furthermore, Sho levels were decreased in the brains of prion-infected, transgenic mice overexpressing Sho and in infected neuroblastoma cells. Time-course experiments revealed that Sho levels were inversely proportional to levels of protease-resistant PrP(Sc). Membrane anchoring and the N-terminal domain of PrP both influenced the inverse relationship between Sho and PrP(Sc). Although increased Sho levels had no discernible effect on prion replication in mice, we conclude that Sho is the first non-PrP marker specific for prion disease. Additional studies using this paradigm may provide insight into the cellular pathways and systems subverted by PrP(Sc) during prion disease.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteínas PrPSc/metabolismo , Enfermedades por Prión/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Línea Celular , Regulación hacia Abajo , Proteínas Ligadas a GPI , Ratones , Ratones Transgénicos , Neuronas/enzimología , Neuronas/metabolismo , Péptido Hidrolasas/metabolismo , Proteínas PrPC/química , Proteínas PrPC/metabolismo , Proteínas PrPSc/química , Enfermedades por Prión/veterinaria , Ovinos , Enfermedades de las Ovejas/metabolismo
16.
Subcell Biochem ; 65: 91-108, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23225001

RESUMEN

This chapter focuses on the structural conversion of natural and recombinant prion proteins in vitro. They key event in prion diseases is the conversion of the cellular prion protein (PrP(C)) into its disease causing isoform PrP(Sc). This conversion is represented by a conformational change from an ß-helical dominated isoform into the mostly ß-sheeted PrP(Sc). Represented is an overview of in vitro conversion systems that result in ß-structured recombinant prion proteins including the current achievements in the generation of synthetic mammalian prions as proof of the protein-only hypothesis. In addition to the conversion of recombinant PrP the chapter features a summary of the protein misfolding cyclic amplification (PMCA) technique which has gained enormous popularity in prion research. Given is a general overview about the technique itself and the broad spectrum of utilization as detection method for prions. The spontaneous generation of prions by the protein misfolding amplification (PMCA) are also discussed.


Asunto(s)
Complejos Multiproteicos/química , Proteínas PrPC/química , Proteínas PrPSc/química , Pliegue de Proteína , Animales , Humanos , Complejos Multiproteicos/metabolismo , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
bioRxiv ; 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36824886

RESUMEN

PINK1 loss-of-function mutations and exposure to mitochondrial toxins are causative for Parkinson's disease (PD) and Parkinsonism, respectively. We demonstrate that pathological α-synuclein deposition, the hallmark pathology of idiopathic PD, induces mitochondrial dysfunction and impairs mitophagy, driving accumulation of the PINK1 substrate pS65-Ubiquitin (pUb) in primary neurons and in vivo. We synthesized MTK458, a brain penetrant small molecule that binds to PINK1 and stabilizes an active heterocomplex, thereby increasing mitophagy. MTK458 mediates clearance of α-synuclein pathology in PFF seeding models in vitro and in vivo and reduces pUb. We developed an ultrasensitive assay to quantify pUb levels in plasma and observed an increase in pUb in PD subjects that correlates with disease progression, paralleling our observations in PD models. Our combined findings from preclinical PD models and patient biofluids suggest that pharmacological activation of PINK1 is worthy of further study as a therapeutic strategy for disease modification in PD. Highlights: Discovery of a plasma Parkinson's Disease biomarker candidate, pS65-Ubiquitin (pUb)Plasma pUb levels correlate with disease status and progression in PD patients.Identification of a potent, brain penetrant PINK1 activator, MTK458MTK458 selectively activates PINK1 by stimulating dimerization and stabilization of the PINK1/TOM complexMTK458 drives clearance of α-synuclein pathology and normalizes pUb in in vivo Parkinson's models.

18.
Biophys J ; 102(10): 2339-44, 2012 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-22677387

RESUMEN

The prion-forming domain of the fungal prion protein HET-s, HET-s(218-289), is known from solid-state NMR studies to have a ß-solenoidal structure; the ß-solenoid has the cross-ß structure characteristic of all amyloids, but is inherently more complex than the generic stacked ß-sheets found in studies of small synthetic peptides. At low pH HET-s(218-289) has also been reported to form an alternative structure, which has not been characterized. We have confirmed by x-ray fiber diffraction that HET-s(218-289) adopts a ß-solenoidal structure at neutral pH, and shown that at low pH, it forms either a ß-solenoid or a stacked ß-sheet structure, depending on the integrity of the protein and the conditions of fibrillization. The low pH stacked-sheet structure is usually formed only by proteolyzed HET-s(218-289), but intact HET-s(218-289) can form stacked sheets when seeded with proteolyzed stacked-sheet HET-s(218-289). The polymorphism of HET-s parallels the structural differences between the infectious brain-derived and the much less infectious recombinant mammalian prion protein PrP. Taken together, these observations suggest that the functional or pathological forms of amyloid proteins are more complex than the simple generic stacked-sheet amyloids commonly formed by short peptides.


Asunto(s)
Amiloide/metabolismo , Proteínas Fúngicas/metabolismo , Podospora/metabolismo , Priones/metabolismo , Proteolisis , Secuencia de Aminoácidos , Animales , Benzotiazoles , Electroforesis en Gel de Poliacrilamida , Fluorescencia , Proteínas Fúngicas/química , Proteínas Fúngicas/ultraestructura , Concentración de Iones de Hidrógeno , Espectrometría de Masas , Datos de Secuencia Molecular , Priones/química , Priones/ultraestructura , Unión Proteica , Tiazoles/metabolismo , Difracción de Rayos X
19.
Biol Chem ; 392(5): 415-21, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21476870

RESUMEN

The conversion of the cellular isoform of the prion protein (PrP(C)) into the pathologic isoform (PrP(Sc)) is the key event in prion diseases. To study the conversion process, an in vitro system based on varying the concentration of low amounts of sodium dodecyl sulfate (SDS) has been employed. In the present study, the conversion of full-length PrP(C) isolated from Chinese hamster ovary cells (CHO-PrP(C)) was examined. CHO-PrP(C) harbors native, posttranslational modifications, including the GPI anchor and two N-linked glyco-sylation sites. The properties of CHO-PrP(C) were compared with those of full-length and N-terminally truncated recombinant PrP. As shown earlier with recombinant PrP (recPrP90-231), transition from a soluble α-helical state as known for native PrP(C) into an aggregated, ß-sheet-rich PrP(Sc)-like state could be induced by dilution of SDS. The aggregated state is partially proteinase K (PK)-resistant, exhibiting a cleavage site similar to that found with PrP(Sc). Compared to recPrP (90-231), fibril formation with CHO-PrP(C) requires lower SDS concentrations (0.0075%), and can be drastically accelerated by seeding with PrP(Sc) purified from brain homogenates of terminally sick hamsters. Our results show that recPrP 90-231 and CHO-PrPC behave qualitatively similar but quantitatively different. The in vivo situation can be simulated closer with CHO-PrP(C) because the specific PK cleave site could be shown and the seed-assisted fibrillization was much more efficient.


Asunto(s)
Proteínas PrPC/química , Proteínas PrPC/metabolismo , Proteínas PrPSc/química , Proteínas PrPSc/metabolismo , Animales , Western Blotting , Células CHO , Dicroismo Circular , Cricetinae , Cricetulus , Electroforesis en Gel de Poliacrilamida , Microscopía Electrónica , Proteínas PrPC/efectos de los fármacos , Proteínas PrPSc/efectos de los fármacos , Procesamiento Proteico-Postraduccional , Estructura Secundaria de Proteína , Dodecil Sulfato de Sodio/farmacología
20.
Proc Natl Acad Sci U S A ; 105(31): 10815-9, 2008 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-18669653

RESUMEN

Misfolding and subsequent aggregation of endogenous proteins constitute essential steps in many human disorders, including Alzheimer and prion diseases. In most prion protein-folding studies, the posttranslational modifications, the lipid anchor in particular, were lacking. Here, we studied a fully posttranslationally modified cellular prion protein, carrying two N-glycosylations and the natural GPI anchor. We used time-resolved FTIR to study the prion protein secondary structure changes when binding to a raft-like lipid membrane via its GPI anchor. We observed that membrane anchoring above a threshold concentration induced refolding of the prion protein to intermolecular beta-sheets. Such transition is not observed in solution and is membrane specific. Excessive membrane anchoring, analyzed with molecular sensitivity, is thought to be a crucial event in the development of prion diseases.


Asunto(s)
Proteínas de la Membrana/genética , Modelos Moleculares , Proteínas PrPC/genética , Conformación Proteica , Pliegue de Proteína , Animales , Cricetinae , Mesocricetus , Espectroscopía Infrarroja por Transformada de Fourier
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA