Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Mol Med ; 27(1): 102, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34496741

RESUMEN

BACKGROUND: The human L39X phospholamban (PLN) cardiomyopathic mutant has previously been reported as a null mutation but the detailed molecular pathways that lead to the complete lack of detectable protein remain to be clarified. Previous studies have shown the implication between an impaired cellular degradation homeostasis and cardiomyopathy development. Therefore, uncovering the underlying mechanism responsible for the lack of PLN protein has important implications in understanding the patient pathology, chronic human calcium dysregulation and aid the development of potential therapeutics. METHODS: A panel of mutant and wild-type reporter tagged PLN modified mRNA (modRNA) constructs were transfected in human embryonic stem cell-derived cardiomyocytes. Lysosomal and proteasomal chemical inhibitors were used together with cell imaging and protein analysis tools in order to dissect degradation pathways associated with expressed PLN constructs. Transcriptional profiling of the cardiomyocytes transfected by wild-type or L39X mutant PLN modRNA was analysed with bulk RNA sequencing. RESULTS: Our modRNA assay system revealed that transfected L39X mRNA was stable and actively translated in vitro but with only trace amount of protein detectable. Proteasomal inhibition of cardiomyocytes transfected with L39X mutant PLN modRNA showed a fourfold increase in protein expression levels. Additionally, RNA sequencing analysis of protein degradational pathways showed a significant distinct transcriptomic signature between wild-type and L39X mutant PLN modRNA transfected cardiomyocytes. CONCLUSION: Our results demonstrate that the cardiomyopathic PLN null mutant L39X is rapidly, actively and specifically degraded by proteasomal pathways. Herein, and to the best of our knowledge, we report for the first time the usage of modified mRNAs to screen for and illuminate alternative molecular pathways found in genes associated with inherited cardiomyopathies.


Asunto(s)
Proteínas de Unión al Calcio/genética , Cardiomiopatías/etiología , Cardiomiopatías/metabolismo , Homocigoto , Mutación , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/genética , Alelos , Sustitución de Aminoácidos , Biomarcadores , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Cardiomiopatías/diagnóstico , Línea Celular , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Humanos , Biosíntesis de Proteínas , Estabilidad del ARN
2.
Small ; 17(26): e2007188, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34050722

RESUMEN

Peptides and proteins have evolved to self-assemble into supramolecular entities through a set of non-covalent interactions. Such structures and materials provide the functional basis of life. Crucially, biomolecular assembly processes can be highly sensitive to and modulated by environmental conditions, including temperature, light, ionic strength and pH, providing the inspiration for the development of new classes of responsive functional materials based on peptide building blocks. Here, it is shown that the stimuli-responsive assembly of amyloidogenic peptide can be used as the basis of environmentally responsive microcapsules which exhibit release characteristics triggered by a change in pH. The microcapsules are biocompatible and biodegradable and may act as vehicles for controlled release of a wide range of biomolecules. Cryo-SEM images reveal the formation of a fibrillar network of the capsule interior with discrete compartments in which cargo molecules can be stored. In addition, the reversible formation of these microcapsules by modulating the solution pH is investigated and their potential application for the controlled release of encapsulated cargo molecules, including antibodies, is shown. These results suggest that the approach described here represents a promising venue for generating pH-responsive functional peptide-based materials for a wide range of potential applications for molecular encapsulation, storage, and release.


Asunto(s)
Péptidos , Cápsulas , Concentración de Iones de Hidrógeno , Temperatura
3.
Proc Natl Acad Sci U S A ; 114(25): 6444-6449, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28584111

RESUMEN

The aggregation of the amyloid ß peptide (Aß) into amyloid fibrils is a defining characteristic of Alzheimer's disease. Because of the complexity of this aggregation process, effective therapeutic inhibitors will need to target the specific microscopic steps that lead to the production of neurotoxic species. We introduce a strategy for generating fibril-specific antibodies that selectively suppress fibril-dependent secondary nucleation of the 42-residue form of Aß (Aß42). We target this step because it has been shown to produce the majority of neurotoxic species during aggregation of Aß42. Starting from large phage display libraries of single-chain antibody fragments (scFvs), the three-stage approach that we describe includes (i) selection of scFvs with high affinity for Aß42 fibrils after removal of scFvs that bind Aß42 in its monomeric form; (ii) ranking, by surface plasmon resonance affinity measurements, of the resulting candidate scFvs that bind to the Aß42 fibrils; and (iii) kinetic screening and analysis to find the scFvs that inhibit selectively the fibril-catalyzed secondary nucleation process in Aß42 aggregation. By applying this approach, we have identified four scFvs that inhibit specifically the fibril-dependent secondary nucleation process. Our method also makes it possible to discard antibodies that inhibit elongation, an important factor because the suppression of elongation does not target directly the production of toxic oligomers and may even lead to its increase. On the basis of our results, we suggest that the method described here could form the basis for rationally designed immunotherapy strategies to combat Alzheimer's and related neurodegenerative diseases.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Anticuerpos/metabolismo , Bacteriófagos/metabolismo , Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Proteínas Amiloidogénicas/metabolismo , Amiloidosis/metabolismo , Humanos , Cinética , Fragmentos de Péptidos/metabolismo , Biblioteca de Péptidos
4.
Glia ; 66(1): 191-205, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29024008

RESUMEN

Neuroinflammation mediated by chronically activated microglia, largely caused by abnormal accumulation of misfolded α-synuclein (αSyn) protein, is known to contribute to the pathophysiology of Parkinson's disease (PD). In this work, based on the immunomodulatory activities displayed by particular heat-shock proteins (HSPs), we tested a novel vaccination strategy that used a combination of αSyn and Grp94 (HSPC4 or Gp96) chaperone and a murine PD model. We used two different procedures, first, the adoptive transfer of splenocytes from αSyn/Grp94-immunized mice to recipient animals, and second, direct immunization with αSyn/Grp94, to study the effects in a chronic mouse MPTP-model of parkinsonism. We found that both approaches promoted a distinct profile in the peripheral system-supported by humoral and cellular immunity-consisting of a Th1-shifted αSyn-specific response accompanied by an immune-regulatory/Th2-skewed general phenotype. Remarkably, this mixed profile sustained by αSyn/Grp94 immunization led to strong suppression of microglial activation in the substantia nigra and striatum, pointing to a newly described positive effect of anti-αSyn Th1-responses in the context of PD. This strategy is the first to target αSyn and report the suppression of PD-associated microgliosis. Overall, we show that the αSyn/Grp94 combination supports a distinct and long-lasting immune profile in the peripheral system, which has an impact at the CNS level by suppressing chronic microglial activation in an MPTP model of PD. Furthermore, our study demonstrates that reshaping peripheral immunity by vaccination with appropriate misfolding protein/HSP combinations could be highly beneficial as a treatment for neurodegenerative misfolding diseases.


Asunto(s)
Gliosis/etiología , Gliosis/terapia , Inmunización/métodos , Intoxicación por MPTP , Glicoproteínas de Membrana/inmunología , alfa-Sinucleína/inmunología , Traslado Adoptivo , Análisis de Varianza , Animales , Antígenos CD4/metabolismo , Enfermedad Crónica , Citocinas/metabolismo , Modelos Animales de Enfermedad , Intoxicación por MPTP/inducido químicamente , Intoxicación por MPTP/complicaciones , Intoxicación por MPTP/inmunología , Intoxicación por MPTP/terapia , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Sustancia Negra/metabolismo , Sustancia Negra/patología , Linfocitos T Reguladores/metabolismo
5.
Proc Natl Acad Sci U S A ; 112(16): E1994-2003, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25855634

RESUMEN

We describe the isolation and detailed structural characterization of stable toxic oligomers of α-synuclein that have accumulated during the process of amyloid formation. Our approach has allowed us to identify distinct subgroups of oligomers and to probe their molecular architectures by using cryo-electron microscopy (cryoEM) image reconstruction techniques. Although the oligomers exist in a range of sizes, with different extents and nature of ß-sheet content and exposed hydrophobicity, they all possess a hollow cylindrical architecture with similarities to certain types of amyloid fibril, suggesting that the accumulation of at least some forms of amyloid oligomers is likely to be a consequence of very slow rates of rearrangement of their ß-sheet structures. Our findings reveal the inherent multiplicity of the process of protein misfolding and the key role the ß-sheet geometry acquired in the early stages of the self-assembly process plays in dictating the kinetic stability and the pathological nature of individual oligomeric species.


Asunto(s)
Amiloide/química , Multimerización de Proteína , alfa-Sinucleína/química , alfa-Sinucleína/toxicidad , Microscopía por Crioelectrón , Interacciones Hidrofóbicas e Hidrofílicas , Imagenología Tridimensional , Modelos Moleculares , Peso Molecular , Estructura Secundaria de Proteína , alfa-Sinucleína/ultraestructura
6.
BMC Biol ; 15(1): 57, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28673288

RESUMEN

BACKGROUND: The aggregation of the protein ɑ-synuclein (ɑS) underlies a range of increasingly common neurodegenerative disorders including Parkinson's disease. One widely explored therapeutic strategy for these conditions is the use of antibodies to target aggregated ɑS, although a detailed molecular-level mechanism of the action of such species remains elusive. Here, we characterize ɑS aggregation in vitro in the presence of two ɑS-specific single-domain antibodies (nanobodies), NbSyn2 and NbSyn87, which bind to the highly accessible C-terminal region of ɑS. RESULTS: We show that both nanobodies inhibit the formation of ɑS fibrils. Furthermore, using single-molecule fluorescence techniques, we demonstrate that nanobody binding promotes a rapid conformational conversion from more stable oligomers to less stable oligomers of ɑS, leading to a dramatic reduction in oligomer-induced cellular toxicity. CONCLUSIONS: The results indicate a novel mechanism by which diseases associated with protein aggregation can be inhibited, and suggest that NbSyn2 and NbSyn87 could have significant therapeutic potential.


Asunto(s)
Anticuerpos de Dominio Único/metabolismo , alfa-Sinucleína/metabolismo , Humanos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Unión Proteica
7.
FASEB J ; 30(2): 564-77, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26443817

RESUMEN

We have investigated the potential role of molecular chaperones as modulators of the immune response by using α-synuclein (αSyn) as an aggregation-prone model protein. We first performed an in vitro immunoscreening with 21 preselected candidate chaperones and selected 2 from this set as displaying immunological activity with differential profiles, Grp94/Gp96 and FKBP4/52. We then immunized mice with both chaperone/α-synuclein combinations using monomeric or oligomeric α-synuclein (MαSyn or OαSyn, respectively), and we characterized the immune response generated in each case. We found that Grp94 promoted αSyn-specific T-helper (Th)1/Th17 and IgG1 antibody responses (up to a 3-fold increase) with MαSyn and OαSyn, respectively, coupled to a Th2-type general phenotype (generating 2.5-fold higher IgG1/IgG2 levels). In addition, we observed that FKBP4 favored a Th1-skewed phenotype with MαSyn but strongly supported a Th2-type phenotype with OαSyn (with a 3-fold higher IL-10/IFN-γ serum levels). Importantly, results from adoptive transfer of splenocytes from immunized animals in a Parkinson's disease mouse model indicates that these effects are robust, stable in time, and physiologically relevant. Taken together, Grp94 and FKBP4 are able to generate differential immune responses to α-synuclein-based immunizations, depending both on the nature of the chaperone and on the aggregation state of α-synuclein. Our work reveals that several chaperones are potential modulators of the immune response and suggests that different chaperones could be exploited to redirect the amyloid-elicited immunity both for basic studies of the immunological processes associated with neurodegeneration and for immunotherapy of pathologies associated with protein misfolding and aggregation.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/fisiología , Proteínas de Unión a Tacrolimus/metabolismo , alfa-Sinucleína/metabolismo , Inmunidad Adaptativa , Animales , Regulación de la Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Inmunidad Innata , Masculino , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Pliegue de Proteína , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/inmunología , alfa-Sinucleína/genética
8.
Biomacromolecules ; 18(10): 3052-3059, 2017 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-28792742

RESUMEN

In nature, a wide range of functional materials is based on proteins. Increasing attention is also turning to the use of proteins as artificial biomaterials in the form of films, gels, particles, and fibrils that offer great potential for applications in areas ranging from molecular medicine to materials science. To date, however, most such applications have been limited to single component materials despite the fact that their natural analogues are composed of multiple types of proteins with a variety of functionalities that are coassembled in a highly organized manner on the micrometer scale, a process that is currently challenging to achieve in the laboratory. Here, we demonstrate the fabrication of multicomponent protein microcapsules where the different components are positioned in a controlled manner. We use molecular self-assembly to generate multicomponent structures on the nanometer scale and droplet microfluidics to bring together the different components on the micrometer scale. Using this approach, we synthesize a wide range of multiprotein microcapsules containing three well-characterized proteins: glucagon, insulin, and lysozyme. The localization of each protein component in multishell microcapsules has been detected by labeling protein molecules with different fluorophores, and the final three-dimensional microcapsule structure has been resolved by using confocal microscopy together with image analysis techniques. In addition, we show that these structures can be used to tailor the release of such functional proteins in a sequential manner. Moreover, our observations demonstrate that the protein release mechanism from multishell capsules is driven by the kinetic control of mass transport of the cargo and by the dissolution of the shells. The ability to generate artificial materials that incorporate a variety of different proteins with distinct functionalities increases the breadth of the potential applications of artificial protein-based materials and provides opportunities to design more refined functional protein delivery systems.


Asunto(s)
Cápsulas/química , Liberación de Fármacos , Glucagón/química , Insulina/química , Muramidasa/química
9.
Biophys J ; 111(11): 2358-2367, 2016 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-27926837

RESUMEN

The conversion of human lysozyme into amyloid fibrils is associated with a rare but fatal hereditary form of nonneuropathic systemic amyloidosis. The accumulation of large amounts of aggregated protein is thought to be initiated by the formation of transient intermediate species of disease-related lysozyme variants, essentially due to the loss of global cooperativity under physiologically relevant conditions. Interestingly, all five naturally occurring, amyloidogenic, single-point mutations are located in the ß-domain of lysozyme, the region that is predominantly unfolded during the formation of the transient intermediate species. Given the lack of known naturally occurring, amyloidogenic, single-point mutations in the α-domain, we chose three specific mutations to address the effects that location may have on native-state dynamics, as studied by hydrogen-deuterium (HD) exchange experiments analyzed by NMR spectroscopy, and mass spectrometry. We compared the effect of a destabilizing α-domain mutation (I23A) with that of the well-characterized I59T ß-domain variant. We also investigated the effect of a mutation that has minor effects on native-state stability at the domain interface (I56V) and compared it with that of a variant with similar stability within the C-helix (I89V). We show that when variants have similar reduced native-state stabilities, the location of the mutation (I23A versus I59T) is crucial to the native-state dynamics, with the α-domain mutation having a significantly lower ability to populate transient intermediate species under physiologically relevant conditions. Interestingly, the mutation at the interface (I56V) has a greater effect in facilitating the formation of transient intermediate species at elevated temperatures compared with the variants containing α-domain mutations, even though this mutation results in only minor changes to the native-state stability of lysozyme. These findings reveal that the location of specific mutations is an important factor in determining the native-state dynamical properties of human lysozyme in the context of its propensity to populate the aggregation-prone transient intermediate species associated with pathogenic amyloid formation.


Asunto(s)
Muramidasa/química , Muramidasa/genética , Mutación , Amiloide/química , Estabilidad de Enzimas , Humanos , Modelos Moleculares , Dominios Proteicos , Multimerización de Proteína , Estructura Secundaria de Proteína
10.
Biochemistry ; 55(22): 3116-22, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27096466

RESUMEN

α-Synuclein is an intrinsically disordered protein whose aggregation is associated with Parkinson's disease and other related neurodegenerative disorders. Recently, two single-domain camelid antibodies (nanobodies) were shown to bind α-synuclein with high affinity. Herein, we investigated how these two nanobodies (NbSyn2 and NbSyn87), which are directed to two distinct epitopes within the C-terminal domain of α-synuclein, affect the conformational properties of this protein. Our results suggest that nanobody NbSyn2, which binds to the five C-terminal residues of α-synuclein (residues 136-140), does not disrupt the transient long-range interactions that generate a degree of compaction within the native structural ensemble of α-synuclein. In contrast, the data that we report indicate that NbSyn87, which targets a central region within the C-terminal domain (residues 118-128), has more substantial effects on the fluctuating secondary and tertiary structure of the protein. These results are consistent with the different effects that the two nanobodies have on the aggregation behavior of α-synuclein in vitro. Our findings thus provide new insights into the type of effects that nanobodies can have on the conformational ensemble of α-synuclein.


Asunto(s)
Epítopos/metabolismo , Anticuerpos de Dominio Único/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Animales , Camélidos del Nuevo Mundo , Epítopos/inmunología , Humanos , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Dominios Proteicos , Anticuerpos de Dominio Único/inmunología , alfa-Sinucleína/inmunología
11.
Chembiochem ; 17(20): 1920-1924, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27472818

RESUMEN

The measurement of molecular interactions with pathological protein aggregates, including amyloid fibrils, is of central importance in the context of the development of diagnostic and therapeutic strategies against protein misfolding disorders. Probing such interactions by conventional methods can, however, be challenging because of the supramolecular nature of protein aggregates, their heterogeneity, and their often dynamic nature. Here we demonstrate that direct measurement of diffusion on a microfluidic platform enables the determination of affinity and kinetics data for ligand binding to amyloid fibrils in solution. This method yields rapid binding information from only microlitres of sample, and is therefore a powerful technique for identifying and characterising molecular species with potential therapeutic or diagnostic application.


Asunto(s)
Amiloide/aislamiento & purificación , Técnicas Analíticas Microfluídicas , Deficiencias en la Proteostasis/diagnóstico , Tiazoles/aislamiento & purificación , Amiloide/antagonistas & inhibidores , Amiloide/química , Benzotiazoles , Sitios de Unión/efectos de los fármacos , Difusión , Humanos , Cinética , Ligandos , Nanopartículas/química , Tamaño de la Partícula , Agregado de Proteínas/efectos de los fármacos , Deficiencias en la Proteostasis/tratamiento farmacológico , Tiazoles/química , Tiazoles/farmacología
12.
Biochim Biophys Acta ; 1844(11): 1907-1919, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25194824

RESUMEN

Protein misfolding disorders, including the neurodegenerative conditions Alzheimer's disease (AD) and Parkinson's disease (PD) represent one of the major medical challenges or our time. The underlying molecular mechanisms that govern protein misfolding and its links with disease are very complex processes, involving the formation of transiently populated but highly toxic molecular species within the crowded environment of the cell and tissue. Nevertheless, much progress has been made in understanding these events in recent years through innovative experiments and therapeutic strategies, and in this review we present an overview of the key roles of antibodies and antibody fragments in these endeavors. We discuss in particular how these species are being used in combination with a variety of powerful biochemical and biophysical methodologies, including a range of spectroscopic and microscopic techniques applied not just in vitro but also in situ and in vivo, both to gain a better understanding of the mechanistic nature of protein misfolding and aggregation and also to design novel therapeutic strategies to combat the family of diseases with which they are associated. This article is part of a Special Issue entitled: Recent advances in molecular engineering of antibody.

13.
Proc Natl Acad Sci U S A ; 108(36): 14746-51, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21876182

RESUMEN

The crucial early stages of amyloid growth, in which normally soluble proteins are converted into fibrillar nanostructures, are challenging to study using conventional techniques yet are critical to the protein aggregation phenomena implicated in many common pathologies. As with all nucleation and growth phenomena, it is difficult to track individual nuclei in traditional macroscopic experiments, which probe the overall temporal evolution of the sample, but do not yield detailed information on the primary nucleation step as they mix independent stochastic events into an ensemble measurement. To overcome this limitation, we have developed microdroplet assays enabling us to detect single primary nucleation events and to monitor their subsequent spatial as well as temporal evolution, both of which we find to be determined by secondary nucleation phenomena. By deforming the droplets to high aspect ratio, we visualize in real-time propagating waves of protein assembly emanating from discrete primary nucleation sites. We show that, in contrast to classical gelation phenomena, the primary nucleation step is characterized by a striking dependence on system size, and the filamentous protein self-assembly process involves a highly nonuniform spatial distribution of aggregates. These findings deviate markedly from the current picture of amyloid growth and uncover a general driving force, originating from confinement, which, together with biological quality control mechanisms, helps proteins remain soluble and therefore functional in nature.


Asunto(s)
Amiloide/química , Modelos Químicos , Amiloide/metabolismo , Animales , Humanos , Solubilidad
14.
Biochemistry ; 51(44): 8771-8, 2012 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-23003198

RESUMEN

The aggregation process of α-synuclein, a protein closely associated with Parkinson's disease, is highly sensitive to sequence variations. It is therefore of great importance to understand the factors that define the aggregation propensity of specific mutational variants as well as their toxic behavior in the cellular environment. In this context, we investigated the extent to which the aggregation behavior of α-synuclein can be altered to resemble that of ß-synuclein, an aggregation-resistant homologue of α-synuclein not associated with disease, by swapping residues between the two proteins. Because of the vast number of possible swaps, we have applied a rational design procedure to single out a mutational variant, called α2ß, in which two short stretches of the sequence in the NAC region have been replaced in α-synuclein from ß-synuclein. We find not only that the aggregation rate of α2ß is close to that of ß-synuclein, being much lower than that of α-synuclein, but also that α2ß effectively changes the cellular toxicity of α-synuclein to a value similar to that of ß-synuclein upon exposure of SH-SY5Y cells to preformed oligomers. Remarkably, control experiments on the corresponding mutational variant of ß-synuclein, called ß2α, confirmed that the mutations that we have identified also shift the aggregation behavior of this protein toward that of α-synuclein. These results demonstrate that it is becoming possible to control in quantitative detail the sequence code that defines the aggregation behavior and toxicity of α-synuclein.


Asunto(s)
Estructura Cuaternaria de Proteína , alfa-Sinucleína/química , Sinucleína beta/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Supervivencia Celular , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia , Células Tumorales Cultivadas , alfa-Sinucleína/genética , alfa-Sinucleína/toxicidad , Sinucleína beta/toxicidad
15.
Nat Commun ; 13(1): 3018, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35641497

RESUMEN

The dysregulated physical interaction between two intracellular membrane proteins, the sarco/endoplasmic reticulum Ca2+ ATPase and its reversible inhibitor phospholamban, induces heart failure by inhibiting calcium cycling. While phospholamban is a bona-fide therapeutic target, approaches to selectively inhibit this protein remain elusive. Here, we report the in vivo application of intracellular acting antibodies (intrabodies), derived from the variable domain of camelid heavy-chain antibodies, to modulate the function of phospholamban. Using a synthetic VHH phage-display library, we identify intrabodies with high affinity and specificity for different conformational states of phospholamban. Rapid phenotypic screening, via modified mRNA transfection of primary cells and tissue, efficiently identifies the intrabody with most desirable features. Adeno-associated virus mediated delivery of this intrabody results in improvement of cardiac performance in a murine heart failure model. Our strategy for generating intrabodies to investigate cardiac disease combined with modified mRNA and adeno-associated virus screening could reveal unique future therapeutic opportunities.


Asunto(s)
Proteínas de Unión al Calcio , Insuficiencia Cardíaca , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Corazón , Ratones , ARN Mensajero
16.
Biochemistry ; 47(42): 11041-54, 2008 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-18816062

RESUMEN

A single-domain fragment, cAb-HuL22, of a camelid heavy-chain antibody specific for the active site of human lysozyme has been generated, and its effects on the properties of the I56T and D67H amyloidogenic variants of human lysozyme, which are associated with a form of systemic amyloidosis, have been investigated by a wide range of biophysical techniques. Pulse-labeling hydrogen-deuterium exchange experiments monitored by mass spectrometry reveal that binding of the antibody fragment strongly inhibits the locally cooperative unfolding of the I56T and D67H variants and restores their global cooperativity to that characteristic of the wild-type protein. The antibody fragment was, however, not stable enough under the conditions used to explore its ability to perturb the aggregation behavior of the lysozyme amyloidogenic variants. We therefore engineered a more stable version of cAb-HuL22 by adding a disulfide bridge between the two beta-sheets in the hydrophobic core of the protein. The binding of this engineered antibody fragment to the amyloidogenic variants of lysozyme inhibited their aggregation into fibrils. These findings support the premise that the reduction in global cooperativity caused by the pathogenic mutations in the lysozyme gene is the determining feature underlying their amyloidogenicity. These observations indicate further that molecular targeting of enzyme active sites, and of protein binding sites in general, is an effective strategy for inhibiting or preventing the aberrant self-assembly process that is often a consequence of protein mutation and the origin of pathogenicity. Moreover, this work further demonstrates the unique properties of camelid single-domain antibody fragments as structural probes for studying the mechanism of aggregation and as potential inhibitors of fibril formation.


Asunto(s)
Amiloide/antagonistas & inhibidores , Camelus/inmunología , Fragmentos de Inmunoglobulinas/metabolismo , Muramidasa/inmunología , Secuencia de Aminoácidos , Amiloide/química , Amiloide/inmunología , Amiloide/metabolismo , Animales , Afinidad de Anticuerpos , Camelus/genética , Dominio Catalítico/inmunología , Humanos , Fragmentos de Inmunoglobulinas/genética , Técnicas In Vitro , Datos de Secuencia Molecular , Muramidasa/antagonistas & inhibidores , Muramidasa/química , Muramidasa/metabolismo , Resonancia Magnética Nuclear Biomolecular , Ingeniería de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
17.
NPJ Parkinsons Dis ; 4: 25, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30155513

RESUMEN

Therapeutics designed to target α-synuclein (α-syn) aggregation may be critical in halting the progression of pathology in Parkinson's disease (PD) patients. Nanobodies are single-domain antibody fragments that bind with antibody specificity, but allow readier genetic engineering and delivery. When expressed intracellularly as intrabodies, anti-α-syn nanobodies fused to a proteasome-targeting proline, aspartate or glutamate, serine, and threonine (PEST) motif can modulate monomeric concentrations of target proteins. Here we aimed to validate and compare the in vivo therapeutic potential of gene therapy delivery of two proteasome-directed nanobodies selectively targeting α-syn in a synuclein overexpression-based PD model: VH14*PEST (non-amyloid component region) and NbSyn87*PEST (C-terminal region). Stereotaxic injections of adeno-associated viral 5-α-syn (AAV5-α-syn) into the substantia nigra (SN) were performed in Sprague-Dawley rats that were sorted into three cohorts based on pre-operative behavioral testing. Rats were treated with unilateral SN injections of vectors for VH14*PEST, NbSyn87*PEST, or injected with saline 3 weeks post lesion. Post-mortem assessments of the SN showed that both nanobodies markedly reduced the level of phosphorylated Serine-129 α-syn labeling relative to saline-treated animals. VH14*PEST showed considerable maintenance of striatal dopaminergic tone in comparison to saline-treated and NbSyn87*PEST-treated animals as measured by tyrosine hydroxylase immunoreactivity (optical density), DAT immunoreactivity (optical density), and dopamine concentration (high-performance liquid chromatography). Microglial accumulation and inflammatory response, assessed by stereological counts of Iba-1-labeled cells, was modestly increased in NbSyn87*PEST-injected rats but not in VH14*PEST-treated or saline-treated animals. Modest behavioral rescue was also observed, although there was pronounced variability among individual animals. These data validate in vivo therapeutic efficacy of vector-delivered intracellular nanobodies targeting α-syn misfolding and aggregation in synucleinopathies such as PD.

18.
Protein Sci ; 27(7): 1262-1274, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29603451

RESUMEN

Intracellular deposits of α-synuclein in the form of Lewy bodies are major hallmarks of Parkinson's disease (PD) and a range of related neurodegenerative disorders. Post-translational modifications (PTMs) of α-synuclein are increasingly thought to be major modulators of its structure, function, degradation and toxicity. Among these PTMs, phosphorylation near the C-terminus at S129 has emerged as a dominant pathogenic modification as it is consistently observed to occur within the brain and cerebrospinal fluid (CSF) of post-mortem PD patients, and its level appears to correlate with disease progression. Phosphorylation at the neighboring tyrosine residue Y125 has also been shown to protect against α-synuclein toxicity in a Drosophila model of PD. In the present study we address the potential roles of C-terminal phosphorylation in modulating the interaction of α-synuclein with other protein partners, using a single domain antibody fragment (NbSyn87) that binds to the C-terminal region of α-synuclein with nanomolar affinity. The results reveal that phosphorylation at S129 has negligible effect on the binding affinity of NbSyn87 to α-synuclein while phosphorylation at Y125, only four residues away, decreases the binding affinity by a factor of 400. These findings show that, despite the fact that α-synuclein is intrinsically disordered in solution, selective phosphorylation can modulate significantly its interactions with other molecules and suggest how this particular form of modification could play a key role in regulating the normal and aberrant function of α-synuclein.


Asunto(s)
Procesamiento Proteico-Postraduccional , Anticuerpos de Dominio Único/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Autopsia , Sitios de Unión , Encéfalo/metabolismo , Humanos , Enfermedad de Parkinson/metabolismo , Fosforilación , Unión Proteica , Serina/metabolismo , Tirosina/metabolismo , alfa-Sinucleína/líquido cefalorraquídeo
19.
J Am Soc Mass Spectrom ; 28(4): 638-646, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27921259

RESUMEN

In the gas phase, protein ions can adopt a broad range of structures, which have been investigated extensively in the past using ion mobility-mass spectrometry (IM-MS)-based methods. Compact ions with low number of charges undergo a Coulomb-driven transition to partially folded species when the charge increases, and finally form extended structures with presumably little or no defined structure when the charge state is high. However, with respect to the secondary structure, IM-MS methods are essentially blind. Infrared (IR) spectroscopy, on the other hand, is sensitive to such structural details and there is increasing evidence that helices as well as ß-sheet-like structures can exist in the gas phase, especially for ions in low charge states. Very recently, we showed that also the fully extended form of highly charged protein ions can adopt a distinct type of secondary structure that features a characteristic C5-type hydrogen bond pattern. Here we use a combination of IM-MS and IR spectroscopy to further investigate the influence of the initial, native conformation on the formation of these structures. Our results indicate that when intramolecular Coulomb-repulsion is large enough to overcome the stabilization energies of the genuine secondary structure, all proteins, regardless of their sequence or native conformation, form C5-type hydrogen bond structures. Furthermore, our results suggest that in highly charged proteins the positioning of charges along the sequence is only marginally influenced by the basicity of individual residues. Graphical Abstract ᅟ.


Asunto(s)
Espectrometría de Masas/métodos , Péptidos/química , Ubiquitina/química , alfa-Sinucleína/química , Secuencia de Aminoácidos , Humanos , Enlace de Hidrógeno , Espectrometría de Movilidad Iónica/métodos , Iones/química , Modelos Moleculares , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Espectrofotometría Infrarroja/métodos
20.
Sci Rep ; 7(1): 15018, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-29101328

RESUMEN

Populating transient and partially unfolded species is a crucial step in the formation and accumulation of amyloid fibrils formed from pathogenic variants of human lysozyme linked with a rare but fatal hereditary systemic amyloidosis. The partially unfolded species possess an unstructured ß-domain and C-helix with the rest of the α-domain remaining native-like. Here we use paramagnetic relaxation enhancement (PRE) measured by NMR spectroscopy to study the transient intermolecular interactions between such intermediate species. Nitroxide spin labels, introduced specifically at three individual lysine residues, generate distinct PRE profiles, indicating the presence of intermolecular interactions between residues within the unfolded ß-domain. This study describes the applicability to PRE NMR measurements of selective lysine labeling, at different sites within a protein, as an alternative to the introduction of spin labels via engineered cysteine residues. These results reveal the importance of the ß-sheet region of lysozyme for initiating self-assembly into amyloid fibrils.


Asunto(s)
Amiloide/metabolismo , Lisina/metabolismo , Muramidasa/metabolismo , Humanos , Resonancia Magnética Nuclear Biomolecular , Estructura Secundaria de Proteína , Marcadores de Spin
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA