Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 210(9): 1447-1458, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36939393

RESUMEN

IgE Abs, best known for their role in allergic reactions, have only rarely been used in immunotherapies. Nevertheless, they offer a potential alternative to the more commonly used IgGs. The affinity of IgE Ag binding influences the type of response from mast cells, so any immunotherapies using IgEs must balance Ag affinity with desired therapeutic effect. One potential way to harness differential binding affinities of IgE is in protein aggregation diseases, where low-affinity binding of endogenous proteins is preferred, but enhanced binding of clusters of disease-associated aggregated proteins could target responses to the sites of disease. For this reason, we sought to create a low-affinity IgE against the prion protein (PrP), which exists in an endogenous monomeric state but can misfold into aggregated states during the development of prion disease. First, we determined that mast cell proteases tryptase and cathepsin G were capable of degrading PrP. Then we engineered a recombinant IgE Ab directed against PrP from the V region of a PrP-specific IgG and tested its activation of the human mast cell line LAD2. The αPrP IgE bound LAD2 through Fc receptors. Crosslinking receptor-bound αPrP IgE activated SYK and ERK phosphorylation, caused Fc receptor internalization, and resulted in degranulation. This work shows that a recombinant αPrP IgE can activate LAD2 cells to release enzymes that can degrade PrP, suggesting that IgE may be useful in targeting diseases that involve protein aggregation.


Asunto(s)
Proteínas Priónicas , Receptores de IgE , Humanos , Receptores de IgE/metabolismo , Proteínas Priónicas/metabolismo , Mastocitos/metabolismo , Péptido Hidrolasas/metabolismo , Agregado de Proteínas , Inmunoglobulina E/metabolismo , Degranulación de la Célula
2.
PLoS Pathog ; 17(6): e1009703, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34181702

RESUMEN

Prion diseases are transmissible neurodegenerative disorders that affect mammals, including humans. The central molecular event is the conversion of cellular prion glycoprotein, PrPC, into a plethora of assemblies, PrPSc, associated with disease. Distinct phenotypes of disease led to the concept of prion strains, which are associated with distinct PrPSc structures. However, the degree to which intra- and inter-strain PrPSc heterogeneity contributes to disease pathogenesis remains unclear. Addressing this question requires the precise isolation and characterization of all PrPSc subpopulations from the prion-infected brains. Until now, this has been challenging. We used asymmetric-flow field-flow fractionation (AF4) to isolate all PrPSc subpopulations from brains of hamsters infected with three prion strains: Hyper (HY) and 263K, which produce almost identical phenotypes, and Drowsy (DY), a strain with a distinct presentation. In-line dynamic and multi-angle light scattering (DLS/MALS) data provided accurate measurements of particle sizes and estimation of the shape and number of PrPSc particles. We found that each strain had a continuum of PrPSc assemblies, with strong correlation between PrPSc quaternary structure and phenotype. HY and 263K were enriched with large, protease-resistant PrPSc aggregates, whereas DY consisted primarily of smaller, more protease-sensitive aggregates. For all strains, a transition from protease-sensitive to protease-resistant PrPSc took place at a hydrodynamic radius (Rh) of 15 nm and was accompanied by a change in glycosylation and seeding activity. Our results show that the combination of AF4 with in-line MALS/DLS is a powerful tool for analyzing PrPSc subpopulations and demonstrate that while PrPSc quaternary structure is a major contributor to PrPSc structural heterogeneity, a fundamental change, likely in secondary/tertiary structure, prevents PrPSc particles from maintaining proteinase K resistance below an Rh of 15 nm, regardless of strain. This results in two biochemically distinctive subpopulations, the proportion, seeding activity, and stability of which correlate with prion strain phenotype.


Asunto(s)
Dispersión Dinámica de Luz/métodos , Fotometría/métodos , Proteínas PrPSc/análisis , Proteínas PrPSc/química , Animales , Cricetinae , Hidrodinámica , Ratones , Estructura Cuaternaria de Proteína
3.
Int J Mol Sci ; 24(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38139358

RESUMEN

A distinctive signature of the prion diseases is the accumulation of the pathogenic isoform of the prion protein, PrPSc, in the central nervous system of prion-affected humans and animals. PrPSc is also found in peripheral tissues, raising concerns about the potential transmission of pathogenic prions through human food supplies and posing a significant risk to public health. Although muscle tissues are considered to contain levels of low prion infectivity, it has been shown that myotubes in culture efficiently propagate PrPSc. Given the high consumption of muscle tissue, it is important to understand what factors could influence the establishment of a prion infection in muscle tissue. Here we used in vitro myotube cultures, differentiated from the C2C12 myoblast cell line (dC2C12), to identify factors affecting prion replication. A range of experimental conditions revealed that PrPSc is tightly associated with proteins found in the systemic extracellular matrix, mostly fibronectin (FN). The interaction of PrPSc with FN decreased prion infectivity, as determined by standard scrapie cell assay. Interestingly, the prion-resistant reserve cells in dC2C12 cultures displayed a FN-rich extracellular matrix while the prion-susceptible myotubes expressed FN at a low level. In agreement with the in vitro results, immunohistopathological analyses of tissues from sheep infected with natural scrapie demonstrated a prion susceptibility phenotype linked to an extracellular matrix with undetectable levels of FN. Conversely, PrPSc deposits were not observed in tissues expressing FN. These data indicate that extracellular FN may act as a natural barrier against prion replication and that the extracellular matrix composition may be a crucial feature determining prion tropism in different tissues.


Asunto(s)
Fibronectinas , Enfermedades por Prión , Priones , Scrapie , Animales , Humanos , Línea Celular , Fibronectinas/uso terapéutico , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/prevención & control , Priones/metabolismo , Scrapie/metabolismo , Ovinos
4.
J Biol Chem ; 295(25): 8460-8469, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32358064

RESUMEN

Prions are lipidated proteins that interact with endogenous lipids and metal ions. They also assemble into multimers and propagate into the infectious scrapie form known as PrPSc The high-resolution structure of the infectious PrPSc state remains unknown, and its analysis largely relies on detergent-based preparations devoid of endogenous ligands. Here we designed polymers that allow isolation of endogenous membrane:protein assemblies in native nanodiscs without exposure to conventional detergents that destabilize protein structures and induce fibrillization. A set of styrene-maleic acid (SMA) polymers including a methylamine derivative facilitated gentle release of the infectious complexes for resolution of multimers, and a thiol-containing version promoted crystallization. Polymer extraction from brain homogenates from Syrian hamsters infected with Hyper prions and WT mice infected with Rocky Mountain Laboratories prions yielded infectious prion nanoparticles including oligomers and microfilaments bound to lipid vesicles. Lipid analysis revealed the brain phospholipids that associate with prion protofilaments, as well as those that are specifically enriched in prion assemblies captured by the methylamine-modified copolymer. A comparison of the infectivity of PrPSc attached to SMA lipid particles in mice and hamsters indicated that these amphipathic polymers offer a valuable tool for high-yield production of intact, detergent-free prions that retain in vivo activity. This native prion isolation method provides an avenue for producing relevant prion:lipid targets and potentially other proteins that form multimeric assemblies and fibrils on membranes.


Asunto(s)
Encéfalo/metabolismo , Lípidos/química , Maleatos/química , Nanoestructuras/química , Poliestirenos/química , Proteínas Priónicas/metabolismo , Animales , Cricetinae , Maleatos/síntesis química , Maleatos/metabolismo , Metilaminas/química , Ratones , Fosfolípidos/química , Fosfolípidos/metabolismo , Poliestirenos/síntesis química , Poliestirenos/metabolismo , Proteínas Priónicas/química , Proteínas Priónicas/aislamiento & purificación , Compuestos de Sulfhidrilo/química
5.
Acta Neuropathol ; 141(6): 841-859, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33881612

RESUMEN

Triggering receptor expressed on myeloid cells 2 (TREM2) is an innate immune cell surface receptor that regulates microglial function and is involved in the pathophysiology of several neurodegenerative diseases. Its soluble form (sTREM2) results from shedding of the TREM2 ectodomain. The role of TREM2 in prion diseases, a group of rapidly progressive dementias remains to be elucidated. In the present study, we analysed the expression of TREM2 and its main sheddase ADAM10 in the brain of sporadic Creutzfeldt-Jakob disease (sCJD) patients and evaluated the role of CSF and plasma sTREM2 as a potential diagnostic marker of prion disease. Our data indicate that, compared to controls, TREM2 is increased in sCJD patient brains at the mRNA and protein levels in a regional and subtype dependent fashion, and expressed in a subpopulation of microglia. In contrast, ADAM10 is increased at the protein, but not the mRNA level, with a restricted neuronal expression. Elevated CSF sTREM2 is found in sCJD, genetic CJD with mutations E200K and V210I in the prion protein gene (PRNP), and iatrogenic CJD, as compared to healthy controls (HC) (AUC = 0.78-0.90) and neurological controls (AUC = 0.73-0.85), while CSF sTREM2 is unchanged in fatal familial insomnia. sTREM2 in the CSF of cases with Alzheimer's disease, and multiple sclerosis was not significantly altered in our series. CSF sTREM2 concentrations in sCJD are PRNP codon 129 and subtype-related, correlate with CSF 14-3-3 positivity, total-tau and YKL-40, and increase with disease progression. In plasma, sTREM2 is increased in sCJD compared with HC (AUC = 0.80), displaying positive correlations with plasma total-tau, neurofilament light, and YKL-40. We conclude that comparative study of TREM2 in brain and biological fluids of prion diseases reveals TREM2 to be altered in human prion diseases with a potential value in target engagement, patient stratification, and disease monitoring.


Asunto(s)
Proteína ADAM10 , Encéfalo , Glicoproteínas de Membrana , Enfermedades por Prión , Receptores Inmunológicos , Proteína ADAM10/sangre , Proteína ADAM10/líquido cefalorraquídeo , Proteína ADAM10/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Biomarcadores/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Humanos , Glicoproteínas de Membrana/sangre , Glicoproteínas de Membrana/líquido cefalorraquídeo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Microglía/metabolismo , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Proteínas Priónicas/metabolismo , Receptores Inmunológicos/sangre , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
6.
Clin Infect Dis ; 70(4): 692-695, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31247065

RESUMEN

We report the cases of 3 patients with fatal, disseminated Mycobacterium chimaera infections following cardiac surgeries. Progressive neurocognitive decline and death were explained by active granulomatous encephalitis, with widespread involvement of other organs. This syndrome is clinically elusive and, thus, may have caused deaths in prior reported series.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Encefalitis , Infecciones por Mycobacterium no Tuberculosas , Infecciones por Mycobacterium , Mycobacterium , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Encefalitis/diagnóstico , Encefalitis/etiología , Humanos , Infecciones por Mycobacterium/diagnóstico , Infecciones por Mycobacterium/etiología
7.
Artículo en Inglés | MEDLINE | ID: mdl-29784843

RESUMEN

Prion diseases are a group of neurodegenerative diseases associated with the misfolding of the cellular prion protein (PrPC) into the infectious form (PrPSc). There are currently no treatments for prion disease. Bile acids have the ability to protect hepatocytes from apoptosis and are neuroprotective in animal models of other protein-folding neurodegenerative diseases, including Huntington's, Parkinson's, and Alzheimer's disease. Importantly, bile acids are approved for clinical use in patients with cirrhosis and have recently been shown to be safe and possibly effective in pilot trials of patients with amyotrophic lateral sclerosis (ALS). We previously reported that the bile acid ursodeoxycholic acid (UDCA), given early in disease, prolonged incubation periods in male RML-infected mice. Here, we expand on this result to include tauro-ursodeoxycholic acid (TUDCA) treatment trials and delayed UDCA treatment. We demonstrate that despite a high dose of TUDCA given early in disease, there was no significant difference in incubation periods between treated and untreated cohorts, regardless of sex. In addition, delayed treatment with a high dose of UDCA resulted in a significant shortening of the average survival time for both male and female mice compared to their sex-matched controls, with evidence of increased BiP, a marker of apoptosis, in treated female mice. Our findings suggest that treatment with high-dose TUDCA provides no therapeutic benefit and that delayed treatment with high-dose UDCA is ineffective and could worsen outcomes.


Asunto(s)
Antiinfecciosos/farmacología , Proteínas PrPSc/efectos de los fármacos , Enfermedades por Prión/tratamiento farmacológico , Ácido Tauroquenodesoxicólico/farmacología , Ácido Ursodesoxicólico/farmacología , Animales , Modelos Animales de Enfermedad , Esquema de Medicación , Femenino , Masculino , Ratones , Proteínas PrPSc/patogenicidad , Enfermedades por Prión/mortalidad , Enfermedades por Prión/patología , Análisis de Supervivencia , Tiempo de Tratamiento , Insuficiencia del Tratamiento
9.
Glia ; 64(6): 937-51, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26880394

RESUMEN

Prion diseases are progressive neurodegenerative disorders affecting humans and various mammals. The prominent neuropathological change in prion diseases is neuroinflammation characterized by activation of neuroglia surrounding prion deposition. The cause and effect of this cellular response, however, is unclear. We investigated innate immune defenses against prion infection using primary mixed neuronal and glial cultures. Conditional prion propagation occurred in glial cultures depending on their immune status. Preconditioning of the cells with the toll-like receptor (TLR) ligand, lipopolysaccharide, resulted in a reduction in prion propagation, whereas suppression of the immune responses with the synthetic glucocorticoid, dexamethasone, increased prion propagation. In response to recombinant prion fibrils, glial cells up-regulated TLRs (TLR1 and TLR2) expression and secreted cytokines (tumor necrosis factor-α, interleukin-1ß, interleukin-6, granulocyte-macrophage colony-stimulating factor, and interferon-ß). Preconditioning of neuronal and glial cultures with recombinant prion fibrils inhibited prion replication and altered microglial and astrocytic populations. Our results provide evidence that, in early stages of prion infection, glial cells respond to prion infection through TLR-mediated innate immunity.


Asunto(s)
Inmunidad Innata/inmunología , Neuroglía/metabolismo , Priones/metabolismo , Receptores Toll-Like/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-1beta/metabolismo , Ratones , Neuroglía/inmunología , Priones/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Virol ; 89(15): 7660-72, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25972546

RESUMEN

UNLABELLED: Prion diseases are fatal neurodegenerative disorders associated with the conversion of cellular prion protein (PrPC) into its aberrant infectious form (PrPSc). There is no treatment available for these diseases. The bile acids tauroursodeoxycholic acid(TUDCA) and ursodeoxycholic acid (UDCA) have been recently shown to be neuroprotective in other protein misfolding disease models, including Parkinson's, Huntington's and Alzheimer's diseases, and also in humans with amyotrophic lateral sclerosis.Here, we studied the therapeutic efficacy of these compounds in prion disease. We demonstrated that TUDCA and UDCA substantially reduced PrP conversion in cell-free aggregation assays, as well as in chronically and acutely infected cell cultures. This effect was mediated through reduction of PrPSc seeding ability, rather than an effect on PrPC. We also demonstrated the ability of TUDCA and UDCA to reduce neuronal loss in prion-infected cerebellar slice cultures. UDCA treatment reduced astrocytosis and prolonged survival in RML prion-infected mice. Interestingly, these effects were limited to the males, implying a gender-specific difference in drug metabolism. Beyond effects on PrPSc, we found that levels of phosphorylated eIF2 were increased at early time points, with correlated reductions in postsynaptic density protein 95. As demonstrated for other neurodegenerative diseases, we now show that TUDCA and UDCA may have a therapeutic role in prion diseases, with effects on both prion conversion and neuroprotection. Our findings, together with the fact that these natural compounds are orally bioavailable, permeable to the blood-brain barrier, and U.S. Food and Drug Administration-approved for use in humans, make these compounds promising alternatives for the treatment of prion diseases. IMPORTANCE: Prion diseases are fatal neurodegenerative diseases that are transmissible to humans and other mammals. There are no disease-modifying therapies available, despite decades of research. Treatment targets have included inhibition of protein accumulation,clearance of toxic aggregates, and prevention of downstream neurodegeneration. No one target may be sufficient; rather, compounds which have a multimodal mechanism, acting on different targets, would be ideal. TUDCA and UDCA are bile acids that may fulfill this dual role. Previous studies have demonstrated their neuroprotective effects in several neurodegenerative disease models, and we now demonstrate that this effect occurs in prion disease, with an added mechanistic target of upstream prion seeding. Importantly, these are natural compounds which are orally bioavailable, permeable to the blood-brain barrier, and U.S.Food and Drug Administration-approved for use in humans with primary biliary cirrhosis. They have recently been proven efficacious in human amyotrophic lateral sclerosis. Therefore, these compounds are promising options for the treatment of prion diseases.


Asunto(s)
Neuronas/metabolismo , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Enfermedades por Prión/metabolismo , Enfermedades por Prión/fisiopatología , Ácido Tauroquenodesoxicólico/metabolismo , Ácido Ursodesoxicólico/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Muerte Celular , Supervivencia Celular , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Especificidad de la Especie
11.
Can J Neurol Sci ; 43(4): 593-5, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26972054

RESUMEN

Frontotemporal brain sagging syndrome is a dementia associated with hypersomnolence, personality changes, and features of intracranial hypotension on magnetic resonance imaging. The literature is sparse with respect to treatment options; many patients simply worsen. We present a case in which this syndrome responded to lumbar dural reduction surgery. Postoperative magnetic resonance imaging indicated normalization of brain sagging and lumbar intrathecal pressure. Although no evidence of cerebrospinal leak was found, extremely thin dura was noted intraoperatively, suggesting that a thin and incompetent dura could result in this low-pressure syndrome. Clinicians who encounter this syndrome should consider dural reduction surgery as a treatment strategy.


Asunto(s)
Trastornos de Somnolencia Excesiva/complicaciones , Demencia Frontotemporal/complicaciones , Demencia Frontotemporal/cirugía , Hipotensión Intracraneal/complicaciones , Procedimientos Neuroquirúrgicos/métodos , Trastornos de la Personalidad/complicaciones , Trastornos de Somnolencia Excesiva/diagnóstico por imagen , Demencia Frontotemporal/diagnóstico por imagen , Humanos , Hipotensión Intracraneal/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Trastornos de la Personalidad/diagnóstico por imagen
12.
J Biol Chem ; 289(15): 10419-10430, 2014 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-24554723

RESUMEN

The formation of fibrillar aggregates has long been associated with neurodegenerative disorders such as Alzheimer and Parkinson diseases. Although fibrils are still considered important to the pathology of these disorders, it is now widely understood that smaller amyloid oligomers are the toxic entities along the misfolding pathway. One characteristic shared by the majority of amyloid oligomers is the ability to disrupt membranes, a commonality proposed to be responsible for their toxicity, although the mechanisms linking this to cell death are poorly understood. Here, we describe the physical basis for the cytotoxicity of oligomers formed by the prion protein (PrP)-derived amyloid peptide PrP(106-126). We show that oligomers of this peptide kill several mammalian cells lines, as well as mouse cerebellar organotypic cultures, and we also show that they exhibit antimicrobial activity. Physical perturbation of model membranes mimicking bacterial or mammalian cells was investigated using atomic force microscopy, polarized total internal reflection fluorescence microscopy, and NMR spectroscopy. Disruption of anionic membranes proceeds through a carpet or detergent model as proposed for other antimicrobial peptides. By contrast, when added to zwitterionic membranes containing cholesterol-rich ordered domains, PrP(106-126) oligomers induce a loss of domain separation and decreased membrane disorder. Loss of raft-like domains may lead to activation of apoptotic pathways, resulting in cell death. This work sheds new light on the physical mechanisms of amyloid cytotoxicity and is the first to clearly show membrane type-specific modes of action for a cytotoxic peptide.


Asunto(s)
Amiloide/química , Fragmentos de Péptidos/química , Priones/química , Animales , Péptidos Catiónicos Antimicrobianos/química , Membrana Celular/química , Cerebelo/patología , Colesterol/química , Detergentes/química , Escherichia coli/crecimiento & desarrollo , Membrana Dobles de Lípidos/química , Lípidos/química , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Microscopía de Fuerza Atómica , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Técnicas de Cultivo de Órganos , Células PC12 , Fosfatidilcolinas/química , Ratas
13.
PLoS Pathog ; 9(11): e1003755, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244171

RESUMEN

Prion diseases are driven by the strain-specific, template-dependent transconformation of the normal cellular prion protein (PrP(C)) into a disease specific isoform PrP(Sc). Cell culture models of prion infection generally use replicating cells resulting in lower levels of prion accumulation compared to animals. Using non-replicating cells allows the accumulation of higher levels of PrP(Sc) and, thus, greater amounts of infectivity. Here, we infect non-proliferating muscle fiber myotube cultures prepared from differentiated myoblasts. We demonstrate that prion-infected myotubes generate substantial amounts of PrP(Sc) and that the level of infectivity produced in these post-mitotic cells, 10(5.5) L.D.50/mg of total protein, approaches that observed in vivo. Exposure of the myotubes to different mouse-adapted agents demonstrates strain-specific replication of infectious agents. Mouse-derived myotubes could not be infected with hamster prions suggesting that the species barrier effect is intact. We suggest that non-proliferating myotubes will be a valuable model system for generating infectious prions and for screening compounds for anti-prion activity.


Asunto(s)
Proliferación Celular , Proteínas PrPSc/metabolismo , Animales , Línea Celular , Cricetinae , Ratones , Fibras Musculares Esqueléticas , Especificidad de la Especie
14.
J Biol Chem ; 288(7): 4772-81, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23283973

RESUMEN

Prion diseases are fatal neurodegenerative disorders associated with the polymerization of the cellular form of prion protein (PrP(C)) into an amyloidogenic ß-sheet infectious form (PrP(Sc)). The sequence of host PrP is the major determinant of host prion disease susceptibility. In mice, the presence of allele a (Prnp(a), encoding the polymorphism Leu-108/Thr-189) or b (Prnp(b), Phe-108/Val-189) is associated with short or long incubation times, respectively, following infection with PrP(Sc). The molecular bases linking PrP sequence, infection susceptibility, and convertibility of PrP(C) into PrP(Sc) remain unclear. Here we show that recombinant PrP(a) and PrP(b) aggregate and respond to seeding differently in vitro. Our kinetic studies reveal differences during the nucleation phase of the aggregation process, where PrP(b) exhibits a longer lag phase that cannot be completely eliminated by seeding the reaction with preformed fibrils. Additionally, PrP(b) is more prone to propagate features of the seeds, as demonstrated by conformational stability and electron microscopy studies of the formed fibrils. We propose a model of polymerization to explain how the polymorphisms at positions 108 and 189 produce the phenotypes seen in vivo. This model also provides insight into phenomena such as species barrier and prion strain generation, two phenomena also influenced by the primary structure of PrP.


Asunto(s)
Fenilalanina/genética , Polimorfismo Genético , Enfermedades por Prión/metabolismo , Priones/genética , Valina/genética , Alelos , Animales , Benzotiazoles , Dicroismo Circular , Relación Dosis-Respuesta a Droga , Cinética , Ratones , Microscopía Electrónica/métodos , Polímeros/química , Enfermedades por Prión/genética , Conformación Proteica , Proteínas Recombinantes/metabolismo , Tiazoles/química , Factores de Tiempo
15.
J Leukoc Biol ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38725289

RESUMEN

While several functions of the endogenous prion protein (PrP) have been studied, the homeostatic function of PrP is still debated. Notably, PrP is highly expressed on mast cells, granular immune cells that regulate inflammation. When activated, mast cells shed PrP though the mechanism and consequences of this are not yet understood. First, we tested several mast cell lines and found that, while PrP was almost always present, the total amount differed greatly. Activation of mast cells induced a cleavage of the N-terminal region of PrP, and this was reduced by protease inhibitors. Exogenous mast cell proteases caused a similar loss of the PrP N-terminus. Additionally, mast cells shed PrP in an ADAM10-dependent fashion even in the absence of activation. Our results suggest that PrP is cleaved from resting mast cells by ADAM10 and from activated mast cells by mast cell proteases. PrP also appears to affect mast cell function, as Prnp-/- BMMC showed lower levels of degranulation and cytokine release, as well as lower levels of both FcεRI and CD117. Finally, we sought to provide clinical relevance by measuring the levels of PrP in bodily fluids of asthmatic patients, a disease that involves the activation of mast cells. We found an N-terminal fragment of PrP could be detected in human sputum and serum and the amount of this PrP fragment was decreased in the serum of patients with asthma.

16.
Mol Neurodegener ; 19(1): 42, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802940

RESUMEN

Microglia play diverse pathophysiological roles in Alzheimer's disease (AD), with genetic susceptibility factors skewing microglial cell function to influence AD risk. CD33 is an immunomodulatory receptor associated with AD susceptibility through a single nucleotide polymorphism that modulates mRNA splicing, skewing protein expression from a long protein isoform (CD33M) to a short isoform (CD33m). Understanding how human CD33 isoforms differentially impact microglial cell function in vivo has been challenging due to functional divergence of CD33 between mice and humans. We address this challenge by studying transgenic mice expressing either of the human CD33 isoforms crossed with the 5XFAD mouse model of amyloidosis and find that human CD33 isoforms have opposing effects on the response of microglia to amyloid-ß (Aß) deposition. Mice expressing CD33M have increased Aß levels, more diffuse plaques, fewer disease-associated microglia, and more dystrophic neurites compared to 5XFAD control mice. Conversely, CD33m promotes plaque compaction and microglia-plaque contacts, and minimizes neuritic plaque pathology, highlighting an AD protective role for this isoform. Protective phenotypes driven by CD33m are detected at an earlier timepoint compared to the more aggressive pathology in CD33M mice that appears at a later timepoint, suggesting that CD33m has a more prominent impact on microglia cell function at earlier stages of disease progression. In addition to divergent roles in modulating phagocytosis, scRNAseq and proteomics analyses demonstrate that CD33m+ microglia upregulate nestin, an intermediate filament involved in cell migration, at plaque contact sites. Overall, our work provides new functional insights into how CD33, as a top genetic susceptibility factor for AD, modulates microglial cell function.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Ratones Transgénicos , Microglía , Isoformas de Proteínas , Lectina 3 Similar a Ig de Unión al Ácido Siálico , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Microglía/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Humanos , Ratones , Isoformas de Proteínas/metabolismo , Péptidos beta-Amiloides/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patología
17.
J Biol Chem ; 287(4): 2398-409, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22134915

RESUMEN

Lewy bodies and Lewy neurites, neuropathological hallmarks of several neurological diseases, are mainly made of filamentous assemblies of α-synuclein. However, other macromolecules including Tau, ubiquitin, glyceraldehyde-3-phosphate dehydrogenase, and glycosaminoglycans are routinely found associated with these amyloid deposits. Glyceraldehyde-3-phosphate dehydrogenase is a glycolytic enzyme that can form fibrillar aggregates in the presence of acidic membranes, but its role in Parkinson disease is still unknown. In this work, the ability of heparin to trigger the amyloid aggregation of this protein at physiological conditions of pH and temperature is demonstrated by infrared and fluorescence spectroscopy, dynamic light scattering, small angle x-ray scattering, circular dichroism, and fluorescence microscopy. Aggregation proceeds through the formation of short rod-like oligomers, which elongates in one dimension. Heparan sulfate was also capable of inducing glyceraldehyde-3-phosphate dehydrogenase aggregation, but chondroitin sulfates A, B, and C together with dextran sulfate had a negligible effect. Aided with molecular docking simulations, a putative binding site on the protein is proposed providing a rational explanation for the structural specificity of heparin and heparan sulfate. Finally, it is demonstrated that in vitro the early oligomers present in the glyceraldehyde-3-phosphate dehydrogenase fibrillation pathway promote α-synuclein aggregation. Taking into account the toxicity of α-synuclein prefibrillar species, the heparin-induced glyceraldehyde-3-phosphate dehydrogenase early oligomers might come in useful as a novel therapeutic strategy in Parkinson disease and other synucleinopathies.


Asunto(s)
Amiloide/química , Gliceraldehído-3-Fosfato Deshidrogenasas/química , Heparina/química , Multimerización de Proteína , alfa-Sinucleína/química , Amiloide/metabolismo , Animales , Sulfatos de Condroitina/química , Sulfatos de Condroitina/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Enfermedad de Parkinson/metabolismo , Conejos , alfa-Sinucleína/metabolismo
18.
Protein Sci ; 31(12): e4477, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36254680

RESUMEN

Prion diseases are fatal neurodegenerative diseases caused by pathogenic misfolding of the prion protein, PrP. They are transmissible between hosts, and sometimes between different species, as with transmission of bovine spongiform encephalopathy to humans. Although PrP is found in a wide range of vertebrates, prion diseases are seen only in certain mammals, suggesting that infectious misfolding was a recent evolutionary development. To explore when PrP acquired the ability to misfold infectiously, we reconstructed the sequences of ancestral versions of PrP from the last common primate, primate-rodent, artiodactyl, placental, bird, and amniote. Recombinant ancestral PrPs were then tested for their ability to form ß-sheet aggregates, either spontaneously or when seeded with infectious prion strains from human, cervid, or rodent species. The ability to aggregate developed after the oldest ancestor (last common amniote), and aggregation capabilities diverged along evolutionary pathways consistent with modern-day susceptibilities. Ancestral bird PrP could not be seeded with modern-day prions, just as modern-day birds are resistant to prion disease. Computational modeling of structures suggested that differences in helix 2 could account for the resistance of ancestral bird PrP to seeding. Interestingly, ancestral primate PrP could be converted by all prion seeds, including both human and cervid prions, raising the possibility that species descended from an ancestral primate have retained the susceptibility to conversion by cervid prions. More generally, the results suggest that susceptibility to prion disease emerged prior to ~100 million years ago, with placental mammals possibly being generally susceptible to disease.


Asunto(s)
Enfermedades por Prión , Priones , Embarazo , Animales , Bovinos , Femenino , Humanos , Proteínas Priónicas/química , Placenta/metabolismo , Priones/metabolismo , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Mamíferos
19.
Nature ; 437(7056): 257-61, 2005 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-16148934

RESUMEN

Neurodegenerative diseases such as Alzheimer's, Parkinson's and the transmissible spongiform encephalopathies (TSEs) are characterized by abnormal protein deposits, often with large amyloid fibrils. However, questions have arisen as to whether such fibrils or smaller subfibrillar oligomers are the prime causes of disease. Abnormal deposits in TSEs are rich in PrP(res), a protease-resistant form of the PrP protein with the ability to convert the normal, protease-sensitive form of the protein (PrP(sen)) into PrP(res) (ref. 3). TSEs can be transmitted between organisms by an enigmatic agent (prion) that contains PrP(res) (refs 4 and 5). To evaluate systematically the relationship between infectivity, converting activity and the size of various PrP(res)-containing aggregates, PrP(res) was partially disaggregated, fractionated by size and analysed by light scattering and non-denaturing gel electrophoresis. Our analyses revealed that with respect to PrP content, infectivity and converting activity peaked markedly in 17-27-nm (300-600 kDa) particles, whereas these activities were substantially lower in large fibrils and virtually absent in oligomers of < or =5 PrP molecules. These results suggest that non-fibrillar particles, with masses equivalent to 14-28 PrP molecules, are the most efficient initiators of TSE disease.


Asunto(s)
Proteínas PrPSc/química , Proteínas PrPSc/patogenicidad , Enfermedades por Prión/metabolismo , Enfermedades por Prión/transmisión , Animales , Encéfalo , Fraccionamiento Químico , Cricetinae , Electroforesis en Gel de Poliacrilamida , Immunoblotting , Microscopía Electrónica de Transmisión , Peso Molecular , Proteínas PrPSc/aislamiento & purificación , Proteínas PrPSc/ultraestructura , Estructura Cuaternaria de Proteína , Dispersión de Radiación
20.
Biomolecules ; 11(1)2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-33466947

RESUMEN

Prion diseases are the hallmark protein folding neurodegenerative disease. Their transmissible nature has allowed for the development of many different cellular models of disease where prion propagation and sometimes pathology can be induced. This review examines the range of simple cell cultures to more complex neurospheres, organoid, and organotypic slice cultures that have been used to study prion disease pathogenesis and to test therapeutics. We highlight the advantages and disadvantages of each system, giving special consideration to the importance of strains when choosing a model and when interpreting results, as not all systems propagate all strains, and in some cases, the technique used, or treatment applied, can alter the very strain properties being studied.


Asunto(s)
Técnicas de Cultivo de Célula , Modelos Biológicos , Organoides/metabolismo , Priones/metabolismo , Animales , Encéfalo/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA