Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Nature ; 616(7957): 598-605, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36991125

RESUMEN

Cytotoxic lymphocyte-derived granzyme A (GZMA) cleaves GSDMB, a gasdermin-family pore-forming protein1,2, to trigger target cell pyroptosis3. GSDMB and the charter gasdermin family member GSDMD4,5 have been inconsistently reported to be degraded by the Shigella flexneri ubiquitin-ligase virulence factor IpaH7.8 (refs. 6,7). Whether and how IpaH7.8 targets both gasdermins is undefined, and the pyroptosis function of GSDMB has even been questioned recently6,8. Here we report the crystal structure of the IpaH7.8-GSDMB complex, which shows how IpaH7.8 recognizes the GSDMB pore-forming domain. We clarify that IpaH7.8 targets human (but not mouse) GSDMD through a similar mechanism. The structure of full-length GSDMB suggests stronger autoinhibition than in other gasdermins9,10. GSDMB has multiple splicing isoforms that are equally targeted by IpaH7.8 but exhibit contrasting pyroptotic activities. Presence of exon 6 in the isoforms dictates the pore-forming, pyroptotic activity in GSDMB. We determine the cryo-electron microscopy structure of the 27-fold-symmetric GSDMB pore and depict conformational changes that drive pore formation. The structure uncovers an essential role for exon-6-derived elements in pore assembly, explaining pyroptosis deficiency in the non-canonical splicing isoform used in recent studies6,8. Different cancer cell lines have markedly different isoform compositions, correlating with the onset and extent of pyroptosis following GZMA stimulation. Our study illustrates fine regulation of GSDMB pore-forming activity by pathogenic bacteria and mRNA splicing and defines the underlying structural mechanisms.


Asunto(s)
Gasderminas , Proteínas Citotóxicas Formadoras de Poros , Animales , Humanos , Ratones , Línea Celular Tumoral , Microscopía por Crioelectrón , Cristalografía por Rayos X , Gasderminas/química , Gasderminas/genética , Gasderminas/metabolismo , Gasderminas/ultraestructura , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/ultraestructura , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas Citotóxicas Formadoras de Poros/ultraestructura , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/ultraestructura , Piroptosis , Shigella flexneri , Especificidad de la Especie , Empalme Alternativo
2.
Nat Cell Biol ; 24(2): 253-267, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35102268

RESUMEN

The microtubule cytoskeleton forms complex macromolecular assemblies with a range of microtubule-associated proteins (MAPs) that have fundamental roles in cell architecture, division and motility. Determining how an individual MAP modulates microtubule behaviour is an important step in understanding the physiological roles of various microtubule assemblies. To characterize how MAPs control microtubule properties and functions, we developed an approach allowing for medium-throughput analyses of MAPs in cell-free conditions using lysates of mammalian cells. Our pipeline allows for quantitative as well as ultrastructural analyses of microtubule-MAP assemblies. Analysing 45 bona fide and potential mammalian MAPs, we uncovered previously unknown activities that lead to distinct and unique microtubule behaviours such as microtubule coiling or hook formation, or liquid-liquid phase separation along the microtubule lattice that initiates microtubule branching. We have thus established a powerful tool for a thorough characterization of a wide range of MAPs and MAP variants, thus opening avenues for the determination of mechanisms underlying their physiological roles and pathological implications.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Imagen Individual de Molécula , Fracciones Subcelulares , Animales , Línea Celular Tumoral , Microscopía por Crioelectrón , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Microscopía por Video , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/ultraestructura , Microtúbulos/genética , Microtúbulos/ultraestructura , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/ultraestructura , Transducción de Señal , Factores de Tiempo , Imagen de Lapso de Tiempo , Tubulina (Proteína)/metabolismo
3.
Nat Commun ; 12(1): 4893, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34385445

RESUMEN

The Tweety homologs (TTYHs) are members of a conserved family of eukaryotic membrane proteins that are abundant in the brain. The three human paralogs were assigned to function as anion channels that are either activated by Ca2+ or cell swelling. To uncover their unknown architecture and its relationship to function, we have determined the structures of human TTYH1-3 by cryo-electron microscopy. All structures display equivalent features of a dimeric membrane protein that contains five transmembrane segments and an extended extracellular domain. As none of the proteins shows attributes reminiscent of an anion channel, we revisited functional experiments and did not find any indication of ion conduction. Instead, we find density in an extended hydrophobic pocket contained in the extracellular domain that emerges from the lipid bilayer, which suggests a role of TTYH proteins in the interaction with lipid-like compounds residing in the membrane.


Asunto(s)
Canales de Cloruro/ultraestructura , Microscopía por Crioelectrón/métodos , Proteínas de la Membrana/ultraestructura , Proteínas de Neoplasias/ultraestructura , Canales de Cloruro/química , Canales de Cloruro/metabolismo , Humanos , Canales Iónicos/química , Canales Iónicos/metabolismo , Canales Iónicos/ultraestructura , Membrana Dobles de Lípidos/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Unión Proteica , Conformación Proteica
4.
Nat Commun ; 12(1): 785, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33542223

RESUMEN

The binding of cytoplasmic Ca2+ to the anion-selective channel TMEM16A triggers a conformational change around its binding site that is coupled to the release of a gate at the constricted neck of an hourglass-shaped pore. By combining mutagenesis, electrophysiology, and cryo-electron microscopy, we identified three hydrophobic residues at the intracellular entrance of the neck as constituents of this gate. Mutation of each of these residues increases the potency of Ca2+ and results in pronounced basal activity. The structure of an activating mutant shows a conformational change of an α-helix that contributes to Ca2+ binding as a likely cause for the basal activity. Although not in physical contact, the three residues are functionally coupled to collectively contribute to the stabilization of the gate in the closed conformation of the pore, thus explaining the low open probability of the channel in the absence of Ca2+.


Asunto(s)
Anoctamina-1/metabolismo , Calcio/metabolismo , Activación del Canal Iónico , Proteínas de Neoplasias/metabolismo , Anoctamina-1/genética , Anoctamina-1/ultraestructura , Sitios de Unión/genética , Cationes Bivalentes/metabolismo , Cloruros/metabolismo , Microscopía por Crioelectrón , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Mutagénesis , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/ultraestructura , Unión Proteica , Conformación Proteica en Hélice alfa
5.
Nat Commun ; 12(1): 786, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33542228

RESUMEN

The anion channel TMEM16A is activated by intracellular Ca2+ in a highly cooperative process. By combining electrophysiology and autocorrelation analysis, we investigated the mechanism of channel activation and the concurrent rearrangement of the gate in the narrow part of the pore. Features in the fluctuation characteristics of steady-state current indicate the sampling of intermediate conformations that are successively occupied during gating. The initial step is related to conformational changes induced by Ca2+ binding, which is ensued by rearrangements that open the pore. Mutations in the gate shift the equilibrium of transitions in a manner consistent with a progressive destabilization of this region during pore opening. We come up with a mechanism of channel activation where the binding of Ca2+ induces conformational changes in the protein that, in a sequential manner, propagate from the binding site and couple to the gate in the narrow pore to allow ion permeation.


Asunto(s)
Anoctamina-1/metabolismo , Calcio/metabolismo , Activación del Canal Iónico , Modelos Moleculares , Proteínas de Neoplasias/metabolismo , Regulación Alostérica , Anoctamina-1/genética , Anoctamina-1/ultraestructura , Sitios de Unión/genética , Cationes Bivalentes/metabolismo , Cloruros/metabolismo , Células HEK293 , Humanos , Cinética , Método de Montecarlo , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/ultraestructura , Técnicas de Placa-Clamp , Distribución de Poisson , Unión Proteica/genética , Conformación Proteica en Hélice alfa
6.
BMC Cancer ; 20(1): 1217, 2020 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-33302909

RESUMEN

BACKGROUND: Metastases are the leading cause of breast cancer-related deaths. The tumor microenvironment impacts cancer progression and metastatic ability. Fibrillar collagen, a major extracellular matrix component, can be studied using the light scattering phenomenon known as second-harmonic generation (SHG). The ratio of forward- to backward-scattered SHG photons (F/B) is sensitive to collagen fiber internal structure and has been shown to be an independent prognostic indicator of metastasis-free survival time (MFS). Here we assess the effects of heterogeneity in the tumor matrix on the possible use of F/B as a prognostic tool. METHODS: SHG imaging was performed on sectioned primary tumor excisions from 95 untreated, estrogen receptor-positive, lymph node negative invasive ductal carcinoma patients. We identified two distinct regions whose collagen displayed different average F/B values, indicative of spatial heterogeneity: the cellular tumor bulk and surrounding tumor-stroma interface. To evaluate the impact of heterogeneity on F/B's prognostic ability, we performed SHG imaging in the tumor bulk and tumor-stroma interface, calculated a 21-gene recurrence score (surrogate for OncotypeDX®, or S-ODX) for each patient and evaluated their combined prognostic ability. RESULTS: We found that F/B measured in tumor-stroma interface, but not tumor bulk, is prognostic of MFS using three methods to select pixels for analysis: an intensity threshold selected by a blinded observer, a histogram-based thresholding method, and an adaptive thresholding method. Using both regression trees and Random Survival Forests for MFS outcome, we obtained data-driven prediction rules that show F/B from tumor-stroma interface, but not tumor bulk, and S-ODX both contribute to predicting MFS in this patient cohort. We also separated patients into low-intermediate (S-ODX < 26) and high risk (S-ODX ≥26) groups. In the low-intermediate risk group, comprised of patients not typically recommended for adjuvant chemotherapy, we find that F/B from the tumor-stroma interface is prognostic of MFS and can identify a patient cohort with poor outcomes. CONCLUSIONS: These data demonstrate that intratumoral heterogeneity in F/B values can play an important role in its possible use as a prognostic marker, and that F/B from tumor-stroma interface of primary tumor excisions may provide useful information to stratify patients by metastatic risk.


Asunto(s)
Neoplasias de la Mama/ultraestructura , Carcinoma Ductal de Mama/ultraestructura , Estrógenos , Colágenos Fibrilares/ultraestructura , Metástasis de la Neoplasia , Proteínas de Neoplasias/ultraestructura , Neoplasias Hormono-Dependientes/ultraestructura , Microscopía de Generación del Segundo Armónico , Neoplasias de la Mama/química , Carcinoma Ductal de Mama/química , Carcinoma Ductal de Mama/secundario , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Neoplasias Hormono-Dependientes/química , Pronóstico , Riesgo , Método Simple Ciego , Células del Estroma/química , Células del Estroma/ultraestructura , Microambiente Tumoral
7.
Nat Commun ; 11(1): 4931, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33004795

RESUMEN

Testis-restricted melanoma antigen (MAGE) proteins are frequently hijacked in cancer and play a critical role in tumorigenesis. MAGEs assemble with E3 ubiquitin ligases and function as substrate adaptors that direct the ubiquitination of novel targets, including key tumor suppressors. However, how MAGEs recognize their targets is unknown and has impeded the development of MAGE-directed therapeutics. Here, we report the structural basis for substrate recognition by MAGE ubiquitin ligases. Biochemical analysis of the degron motif recognized by MAGE-A11 and the crystal structure of MAGE-A11 bound to the PCF11 substrate uncovered a conserved substrate binding cleft (SBC) in MAGEs. Mutation of the SBC disrupted substrate recognition by MAGEs and blocked MAGE-A11 oncogenic activity. A chemical screen for inhibitors of MAGE-A11:substrate interaction identified 4-Aminoquinolines as potent inhibitors of MAGE-A11 that show selective cytotoxicity. These findings provide important insights into the large family of MAGE ubiquitin ligases and identify approaches for developing cancer-specific therapeutics.


Asunto(s)
Antígenos de Neoplasias/ultraestructura , Proteínas de Neoplasias/ultraestructura , Neoplasias/tratamiento farmacológico , Ubiquitina-Proteína Ligasas/metabolismo , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Secuencias de Aminoácidos , Aminoquinolinas/farmacología , Aminoquinolinas/uso terapéutico , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinogénesis/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Células HEK293 , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Mutagénesis , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patología , Prueba de Estudio Conceptual , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Dominios Proteicos/genética , Mapeo de Interacción de Proteínas , Relación Estructura-Actividad , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/genética , Ubiquitinación/efectos de los fármacos , Ubiquitinación/genética
8.
J Biol Chem ; 295(28): 9502-9512, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32409586

RESUMEN

Six-transmembrane epithelial antigen of the prostate 1 (STEAP1) is an integral membrane protein that is highly up-regulated on the cell surface of several human cancers, making it a promising therapeutic target to manage these diseases. It shares sequence homology with three enzymes (STEAP2-STEAP4) that catalyze the NADPH-dependent reduction of iron(III). However, STEAP1 lacks an intracellular NADPH-binding domain and does not exhibit cellular ferric reductase activity. Thus, both the molecular function of STEAP1 and its role in cancer progression remain elusive. Here, we present a ∼3.0-Šcryo-EM structure of trimeric human STEAP1 bound to three antigen-binding fragments (Fabs) of the clinically used antibody mAb120.545. The structure revealed that STEAP1 adopts a reductase-like conformation and interacts with the Fabs through its extracellular helices. Enzymatic assays in human cells revealed that STEAP1 promotes iron(III) reduction when fused to the intracellular NADPH-binding domain of its family member STEAP4, suggesting that STEAP1 functions as a ferric reductase in STEAP heterotrimers. Our work provides a foundation for deciphering the molecular mechanisms of STEAP1 and may be useful in the design of new therapeutic strategies to target STEAP1 in cancer.


Asunto(s)
Antígenos de Neoplasias , Proteínas de Neoplasias , Neoplasias/enzimología , Oxidorreductasas , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Antígenos de Neoplasias/ultraestructura , Antineoplásicos Inmunológicos/química , Microscopía por Crioelectrón , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/ultraestructura , Neoplasias/ultraestructura , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Oxidorreductasas/ultraestructura , Dominios Proteicos
9.
Nat Commun ; 10(1): 5433, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31780715

RESUMEN

The human ATP-binding cassette transporter ABCG2 is a key to anticancer resistance and physiological detoxification. However, the molecular mechanism of substrate transport remains enigmatic. A hydrophobic di-leucine motif in the ABCG2 core separates a large intracellular cavity from a smaller upper cavity. We show that the di-leucine motif acts as a valve that controls drug extrusion. Moreover, the extracellular structure engages the re-entry helix and all extracellular loops to form a roof architecture on top of the upper cavity. Disulfide bridges and a salt bridge limit roof flexibility, but provide a lid-like function to control drug release. We propose that drug translocation from the central to the upper cavities through the valve is driven by a squeezing motion, suggesting that ABCG2 operates similar to a peristaltic pump. Finally, the roof contains essential residues, offering therapeutic options to block ABCG2 by either targeting the valve or essential residues in the roof.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Proteínas de Neoplasias/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/ultraestructura , Antineoplásicos/metabolismo , Resistencia a Antineoplásicos/genética , Células HEK293 , Humanos , Mitoxantrona/metabolismo , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/ultraestructura
10.
Nature ; 563(7731): 426-430, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30405239

RESUMEN

ABCG2 is a transporter protein of the ATP-binding-cassette (ABC) family that is expressed in the plasma membrane in cells of various tissues and tissue barriers, including the blood-brain, blood-testis and maternal-fetal barriers1-4. Powered by ATP, it translocates endogenous substrates, affects the pharmacokinetics of many drugs and protects against a wide array of xenobiotics, including anti-cancer drugs5-12. Previous studies have revealed the architecture of ABCG2 and the structural basis of its inhibition by small molecules and antibodies13,14. However, the mechanisms of substrate recognition and ATP-driven transport are unknown. Here we present high-resolution cryo-electron microscopy (cryo-EM) structures of human ABCG2 in a substrate-bound pre-translocation state and an ATP-bound post-translocation state. For both structures, we used a mutant containing a glutamine replacing the catalytic glutamate (ABCG2EQ), which resulted in reduced ATPase and transport rates and facilitated conformational trapping for structural studies. In the substrate-bound state, a single molecule of estrone-3-sulfate (E1S) is bound in a central, hydrophobic and cytoplasm-facing cavity about halfway across the membrane. Only one molecule of E1S can bind in the observed binding mode. In the ATP-bound state, the substrate-binding cavity has collapsed while an external cavity has opened to the extracellular side of the membrane. The ATP-induced conformational changes include rigid-body shifts of the transmembrane domains, pivoting of the nucleotide-binding domains (NBDs), and a change in the relative orientation of the NBD subdomains. Mutagenesis and in vitro characterization of transport and ATPase activities demonstrate the roles of specific residues in substrate recognition, including a leucine residue that forms a 'plug' between the two cavities. Our results show how ABCG2 harnesses the energy of ATP binding to extrude E1S and other substrates, and suggest that the size and binding affinity of compounds are important for distinguishing substrates from inhibitors.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/ultraestructura , Adenosina Trifosfato/metabolismo , Microscopía por Crioelectrón , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestructura , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/ultraestructura , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Sitios de Unión , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutación , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Unión Proteica , Conformación Proteica , Especificidad por Sustrato
11.
Nature ; 557(7703): 62-67, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29695864

RESUMEN

Gasdermins mediate inflammatory cell death after cleavage by caspases or other, unknown enzymes. The cleaved N-terminal fragments bind to acidic membrane lipids to form pores, but the mechanism of pore formation remains unresolved. Here we present the cryo-electron microscopy structures of the 27-fold and 28-fold single-ring pores formed by the N-terminal fragment of mouse GSDMA3 (GSDMA3-NT) at 3.8 and 4.2 Å resolutions, and of a double-ring pore at 4.6 Å resolution. In the 27-fold pore, a 108-stranded anti-parallel ß-barrel is formed by two ß-hairpins from each subunit capped by a globular domain. We identify a positively charged helix that interacts with the acidic lipid cardiolipin. GSDMA3-NT undergoes radical conformational changes upon membrane insertion to form long, membrane-spanning ß-strands. We also observe an unexpected additional symmetric ring of GSDMA3-NT subunits that does not insert into the membrane in the double-ring pore, which may represent a pre-pore state of GSDMA3-NT. These structures provide a basis that explains the activities of several mutant gasdermins, including defective mutants that are associated with cancer.


Asunto(s)
Microscopía por Crioelectrón , Proteínas/química , Proteínas/ultraestructura , Animales , Membrana Celular/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lípidos de la Membrana/metabolismo , Ratones , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/ultraestructura , Mutación , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/ultraestructura , Neoplasias/genética , Perforina/química , Perforina/metabolismo , Proteínas de Unión a Fosfato , Conformación Proteica , Pliegue de Proteína , Multimerización de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas/genética , Proteínas/metabolismo , Relación Estructura-Actividad
12.
Nature ; 546(7659): 504-509, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28554189

RESUMEN

ABCG2 is a constitutively expressed ATP-binding cassette (ABC) transporter that protects many tissues against xenobiotic molecules. Its activity affects the pharmacokinetics of commonly used drugs and limits the delivery of therapeutics into tumour cells, thus contributing to multidrug resistance. Here we present the structure of human ABCG2 determined by cryo-electron microscopy, providing the first high-resolution insight into a human multidrug transporter. We visualize ABCG2 in complex with two antigen-binding fragments of the human-specific, inhibitory antibody 5D3 that recognizes extracellular loops of the transporter. We observe two cholesterol molecules bound in the multidrug-binding pocket that is located in a central, hydrophobic, inward-facing translocation pathway between the transmembrane domains. Combined with functional in vitro analyses, our results suggest a multidrug recognition and transport mechanism of ABCG2, rationalize disease-causing single nucleotide polymorphisms and the allosteric inhibition by the 5D3 antibody, and provide the structural basis of cholesterol recognition by other G-subfamily ABC transporters.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/ultraestructura , Microscopía por Crioelectrón , Proteínas de Neoplasias/química , Proteínas de Neoplasias/ultraestructura , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/ultraestructura , Secuencia de Aminoácidos , Anticuerpos/química , Anticuerpos/inmunología , Anticuerpos/ultraestructura , Sitios de Unión , Transporte Biológico , Colesterol/química , Colesterol/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/ultraestructura , Modelos Moleculares , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Polimorfismo de Nucleótido Simple/genética , Dominios Proteicos
13.
Am J Physiol Cell Physiol ; 310(8): C643-62, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26825122

RESUMEN

Ca(2+)entry into the cell via store-operated Ca(2+)release-activated Ca(2+)(CRAC) channels triggers diverse signaling cascades that affect cellular processes like cell growth, gene regulation, secretion, and cell death. These store-operated Ca(2+)channels open after depletion of intracellular Ca(2+)stores, and their main features are fully reconstituted by the two molecular key players: the stromal interaction molecule (STIM) and Orai. STIM represents an endoplasmic reticulum-located Ca(2+)sensor, while Orai forms a highly Ca(2+)-selective ion channel in the plasma membrane. Functional as well as mutagenesis studies together with structural insights about STIM and Orai proteins provide a molecular picture of the interplay of these two key players in the CRAC signaling cascade. This review focuses on the main experimental advances in the understanding of the STIM1-Orai choreography, thereby establishing a portrait of key mechanistic steps in the CRAC channel signaling cascade. The focus is on the activation of the STIM proteins, the subsequent coupling of STIM1 to Orai1, and the consequent structural rearrangements that gate the Orai channels into the open state to allow Ca(2+)permeation into the cell.


Asunto(s)
Señalización del Calcio , Calcio/química , Proteínas de Neoplasias/química , Proteínas de Neoplasias/ultraestructura , Proteína ORAI1/química , Proteína ORAI1/ultraestructura , Molécula de Interacción Estromal 1/química , Molécula de Interacción Estromal 1/ultraestructura , Sitios de Unión , Calcio/metabolismo , Humanos , Activación del Canal Iónico , Transporte Iónico , Proteínas de la Membrana , Modelos Biológicos , Modelos Químicos , Modelos Moleculares , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Unión Proteica , Conformación Proteica , Molécula de Interacción Estromal 1/metabolismo
14.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 8): 1725-35, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26249353

RESUMEN

ABCG2 is an efflux drug transporter that plays an important role in drug resistance and drug disposition. In this study, the first three-dimensional structure of human full-length ABCG2 analysed by electron crystallography from two-dimensional crystals in the absence of nucleotides and transported substrates is reported at 2 nm resolution. In this state, ABCG2 forms a symmetric homodimer with a noncrystallographic twofold axis perpendicular to the two-dimensional crystal plane, as confirmed by subtomogram averaging. This configuration suggests an inward-facing configuration similar to murine ABCB1, with the nucleotide-binding domains (NBDs) widely separated from each other. In the three-dimensional map, densities representing the long cytoplasmic extensions from the transmembrane domains that connect the NBDs are clearly visible. The structural data have allowed the atomic model of ABCG2 to be refined, in which the two arms of the V-shaped ABCG2 homodimeric complex are in a more closed and narrower conformation. The structural data and the refined model of ABCG2 are compatible with the biochemical analysis of the previously published mutagenesis studies, providing novel insight into the structure and function of the transporter.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Microscopía por Crioelectrón , Proteínas de Neoplasias/química , Estructura Cuaternaria de Proteína , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/ultraestructura , Mama/metabolismo , Neoplasias de la Mama/metabolismo , Microscopía por Crioelectrón/métodos , Cristalización/métodos , Femenino , Humanos , Modelos Moleculares , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/ultraestructura , Multimerización de Proteína
15.
Sci Rep ; 4: 5447, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24965094

RESUMEN

CCL5 (RANTES) is an inflammatory chemokine which binds to chemokine receptor CCR5 and induces signaling. The CCL5:CCR5 associated chemotactic signaling is of critical biological importance and is a potential HIV-1 therapeutic axis. Several studies provided growing evidence for the expression of CCL5 and CCR5 in non-hematological malignancies. Therefore, the delineation of the CCL5:CCR5 complex structure can pave the way for novel CCR5-targeted drugs. We employed a computational protocol which is primarily based on free energy calculations and molecular dynamics simulations, and report, what is to our knowledge, the first computationally derived CCL5:CCR5 complex structure which is in excellent agreement with experimental findings and clarifies the functional role of CCL5 and CCR5 residues which are associated with binding and signaling. A wealth of polar and non-polar interactions contributes to the tight CCL5:CCR5 binding. The structure of an HIV-1 gp120 V3 loop in complex with CCR5 has recently been derived through a similar computational protocol. A comparison between the CCL5 : CCR5 and the HIV-1 gp120 V3 loop : CCR5 complex structures depicts that both the chemokine and the virus primarily interact with the same CCR5 residues. The present work provides insights into the blocking mechanism of HIV-1 by CCL5.


Asunto(s)
Quimiocina CCL5/química , Quimiocina CCL5/ultraestructura , Proteína gp120 de Envoltorio del VIH/química , Modelos Químicos , Receptores CCR5/química , Receptores CCR5/ultraestructura , Animales , Sitios de Unión , Simulación por Computador , Proteína gp120 de Envoltorio del VIH/ultraestructura , Humanos , Modelos Moleculares , Proteínas de Neoplasias/química , Proteínas de Neoplasias/ultraestructura , Unión Proteica , Conformación Proteica
16.
Opt Express ; 21(22): 25655-63, 2013 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-24216790

RESUMEN

High-resolution microscopy for biological specimens was performed using cathodoluminescence (CL) of Y(2)O(3):Eu, Zn nanophosphors, which have high CL intensity due to the incorporation of Zn. The intensity of Y(2)O(3):Eu nanophosphors at low acceleration voltage (3 kV) was increased by adding Zn. The CL intensity was high enough for imaging even with a phosphor size as small as about 30 nm. The results show the possibility of using CL microscopy for biological specimens at single-protein-scale resolution. CL imaging of HeLa cells containing laser-ablated Y(2)O(3):Eu, Zn nanophosphors achieved a spatial resolution of a few tens of nanometers. Y(2)O(3):Eu, Zn nanophosphors in HeLa cells were also imaged with 254 nm ultraviolet light excitation. The results suggest that correlative microscopy using CL, secondary electrons and fluorescence imaging could enable multi-scale investigation of molecular localization from the nanoscale to the microscale.


Asunto(s)
Aumento de la Imagen/métodos , Mediciones Luminiscentes/métodos , Nanopartículas del Metal , Microscopía Fluorescente/métodos , Imagen Molecular/métodos , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/ultraestructura , Medios de Contraste , Células HeLa , Humanos , Iluminación/métodos
18.
J Struct Biol ; 177(1): 160-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22119847

RESUMEN

End binding proteins (EBs) track growing microtubule ends and play a master role in organizing dynamic protein networks. Mammalian cells express up to three different EBs (EB1, EB2, and EB3). Besides forming homodimers, EB1 and EB3 also assemble into heterodimers. One group of EB-binding partners encompasses proteins that harbor CAP-Gly domains. The binding properties of the different EBs towards CAP-Gly proteins have not been systematically investigated. This information is, however, important to compare and contrast functional differences. Here we analyzed the interactions between CLIP-170 and p150(glued) CAP-Gly domains with the three EB homodimers and the EB1-EB3 heterodimer. Using isothermal titration calorimetry we observed that some EBs bind to the individual CAP-Gly domains with similar affinities while others interact with their targets with pronounced differences. We further found that the two types of CAP-Gly domains use alternative mechanisms to target the C-terminal domains of EBs. We succeeded to solve the crystal structure of a complex composed of a heterodimer of EB1 and EB3 C-termini together with the CAP-Gly domain of p150(glued). Together, our results provide mechanistic insights into the interaction properties of EBs and offer a molecular framework for the systematic investigation of their functional differences in cells.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X/métodos , Complejo Dinactina , Humanos , Proteínas Asociadas a Microtúbulos/ultraestructura , Microtúbulos/metabolismo , Datos de Secuencia Molecular , Proteínas de Neoplasias/ultraestructura , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína
19.
J Biochem ; 148(6): 659-67, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20823370

RESUMEN

It is well established that promyelocytic leukaemia nuclear bodies (PML NBs) play important roles in DNA damage responses (DDR). After irradiation, PML NBs dynamically recruit or release important proteins involved in cell-cycle regulation, DNA repair and apoptosis. As PML protein is the key molecule of PML NBs' dynamic assembling, we aimed to characterize the PML-interacting proteins in (60)Co-irradiated MCF-7 cells. A proteomic approach using CoIP, mono-dimensional electrophoresis and tandem mass spectrometry, allowed us to identify a total of 124 proteins that may associate with PML after irradiation. Bioinformatic analysis of the identified proteins showed that most of them were related to characterized PML functions, such as transcriptional regulation, cell-cycle regulation, cell-death regulation and response to stress. Four proteins, B23, MVP, G3BP1 and DHX9, were verified to co-localize with PML differentially before and after ionizing radiation (IR) treatment. The proteins identified in this study will significantly improve our understanding of the dynamic organization and multiple functions of PML NBs in DDR.


Asunto(s)
Apoptosis/efectos de la radiación , Estructuras del Núcleo Celular , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , ADN de Neoplasias/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Proteómica , Factores de Transcripción , Proteínas Supresoras de Tumor/efectos de la radiación , Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/fisiología , Línea Celular Tumoral , Estructuras del Núcleo Celular/metabolismo , Estructuras del Núcleo Celular/efectos de la radiación , Estructuras del Núcleo Celular/ultraestructura , Femenino , Humanos , Leucemia Promielocítica Aguda/metabolismo , Microscopía Fluorescente , Proteínas de Neoplasias/ultraestructura , Proteínas Nucleares/metabolismo , Proteínas Nucleares/ultraestructura , Proteína de la Leucemia Promielocítica , Factores de Transcripción/metabolismo , Factores de Transcripción/ultraestructura , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/fisiología , Proteínas Supresoras de Tumor/ultraestructura
20.
Cell Calcium ; 46(4): 227-32, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19733393

RESUMEN

Ca(2+) entry in non-excitable cells is mainly carried by store-operated channels among which the CRAC channel is best characterized. Its two limiting molecular components are represented by the Ca(2+) sensor protein STIM1 located in the endoplasmic reticulum and Orai1 in the plasma membrane. STIM1 senses a decrease of the Ca(2+) content in internal stores and triggers its accumulation into puncta like structures resulting in coupling to as well as activation of Orai1 channels. The STIM1-Orai coupling process is determined by an interaction via their C-termini. This review highlights recent developments on domains particularly within the cytosolic part of STIM1 that govern this interaction.


Asunto(s)
Canales de Calcio/química , Canales de Calcio/metabolismo , Canales de Calcio/ultraestructura , Calcio/química , Calcio/metabolismo , Activación del Canal Iónico , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/ultraestructura , Dominios y Motivos de Interacción de Proteínas , Animales , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Humanos , Proteína ORAI1 , Unión Proteica , Molécula de Interacción Estromal 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...