Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(39): 24258-24268, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32913056

RESUMEN

The small GTPase KRAS is localized at the plasma membrane where it functions as a molecular switch, coupling extracellular growth factor stimulation to intracellular signaling networks. In this process, KRAS recruits effectors, such as RAF kinase, to the plasma membrane where they are activated by a series of complex molecular steps. Defining the membrane-bound state of KRAS is fundamental to understanding the activation of RAF kinase and in evaluating novel therapeutic opportunities for the inhibition of oncogenic KRAS-mediated signaling. We combined multiple biophysical measurements and computational methodologies to generate a consensus model for authentically processed, membrane-anchored KRAS. In contrast to the two membrane-proximal conformations previously reported, we identify a third significantly populated state using a combination of neutron reflectivity, fast photochemical oxidation of proteins (FPOP), and NMR. In this highly populated state, which we refer to as "membrane-distal" and estimate to comprise ∼90% of the ensemble, the G-domain does not directly contact the membrane but is tethered via its C-terminal hypervariable region and carboxymethylated farnesyl moiety, as shown by FPOP. Subsequent interaction of the RAF1 RAS binding domain with KRAS does not significantly change G-domain configurations on the membrane but affects their relative populations. Overall, our results are consistent with a directional fly-casting mechanism for KRAS, in which the membrane-distal state of the G-domain can effectively recruit RAF kinase from the cytoplasm for activation at the membrane.


Asunto(s)
Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Quinasas raf/metabolismo , Membrana Celular/metabolismo , Simulación de Dinámica Molecular
2.
J Biol Chem ; 294(6): 2193-2207, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30559287

RESUMEN

The gene encoding the GTPase KRAS is frequently mutated in pancreatic, lung, and colorectal cancers. The KRAS fraction in the plasma membrane (PM) correlates with activation of the mitogen-activated protein kinase (MAPK) pathway and subsequent cellular proliferation. Understanding KRAS's interaction with the PM is challenging given the complexity of the cellular environment. To gain insight into key components necessary for KRAS signal transduction at the PM, we used synthetic membranes such as liposomes and giant unilamellar vesicles. Using surface plasmon resonance (SPR) spectroscopy, we demonstrated that KRAS and Raf-1 proto-oncogene Ser/Thr kinase (RAF1) domains interact with these membranes primarily through electrostatic interactions with negatively charged lipids reinforced by additional interactions involving phosphatidyl ethanolamine and cholesterol. We found that the RAF1 region spanning RBD through CRD (RBDCRD) interacts with the membrane significantly more strongly than the isolated RBD or CRD domains and synergizes KRAS partitioning to the membrane. We also found that calmodulin and phosphodiesterase 6 delta (PDE6δ), but not galectin3 previously proposed to directly interact with KRAS, passively sequester KRAS and prevent it from partitioning into the PM. RAF1 RBDCRD interacted with membranes preferentially at nonraft lipid domains. Moreover, a C-terminal O-methylation was crucial for KRAS membrane localization. These results contribute to a better understanding of how the KRAS-membrane interaction is tuned by multiple factors whose identification could inform drug discovery efforts to disrupt this critical interaction in diseases such as cancer.


Asunto(s)
Membrana Celular/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Calmodulina/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Membranas Artificiales , Dominios Proteicos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-raf , Transducción de Señal , Electricidad Estática
3.
Proc Natl Acad Sci U S A ; 113(44): E6766-E6775, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27791178

RESUMEN

Farnesylation and carboxymethylation of KRAS4b (Kirsten rat sarcoma isoform 4b) are essential for its interaction with the plasma membrane where KRAS-mediated signaling events occur. Phosphodiesterase-δ (PDEδ) binds to KRAS4b and plays an important role in targeting it to cellular membranes. We solved structures of human farnesylated-methylated KRAS4b in complex with PDEδ in two different crystal forms. In these structures, the interaction is driven by the C-terminal amino acids together with the farnesylated and methylated C185 of KRAS4b that binds tightly in the central hydrophobic pocket present in PDEδ. In crystal form II, we see the full-length structure of farnesylated-methylated KRAS4b, including the hypervariable region. Crystal form I reveals structural details of farnesylated-methylated KRAS4b binding to PDEδ, and crystal form II suggests the potential binding mode of geranylgeranylated-methylated KRAS4b to PDEδ. We identified a 5-aa-long sequence motif (Lys-Ser-Lys-Thr-Lys) in KRAS4b that may enable PDEδ to bind both forms of prenylated KRAS4b. Structure and sequence analysis of various prenylated proteins that have been previously tested for binding to PDEδ provides a rationale for why some prenylated proteins, such as KRAS4a, RalA, RalB, and Rac1, do not bind to PDEδ. Comparison of all four available structures of PDEδ complexed with various prenylated proteins/peptides shows the presence of additional interactions due to a larger protein-protein interaction interface in KRAS4b-PDEδ complex. This interface might be exploited for designing an inhibitor with minimal off-target effects.


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas/química , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Dominios y Motivos de Interacción de Proteínas , Prenilación de Proteína/fisiología , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , 3',5'-GMP Cíclico Fosfodiesterasas/genética , Secuencia de Aminoácidos , Sitios de Unión , Membrana Celular/metabolismo , Cristalografía por Rayos X , Genes ras , Humanos , Metilación , Modelos Moleculares , Conformación Molecular , Mutación , Unión Proteica/fisiología , Proteínas Proto-Oncogénicas p21(ras)/genética , Análisis de Secuencia , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP ral/metabolismo
4.
Biophys J ; 114(1): 137-145, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29320680

RESUMEN

Ras is a membrane-anchored signaling protein that serves as a hub for many signaling pathways and also plays a prominent role in cancer. The intrinsic behavior of Ras on the membrane has captivated the biophysics community in recent years, especially the possibility that it may form dimers. In this article, we describe results from a comprehensive series of experiments using fluorescence correlation spectroscopy and single-molecule tracking to probe the possible dimerization of natively expressed and fully processed K-Ras4B in supported lipid bilayer membranes. Key to these studies is the fact that K-Ras4B has its native membrane anchor, including both the farnesylation and methylation of the terminal cysteine, enabling detailed exploration of possible effects of cholesterol and lipid composition on K-Ras4B membrane organization. The results from all conditions studied indicate that full-length K-Ras4B lacks intrinsic dimerization capability. This suggests that any lateral organization of Ras in living cell membranes likely stems from interactions with other factors.


Asunto(s)
Membrana Celular/química , Proteínas Proto-Oncogénicas p21(ras)/química , Humanos , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Propiedades de Superficie
5.
Am J Physiol Lung Cell Mol Physiol ; 306(1): L10-22, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24213919

RESUMEN

Secretoglobin (SCGB) 3A2 is a member of the SCGB gene superfamily of small secreted proteins, predominantly expressed in lung airways. We hypothesize that human SCGB3A2 may exhibit anti-inflammatory, growth factor, and antifibrotic activities and be of clinical utility. Recombinant human SCGB3A2 was expressed, purified, and biochemically characterized as a first step to its development as a therapeutic agent in clinical settings. Human SCGB3A2, as well as mouse SCGB3A2, readily formed a dimer in solution and exhibited novel phospholipase A2 inhibitory activity. This is the first demonstration of any quantitative biochemical measurement for the evaluation of SCGB3A2 protein. In the mouse as an experimental animal, human SCGB3A2 exhibited growth factor activity by promoting embryonic lung development in both ex vivo and in vivo systems and antifibrotic activity in the bleomycin-induced lung fibrosis model. The results suggested that human SCGB3A2 can function as a growth factor and an antifibrotic agent in humans. When SCGB3A2 was administered to pregnant female mice through the tail vein, the protein was detected in the dam's serum and lung, as well as the placenta, amniotic fluids, and embryonic lungs at 10 min postadministration, suggesting that SCGB3A2 readily crosses the placenta. The results warrant further development of recombinant SCGB3A2 as a therapeutic agent in treating patients suffering from lung diseases or preterm infants with respiratory distress.


Asunto(s)
Pulmón/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Secretoglobinas/administración & dosificación , Animales , Disponibilidad Biológica , Bleomicina , Evaluación Preclínica de Medicamentos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Pulmón/embriología , Ratones , Ratones Endogámicos C57BL , Inhibidores de Fosfolipasa A2/administración & dosificación , Inhibidores de Fosfolipasa A2/química , Inhibidores de Fosfolipasa A2/farmacocinética , Fosfolipasas A2/química , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Secretoglobinas/química , Secretoglobinas/farmacocinética , Técnicas de Cultivo de Tejidos
6.
ACS Pharmacol Transl Sci ; 7(7): 1983-1995, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39022364

RESUMEN

The KRAS gene plays a pivotal role in numerous cancers by encoding a GTPase that upon association with the plasma membrane activates the MAPK pathway, promoting cellular proliferation. In our study, we investigated small molecules that disrupt KRAS's membrane interaction, hypothesizing that such disruption could in turn inhibit mutant RAS signaling. Native mass spectrometry screening of KRAS-FMe identified compounds with a preference for interacting with the hypervariable region (HVR), and surface plasmon resonance (SPR) further refined our selection to graveoline as a compound exhibiting preferential HVR binding. Subsequent nuclear magnetic resonance (NMR) analysis showed that graveoline's interaction with KRAS depends on C-terminal O-methylation. Moreover, our findings revealed multiple interaction sites, suggesting weak engagement with the KRAS G domain. Using nanodiscs as a membrane mimetic, further characterization through NMR and Förster resonance energy transfer (FRET) studies demonstrated graveoline's ability to perturb KRAS membrane interaction in a biochemical setting. Our biophysical approach sheds light on the intricate molecular mechanisms underlying KRAS-ligand interactions, providing valuable insights into understanding the KRAS-associated pathophysiology. These findings contribute to the translational aspect of our study, offering potential avenues for further research targeting KRAS membrane association with the potential to lead to a new class of RAS therapeutics.

7.
Commun Biol ; 7(1): 242, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38418613

RESUMEN

The oncogene RAS, extensively studied for decades, presents persistent gaps in understanding, hindering the development of effective therapeutic strategies due to a lack of precise details on how RAS initiates MAPK signaling with RAF effector proteins at the plasma membrane. Recent advances in X-ray crystallography, cryo-EM, and super-resolution fluorescence microscopy offer structural and spatial insights, yet the molecular mechanisms involving protein-protein and protein-lipid interactions in RAS-mediated signaling require further characterization. This study utilizes single-molecule experimental techniques, nuclear magnetic resonance spectroscopy, and the computational Machine-Learned Modeling Infrastructure (MuMMI) to examine KRAS4b and RAF1 on a biologically relevant lipid bilayer. MuMMI captures long-timescale events while preserving detailed atomic descriptions, providing testable models for experimental validation. Both in vitro and computational studies reveal that RBDCRD binding alters KRAS lateral diffusion on the lipid bilayer, increasing cluster size and decreasing diffusion. RAS and membrane binding cause hydrophobic residues in the CRD region to penetrate the bilayer, stabilizing complexes through ß-strand elongation. These cooperative interactions among lipids, KRAS4b, and RAF1 are proposed as essential for forming nanoclusters, potentially a critical step in MAP kinase signal activation.


Asunto(s)
Membrana Dobles de Lípidos , Lípidos de la Membrana , Lípidos de la Membrana/metabolismo , Membrana Dobles de Lípidos/metabolismo , Membrana Celular/metabolismo , Membranas/metabolismo , Transducción de Señal
8.
J Biol Chem ; 286(22): 19682-92, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21478551

RESUMEN

With increasing worldwide rates of morbidity and mortality of pulmonary fibrosis, the development of effective therapeutics for this disease is of great interest. Secretoglobin (SCGB) 3A2, a novel cytokine-like molecule predominantly expressed in pulmonary airways epithelium, exhibits anti-inflammatory and growth factor activities. In the current study SCGB3A2 was found to inhibit TGFß-induced differentiation of fibroblasts to myofibroblasts, a hallmark of the fibrogenic process, using pulmonary fibroblasts isolated from adult mice. This induction was through increased phosphorylation of STAT1 and expression of SMAD7 and decreased phosphorylation of SMAD2 and SMAD3. To demonstrate the effect of SCGB3A2 on the TGFß signaling in vivo, a bleomycin-induced pulmonary fibrosis mouse model was used. Mice were administered bleomycin intratracheally followed by intravenous injection of recombinant SCGB3A2. Histological examination in conjunction with inflammatory cell counts in bronchoalveolar lavage fluids demonstrated that SCGB3A2 suppressed bleomycin-induced pulmonary fibrosis. Microarray analysis was carried out using RNAs from lungs of bleomycin-treated mice with or without SCGB3A2 and normal mice treated with SCGB3A2. The results demonstrated that SCGB3A2 affects TGFß signaling and reduces the expression of genes involved in fibrosis. This study suggests the potential utility of SCGB3A2 for targeting TGFß signaling in the treatment of pulmonary fibrosis.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Bleomicina/efectos adversos , Regulación hacia Abajo/efectos de los fármacos , Proteínas/metabolismo , Fibrosis Pulmonar/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Bleomicina/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Regulación hacia Abajo/genética , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Humanos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas/genética , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Secretoglobinas , Transducción de Señal/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/genética
9.
Biomol NMR Assign ; 16(1): 1-8, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34686998

RESUMEN

RAS proteins cycling between the active-form (GTP-bound) and inactive-form (GDP-bound) play a key role in cell signaling pathways that control cell survival, proliferation, and differentiation. Mutations at codon 12, 13, and 61 in RAS are known to attenuate its GTPase activity favoring the RAS active state and constitutively active downstream signaling. This hyperactivation accounts for various malignancies including pancreatic, lung, and colorectal cancers. Active KRAS is found to exist in equilibrium between two rapidly interconverting conformational states (State1-State2) in solution. Due to this dynamic feature of the protein, the 1H-15N correlation cross-peak signals of several amino acid (AA) residues of KRAS belonging to the flexible loop regions are absent from its 2D 1H-15N HSQC spectrum within and near physiological solution pH. A threonine to serine mutation at position 35 (T35S) shifts the interconverting equilibrium to State1 conformation and enables the emergence of such residues in the 2D 1H-15N HSQC spectrum due to gained conformational rigidity. We report here the 1HN, 15N, and 13C backbone resonance assignments for the 19.2 kDa (AA 1-169) protein constructs of KRAS-GppNHp harboring T35S mutation (KRAST35S/C118S-GppNHp) and of its oncogenic counterpart harboring the Q61L mutation (KRAST35S/Q61L/C118S-GppNHp) using heteronuclear, multidimensional NMR spectroscopy at 298 K. High resolution NMR data allowed the unambiguous assignments of 1H-15N correlation cross-peaks for all the residues except for Met1. Furthermore, 2D 1H-15N HSQC overlay of two proteins assisted in determination of Q61L mutation-induced chemical shift perturbations for select residues in the regions of P-loop, Switch-II, and helix α3.


Asunto(s)
Proteínas Proto-Oncogénicas p21(ras) , Guanosina Trifosfato/metabolismo , Humanos , Espectroscopía de Resonancia Magnética , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Proto-Oncogénicas p21(ras)/genética
10.
Protein Expr Purif ; 76(2): 238-47, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21146612

RESUMEN

Purifying proteins from recombinant sources is often difficult, time-consuming, and costly. We have recently instituted a series of improvements in our protein purification pipeline that allows much more accurate choice of expression host and conditions and purification protocols. The key elements are parallel cloning, small scale parallel expression and lysate preparation, and small scale parallel protein purification. Compared to analyzing expression data only, results from multiple small scale protein purifications predict success at scale-up with greatly improved reliability. Using these new procedures we purified eight of nine proteins from xenotropic murine leukemia virus-related virus (XMRV) on the first attempt at large scale.


Asunto(s)
Clonación Molecular/métodos , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Virus Relacionado con el Virus Xenotrópico de la Leucemia Murina/química , Animales , Baculoviridae/genética , Secuencia de Bases , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Datos de Secuencia Molecular , Proteínas Recombinantes/genética , Spodoptera , Virus Relacionado con el Virus Xenotrópico de la Leucemia Murina/genética , Virus Relacionado con el Virus Xenotrópico de la Leucemia Murina/metabolismo
11.
Methods Mol Biol ; 2262: 105-116, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33977473

RESUMEN

Recombinant mammalian proteins are routinely produced in E. coli and thus lack post-translational modifications. KRAS4b is processed at both the N- and C-terminus, resulting in an acetylation of the N-terminus (at Thr, after aminopeptidase removal of the original N-term Met) and farnesylation/carboxymethylation of the C-terminal Cys (after proteolytic cleavage of the original C-terminal three amino acids, Val-Iso-Met). Processing of KRAS enables it to associate with the plasma membrane and fulfill its function in cell signaling. We describe here the production of recombinant KRAS4b from our modified baculovirus/insect cell expression system that accurately incorporates these in vivo modifications to allow experiments that anchor KRAS4b to membrane mimetics (e.g., nanodiscs and liposomes).


Asunto(s)
Membrana Celular/metabolismo , Prenilación de Proteína , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Acetilación , Secuencia de Aminoácidos , Humanos , Metilación , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/aislamiento & purificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación
12.
Sci Rep ; 9(1): 10512, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31324887

RESUMEN

Although post-translational modification of the C-terminus of RAS has been studied extensively, little is known about N-terminal processing. Mass spectrometric characterization of KRAS expressed in mammalian cells showed cleavage of the initiator methionine (iMet) and N-acetylation of the nascent N-terminus. Interestingly, structural studies on GDP- and GMPPNP-bound KRAS lacking the iMet and N-acetylation resulted in Mg2+-free structures of KRAS with flexible N-termini. In the Mg2+-free KRAS-GDP structure, the flexible N-terminus causes conformational changes in the interswitch region resulting in a fully open conformation of switch I. In the Mg2+-free KRAS-GMPPNP structure, the flexible N-terminus causes conformational changes around residue A59 resulting in the loss of Mg2+ and switch I in the inactive state 1 conformation. Structural studies on N-acetylated KRAS-GDP lacking the iMet revealed the presence of Mg2+ and a conformation of switch regions also observed in the structure of GDP-bound unprocessed KRAS with the iMet. In the absence of the iMet, the N-acetyl group interacts with the central beta-sheet and stabilizes the N-terminus and the switch regions. These results suggest there is crosstalk between the N-terminus and the Mg2+ binding site, and that N-acetylation plays an important role by stabilizing the N-terminus of RAS upon excision of the iMet.


Asunto(s)
Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas p21(ras)/química , Acetilación , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Guanosina Difosfato/metabolismo , Guanilil Imidodifosfato/metabolismo , Humanos , Enlace de Hidrógeno , Magnesio/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
14.
Sci Signal ; 10(498)2017 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-28951536

RESUMEN

There is intense interest in developing therapeutic strategies for RAS proteins, the most frequently mutated oncoprotein family in cancer. Development of effective anti-RAS therapies will be aided by the greater appreciation of RAS isoform-specific differences in signaling events that support neoplastic cell growth. However, critical issues that require resolution to facilitate the success of these efforts remain. In particular, the use of well-validated anti-RAS antibodies is essential for accurate interpretation of experimental data. We evaluated 22 commercially available anti-RAS antibodies with a set of distinct reagents and cell lines for their specificity and selectivity in recognizing the intended RAS isoforms and mutants. Reliability varied substantially. For example, we found that some pan- or isoform-selective anti-RAS antibodies did not adequately recognize their intended target or showed greater selectivity for another; some were valid for detecting G12D and G12V mutant RAS proteins in Western blotting, but none were valid for immunofluorescence or immunohistochemical analyses; and some antibodies recognized nonspecific bands in lysates from "Rasless" cells expressing the oncoprotein BRAFV600E Using our validated antibodies, we identified RAS isoform-specific siRNAs and shRNAs. Our results may help to ensure the accurate interpretation of future RAS studies.


Asunto(s)
Antineoplásicos Inmunológicos/inmunología , Mutación , Proteínas Oncogénicas/inmunología , Proteínas ras/inmunología , Animales , Antineoplásicos Inmunológicos/análisis , Línea Celular Tumoral , Fibroblastos , Humanos , Hibridomas , Ratones , Proteínas Oncogénicas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , ARN Interferente Pequeño , Proteínas ras/genética
15.
Sci Rep ; 5: 15916, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26522388

RESUMEN

Prenylated proteins play key roles in several human diseases including cancer, atherosclerosis and Alzheimer's disease. KRAS4b, which is frequently mutated in pancreatic, colon and lung cancers, is processed by farnesylation, proteolytic cleavage and carboxymethylation at the C-terminus. Plasma membrane localization of KRAS4b requires this processing as does KRAS4b-dependent RAF kinase activation. Previous attempts to produce modified KRAS have relied on protein engineering approaches or in vitro farnesylation of bacterially expressed KRAS protein. The proteins produced by these methods do not accurately replicate the mature KRAS protein found in mammalian cells and the protein yield is typically low. We describe a protocol that yields 5-10 mg/L highly purified, farnesylated, and methylated KRAS4b from insect cells. Farnesylated and methylated KRAS4b is fully active in hydrolyzing GTP, binds RAF-RBD on lipid Nanodiscs and interacts with the known farnesyl-binding protein PDEδ.


Asunto(s)
Lípidos/fisiología , Prenilación de Proteína/fisiología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Biofisica/métodos , Membrana Celular/metabolismo , Células Cultivadas , Guanosina Trifosfato/metabolismo , Humanos , Insectos/metabolismo , Metilación , Unión Proteica/fisiología , Quinasas raf/metabolismo
16.
J Immunol Methods ; 356(1-2): 39-46, 2010 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-20211626

RESUMEN

Detection of antibodies to Kaposi's sarcoma-associated herpesvirus (KSHV or Human herpesvirus 8) is a topic of ongoing controversy. KSHV expresses multiple antigens and host responses are highly variable. We have previously described an algorithm for determining KSHV infection based on K8.1 ELISA and LANA immunofluorescence assay (IFA). Here we describe the development of a recombinant ELISA for LANA and an improved testing strategy using ELISAs for LANA and K8.1. We assessed mammalian and baculovirus expression systems for the production of full-length recombinant LANA. We evaluated the performance of LANA ELISAs using human serum samples from several sources including blood donors and clinical patients diagnosed with Kaposi's sarcoma and compared them to LANA IFA. Both LANA ELISAs exhibited comparable sensitivity and specificity to LANA IFA but showed considerably greater reliability. The LANA ELISA can thus be used in conjunction with the previously described K8.1 ELISA to enable the highly sensitive and specific detection of antibodies to KSHV. Use of this testing strategy will provide a more accurate and reliable diagnostic assessment of KSHV status.


Asunto(s)
Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 8/inmunología , Proteínas Nucleares/inmunología , Sarcoma de Kaposi/inmunología , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Técnica del Anticuerpo Fluorescente/métodos , Infecciones por Herpesviridae/complicaciones , Humanos , Proteínas Recombinantes/inmunología , Reproducibilidad de los Resultados , Sarcoma de Kaposi/etiología , Spodoptera
18.
J Infect Dis ; 196 Suppl 2: S421-9, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17940979

RESUMEN

BACKGROUND: Virus-like particles (VLPs) of Ebola virus (EBOV) and Marburg virus (MARV) produced in human 293T embryonic kidney cells have been shown to be effective vaccines against filoviral infection. In this study, we explored alternative strategies for production of filovirus-like particle-based vaccines, to accelerate the development process. The goal of this work was to increase the yield of VLPs, while retaining their immunogenic properties. METHODS: Ebola and Marburg VLPs (eVLPs and mVLPs, respectively) were generated by use of recombinant baculovirus constructs expressing glycoprotein, VP40 matrix protein, and nucleoprotein from coinfected insect cells. The baculovirus-derived eVLPs and mVLPs were characterized biochemically, and then the immune responses produced by the eVLPs in insect cells were studied further. RESULTS: The baculovirus-derived eVLPs elicited maturation of human myeloid dendritic cells (DCs), indicating their immunogenic properties. Mice vaccinated with insect cell-derived eVLPs generated antibody and cellular responses equivalent to those vaccinated with mammalian 293T cell-derived eVLPs and were protected from EBOV challenge in a dose-dependent manner. CONCLUSION: Together, these data suggest that filovirus-like particles produced by baculovirus expression systems, which are amenable to large-scale production, are highly immunogenic and are suitable as safe and effective vaccines for the prevention of filoviral infection.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Filoviridae/inmunología , Fiebre Hemorrágica Ebola/inmunología , Enfermedad del Virus de Marburg/inmunología , Animales , Línea Celular , Modelos Animales de Enfermedad , Ebolavirus/inmunología , Ebolavirus/fisiología , Femenino , Filoviridae/inmunología , Humanos , Marburgvirus/inmunología , Marburgvirus/fisiología , Ratones , Ratones Endogámicos C57BL , Roedores , Replicación Viral
19.
Proc Natl Acad Sci U S A ; 103(42): 15552-7, 2006 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-17028174

RESUMEN

Birt-Hogg-Dubé syndrome, a hamartoma disorder characterized by benign tumors of the hair follicle, lung cysts, and renal neoplasia, is caused by germ-line mutations in the BHD(FLCN) gene, which encodes a tumor-suppressor protein, folliculin (FLCN), with unknown function. The tumor-suppressor proteins encoded by genes responsible for several other hamartoma syndromes, LKB1, TSC1/2, and PTEN, have been shown to be involved in the mammalian target of rapamycin (mTOR) signaling pathway. Here, we report the identification of the FLCN-interacting protein, FNIP1, and demonstrate its interaction with 5' AMP-activated protein kinase (AMPK), a key molecule for energy sensing that negatively regulates mTOR activity. FNIP1 was phosphorylated by AMPK, and its phosphorylation was reduced by AMPK inhibitors, which resulted in reduced FNIP1 expression. AMPK inhibitors also reduced FLCN phosphorylation. Moreover, FLCN phosphorylation was diminished by rapamycin and amino acid starvation and facilitated by FNIP1 overexpression, suggesting that FLCN may be regulated by mTOR and AMPK signaling. Our data suggest that FLCN, mutated in Birt-Hogg-Dubé syndrome, and its interacting partner FNIP1 may be involved in energy and/or nutrient sensing through the AMPK and mTOR signaling pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Complejos Multienzimáticos/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas , Proteínas Proto-Oncogénicas , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor , Proteínas Quinasas Activadas por AMP , Animales , Proteínas Portadoras/genética , Línea Celular , Clonación Molecular , Activación Enzimática , Humanos , Datos de Secuencia Molecular , Complejos Multiproteicos , Unión Proteica , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Síndrome , Serina-Treonina Quinasas TOR , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
20.
Protein Expr Purif ; 40(2): 424-8, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15766886

RESUMEN

Secretion of a recombinant protein from the yeast Pichia pastoris requires the presence of a signal peptide at the amino terminus. Maintaining the full amino acid sequence of the signal peptide is thought to be important for proper signal processing and protein secretion. We show that at least for one protein, a synthetic human interferon, the presence of a Gateway recombination site within the signal peptide is fully compatible with high levels of protein secretion. The amino termini of the secreted interferon proteins cloned with Gateway and cloned with restriction enzymes and ligase are identical, and the proteins were highly active in biological assays. Compatibility with Gateway cloning simplifies construction of plasmids directing secretion of recombinant proteins from P. pastoris.


Asunto(s)
Clonación Molecular/métodos , Pichia/genética , Señales de Clasificación de Proteína/genética , Proteínas Recombinantes/metabolismo , Secuencia de Bases , ADN Ligasas , Enzimas de Restricción del ADN , Vectores Genéticos , Humanos , Interferones/genética , Interferones/metabolismo , Proteínas Recombinantes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA