Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Biophys J ; 123(2): 172-183, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38071428

RESUMEN

Heat shock protein 90 (Hsp90) serves as a crucial regulator of cellular proteostasis by stabilizing and regulating the activity of numerous substrates, many of which are oncogenic proteins. Therefore, Hsp90 is a drug target for cancer therapy. Hsp90 comprises three structural domains, a highly conserved amino-terminal domain (NTD), a middle domain (MD), and a carboxyl-terminal domain (CTD). The CTD is responsible for protein dimerization, is crucial for Hsp90's activity, and has therefore been targeted for inhibiting Hsp90. Here we addressed the question of whether the CTD dimerization in Hsp90, in the absence of bound nucleotides, is modulated by allosteric effects from the other domains. We studied full length (FL) and isolated CTD (isoC) yeast Hsp90 spin-labeled with a Gd(III) tag by double electron-electron resonance measurements to track structural differences and to determine the apparent dissociation constant (Kd). We found the distance distributions for both the FL and isoC to be similar, indicating that the removal of the NTD and MD does not significantly affect the structure of the CTD dimer. The low-temperature double electron-electron resonance-derived Kd values, as well as those obtained at room temperature using microscale thermophoresis and native mass spectrometry, collectively suggested the presence of some allosteric effects from the NTDs and MDs on the CTD dimerization stability in the apo state. This was evidenced by a moderate increase in the Kd for the isoC compared with the FL mutants. Our results reveal a fine regulation of the CTD dimerization by allosteric modulation, which may have implications for drug targeting strategies in cancer therapy.


Asunto(s)
Neoplasias , Saccharomyces cerevisiae , Humanos , Dimerización , Saccharomyces cerevisiae/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Multimerización de Proteína , Unión Proteica
2.
Life Sci Alliance ; 6(10)2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37460146

RESUMEN

Protein degradation is one of the essential mechanisms that enables reshaping of the proteome landscape in response to various stimuli. The largest E3 ubiquitin ligase family that targets proteins to degradation by catalyzing ubiquitination is the cullin-RING ligases (CRLs). Many of the proteins that are regulated by CRLs are central to tumorigenesis and tumor progression, and dysregulation of the CRL family is frequently associated with cancer. The CRL family comprises ∼300 complexes, all of which are regulated by the COP9 signalosome complex (CSN). Therefore, CSN is considered an attractive target for therapeutic intervention. Research efforts for targeted CSN inhibition have been directed towards inhibition of the complex enzymatic subunit, CSN5. Here, we have taken a fresh approach focusing on CSNAP, the smallest CSN subunit. Our results show that the C-terminal region of CSNAP is tightly packed within the CSN complex, in a groove formed by CSN3 and CSN8. We show that a 16 amino acid C-terminal peptide, derived from this CSN-interacting region, can displace the endogenous CSNAP subunit from the complex. This, in turn, leads to a CSNAP null phenotype that attenuates CSN activity and consequently CRLs function. Overall, our findings emphasize the potential of a CSNAP-based peptide for CSN inhibition as a new therapeutic avenue.


Asunto(s)
Ubiquitina-Proteína Ligasas , Complejo del Señalosoma COP9/genética , Complejo del Señalosoma COP9/metabolismo , Ubiquitinación , Ubiquitina-Proteína Ligasas/metabolismo , Fenotipo
3.
Pharmaceutics ; 13(10)2021 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-34683850

RESUMEN

Human serum albumin (HSA) is efficiently taken up by cancer cells as a source of carbon and energy. In this study, we prepared a monomodified derivative of HSA covalently linked to an EDTA derivative and investigated its efficacy to shuttle weakly anti-proliferative EDTA associating ligands such as vanadium, into a cancer cell line. HSA-S-MAL-(CH2)2-NH-CO-EDTA was found to associate both with the vanadium anion (+5) and the vanadium cation (+4) with more than thrice the associating affinity of those ligands toward EDTA. Both conjugates internalized into glioma tumor cell line via caveolae-mediated endocytosis pathway and showed potent anti-proliferative capacities. IC50 values were in the range of 0.2 to 0.3 µM, potentiating the anti-proliferative efficacies of vanadium (+4) and vanadium (+5) twenty to thirty fold, respectively. HSA-EDTA-VO++ in particular is a cancer permeable prodrug conjugate. The associated vanadium (+4) is not released, nor is it active anti-proliferatively prior to its engagement with the cancerous cells. The bound vanadium (+4) dissociates from the conjugate under acidic conditions with half maximal value at pH 5.8. In conclusion, the anti-proliferative activity feature of vanadium can be amplified and directed toward a cancer cell line. This is accomplished using a specially designed HSA-EDTA-shuttling vehicle, enabling vanadium to be anti-proliferatively active at the low micromolar range of concentration.

4.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33846247

RESUMEN

The P-loop Walker A motif underlies hundreds of essential enzyme families that bind nucleotide triphosphates (NTPs) and mediate phosphoryl transfer (P-loop NTPases), including the earliest DNA/RNA helicases, translocases, and recombinases. What were the primordial precursors of these enzymes? Could these large and complex proteins emerge from simple polypeptides? Previously, we showed that P-loops embedded in simple ßα repeat proteins bind NTPs but also, unexpectedly so, ssDNA and RNA. Here, we extend beyond the purely biophysical function of ligand binding to demonstrate rudimentary helicase-like activities. We further constructed simple 40-residue polypeptides comprising just one ß-(P-loop)-α element. Despite their simplicity, these P-loop prototypes confer functions such as strand separation and exchange. Foremost, these polypeptides unwind dsDNA, and upon addition of NTPs, or inorganic polyphosphates, release the bound ssDNA strands to allow reformation of dsDNA. Binding kinetics and low-resolution structural analyses indicate that activity is mediated by oligomeric forms spanning from dimers to high-order assemblies. The latter are reminiscent of extant P-loop recombinases such as RecA. Overall, these P-loop prototypes compose a plausible description of the sequence, structure, and function of the earliest P-loop NTPases. They also indicate that multifunctionality and dynamic assembly were key in endowing short polypeptides with elaborate, evolutionarily relevant functions.


Asunto(s)
Dominio AAA/genética , Dominio AAA/fisiología , Secuencias de Aminoácidos/fisiología , Secuencia de Aminoácidos/genética , ADN Helicasas/metabolismo , ADN Helicasas/fisiología , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , Modelos Moleculares , Nucleósido-Trifosfatasa/química , Péptidos/química , Fosfatos/química , Conformación Proteica en Hélice alfa/fisiología , Conformación Proteica en Lámina beta/fisiología , Proteínas/química , ARN/química , Rec A Recombinasas/metabolismo
5.
Biochim Biophys Acta Mol Basis Dis ; 1867(3): 166041, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33338594

RESUMEN

Accumulating evidence arising from numerous clinical studies indicate that assembled and functional 20S proteasome complexes circulate freely in plasma. Elevated levels of this core proteolytic complex have been found in the plasma of patients suffering from blood, skin and solid cancers, autoimmune disorders, trauma and sepsis. Moreover, in various diseases, there is a positive correlation between circulating 20S proteasome (c20S) levels and treatment efficacy and survival rates, suggesting the involvement of this under-studied c20S complex in pathophysiology. However, many aspects of this system remain enigmatic, as we still do not know the origin, biological role or mechanisms of extracellular transport and regulation of c20S proteasomes. In this review, we provide an overview of the current understanding of the c20S proteasome system and discuss the remaining gaps in knowledge.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/sangre , Animales , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/metabolismo , Quemaduras/sangre , Quemaduras/metabolismo , Neoplasias Hematológicas/sangre , Neoplasias Hematológicas/metabolismo , Humanos , Neoplasias/sangre , Neoplasias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Sepsis/sangre , Sepsis/metabolismo
6.
J Mol Biol ; 431(24): 4796-4816, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31520601

RESUMEN

Methionine S-adenosyltransferases (MATs) are predominantly homotetramers, comprised of dimers of dimers. The larger, highly conserved intradimeric interface harbors two active sites, making the dimer the obligatory functional unit. However, functionality of the smaller, more diverged, and recently evolved interdimeric interface is largely unknown. Here, we show that the interdimeric interface of Ureaplasmaurealiticum MAT has evolved to control the catalytic activity and structural integrity of the homotetramer in response to product accumulation. When all four active sites are occupied with the product, S-adenosylmethionine (SAM), binding of four additional SAM molecules to the interdimeric interface prompts a ∼45° shift in the dimer orientation and a concomitant ∼60% increase in the interface area. This rearrangement inhibits the enzymatic activity by locking the flexible active site loops in a closed state and renders the tetramer resistant to proteolytic degradation. Our findings suggest that the interdimeric interface of MATs is subject to rapid evolutionary changes that tailor the molecular properties of the entire homotetramer to the specific needs of the organism.


Asunto(s)
Metionina Adenosiltransferasa/química , Multimerización de Proteína , Ureaplasma/enzimología , Sitios de Unión , Espectrometría de Masas , Metionina Adenosiltransferasa/metabolismo , Modelos Moleculares , Conformación Proteica , Estabilidad Proteica , Proteolisis , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Relación Estructura-Actividad
7.
PLoS Comput Biol ; 15(8): e1007207, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31442220

RESUMEN

Antibodies developed for research and clinical applications may exhibit suboptimal stability, expressibility, or affinity. Existing optimization strategies focus on surface mutations, whereas natural affinity maturation also introduces mutations in the antibody core, simultaneously improving stability and affinity. To systematically map the mutational tolerance of an antibody variable fragment (Fv), we performed yeast display and applied deep mutational scanning to an anti-lysozyme antibody and found that many of the affinity-enhancing mutations clustered at the variable light-heavy chain interface, within the antibody core. Rosetta design combined enhancing mutations, yielding a variant with tenfold higher affinity and substantially improved stability. To make this approach broadly accessible, we developed AbLIFT, an automated web server that designs multipoint core mutations to improve contacts between specific Fv light and heavy chains (http://AbLIFT.weizmann.ac.il). We applied AbLIFT to two unrelated antibodies targeting the human antigens VEGF and QSOX1. Strikingly, the designs improved stability, affinity, and expression yields. The results provide proof-of-principle for bypassing laborious cycles of antibody engineering through automated computational affinity and stability design.


Asunto(s)
Afinidad de Anticuerpos , Diseño de Fármacos , Región Variable de Inmunoglobulina/genética , Ingeniería de Proteínas/métodos , Animales , Afinidad de Anticuerpos/genética , Biología Computacional , Células HEK293 , Humanos , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/genética , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/química , Cadenas Ligeras de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/química , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/antagonistas & inhibidores , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/inmunología , Biblioteca de Péptidos , Ingeniería de Proteínas/estadística & datos numéricos , Estabilidad Proteica , Programas Informáticos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/inmunología
8.
J Proteomics ; 193: 1-9, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30557664

RESUMEN

Proteasome dysfunction is emerging as a novel pathomechanism for the development of chronic obstructive pulmonary disease (COPD), a major leading cause of death in the world. Cigarette smoke, one of the main risk factors for COPD, impairs proteasome function in vitro and in vivo. In the present study, we dissected the molecular changes induced by cigarette smoke on the proteasome in lung epithelial cells and mouse lungs. 26S proteasome pull-down, MS interactome, and stoichiometry analyses indicated that 26S proteasome complexes become instable in cigarette smoke-treated lung epithelial cells as well as in lungs of mice after three day smoke exposure. The interactome of the 26S was clearly altered in mouse lungs upon smoke exposure but not in cells after 24 h of smoke exposure. Using native MS analysis of purified 20S proteasomes, we observed some destabilization of 20S complexes purified from cigarette smoke-exposed cells in the absence of any dominant and inhibitory modification of proteasomal proteins. Taken together, our results suggest that cigarette smoke induces minor but detectable changes in the stability of 20S and 26S proteasome complexes which might contribute to imbalanced proteostasis in a chronic setting as observed in chronic lung diseases associated with cigarette smoking.


Asunto(s)
Fumar Cigarrillos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteostasis , Animales , Fumar Cigarrillos/patología , Células HEK293 , Humanos , Espectrometría de Masas , Ratones , Factores de Tiempo
9.
Proc Natl Acad Sci U S A ; 115(51): E11943-E11950, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30504143

RESUMEN

Abundant and essential motifs, such as phosphate-binding loops (P-loops), are presumed to be the seeds of modern enzymes. The Walker-A P-loop is absolutely essential in modern NTPase enzymes, in mediating binding, and transfer of the terminal phosphate groups of NTPs. However, NTPase function depends on many additional active-site residues placed throughout the protein's scaffold. Can motifs such as P-loops confer function in a simpler context? We applied a phylogenetic analysis that yielded a sequence logo of the putative ancestral Walker-A P-loop element: a ß-strand connected to an α-helix via the P-loop. Computational design incorporated this element into de novo designed ß-α repeat proteins with relatively few sequence modifications. We obtained soluble, stable proteins that unlike modern P-loop NTPases bound ATP in a magnesium-independent manner. Foremost, these simple P-loop proteins avidly bound polynucleotides, RNA, and single-strand DNA, and mutations in the P-loop's key residues abolished binding. Binding appears to be facilitated by the structural plasticity of these proteins, including quaternary structure polymorphism that promotes a combined action of multiple P-loops. Accordingly, oligomerization enabled a 55-aa protein carrying a single P-loop to confer avid polynucleotide binding. Overall, our results show that the P-loop Walker-A motif can be implemented in small and simple ß-α repeat proteins, primarily as a polynucleotide binding motif.


Asunto(s)
Sitios de Unión , Fosfatos/química , Dominios y Motivos de Interacción de Proteínas , Proteínas/química , Adenosina Trifosfato/química , Secuencia de Aminoácidos , Dominio Catalítico , ADN , Evolución Molecular , Magnesio , Modelos Moleculares , Mutación , Nucleósido-Trifosfatasa/química , Filogenia , Polinucleótidos , Unión Proteica , Conformación Proteica , ARN , Proteínas de Unión al ARN/química , Alineación de Secuencia , Homología de Secuencia de Aminoácido
11.
Cell Death Differ ; 24(12): 2187-2198, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28885617

RESUMEN

The tumor suppressor p53 is a transcription factor that regulates the expression of a range of target genes in response to cellular stress. Adding to the complexity of understanding its cellular function is that in addition to the full-length protein, several p53 isoforms are produced in humans, harboring diverse expression patterns and functionalities. One isoform, Δ40p53, which lacks the first transactivation domain including the binding region for the negative regulator MDM2, was shown to be a product of alternative translation initiation. Here we report the discovery of an alternative cellular mechanism for Δ40p53 formation. We show that the 20S proteasome specifically cleaves the full-length protein (FLp53) to generate the Δ40p53 isoform. Moreover, we demonstrate that a dimer of FLp53 interacts with a Δ40p53 dimer, creating a functional hetero-tetramer. Consequently, the co-expression of both isoforms attenuates the transcriptional activity of FLp53 in a dominant negative manner. Finally, we demonstrate that following oxidative stress, at the time when the 20S proteasome becomes the major degradation machinery and FLp53 is activated, the formation of Δ40p53 is enhanced, creating a negative feedback loop that balances FLp53 activation. Overall, our results suggest that Δ40p53 can be generated by a 20S proteasome-mediated post-translational mechanism so as to control p53 function. More generally, the discovery of a specific cleavage function for the 20S proteasome may represent a more general cellular regulatory mechanism to produce proteins with distinct functional properties.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Proteína p53 Supresora de Tumor/metabolismo , Animales , Células HEK293 , Humanos , Ratas , Proteínas Recombinantes/metabolismo
12.
Proc Natl Acad Sci U S A ; 114(11): 2904-2909, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28242691

RESUMEN

Membrane protein function can be affected by the physical state of the lipid bilayer and specific lipid-protein interactions. For Na,K-ATPase, bilayer properties can modulate pump activity, and, as observed in crystal structures, several lipids are bound within the transmembrane domain. Furthermore, Na,K-ATPase activity depends on phosphatidylserine (PS) and cholesterol, which stabilize the protein, and polyunsaturated phosphatidylcholine (PC) or phosphatidylethanolamine (PE), known to stimulate Na,K-ATPase activity. Based on lipid structural specificity and kinetic mechanisms, specific interactions of both PS and PC/PE have been inferred. Nevertheless, specific binding sites have not been identified definitively. We address this question with native mass spectrometry (MS) and site-directed mutagenesis. Native MS shows directly that one molecule each of 18:0/18:1 PS and 18:0/20:4 PC can bind specifically to purified human Na,K-ATPase (α1ß1). By replacing lysine residues at proposed phospholipid-binding sites with glutamines, the two sites have been identified. Mutations in the cytoplasmic αL8-9 loop destabilize the protein but do not affect Na,K-ATPase activity, whereas mutations in transmembrane helices (TM), αTM2 and αTM4, abolish the stimulation of activity by 18:0/20:4 PC but do not affect stability. When these data are linked to crystal structures, the underlying mechanism of PS and PC/PE effects emerges. PS (and cholesterol) bind between αTM 8, 9, 10, near the FXYD subunit, and maintain topological integrity of the labile C terminus of the α subunit (site A). PC/PE binds between αTM2, 4, 6, and 9 and accelerates the rate-limiting E1P-E2P conformational transition (site B). We discuss the potential physiological implications.


Asunto(s)
Sitios de Unión , Fosfolípidos/química , Fosfolípidos/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/química , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Activación Enzimática , Humanos , Espectrometría de Masas , Modelos Moleculares , Conformación Molecular , Unión Proteica , Estabilidad Proteica
13.
Am J Physiol Cell Physiol ; 311(5): C793-C804, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27605452

RESUMEN

Degradation by the proteasome is the fate for a large portion of cellular proteins, and it plays a major role in maintaining protein homeostasis, as well as in regulating many cellular processes like cell cycle progression. A decrease in proteasome activity has been linked to aging and several age-related neurodegenerative pathologies and highlights the importance of the ubiquitin proteasome system regulation. While the proteasome has been traditionally viewed as a constitutive element of proteolysis, major studies have highlighted how different regulatory mechanisms can impact its activity. Importantly, alterations of proteasomal activity may have major impacts for its function and in therapeutics. On one hand, increasing proteasome activity could be beneficial to prevent the age-related downfall of protein homeostasis, whereas inhibiting or reducing its activity can prevent the proliferation of cancer cells.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas/metabolismo , Animales , Proliferación Celular/fisiología , Homeostasis/fisiología , Humanos , Neoplasias/metabolismo , Neoplasias/patología
14.
EMBO J ; 35(12): 1254-75, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27220849

RESUMEN

Membrane-less organelles in cells are large, dynamic protein/protein or protein/RNA assemblies that have been reported in some cases to have liquid droplet properties. However, the molecular interactions underlying the recruitment of components are not well understood. Herein, we study how the ability to form higher-order assemblies influences the recruitment of the speckle-type POZ protein (SPOP) to nuclear speckles. SPOP, a cullin-3-RING ubiquitin ligase (CRL3) substrate adaptor, self-associates into higher-order oligomers; that is, the number of monomers in an oligomer is broadly distributed and can be large. While wild-type SPOP localizes to liquid nuclear speckles, self-association-deficient SPOP mutants have a diffuse distribution in the nucleus. SPOP oligomerizes through its BTB and BACK domains. We show that BTB-mediated SPOP dimers form linear oligomers via BACK domain dimerization, and we determine the concentration-dependent populations of the resulting oligomeric species. Higher-order oligomerization of SPOP stimulates CRL3(SPOP) ubiquitination efficiency for its physiological substrate Gli3, suggesting that nuclear speckles are hotspots of ubiquitination. Dynamic, higher-order protein self-association may be a general mechanism to concentrate functional components in membrane-less cellular bodies.


Asunto(s)
Núcleo Celular/metabolismo , Sustancias Macromoleculares/metabolismo , Proteínas Nucleares/metabolismo , Multimerización de Proteína , Proteínas Represoras/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Dominios Proteicos , Ubiquitinación , Proteína Gli3 con Dedos de Zinc
15.
J Am Soc Mass Spectrom ; 27(6): 1062-70, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27048419

RESUMEN

Missense mutations that lead to the expression of mutant proteins carrying single amino acid substitutions are the cause of numerous diseases. Unlike gene lesions, insertions, deletions, nonsense mutations, or modified RNA splicing, which affect the length of a polypeptide, or determine whether a polypeptide is translated at all, missense mutations exert more subtle effects on protein structure, which are often difficult to evaluate. Here, we took advantage of the spectral resolution afforded by the EMR Orbitrap platform, to generate a mass spectrometry-based approach relying on simultaneous measurements of the wild-type protein and the missense variants. This approach not only considerably shortens the analysis time due to the concurrent acquisition but, more importantly, enables direct comparisons between the wild-type protein and the variants, allowing identification of even subtle structural changes. We demonstrate our approach using the Parkinson's-associated protein, DJ-1. Together with the wild-type protein, we examined two missense mutants, DJ-1A104T and DJ-1D149A, which lead to early-onset familial Parkinson's disease. Gas-phase, thermal, and chemical stability assays indicate clear alterations in the conformational stability of the two mutants: the structural stability of DJ-1D149A is reduced, whereas that of DJ-1A104T is enhanced. Overall, we anticipate that the methodology presented here will be applicable to numerous other missense mutants, promoting the structural investigations of multiple variants of the same protein. Graphical Abstract ᅟ.


Asunto(s)
Espectrometría de Masas , Mutación Missense , Proteína Desglicasa DJ-1/química , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas Oncogénicas , Enfermedad de Parkinson , Proteína Desglicasa DJ-1/genética
16.
Sci Signal ; 8(379): ra53, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26038598

RESUMEN

Despite initial responses to targeted kinase inhibitors, lung cancer patients presenting with primary epidermal growth factor receptor (EGFR) mutations acquire resistance, often due to a second-site mutation (T790M). However, clinical trials found no survival benefits in patients treated with a monoclonal antibody (mAb) to EGFR that should block activation of the mutated receptor and thus bypass resistance to molecules that target the catalytic or ATP-binding site. Using cell lines with the T790M mutation, we discovered that prolonged exposure to mAbs against only the EGFR triggered network rewiring by (i) stimulating the extracellular signal-regulated kinase (ERK) pathway; (ii) inducing the transcription of HER2 (human epidermal growth factor receptor 2) and HER3, which encode other members of the EGFR family, and the gene encoding HGF, which is the ligand for the receptor tyrosine kinase MET; and (iii) stimulating the interaction between MET and HER3, which promoted MET activity. Supplementing the EGFR-specific mAb with those targeting HER2 and HER3 suppressed these compensatory feedback loops in cultured lung cancer cells. The triple mAb combination targeting all three receptors prevented the activation of ERK, accelerated the degradation of the receptors, inhibited the proliferation of tumor cells but not of normal cells, and markedly reduced the growth of tumors in mice xenografted with cells that were resistant to combined treatment with erlotinib and the single function-blocking EGFR mAb. These findings uncovered feedback loops that enable resistance to treatment paradigms that use a single antibody and indicate a new strategy for the treatment of lung cancer patients.


Asunto(s)
Anticuerpos Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Clorhidrato de Erlotinib , Neoplasias Pulmonares/tratamiento farmacológico , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-3/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Protein Sci ; 24(8): 1193-209, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25970732

RESUMEN

Mass spectrometry (MS) is becoming increasingly popular in the field of structural biology for analyzing protein three-dimensional-structures and for mapping protein-protein interactions. In this review, the specific contributions of chemical crosslinking and native MS are outlined to reveal the structural features of proteins and protein assemblies. Both strategies are illustrated based on the examples of the tetrameric tumor suppressor protein p53 and multisubunit vinculin-Arp2/3 hybrid complexes. We describe the distinct advantages and limitations of each technique and highlight synergistic effects when both techniques are combined. Integrating both methods is especially useful for characterizing large protein assemblies and for capturing transient interactions. We also point out the future directions we foresee for a combination of in vivo crosslinking and native MS for structural investigation of intact protein assemblies.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Espectrometría de Masas/métodos , Mapeo de Interacción de Proteínas/métodos , Proteínas/química , Proteínas/metabolismo , Animales , Humanos , Modelos Moleculares , Conformación Proteica , Proteómica/métodos , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo
18.
Nat Commun ; 6: 6609, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25833141

RESUMEN

The Parkinson's-associated protein, DJ-1, is a highly conserved homodimer, ubiquitously expressed in cells. Here we demonstrate that DJ-1 is a 20S proteasome regulator. We show that DJ-1 physically binds the 20S proteasome and inhibits its activity, rescuing partially unfolded proteins from degradation. Consequently, DJ-1 stabilizes the cellular levels of 20S proteasome substrates, as we show for α-synuclein and p53. Furthermore, we demonstrate that following oxidative stress, DJ-1 is involved in the Nrf2-dependent oxidative stress response that leads to the upregulation of both the 20S proteasome and its regulator, NQO1. Overall, our results suggest a regulatory circuit in which DJ-1, under conditions of oxidative stress, both upregulates and inhibits the 20S proteasome, providing a rigorous control mechanism at a time when the 20S proteasome becomes the major proteolytic machinery. Such a tight regulation of the 20S proteasome may sustain the balance between the need to rapidly eliminate oxidatively damaged proteins and maintain the abundance of native, intrinsically unstructured proteins, which coordinate regulatory and signalling events.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Estrés Oxidativo , Peroxirredoxinas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Línea Celular , Células HEK293 , Células HeLa , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Mutación , Proteína Desglicasa DJ-1 , Ratas , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
19.
Cell Host Microbe ; 16(3): 351-63, 2014 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-25211077

RESUMEN

Autotransporters deliver virulence factors to the bacterial surface by translocating an effector passenger domain through a membrane-anchored barrel structure. Although passenger domains are diverse, those found in enteric bacteria autotransporters, including AIDA-I in diffusely adhering Escherichia coli (DAEC) and TibA in enterotoxigenic E. coli, are commonly glycosylated. We show that AIDA-I is heptosylated within the bacterial cytoplasm by autotransporter adhesin heptosyltransferase (AAH) and its paralogue AAH2. AIDA-I heptosylation determines DAEC adhesion to host cells. AAH/AAH2 define a bacterial autotransporter heptosyltransferase (BAHT) family that contains ferric ion and adopts a dodecamer assembly. Structural analyses of the heptosylated TibA passenger domain reveal 35 heptose conjugates forming patterned and solenoid-like arrays on the surface of a ß helix. Additionally, CARC, the AIDA-like autotransporter from Citrobacter rodentium, is essential for colonization in mice and requires heptosylation by its cognate BAHT. Our study establishes a bacterial glycosylation system that regulates virulence and is essential for pathogenesis.


Asunto(s)
Adhesinas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Glicosiltransferasas/metabolismo , Hierro/metabolismo , Adhesinas de Escherichia coli/genética , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana , Citrobacter rodentium/enzimología , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidad , Citrobacter rodentium/fisiología , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli/genética , Escherichia coli/patogenicidad , Escherichia coli/fisiología , Infecciones por Escherichia coli/microbiología , Glicosilación , Glicosiltransferasas/química , Glicosiltransferasas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Alineación de Secuencia , Virulencia
20.
Mol Cell Biol ; 34(6): 1066-76, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24421388

RESUMEN

The COP9 signalosome (CSN) is an evolutionarily conserved protein complex that participates in the regulation of the ubiquitin/26S proteasome pathway by controlling the function of cullin-RING-ubiquitin ligases. Impressive progress has been made in deciphering its critical role in diverse cellular and developmental processes. However, little is known about the underlying regulatory principles that coordinate its function. Through biochemical and fluorescence microscopy analyses, we determined that the complex is localized in the cytoplasm, nucleoplasm, and chromatin-bound fractions, each differing in the composition of posttranslationally modified subunits, depending on its location within the cell. During the cell cycle, the segregation between subcellular localizations remains steady. However, upon UV damage, a dose-dependent temporal shuttling of the CSN complex into the nucleus was seen, accompanied by upregulation of specific phosphorylations within CSN1, CSN3, and CSN8. Taken together, our results suggest that the specific spatiotemporal composition of the CSN is highly controlled, enabling the complex to rapidly adapt and respond to DNA damage.


Asunto(s)
Daño del ADN/genética , Complejos Multiproteicos/genética , Péptido Hidrolasas/genética , Procesamiento Proteico-Postraduccional/genética , Complejo del Señalosoma COP9 , Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Núcleo Celular/genética , Cromatina/genética , Citoplasma/genética , Células HEK293 , Células HeLa , Humanos , Fosforilación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA