Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros











Intervalo de año de publicación
1.
Ann Clin Transl Neurol ; 9(8): 1252-1263, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35903975

RESUMEN

OBJECTIVE: Hereditary transthyretin-mediated amyloidosis is a treatable condition caused by amyloidogenic variants in the transthyretin-gene resulting in severe peripheral neuropathy or cardiomyopathy. Only about a third of over 130 known variants are clearly pathogenic, most are classified as variants of uncertain significance. A clear delineation of these into pathogenic or non-pathogenic is highly desirable but hampered by low frequency and penetrance. We thus sought to characterize their amylogenic potential by an unbiased in vitro approach. METHODS: Thioflavin T and turbidity assays were used to compare the potential of mammalian cell expressed wt-transthyretin and 12 variant proteins (either variants of uncertain significance, benign, pathogenic) to aggregate and produce amyloid fibrils in vitro. As proof of principle, the assays were applied to transthyretin-Ala65Val, a variant that was newly detected in a family with peripheral neuropathy and amyloid deposits in biopsies. In silico analysis was performed to compare the position of the benign and pathogenic variants. RESULTS: Transthyretin-Ala65Val showed a significantly higher amyloidogenic potential than wt-transthyretin, in both turbidity- and Thioflavin T-assays, comparable to known pathogenic variants. The other eight tested variants did not show an increased amyloidogenic potential. In silico structural analysis further confirmed differences between pathogenic and benign variants in position and interactions. INTERPRETATION: We propose a biochemical approach to assess amyloidogenic potential of transthyretin variants. As exemplified by transthyretin-Ala65Val, data of three assays together with histopathology clearly demonstrates its amyloidogenicity.


Asunto(s)
Neuropatías Amiloides Familiares , Prealbúmina , Amiloide/genética , Amiloide/metabolismo , Neuropatías Amiloides Familiares/genética , Neuropatías Amiloides Familiares/metabolismo , Humanos , Prealbúmina/genética
2.
J Biol Chem ; 298(4): 101739, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35182525

RESUMEN

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as a global threat to human health has highlighted the need for the development of novel therapies targeting current and emerging coronaviruses with pandemic potential. The coronavirus main protease (Mpro, also called 3CLpro) is a validated drug target against coronaviruses and has been heavily studied since the emergence of SARS-CoV-2 in late 2019. Here, we report the biophysical and enzymatic characterization of native Mpro, then characterize the steady-state kinetics of several commonly used FRET substrates, fluorogenic substrates, and six of the 11 reported SARS-CoV-2 polyprotein cleavage sequences. We then assessed the suitability of these substrates for high-throughput screening. Guided by our assessment of these substrates, we developed an improved 5-carboxyfluorescein-based FRET substrate, which is better suited for high-throughput screening and is less susceptible to interference and false positives than existing substrates. This study provides a useful framework for the design of coronavirus Mpro enzyme assays to facilitate the discovery and development of therapies targeting Mpro.


Asunto(s)
Proteasas 3C de Coronavirus , Pruebas de Enzimas , Fluoresceínas , SARS-CoV-2 , Antivirales/química , Proteasas 3C de Coronavirus/química , Proteasas 3C de Coronavirus/aislamiento & purificación , Proteasas 3C de Coronavirus/metabolismo , Pruebas de Enzimas/métodos , Fluoresceínas/química , Fluoresceínas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Inhibidores de Proteasas/química , SARS-CoV-2/enzimología , SARS-CoV-2/genética , Tratamiento Farmacológico de COVID-19
3.
Sci Rep ; 11(1): 15520, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330984

RESUMEN

Boron neutron capture therapy (BNCT) is a two-step therapeutic process that utilizes Boron-10 in combination with low energy neutrons to effectively eliminate targeted cells. This therapy is primarily used for difficult to treat head and neck carcinomas; recent advances have expanded this method to cover a broader range of carcinomas. However, it still remains an unconventional therapy where one of the barriers for widespread adoption is the adequate delivery of Boron-10 to target cells. In an effort to address this issue, we examined a unique nanoparticle drug delivery system based on a highly stable and modular proteinaceous nanotube. Initially, we confirmed and structurally analyzed ortho-carborane binding into the cavities of the nanotube. The high ratio of Boron to proteinaceous mass and excellent thermal stability suggest the nanotube system as a suitable candidate for drug delivery into cancer cells. The full physicochemical characterization of the nanotube then allowed for further mechanistic molecular dynamic studies of the ortho-carborane uptake and calculations of corresponding energy profiles. Visualization of the binding event highlighted the protein dynamics and the importance of the interhelical channel formation to allow movement of the boron cluster into the nanotube. Additionally, cell assays showed that the nanotube can penetrate outer membranes of cancer cells followed by localization around the cells' nuclei. This work uses an integrative approach combining experimental data from structural, molecular dynamics simulations and biological experiments to thoroughly present an alternative drug delivery device for BNCT which offers additional benefits over current delivery methods.


Asunto(s)
Terapia por Captura de Neutrón de Boro/métodos , Portadores de Fármacos/química , Nanotubos/química , Boro/química , Isótopos/química
4.
BMC Biol ; 19(1): 120, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34107975

RESUMEN

BACKGROUND: The Hydra head organizer acts as a signaling center that initiates and maintains the primary body axis in steady state polyps and during budding or regeneration. Wnt/beta-Catenin signaling functions as a primary cue controlling this process, but how Wnt ligand activity is locally restricted at the protein level is poorly understood. Here we report a proteomic analysis of Hydra head tissue leading to the identification of an astacin family proteinase as a Wnt processing factor. RESULTS: Hydra astacin-7 (HAS-7) is expressed from gland cells as an apical-distal gradient in the body column, peaking close beneath the tentacle zone. HAS-7 siRNA knockdown abrogates HyWnt3 proteolysis in the head tissue and induces a robust double axis phenotype, which is rescued by simultaneous HyWnt3 knockdown. Accordingly, double axes are also observed in conditions of increased Wnt activity as in transgenic actin::HyWnt3 and HyDkk1/2/4 siRNA treated animals. HyWnt3-induced double axes in Xenopus embryos could be rescued by coinjection of HAS-7 mRNA. Mathematical modelling combined with experimental promotor analysis indicate an indirect regulation of HAS-7 by beta-Catenin, expanding the classical Turing-type activator-inhibitor model. CONCLUSIONS: We show the astacin family protease HAS-7 maintains a single head organizer through proteolysis of HyWnt3. Our data suggest a negative regulatory function of Wnt processing astacin proteinases in the global patterning of the oral-aboral axis in Hydra.


Asunto(s)
Hydra , Animales , Tipificación del Cuerpo , Cabeza , Hydra/genética , Metaloendopeptidasas , Proteolisis , Proteómica , ARN Interferente Pequeño , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
5.
Hypertension ; 76(4): 1176-1184, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32623922

RESUMEN

Preeclampsia is a potentially life-threatening multisystem disease affecting 4% to 8% of pregnant women after the 20th week of gestation. An excess of placental expressed antiangiogenic soluble VEGF (vascular endothelial growth factor)-receptor 1 (soluble FMS-like tyrosine kinase 1) scavenges VEGF and PlGF (placental growth factor), causing generalized endothelial dysfunction. Interventions to restore the angiogenic balance in preeclamptic pregnancies are intensively studied and improve maternal and neonatal outcomes. Especially extracorporeal strategies to remove sFlt-1 are promising in human pregnancy. However, available apheresis systems adsorb sFlt-1 unspecifically and with low efficiency. Affinity-enhanced ligands are needed to improve performance and compatibility of apheresis treatments. Using computerized molecular modeling, we developed multimeric VEGF molecules comprised of single-chain VEGF165 dimers (scVEGF165). A short peptide linker hampers intrachain dimerization to induce assembly preferably as tetrameric molecules as visualized in negative staining electron microscopy. scVEGF165 multimers possess 1.2-fold higher affinity for sFlt-1 as compared to the available antibodies or monomeric VEGF. Consequently, scVEGF multimers have the ability to competitively release sFlt-1 bound PlGF and, in particular, VEGF. In ex vivo adsorption experiments using serum samples from patients with preeclampsia, scVEGF multimers reduce sFlt-1 levels by 85% and increase PlGF and VEGF levels by 20- and 9-fold, respectively. Finally, performance and stability of sFlt-1 capturing scVEGF165 multimers were scrutinized on different matrices of which biocompatible agarose matrix yielded optimal results. We introduce the first VEGF-based highly efficient sFlt-1 apheresis system that is directly applicable in vivo due to utilization of inert agarose matrix, using a homomultimeric form of VEGF165 to restore the angiogenic balance in preeclampsia.


Asunto(s)
Modelos Teóricos , Factor de Crecimiento Placentario/sangre , Preeclampsia/sangre , Factor A de Crecimiento Endotelial Vascular/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Biomarcadores/sangre , Femenino , Humanos , Embarazo
6.
Biophys J ; 118(11): 2726-2740, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32413313

RESUMEN

Oligoadenylate synthetases (OASs) are a family of interferon-inducible enzymes that require double-stranded RNA (dsRNA) as a cofactor. Upon binding dsRNA, OAS undergoes a conformational change and is activated to polymerize ATP into 2'-5'-oligoadenylate chains. The OAS family consists of several isozymes, with unique domain organizations to potentially interact with dsRNA of variable length, providing diversity in viral RNA recognition. In addition, oligomerization of OAS isozymes, potentially OAS1 and OAS2, is hypothesized to be important for 2'-5'-oligoadenylate chain building. In this study, we present the solution conformation of dimeric human OAS2 using an integrated approach involving small-angle x-ray scattering, analytical ultracentrifugation, and dynamic light scattering techniques. We also demonstrate OAS2 dimerization using immunoprecipitation approaches in human cells. Whereas mutation of a key active-site aspartic acid residue prevents OAS2 activity, a C-terminal mutation previously hypothesized to disrupt OAS self-association had only a minor effect on OAS2 activity. Finally, we also present the solution structure of OAS1 monomer and dimer, comparing their hydrodynamic properties with OAS2. In summary, our work presents the first, to our knowledge, dimeric structural models of OAS2 that enhance our understanding of the oligomerization and catalytic function of OAS enzymes.


Asunto(s)
2',5'-Oligoadenilato Sintetasa , Ligasas , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , Nucleótidos de Adenina , Humanos , Hidrodinámica , Oligorribonucleótidos , ARN Bicatenario
7.
Biochim Biophys Acta Biomembr ; 1862(6): 183225, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32126231

RESUMEN

NhaP2 is a K+/H+ antiporter from Vibrio cholerae which consists of a transmembrane domain and a cytoplasmic domain of approximately 200 amino acids, both of which are required for cholera infectivity. Here we present the solution structure for a 165 amino acid minimal cytoplasmic domain (P2MIN) form of the protein. The structure reveals a compact N-terminal domain which resembles a Regulator of Conductance of K+ channels (RCK) domain connected to a more open C-terminal domain via a flexible 20 amino acid linker. NMR titration experiments showed that the protein binds ATP through its N-terminal domain, which was further supported by waterLOGSY and Saturation Transfer Difference NMR experiments. The two-domain organisation of the protein was confirmed by BIOSAXS, which also revealed that there are no detectable-ATP-induced conformational changes in the protein structure. Finally, in contrast to all known RCK domain structures solved to date, the current work shows that the protein is a monomer.


Asunto(s)
Proteínas Bacterianas/química , Antiportadores de Potasio-Hidrógeno/química , Dominios Proteicos , Vibrio cholerae/química , Adenosina Trifosfato/metabolismo , Antiportadores/química , Antiportadores/metabolismo , Proteínas Bacterianas/metabolismo , Sitios de Unión , Citoplasma/química , Resonancia Magnética Nuclear Biomolecular , Antiportadores de Potasio-Hidrógeno/metabolismo , Conformación Proteica
8.
Immunol Cell Biol ; 97(6): 526-537, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30680791

RESUMEN

Human IgG1 and IgG3 antibodies (Abs) can mediate Ab-dependent cellular cytotoxicity (ADCC), and engineering of the Ab Fc (point mutation; defucosylation) has been shown to affect ADCC by modulating affinity for FcRγIIIa. In the absence of a CH 1 domain, many camelid heavy-chain Abs (HCAbs) naturally bear very long and flexible hinge regions connecting their VH H and CH 2 domains. To better understand the influence of hinge length and structure on HCAb ADCC, we produced a series of hinge-engineered epidermal growth factor receptor (EGFR)-specific chimeric camelid VH H-human Fc Abs and characterized their affinities for recombinant EGFR and FcRγIIIa, their binding to EGFR-positive tumor cells, and their ability to elicit ADCC. In the case of one chimeric HCAb (EG2-hFc), we found that variants bearing longer hinges (IgG3 or camelid hinge regions) showed dramatically improved ADCC in comparison with a variant bearing the human IgG1 hinge, in similar fashion to a variant with reduced CH 2 fucosylation. Conversely, an EG2-hFc variant bearing a truncated human IgG1 upper hinge region failed to elicit ADCC. However, there was no consistent association between hinge length and ADCC for four similarly engineered chimeric HCAbs directed against distinct EGFR epitopes. These findings demonstrate that the ADCC of some HCAbs can be modulated simply by varying the length of the Ab hinge. Although this effect appears to be heavily epitope-dependent, this strategy may be useful to consider during the design of VH H-based therapeutic Abs for cancer.


Asunto(s)
Adenocarcinoma/terapia , Anticuerpos Monoclonales/metabolismo , Neoplasias de la Mama/terapia , Inmunoterapia/métodos , Proteínas Recombinantes de Fusión/genética , Adenocarcinoma/inmunología , Animales , Anticuerpos Monoclonales/genética , Afinidad de Anticuerpos , Citotoxicidad Celular Dependiente de Anticuerpos , Neoplasias de la Mama/inmunología , Camelidae , Línea Celular Tumoral , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Femenino , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Mutación/genética , Unión Proteica , Ingeniería de Proteínas
9.
Chemosphere ; 215: 206-213, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30317091

RESUMEN

Halogenated polycyclic aromatic hydrocarbons (HPAHs) were identified in biological samples from the Alberta Oil-Sands Region (AOSR) using gas chromatography coupled with high-resolution time-of-flight mass spectrometry (GC-HRTOF-MS) at a resolving power of 25,000. Knowledge of the electron ionization (EI) fragmentation behavior of individual HPAH isomers, achieved by injecting authentic standards in full-scan MS mode, was paramount in identifying a suite of HPAHs in samples from the AOSR. Confirmation of compounds in biological samples was based on the measured mass accuracy (±3 ppm) of 2 characteristic ions prominent in the EI mass spectra of each compound. Numerous compounds were detected in the high resolution total ion chromatogram in liver extracts of 4 biological species from the AOSR: river otter (Lontra Canadensis), northern pike (Esox lucius), lake whitefish (Coregonus clupeaformis) and snails (Gastropod sp.) many of which remain unidentified. Careful examination of the high-resolution accurate mass data suggests that dichloro-anthracene/phenanthrene, bromo-anthracene/phenanthrene and dibromo-fluorene were present in the biological samples. Lipid corrected concentrations of dichloro-PAHs were estimated to be 16.3 ±â€¯11.4 (n = 4) and 5.5 (n = 1) ng/g in lake whitefish and river otter, respectively. Concentrations of mono-bromo-PAHs were an order of magnitude greater in snails (170.5 ng/g) than in northern pike (12.5 ng/g) while concentrations of dibromo-PAHs were 4 times greater in snails than in northern pike. The detection of these compounds in biota implies that these compounds are bioaccumulative. The liver-based biomagnification factor of the dichloro-PAH congener in the river otter/lake whitefish feeding relationship is much smaller than 1 implying that this compound does not biomagnify.


Asunto(s)
Biota , Cromatografía de Gases y Espectrometría de Masas/métodos , Hidrocarburos Policíclicos Aromáticos/análisis , Alberta , Grupos de Población Animal , Animales , Antracenos/análisis , Fluorenos/análisis , Halogenación , Hígado/metabolismo , Fenantrenos/análisis
10.
J Chromatogr A ; 1579: 106-114, 2018 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-30391036

RESUMEN

Polycyclic aromatic compounds (PACs) consists of multiple compounds and the number of theoretically possible isomers can reach into the thousands. Currently each PAC group is quantified collectively as a single group of compounds. However, individual PACs can reveal important information on how the PACs were formed and this information may be used to determine sources of PACs in environmental samples, It is hypothesized that many of the limitations with characterizing alkylated PACs with one dimensional gas-chromatography (1D GC) can be circumvented using GC × GC (two dimensional gas chromatography). Here we apply comprehensive GCxGC coupled to high-resolution time of flight mass spectrometry (GC × GC-HFTOF-MS) to aid in the separation, identification and quantitation of APACs in three environmental matrices: mussel tissue (Mytilus edulis), lubricating oil and coal. In the absence of authentic analytical standards, differences in the mass spectral fragmentation pattern of isomers were used to confirm the identity of isomers within a PAC group. The method was validated according to the EURACHEM guidelines and used to quantify a biological standard reference material (SRM 2974a). The method met all the standard method performance requirements such as trueness, precision and measurement of uncertainty and is fit for quantifying these compounds in biota. Furthermore, the method was used to identify and quantify additional PAC compounds in the SRM 2974a material which to date have not been certified. With appropriate statistical analytical tools, the described GC × GC method can be used as a tool for more robust source fingerprinting and source apportionment of PACs in the environment.


Asunto(s)
Monitoreo del Ambiente/métodos , Cromatografía de Gases y Espectrometría de Masas , Compuestos Policíclicos/análisis , Animales , Carbón Mineral/análisis , Isomerismo , Peso Molecular , Mytilus edulis/química , Aceites/química
11.
J Struct Biol ; 203(3): 281-287, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29879486

RESUMEN

Coiled coils are well described as powerful oligomerization motifs and exhibit a large diversity of functions, including gene regulation, cell division, membrane fusion and drug extrusion. The archaea S-layer originated right-handed coiled coil -RHCC-NT- is characterized by extreme stability and is free of cysteine and histidine moieties. In the current study, we have followed a multidisciplinary approach to investigate the capacity of RHCC-NT to bind a variety of ionic complex metal ions. At the outside of the RHCC-NT, one mercury ion forms an electrostatic interaction with the S-methyl moiety of the single methionine residue present in each coil. We demonstrate that RHCC-NT is reducing and incorporating metallic mercury in the large-sized interior cavities which are lined up along the tetrameric channel.


Asunto(s)
Archaea/química , Nanotubos/química , Conformación Proteica , Proteínas/química , Mercurio , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína , Proteínas/ultraestructura , Electricidad Estática
12.
Biophys J ; 113(12): 2609-2620, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262356

RESUMEN

High mobility group AT-hook 2 (HMGA2) protein is composed of three AT-hook domains. HMGA2 expresses at high levels in both embryonic stem cells and cancer cells, where it interacts with and stabilizes replication forks (RFs), resulting in elevated cell proliferation rates. In this study, we demonstrated that HMGA2 knockdown reduces cell proliferation. To understand the features required for interaction between HMGA2 and RFs, we studied the solution structure of HMGA2, free and in complex with RFs, using an integrated host of biophysical techniques. Circular dichroism and NMR experiments confirmed the disordered state of unbound HMGA2. Dynamic light scattering and sedimentation velocity experiments demonstrated that HMGA2 and RF are monodisperse in solution, and form an equimolar complex. Small-angle x-ray scattering studies revealed that HMGA2 binds in a side-by-side orientation to RF where 3 AT-hooks act as a clamp to wrap around a distorted RF. Thus, our data provide insights into how HMGA2 interacts with stalled RFs and the function of the process.


Asunto(s)
Replicación del ADN , ADN/química , ADN/metabolismo , Proteína HMGA2/metabolismo , Proliferación Celular , ADN/biosíntesis , Técnicas de Silenciamiento del Gen , Células HEK293 , Proteína HMGA2/química , Proteína HMGA2/deficiencia , Proteína HMGA2/genética , Humanos , Modelos Moleculares , Conformación de Ácido Nucleico , Unión Proteica , Conformación Proteica
13.
PLoS Biol ; 15(7): e2001492, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28704364

RESUMEN

The collagen binding integrin α2ß1 plays a crucial role in hemostasis, fibrosis, and cancer progression amongst others. It is specifically inhibited by rhodocetin (RC), a C-type lectin-related protein (CLRP) found in Malayan pit viper (Calloselasma rhodostoma) venom. The structure of RC alone reveals a heterotetramer arranged as an αß and γδ subunit in a cruciform shape. RC specifically binds to the collagen binding A-domain of the integrin α2 subunit, thereby blocking collagen-induced platelet aggregation. However, until now, the molecular basis for this interaction has remained unclear. Here, we present the molecular structure of the RCγδ-α2A complex solved to 3.0 Å resolution. Our findings show that RC undergoes a dramatic structural reorganization upon binding to α2ß1 integrin. Besides the release of the nonbinding RCαß tandem, the RCγ subunit interacts with loop 2 of the α2A domain as result of a dramatic conformational change. The RCδ subunit contacts the integrin α2A domain in the "closed" conformation through its helix C. Combined with epitope-mapped antibodies, conformationally locked α2A domain mutants, point mutations within the α2A loop 2, and chemical modifications of the purified toxin protein, this molecular structure of RCγδ-α2A complex explains the inhibitory mechanism and specificity of RC for α2ß1 integrin.


Asunto(s)
Venenos de Crotálidos/química , Integrina alfa2beta1/química , Venenos de Crotálidos/farmacología , Cristalografía por Rayos X , Integrina alfa2beta1/antagonistas & inhibidores , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína
15.
Nat Commun ; 7: 13515, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27901020

RESUMEN

Netrins, a family of laminin-related molecules, have been proposed to act as guidance cues either during nervous system development or the establishment of the vascular system. This was clearly demonstrated for netrin-1 via its interaction with the receptors DCC and UNC5s. However, mainly based on shared homologies with netrin-1, netrin-4 was also proposed to play a role in neuronal outgrowth and developmental/pathological angiogenesis via interactions with netrin-1 receptors. Here, we present the high-resolution structure of netrin-4, which shows unique features in comparison with netrin-1, and show that it does not bind directly to any of the known netrin-1 receptors. We show that netrin-4 disrupts laminin networks and basement membranes (BMs) through high-affinity binding to the laminin γ1 chain. We hypothesize that this laminin-related function is essential for the previously described effects on axon growth promotion and angiogenesis. Our study unveils netrin-4 as a non-enzymatic extracellular matrix protein actively disrupting pre-existing BMs.


Asunto(s)
Orientación del Axón/fisiología , Membrana Basal/metabolismo , Laminina/fisiología , Neovascularización Fisiológica/fisiología , Netrinas/fisiología , Animales , Axones/fisiología , Pollos , Membrana Corioalantoides/fisiología , Cristalografía por Rayos X , Femenino , Células HEK293 , Humanos , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Netrinas/ultraestructura , Unión Proteica , Multimerización de Proteína , Ratas , Ratas Sprague-Dawley , Células de Schwann , Ensayos Antitumor por Modelo de Xenoinjerto
16.
PLoS One ; 11(3): e0152386, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27029048

RESUMEN

Recombinant proteins are commonly expressed in eukaryotic expression systems to ensure the formation of disulfide bridges and proper glycosylation. Although many proteins can be expressed easily, some proteins, sub-domains, and mutant protein versions can cause problems. Here, we investigated expression levels of recombinant extracellular, intracellular as well as transmembrane proteins tethered to different polypeptides in mammalian cell lines. Strikingly, fusion of proteins to the prokaryotic maltose-binding protein (MBP) generally enhanced protein production. MBP fusion proteins consistently exhibited the most robust increase in protein production in comparison to commonly used tags, e.g., the Fc, Glutathione S-transferase (GST), SlyD, and serum albumin (ser alb) tag. Moreover, proteins tethered to MBP revealed reduced numbers of dying cells upon transient transfection. In contrast to the Fc tag, MBP is a stable monomer and does not promote protein aggregation. Therefore, the MBP tag does not induce artificial dimerization of tethered proteins and provides a beneficial fusion tag for binding as well as cell adhesion studies. Using MBP we were able to secret a disease causing laminin ß2 mutant protein (congenital nephrotic syndrome), which is normally retained in the endoplasmic reticulum. In summary, this study establishes MBP as a versatile expression tag for protein production in eukaryotic expression systems.


Asunto(s)
Proteínas de Unión a Maltosa/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Animales , Adhesión Celular , Línea Celular Tumoral , Expresión Génica , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Proteínas de Unión a Maltosa/genética , Ratones , Proteínas Recombinantes de Fusión/genética
17.
Cancer Cell ; 29(2): 173-85, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26859457

RESUMEN

Netrin-1 has been shown to be up-regulated in a fraction of human cancers as a mechanism to allow these tumors to escape the pro-apoptotic activity of some of its main dependence receptors, the UNC5 homologs (UNC5H). Here we identify the V-2 domain of netrin-1 to be important for its interaction with the Ig1/Ig2 domains of UNC5H2. We generate a humanized anti-netrin-1 antibody that disrupts the interaction between netrin-1 and UNC5H2 and triggers death of netrin-1-expressing tumor cells in vitro. We also present evidence that combining the anti-netrin-1 antibody with epidrugs such as decitabine could be effective in treating tumors showing no or modest netrin-1 expression. These results support that this antibody is a promising drug candidate.


Asunto(s)
Neoplasias/terapia , Factores de Crecimiento Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Neoplasias/metabolismo , Neoplasias/patología , Factores de Crecimiento Nervioso/inmunología , Receptores de Netrina , Netrina-1 , Unión Proteica , Proteínas Supresoras de Tumor/inmunología
18.
J Biol Chem ; 291(10): 5355-72, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26740632

RESUMEN

RNA helicase associated with AU-rich element (RHAU) is an ATP-dependent RNA helicase that demonstrates high affinity for quadruplex structures in DNA and RNA. To elucidate the significance of these quadruplex-RHAU interactions, we have performed RNA co-immunoprecipitation screens to identify novel RNAs bound to RHAU and characterize their function. In the course of this study, we have identified the non-coding RNA BC200 (BCYRN1) as specifically enriched upon RHAU immunoprecipitation. Although BC200 does not adopt a quadruplex structure and does not bind the quadruplex-interacting motif of RHAU, it has direct affinity for RHAU in vitro. Specifically designed BC200 truncations and RNase footprinting assays demonstrate that RHAU binds to an adenosine-rich region near the 3'-end of the RNA. RHAU truncations support binding that is dependent upon a region within the C terminus and is specific to RHAU isoform 1. Tests performed to assess whether BC200 interferes with RHAU helicase activity have demonstrated the ability of BC200 to act as an acceptor of unwound quadruplexes via a cytosine-rich region near the 3'-end of the RNA. Furthermore, an interaction between BC200 and the quadruplex-containing telomerase RNA was confirmed by pull-down assays of the endogenous RNAs. This leads to the possibility that RHAU may direct BC200 to bind and exert regulatory functions at quadruplex-containing RNA or DNA sequences.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , ARN Largo no Codificante/metabolismo , Secuencia de Bases , Sitios de Unión , ARN Helicasas DEAD-box/genética , G-Cuádruplex , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Datos de Secuencia Molecular , Unión Proteica , ARN Largo no Codificante/química , ARN Largo no Codificante/genética
19.
Nanomedicine ; 11(4): 913-25, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25680541

RESUMEN

Malignant glioma are often fatal and pose a significant therapeutic challenge. Here we have employed α-helical right handed coiled coils (RHCC) which self-assemble into tetrameric nanotubes that stably associate with platinum (Pt) (IV) compound. This Pt(IV)-RHCC complex showed superior in vitro and in vivo toxicity in human malignant glioma cells at up to 5 fold lower platinum concentrations when compared to free Pt(IV). Pt(IV)-RHCC nanotubes activated multiple cell death pathways in GB cells without affecting astrocytes in vitro or causing damage to normal mouse brain. This Pt(IV)-RHCC nanotubes may serve as a promising new therapeutic tool for low dose Pt(IV) prodrug application for highly efficient and selective treatment of human brain tumors. FROM THE CLINICAL EDITOR: The prognosis of malignant glioma remains poor despite medical advances. Platinum, one of the chemotherapeutic agents used, has significant systemic side effects. In this article, the authors employed α-helical right handed coiled coil (RHCC) protein nanotubes as a carrier for cisplatin. It was shown that the new compound achieved higher tumor kill rate but lower toxicity to normal cells and thus may hold promise to be a highly efficient treatment for the future.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Nanotubos/química , Compuestos de Platino/farmacología , Profármacos/farmacología , Animales , Antineoplásicos/química , Astrocitos/metabolismo , Astrocitos/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones , Compuestos de Platino/química , Profármacos/química
20.
BMC Biol ; 12: 44, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24885675

RESUMEN

BACKGROUND: Wnt proteins are a family of secreted signaling molecules that regulate key developmental processes in metazoans. The molecular basis of Wnt binding to Frizzled and LRP5/6 co-receptors has long been unknown due to the lack of structural data on Wnt ligands. Only recently, the crystal structure of the Wnt8-Frizzled8-cysteine-rich-domain (CRD) complex was solved, but the significance of interaction sites that influence Wnt signaling has not been assessed. RESULTS: Here, we present an extensive structure-function analysis of mouse Wnt3a in vitro and in vivo. We provide evidence for the essential role of serine 209, glycine 210 (site 1) and tryptophan 333 (site 2) in Fz binding. Importantly, we discovered that valine 337 in the site 2 binding loop is critical for signaling without contributing to binding. Mutations in the presumptive second CRD binding site (site 3) partly abolished Wnt binding. Intriguingly, most site 3 mutations increased Wnt signaling, probably by inhibiting Wnt-CRD oligomerization. In accordance, increasing amounts of soluble Frizzled8-CRD protein modulated Wnt3a signaling in a biphasic manner. CONCLUSIONS: We propose a concentration-dependent switch in Wnt-CRD complex formation from an inactive aggregation state to an activated high mobility state as a possible modulatory mechanism in Wnt signaling gradients.


Asunto(s)
Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Vía de Señalización Wnt , Proteína Wnt3A/química , Proteína Wnt3A/metabolismo , Secuencia de Aminoácidos , Animales , Embrión no Mamífero/metabolismo , Células HEK293 , Humanos , Ratones , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Mutación Puntual/genética , Unión Proteica , Estructura Terciaria de Proteína , Solubilidad , Relación Estructura-Actividad , Pez Cebra/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA