Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 75(4): 823-834.e5, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31302001

RESUMEN

Sirt3, as a major mitochondrial nicotinamide adenine dinucleotide (NAD)-dependent deacetylase, is required for mitochondrial metabolic adaption to various stresses. However, how to regulate Sirt3 activity responding to metabolic stress remains largely unknown. Here, we report Sirt3 as a SUMOylated protein in mitochondria. SUMOylation suppresses Sirt3 catalytic activity. SUMOylation-deficient Sirt3 shows elevated deacetylation on mitochondrial proteins and increased fatty acid oxidation. During fasting, SUMO-specific protease SENP1 is accumulated in mitochondria and quickly de-SUMOylates and activates Sirt3. SENP1 deficiency results in hyper-SUMOylation of Sirt3 and hyper-acetylation of mitochondrial proteins, which reduces mitochondrial metabolic adaption responding to fasting. Furthermore, we find that fasting induces SENP1 translocation into mitochondria to activate Sirt3. The studies on mice show that Sirt3 SUMOylation mutation reduces fat mass and antagonizes high-fat diet (HFD)-induced obesity via increasing oxidative phosphorylation and energy expenditure. Our results reveal that SENP1-Sirt3 signaling modulates Sirt3 activation and mitochondrial metabolism during metabolic stress.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Mitocondrias/metabolismo , Mutación , Obesidad/metabolismo , Transducción de Señal , Sirtuina 3/metabolismo , Sumoilación , Acetilación , Animales , Cisteína Endopeptidasas/genética , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Mutantes , Mitocondrias/genética , Mitocondrias/patología , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , Sirtuina 3/genética
2.
Proc Natl Acad Sci U S A ; 120(13): e2221049120, 2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-36940332

RESUMEN

Smart adhesives that can be applied and removed on demand play an important role in modern life and manufacturing. However, current smart adhesives made of elastomers suffer from the long-standing challenges of the adhesion paradox (rapid decrease in adhesion strength on rough surfaces despite adhesive molecular interactions) and the switchability conflict (trade-off between adhesion strength and easy detachment). Here, we report the use of shape-memory polymers (SMPs) to overcome the adhesion paradox and switchability conflict on rough surfaces. Utilizing the rubbery-glassy phase transition in SMPs, we demonstrate, through mechanical testing and mechanics modeling, that the conformal contact in the rubbery state followed by the shape-locking effect in the glassy state results in the so-called rubber-to-glass (R2G) adhesion (defined as making contact in the rubbery state to a certain indentation depth followed by detachment in the glassy state), with extraordinary adhesion strength (>1 MPa) proportional to the true surface area of a rough surface, overcoming the classic adhesion paradox. Furthermore, upon transitioning back to the rubbery state, the SMP adhesives can detach easily due to the shape-memory effect, leading to a simultaneous improvement in adhesion switchability (up to 103, defined as the ratio of the SMP R2G adhesion to its rubbery-state adhesion) as the surface roughness increases. The working principle and the mechanics model of R2G adhesion provide guidelines for developing stronger and more switchable adhesives adaptable to rough surfaces, thereby enhancing the capabilities of smart adhesives, and impacting various fields such as adhesive grippers and climbing robots.

3.
Blood ; 141(7): 766-786, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36322939

RESUMEN

Extramedullary infiltration (EMI) is a concomitant manifestation that may indicate poor outcome of acute myeloid leukemia (AML). The underlying mechanism remains poorly understood and therapeutic options are limited. Here, we employed single-cell RNA sequencing on bone marrow (BM) and EMI samples from a patient with AML presenting pervasive leukemia cutis. A complement C1Q+ macrophage-like leukemia subset, which was enriched within cutis and existed in BM before EMI manifestations, was identified and further verified in multiple patients with AML. Genomic and transcriptional profiling disclosed mutation and gene expression signatures of patients with EMI that expressed high levels of C1Q. RNA sequencing and quantitative proteomic analysis revealed expression dynamics of C1Q from primary to relapse. Univariate and multivariate analysis demonstrated adverse prognosis significance of C1Q expression. Mechanistically, C1Q expression, which was modulated by transcription factor MAF BZIP transcription factor B, endowed leukemia cells with tissue infiltration ability, which could establish prominent cutaneous or gastrointestinal EMI nodules in patient-derived xenograft and cell line-derived xenograft models. Fibroblasts attracted migration of the C1Q+ leukemia cells through C1Q-globular C1Q receptor recognition and subsequent stimulation of transforming growth factor ß1. This cell-to-cell communication also contributed to survival of C1Q+ leukemia cells under chemotherapy stress. Thus, C1Q served as a marker for AML with adverse prognosis, orchestrating cancer infiltration pathways through communicating with fibroblasts and represents a compelling therapeutic target for EMI.


Asunto(s)
Complemento C1q , Leucemia Mieloide Aguda , Humanos , Proteómica , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Médula Ósea/metabolismo , Pronóstico , Enfermedad Crónica , Recurrencia
4.
Nature ; 574(7780): 722-725, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31645759

RESUMEN

The enzyme protochlorophyllide oxidoreductase (POR) catalyses a light-dependent step in chlorophyll biosynthesis that is essential to photosynthesis and, ultimately, all life on Earth1-3. POR, which is one of three known light-dependent enzymes4,5, catalyses reduction of the photosensitizer and substrate protochlorophyllide to form the pigment chlorophyllide. Despite its biological importance, the structural basis for POR photocatalysis has remained unknown. Here we report crystal structures of cyanobacterial PORs from Thermosynechococcus elongatus and Synechocystis sp. in their free forms, and in complex with the nicotinamide coenzyme. Our structural models and simulations of the ternary protochlorophyllide-NADPH-POR complex identify multiple interactions in the POR active site that are important for protochlorophyllide binding, photosensitization and photochemical conversion to chlorophyllide. We demonstrate the importance of active-site architecture and protochlorophyllide structure in driving POR photochemistry in experiments using POR variants and protochlorophyllide analogues. These studies reveal how the POR active site facilitates light-driven reduction of protochlorophyllide by localized hydride transfer from NADPH and long-range proton transfer along structurally defined proton-transfer pathways.


Asunto(s)
Clorofila/biosíntesis , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/química , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Synechococcus/enzimología , Synechocystis/enzimología , Catálisis , Clorofila/química , Estructura Molecular , Fotoquímica , Protoclorofilida/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato
5.
Blood ; 139(22): 3314-3324, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35148377

RESUMEN

The von Willebrand factor (VWF) propeptide (domains D1D2) is essential for the assembly of VWF multimers and its tubular storage in Weibel-Palade bodies. However, detailed molecular mechanism underlying this propeptide dependence is unclear. Here, we prepared Weibel-Palade body-like tubules using the N-terminal fragment of VWF and solved the cryo-electron microscopy structures of the tubule at atomic resolution. Detailed structural and biochemical analysis indicate that the propeptide forms a homodimer at acidic pH through the D2:D2 binding interface and then recruits 2 D'D3 domains, forming an intertwined D1D2D'D3 homodimer in essence. Stacking of these homodimers by the intermolecular D1:D2 interfaces brings 2 D3 domains face-to-face and facilitates their disulfide linkages and multimerization of VWF. Sequential stacking of these homodimers leads to a right-hand helical tubule for VWF storage. The clinically identified VWF mutations in the propeptide disrupted different steps of the assembling process, leading to diminished VWF multimers in von Willebrand diseases (VWD). Overall, these results indicate that the propeptide serves as a pH-sensing template for VWF multimerization and tubular storage. This sheds light on delivering normal propeptide as a template to rectify the defects in multimerization of VWD mutants.


Asunto(s)
Enfermedades de von Willebrand , Factor de von Willebrand , Microscopía por Crioelectrón , Humanos , Dominios Proteicos , Cuerpos de Weibel-Palade/metabolismo , Enfermedades de von Willebrand/genética , Factor de von Willebrand/metabolismo
6.
J Chem Inf Model ; 63(15): 4780-4790, 2023 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-37486605

RESUMEN

The sirtuin enzyme family members, SIRT1 and SIRT2, play both tumor-promoting and tumor-suppressing roles, depending on the context and experimental conditions. Compounds that inhibit either SIRT1 or SIRT2 show promising antitumor effects in several types of cancer models, both in vitro and in vivo. The simultaneous inhibition of SIRT1 and SIRT2 is helpful in treating cancer by completely blocking p53 deacetylation, leading to cell death. However, only a few SIRT1/2 dual inhibitors have been developed. Here, we report the discovery of a novel series of SIRT1/2 dual inhibitors via a rational drug design that involved virtual screening and a substructure search. Eleven of the derived compounds exhibited high inhibitory activities, with IC50 < 5 µM and high specificity for both SIRT1 and SIRT2. Compounds hsa55 and PS9 strongly induced apoptosis and showed antiproliferative effects against human leukemia cell lines, which could be due to their ability to increase of p53 and α-tubulin acetylation, as we observed in MOLM-13 cells. Therefore, the new scaffolds of these compounds and their efficacy in leukemia cell lines provide important clues for the further development of novel anti-leukemia drugs.


Asunto(s)
Neoplasias , Sirtuina 2 , Humanos , Sirtuina 2/química , Sirtuina 1 , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis
7.
Proc Natl Acad Sci U S A ; 117(2): 872-876, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31888984

RESUMEN

Virtually all proton-pumping terminal respiratory oxygen reductases are members of the heme-copper oxidoreductase superfamily. Most of these enzymes use reduced cytochrome c as a source of electrons, but a group of enzymes have evolved to directly oxidize membrane-bound quinols, usually menaquinol or ubiquinol. All of the quinol oxidases have an additional transmembrane helix (TM0) in subunit I that is not present in the related cytochrome c oxidases. The current work reports the 3.6-Å-resolution X-ray structure of the cytochrome aa3 -600 menaquinol oxidase from Bacillus subtilis containing 1 equivalent of menaquinone. The structure shows that TM0 forms part of a cleft to accommodate the menaquinol-7 substrate. Crystals which have been soaked with the quinol-analog inhibitor HQNO (N-oxo-2-heptyl-4-hydroxyquinoline) or 3-iodo-HQNO reveal a single binding site where the inhibitor forms hydrogen bonds to amino acid residues shown previously by spectroscopic methods to interact with the semiquinone state of menaquinone, a catalytic intermediate.


Asunto(s)
Bacillus subtilis/metabolismo , Cobre/química , Complejo IV de Transporte de Electrones/química , Hemo/química , Hidroquinonas/química , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Grupo Citocromo b/química , Transporte de Electrón , Enlace de Hidrógeno , Modelos Moleculares , Naftoles/metabolismo , Oxidorreductasas , Conformación Proteica , Subunidades de Proteína/química , Bombas de Protones/química , Bombas de Protones/metabolismo , Terpenos/metabolismo , Vitamina K 2/análogos & derivados , Vitamina K 2/química
8.
Acta Biochim Biophys Sin (Shanghai) ; 54(8): 1133-1139, 2022 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-35866602

RESUMEN

The coronavirus papain-like protease (PLpro) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for viral polypeptide cleavage and the deISGylation of interferon-stimulated gene 15 (ISG15), which enable it to participate in virus replication and host innate immune pathways. Therefore, PLpro is considered an attractive antiviral drug target. Here, we show that parthenolide, a germacrane sesquiterpene lactone, has SARS-CoV-2 PLpro inhibitory activity. Parthenolide covalently binds to Cys-191 or Cys-194 of the PLpro protein, but not the Cys-111 at the PLpro catalytic site. Mutation of Cys-191 or Cys-194 reduces the activity of PLpro. Molecular docking studies show that parthenolide may also form hydrogen bonds with Lys-192, Thr-193, and Gln-231. Furthermore, parthenolide inhibits the deISGylation but not the deubiquitinating activity of PLpro in vitro. These results reveal that parthenolide inhibits PLpro activity by allosteric regulation.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Proteasas Similares a la Papaína de Coronavirus , Antivirales/farmacología , Humanos , Interferones , Lactonas , Simulación del Acoplamiento Molecular , Papaína/química , Papaína/metabolismo , Péptido Hidrolasas/metabolismo , SARS-CoV-2 , Sesquiterpenos , Sesquiterpenos de Germacrano , Ubiquitina/metabolismo
9.
Protein Expr Purif ; 179: 105788, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33221504

RESUMEN

MEF2D-fusions have recently been identified as one of the major oncogenic drivers in precursor B-cell acute lymphoblastic leukemia (B-ALL). More importantly, they are often associated with patients with poor prognosis in B-ALL. To have a better understanding of the pathogenic mechanism underpinning MEF2D-fusions-driven leukemogenesis, it's essential to uncover the related structure information. In this study, we expressed and purified the MEF2D N-terminal DNA binding domain. The recombinant protein was engineered by cloning the encoding gene into the expression vector pET-32 m. A series of chromatographic steps involving affinity, ion-exchange and gel-filtration chromatography were used to achieve a final purity of >95%. For the crystallization of the MEF2D-DNA complex, a double-stranded DNA encoding 5'-AACTATTTATAAGA-3' and 5'-TTCTTATAAATAGT-3' was used (Wu et al., 2010) [1]. The MEF2D-DNA crystal with the size of about 20 µm × 20 µm × 20 µm was obtained at a final concentration of 12 mg/ml at the reservoir condition containing 30% PEG1500. The X-ray examination showed that the MEF2D-DNA crystal diffracted to 4.5 Å resolution, and belonged to space group P1, with unit-cell parameters of a = 77.2 Å, b = 77.2 Å, c = 231.4 Å.


Asunto(s)
ADN , Proteínas Recombinantes , Sitios de Unión/genética , Cromatografía Liquida , ADN/química , ADN/metabolismo , Humanos , Factores de Transcripción MEF2/química , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/aislamiento & purificación , Factores de Transcripción MEF2/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Difracción de Rayos X
10.
J Biol Chem ; 294(7): 2353-2364, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30563843

RESUMEN

The renin-angiotensin cascade is a hormone system that regulates blood pressure and fluid balance. Renin-mediated cleavage of the angiotensin I peptide from the N terminus of angiotensinogen (AGT) is the rate-limiting step of this cascade; however, the detailed molecular mechanism underlying this step is unclear. Here, we solved the crystal structures of glycosylated human AGT (2.30 Å resolution), its encounter complex with renin (2.55 Å), AGT cleaved in its reactive center loop (RCL; 2.97 Å), and spent AGT from which the N-terminal angiotensin peptide was removed (2.63 Å). These structures revealed that AGT undergoes profound conformational changes and binds renin through a tail-into-mouth allosteric mechanism that inserts the N terminus into a pocket equivalent to a hormone-binding site on other serpins. These changes fully extended the N-terminal tail, with the scissile bond for angiotensin release docked in renin's active site. Insertion of the N terminus into this pocket accompanied a complete unwinding of helix H of AGT, which, in turn, formed key interactions with renin in the complementary binding interface. Mutagenesis and kinetic analyses confirmed that renin-mediated production of angiotensin I is controlled by interactions of amino acid residues and glycan components outside renin's active-site cleft. Our findings indicate that AGT adapts unique serpin features for hormone delivery and binds renin through concerted movements in the N-terminal tail and in its main body to modulate angiotensin release. These insights provide a structural basis for the development of agents that attenuate angiotensin release by targeting AGT's hormone binding pocket.


Asunto(s)
Angiotensinógeno/química , Renina/química , Regulación Alostérica , Angiotensina I , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Cristalografía por Rayos X , Humanos , Dominios Proteicos , Renina/genética , Renina/metabolismo
11.
Cell Mol Life Sci ; 76(19): 3899-3914, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30993352

RESUMEN

The P3H1/CRTAP/PPIB complex is essential for prolyl 3-hydroxylation and folding of procollagens in the endoplasmic reticulum (ER). Deficiency in any component of this ternary complex is associated with the misfolding of collagen and the onset of osteogenesis imperfecta. However, little structure information is available about how this ternary complex is assembled and retained in the ER. Here, we assessed the role of the KDEL sequence of P3H1 and probed the spatial interactions of PPIB in the complex. We show that the KDEL sequence is essential for retaining the P3H1 complex in the ER. Its removal resulted in co-secretion of P3H1 and CRTAP out of the cell, which was mediated by the binding of P3H1 N-terminal domain with CRTAP. The secreted P3H1/CRTAP can readily bind PPIB with their C-termini close to PPIB in the ternary complex. Cysteine modification, crosslinking, and mass spectrometry experiments identified PPIB surface residues involved in the complex formation, and showed that the surface of PPIB is extensively covered by the binding of P3H1 and CRTAP. Most importantly, we demonstrated that one disease-associated pathological PPIB mutation on the binding interface did not affect the PPIB prolyl-isomerase activity, but disrupted the formation of P3H1/CRTAP/PPIB ternary complex. This suggests that defects in the integrity of the P3H1 ternary complex are associated with pathological collagen misfolding. Taken together, these results provide novel structural information on how PPIB interacts with other components of the P3H1 complex and indicate that the integrity of P3H1 complex is required for proper collagen formation.


Asunto(s)
Ciclofilinas/química , Proteínas de la Matriz Extracelular/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteoglicanos/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Ciclofilinas/genética , Ciclofilinas/metabolismo , Proteínas de la Matriz Extracelular/genética , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Modelos Moleculares , Chaperonas Moleculares , Mutación , Prolil Hidroxilasas , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteoglicanos/química , Proteoglicanos/genética , Eliminación de Secuencia
12.
Inflammopharmacology ; 27(1): 157-166, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29946770

RESUMEN

OBJECTIVE: The present study aimed to evaluate the anti-inflammatory and analgesic activities of the ginsenoside metabolite compound K (CK) and its mechanisms. METHODS: Mice model of xylene-induced ear swelling and rat model of carrageenan-induced paw swelling were used to evaluate the effect of CK on acute inflammation. The analgesic effect of CK was evaluated on heat-, acetic acid-, and carrageenan-induced hyperalgesia. The levels of prostaglandin E2 (PGE2), cyclooxygenase-1 (COX-1), and COX-2 in carrageenan-induced rat paw swelling and gastric mucosa were detected by enzyme-linked immunosorbent assay (ELISA). COX-1 and COX-2 expressions in carrageenan-induced rat paw swelling and gastric mucosa were detected by western blotting. In vitro effect of CK (10-9, 10-8, 10-7, 10-6, 10-5 M) on COX-1 and COX-2 activities was evaluated by measuring the production of 6-keto-PGF1α and PGE2 in rat peritoneal macrophages. RESULTS: CK at doses of 7, 14, 28, 56, 112, and 224 mg/kg alleviated xylene-induced ear oedema, whereas CK at 40, 80, and 160 mg/kg alleviated carrageenan-induced paw oedema. CK at 224 mg/kg showed an analgesic effect against acetic acid-induced pain. CK at 40, 80, and 160 mg/kg significantly increased rat inflammatory pain threshold, but had no effect on heat-induced pain threshold. CK at 10, 20, 40, 80, and 160 mg/kg reduced PGE2 level in the paw tissue, but showed no effect on that in the gastric mucosa. CK at 20, 40, 80, and 160 mg/kg decreased COX-2 expression in the paw tissue and gastric mucosa, but exhibited no effect on COX-1 expression or on COX-1 and COX-2 activities. CONCLUSION: CK exerted anti-inflammatory and analgesic effects, possibly by reducing the catalytic synthesis of PGE2 via downregulation of COX-2 expression.


Asunto(s)
Analgésicos/farmacología , Antiinflamatorios/farmacología , Ciclooxigenasa 2/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Ginsenósidos/farmacología , Inflamación/tratamiento farmacológico , Animales , Carragenina/farmacología , Ciclooxigenasa 1/metabolismo , Dinoprostona/metabolismo , Edema/inducido químicamente , Edema/tratamiento farmacológico , Edema/metabolismo , Femenino , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Dolor/metabolismo , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
13.
Inflammopharmacology ; 27(2): 271-279, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29675711

RESUMEN

OBJECTIVE: To investigate the effects of ß-AR signaling on fibroblast-like synoviocytes (FLS) from adjuvant-induced arthritis (AA) rats and the partial mechanisms focused on ß-AR desensitization mediated by GRK2 and ß-arrestin2. METHODS: Animals were divided into a control group and an AA model group, and FLSs were cultured. Arthritis index, histopathology of joints, epinephrine (Epi) and norepinephrine (NE) were detected in vivo. The effect of the ß-AR agonist isoprenaline (ISO) and the ß2-AR agonist salbutamol on FLS cell viability were detected by CCK8. Cytokines TNF-α, IL-1ß, OPG and RANKL were examined by ELISA. The expression of ß2-AR was detected by immunofluorescence and flow cytometry. The cytomembrane expression and desensitization of ß2-AR, GRK2, and ß-arrestin2 were measured by flow cytometry and western blot. RESULTS: The concentration of NE increased to a peak on day 21, which was consistent with the arthritis index. The levels of Epi and NE in synovial tissues were decreased. ISO inhibited FLS cell viability and TNF-α, IL-1ß, and RANKL secretion, and promoted OPG secretion. ß2-AR mediated the effects of ISO on FLS cell viability. ß2-AR signaling was weaker in AA rats compared to the controls. Elevated GRK2 and ß-arrestin2 in cytomembranes promoted ß2-AR desensitization and may decrease the anti-inflammatory effect of ß2-AR signaling. CONCLUSION: The activation of ß2-AR signaling exerts its anti-inflammatory activities on FLS. ß2-AR signaling decreased in the AA model, which might be related to the increased membrane expression of GRK2 and ß-arrestin2, and promoted the excessive desensitization of ß2-AR. Decreased ß2-AR signaling may be relevant to the exacerbation of arthritis inflammation.


Asunto(s)
Adyuvantes Farmacéuticos/análisis , Artritis Experimental/metabolismo , Fibroblastos/metabolismo , Inflamación/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/fisiología , Sinoviocitos/metabolismo , Adyuvantes Farmacéuticos/efectos adversos , Animales , Artritis Experimental/inducido químicamente , Supervivencia Celular/fisiología , Células Cultivadas , Citocinas/metabolismo , Epinefrina/metabolismo , Interleucina-1beta/metabolismo , Masculino , Norepinefrina/metabolismo , Ligando RANK/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Biol Chem ; 291(48): 24900-24911, 2016 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-27681598

RESUMEN

Lamprey angiotensinogen (l-ANT) is a hormone carrier in the regulation of blood pressure, but it is also a heparin-dependent thrombin inhibitor in lamprey blood coagulation system. The detailed mechanisms on how angiotensin is carried by l-ANT and how heparin binds l-ANT and mediates thrombin inhibition are unclear. Here we have solved the crystal structure of cleaved l-ANT at 2.7 Šresolution and characterized its properties in heparin binding and protease inhibition. The structure reveals that l-ANT has a conserved serpin fold with a labile N-terminal angiotensin peptide and undergoes a typical stressed-to-relaxed conformational change when the reactive center loop is cleaved. Heparin binds l-ANT tightly with a dissociation constant of ∼10 nm involving ∼8 monosaccharides and ∼6 ionic interactions. The heparin binding site is located in an extensive positively charged surface area around helix D involving residues Lys-148, Lys-151, Arg-155, and Arg-380. Although l-ANT by itself is a poor thrombin inhibitor with a second order rate constant of 500 m-1 s-1, its interaction with thrombin is accelerated 90-fold by high molecular weight heparin following a bell-shaped dose-dependent curve. Short heparin chains of 6-20 monosaccharide units are insufficient to promote thrombin inhibition. Furthermore, an l-ANT mutant with the P1 Ile mutated to Arg inhibits thrombin nearly 1500-fold faster than the wild type, which is further accelerated by high molecular weight heparin. Taken together, these results suggest that heparin binds l-ANT at a conserved heparin binding site around helix D and promotes the interaction between l-ANT and thrombin through a template mechanism conserved in vertebrates.


Asunto(s)
Angiotensinas/química , Proteínas de Peces/química , Heparina/química , Lampreas , Trombina/química , Angiotensinas/genética , Angiotensinas/metabolismo , Animales , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Heparina/genética , Heparina/metabolismo , Mutación , Dominios Proteicos , Estructura Secundaria de Proteína , Trombina/genética , Trombina/metabolismo
15.
J Biol Chem ; 291(30): 15674-86, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27246852

RESUMEN

The Z mutation (E342K) of α1-antitrypsin (α1-AT), carried by 4% of Northern Europeans, predisposes to early onset of emphysema due to decreased functional α1-AT in the lung and to liver cirrhosis due to accumulation of polymers in hepatocytes. However, it remains unclear why the Z mutation causes intracellular polymerization of nascent Z α1-AT and why 15% of the expressed Z α1-AT is secreted into circulation as functional, but polymerogenic, monomers. Here, we solve the crystal structure of the Z-monomer and have engineered replacements to assess the conformational role of residue Glu-342 in α1-AT. The results reveal that Z α1-AT has a labile strand 5 of the central ß-sheet A (s5A) with a consequent equilibrium between a native inhibitory conformation, as in its crystal structure here, and an aberrant conformation with s5A only partially incorporated into the central ß-sheet. This aberrant conformation, induced by the loss of interactions from the Glu-342 side chain, explains why Z α1-AT is prone to polymerization and readily binds to a 6-mer peptide, and it supports that annealing of s5A into the central ß-sheet is a crucial step in the serpins' metastable conformational formation. The demonstration that the aberrant conformation can be rectified through stabilization of the labile s5A by binding of a small molecule opens a potential therapeutic approach for Z α1-AT deficiency.


Asunto(s)
Mutación Missense , Deficiencia de alfa 1-Antitripsina , alfa 1-Antitripsina/química , Sustitución de Aminoácidos , Cristalografía por Rayos X , Humanos , Estabilidad Proteica , Estructura Secundaria de Proteína , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
16.
J Biol Chem ; 290(15): 9906-18, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25713144

RESUMEN

The anticoagulant serpin, protein Z-dependent protease inhibitor (ZPI), circulates in blood as a tight complex with its cofactor, protein Z (PZ), enabling it to function as a rapid inhibitor of membrane-associated factor Xa. Here, we show that N,N'-dimethyl-N-(acetyl)-N'-(7-nitrobenz-3-oxa-1,3-diazol-4-yl)ethylenediamine (NBD)-fluorophore-labeled K239C ZPI is a sensitive, moderately perturbing reporter of the ZPI-PZ interaction and utilize the labeled ZPI to characterize in-depth the thermodynamics and kinetics of wild-type and variant ZPI-PZ interactions. NBD-labeled K239C ZPI bound PZ with ∼3 nM KD and ∼400% fluorescence enhancement at physiologic pH and ionic strength. The NBD-ZPI-PZ interaction was markedly sensitive to ionic strength and pH but minimally affected by temperature, consistent with the importance of charged interactions. NBD-ZPI-PZ affinity was reduced ∼5-fold by physiologic calcium levels to resemble NBD-ZPI affinity for γ-carboxyglutamic acid/EGF1-domainless PZ. Competitive binding studies with ZPI variants revealed that in addition to previously identified Asp-293 and Tyr-240 hot spot residues, Met-71, Asp-74, and Asp-238 made significant contributions to PZ binding, whereas Lys-239 antagonized binding. Rapid kinetic studies indicated a multistep binding mechanism with diffusion-limited association and slow complex dissociation. ZPI complexation with factor Xa or cleavage decreased ZPI-PZ affinity 2-7-fold by increasing the rate of PZ dissociation. A catalytic role for PZ was supported by the correlation between a decreased rate of PZ dissociation from the K239A ZPI-PZ complex and an impaired ability of PZ to catalyze the K239A ZPI-factor Xa reaction. Together, these results reveal the energetic basis of the ZPI-PZ interaction and suggest an important role for ZPI Lys-239 in PZ catalytic action.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Lisina/metabolismo , Serpinas/metabolismo , Termodinámica , Algoritmos , Azoles/química , Sitios de Unión/genética , Unión Competitiva , Proteínas Sanguíneas/química , Proteínas Sanguíneas/genética , Calcio/metabolismo , Factor Xa/química , Factor Xa/metabolismo , Colorantes Fluorescentes/química , Humanos , Concentración de Iones de Hidrógeno , Cinética , Lisina/química , Lisina/genética , Mutación , Nitrobencenos/química , Concentración Osmolar , Unión Proteica , Serpinas/química , Serpinas/genética , Espectrometría de Fluorescencia , Temperatura
17.
J Biol Chem ; 290(5): 2689-98, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25516596

RESUMEN

Surface expression of voltage-gated Ca(2+) (Cav) channels is important for their function in calcium homeostasis in the physiology of excitable cells, but whether or not and how the α1 pore-forming subunits of Cav channels are trafficked to plasma membrane in the absence of the known Cav auxiliary subunits, ß and α2δ, remains mysterious. Here we showed that 14-3-3 proteins promoted functional surface expression of the Cav2.2 α1B channel in transfected tsA-201 cells in the absence of any known Cav auxiliary subunit. Both the surface to total ratio of the expressed α1B protein and the current density of voltage step-evoked Ba(2+) current were markedly suppressed by the coexpression of a 14-3-3 antagonist construct, pSCM138, but not its inactive control, pSCM174, as determined by immunofluorescence assay and whole cell voltage clamp recording, respectively. By contrast, coexpression with 14-3-3τ significantly enhanced the surface expression and current density of the Cav2.2 α1B channel. Importantly, we found that between the two previously identified 14-3-3 binding regions at the α1B C terminus, only the proximal region (amino acids 1706-1940), closer to the end of the last transmembrane domain, was retained by the endoplasmic reticulum and facilitated by 14-3-3 to traffic to plasma membrane. Additionally, we showed that the 14-3-3/Cav ß subunit coregulated the surface expression of Cav2.2 channels in transfected tsA-201 cells and neurons. Altogether, our findings reveal a previously unidentified regulatory function of 14-3-3 proteins in promoting the surface expression of Cav2.2 α1B channels.


Asunto(s)
Proteínas 14-3-3/metabolismo , Canales de Calcio Tipo N/metabolismo , Proteínas 14-3-3/genética , Animales , Western Blotting , Canales de Calcio Tipo N/genética , Membrana Celular/metabolismo , Células Cultivadas , Electrofisiología , Retículo Endoplásmico/metabolismo , Inmunoprecipitación , Unión Proteica , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley
18.
Nature ; 468(7320): 108-11, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-20927107

RESUMEN

Blood pressure is critically controlled by angiotensins, which are vasopressor peptides specifically released by the enzyme renin from the tail of angiotensinogen-a non-inhibitory member of the serpin family of protease inhibitors. Although angiotensinogen has long been regarded as a passive substrate, the crystal structures solved here to 2.1 Å resolution show that the angiotensin cleavage site is inaccessibly buried in its amino-terminal tail. The conformational rearrangement that makes this site accessible for proteolysis is revealed in our 4.4 Å structure of the complex of human angiotensinogen with renin. The co-ordinated changes involved are seen to be critically linked by a conserved but labile disulphide bridge. Here we show that the reduced unbridged form of angiotensinogen is present in the circulation in a near 40:60 ratio with the oxidized sulphydryl-bridged form, which preferentially interacts with receptor-bound renin. We propose that this redox-responsive transition of angiotensinogen to a form that will more effectively release angiotensin at a cellular level contributes to the modulation of blood pressure. Specifically, we demonstrate the oxidative switch of angiotensinogen to its more active sulphydryl-bridged form in the maternal circulation in pre-eclampsia-the hypertensive crisis of pregnancy that threatens the health and survival of both mother and child.


Asunto(s)
Angiotensinógeno/química , Angiotensinógeno/metabolismo , Angiotensinas/metabolismo , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Angiotensinógeno/sangre , Angiotensinas/química , Presión Sanguínea , Cristalografía por Rayos X , Disulfuros/química , Disulfuros/metabolismo , Femenino , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Oxidación-Reducción , Estrés Oxidativo , Preeclampsia/sangre , Preeclampsia/metabolismo , Embarazo , Conformación Proteica , Renina/química , Renina/metabolismo
19.
J Am Soc Nephrol ; 26(2): 400-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25012180

RESUMEN

Anion exchanger-1 (AE1) mediates chloride-bicarbonate exchange across the plasma membranes of erythrocytes and, via a slightly shorter transcript, kidney epithelial cells. On an omnivorous human diet, kidney AE1 is mainly active basolaterally in α-intercalated cells of the collecting duct, where it is functionally coupled with apical proton pumps to maintain normal acid-base homeostasis. The C-terminal tail of AE1 has an important role in its polarized membrane residency. We have identified the ß1 subunit of Na(+),K(+)-ATPase (sodium pump) as a binding partner for AE1 in the human kidney. Kidney AE1 and ß1 colocalized in renal α-intercalated cells and coimmunoprecipitated (together with the catalytic α1 subunit of the sodium pump) from human kidney membrane fractions. ELISA and fluorescence titration assays confirmed that AE1 and ß1 interact directly, with a Kd value of 0.81 µM. GST-AE1 pull-down assays using human kidney membrane proteins showed that the last 11 residues of AE1 are important for ß1 binding. siRNA-induced knockdown of ß1 in cell culture resulted in a significant reduction in kidney AE1 levels at the cell membrane, whereas overexpression of kidney AE1 increased cell surface sodium pump levels. Notably, membrane staining of ß1 was reduced throughout collecting ducts of AE1-null mouse kidney, where increased fractional excretion of sodium has been reported. These data suggest a requirement of ß1 for proper kidney AE1 membrane residency, and that activities of AE1 and the sodium pump are coregulated in kidney.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/fisiología , Membrana Celular/metabolismo , Riñón/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/deficiencia , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Línea Celular , Membrana Celular/patología , Células Cultivadas , Homeostasis/fisiología , Humanos , Riñón/patología , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Unión Proteica , ARN Interferente Pequeño/farmacología , ATPasa Intercambiadora de Sodio-Potasio/efectos de los fármacos
20.
Proc Biol Sci ; 281(1779): 20132747, 2014 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-24478298

RESUMEN

The hormone thyroxine that regulates mammalian metabolism is carried and stored in the blood by thyroxine-binding globulin (TBG). We demonstrate here that the release of thyroxine from TBG occurs by a temperature-sensitive mechanism and show how this will provide a homoeostatic adjustment of the concentration of thyroxine to match metabolic needs, as with the hypothermia and torpor of small animals. In humans, a rise in temperature, as in infections, will trigger an accelerated release of thyroxine, resulting in a predictable 23% increase in the concentration of free thyroxine at 39°C. The in vivo relevance of this fever-response is affirmed in an environmental adaptation in aboriginal Australians. We show how two mutations incorporated in their TBG interact in a way that will halve the surge in thyroxine release, and hence the boost in metabolic rate that would otherwise occur as body temperatures exceed 37°C. The overall findings open insights into physiological changes that accompany variations in body temperature, as notably in fevers.


Asunto(s)
Temperatura Corporal , Tiroxina/metabolismo , Adaptación Fisiológica , Animales , Fiebre/sangre , Fiebre/metabolismo , Humanos , Hipotermia/sangre , Hipotermia/metabolismo , Mamíferos/sangre , Mamíferos/metabolismo , Mamíferos/fisiología , Modelos Moleculares , Nativos de Hawái y Otras Islas del Pacífico/genética , Unión Proteica , Conformación Proteica , Tiroxina/sangre , Tiroxina/química , Globulina de Unión a Tiroxina/genética , Globulina de Unión a Tiroxina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA