Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Protein Sci ; 33(4): e4956, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38511511

RESUMEN

Copper ion dys-homeostasis is linked to neurodegenerative diseases involving amyloid formation. Even if many amyloidogenic proteins can bind copper ions as monomers, little is known about copper interactions with the resulting amyloid fibers. Here, we investigate copper interactions with α-synuclein, the amyloid-forming protein in Parkinson's disease. Copper (Cu(II)) binds tightly to monomeric α-synuclein in vitro involving the N-terminal amine and the side chain of His50. Using purified protein and biophysical methods in vitro, we reveal that copper ions are readily incorporated into the formed amyloid fibers when present at the start of aggregation reactions, and the metal ions also bind if added to pre-formed amyloids. Efficient incorporation is observed for α-synuclein variants with perturbation of either one of the high-affinity monomer copper-binding residues (i.e., N-terminus or His50) whereas a variant with both N-terminal acetylation and His50 substituted with Ala does not incorporate any copper into the amyloids. Both the morphology of the resulting α-synuclein amyloids (amyloid fiber pitch, secondary structure, proteinase sensitivity) and the copper chemical properties (redox activity, chemical potential) are altered when copper is incorporated into amyloids. We speculate that copper chelation by α-synuclein amyloids contributes to the observed copper dys-homeostasis (e.g., reduced bioavailable levels) in Parkinson's disease patients. At the same time, amyloid-copper interactions may be protective to neuronal cells as they will shield aberrantly free copper ions from promotion of toxic reactive oxygen species.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Humanos , alfa-Sinucleína/química , Enfermedad de Parkinson/metabolismo , Cobre/química , Amiloide/metabolismo , Proteínas Amiloidogénicas/metabolismo , Iones
2.
J Inorg Biochem ; 247: 112335, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37487298

RESUMEN

The mediator of ERBB2-driven cell motility protein 1, Memo1, plays important roles in cancer signaling pathways. We recently reported Memo1 to coordinate reduced copper ions and protect them from reactive oxygen species (ROS) generation in vitro. We here assess if this Memo1 activity is at play in breast cancer cells. Copper additions to MDA-MB-231 cells promoted cell death, and this toxicity was exaggerated when Memo1 expression was reduced by silencing RNA. Using three different commercial ROS probes, we revealed that copper additions increased intracellular ROS levels, and these were further elevated when Memo1 expression was silenced. We propose that, in addition to other functions, Memo1 protects cancer cells from unwanted copper-mediated redox reactions. This may be a required safety mechanism in cancer cells as they have a high demand for copper.


Asunto(s)
Neoplasias de la Mama , Cobre , Humanos , Femenino , Especies Reactivas de Oxígeno/metabolismo , Cobre/farmacología , Cobre/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Oxidación-Reducción , Transducción de Señal , Línea Celular Tumoral
3.
Proc Natl Acad Sci U S A ; 119(37): e2206905119, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36067318

RESUMEN

The protein mediator of ERBB2-driven cell motility 1 (Memo1) is connected to many signaling pathways that play key roles in cancer. Memo1 was recently postulated to bind copper (Cu) ions and thereby promote the generation of reactive oxygen species (ROS) in cancer cells. Since the concentration of Cu as well as ROS are increased in cancer cells, both can be toxic if not well regulated. Here, we investigated the Cu-binding capacity of Memo1 using an array of biophysical methods at reducing as well as oxidizing conditions in vitro. We find that Memo1 coordinates two reduced Cu (Cu(I)) ions per protein, and, by doing so, the metal ions are shielded from ROS generation. In support of biological relevance, we show that the cytoplasmic Cu chaperone Atox1, which delivers Cu(I) in the secretory pathway, can interact with and exchange Cu(I) with Memo1 in vitro and that the two proteins exhibit spatial proximity in breast cancer cells. Thus, Memo1 appears to act as a Cu(I) chelator (perhaps shuttling the metal ion to Atox1 and the secretory path) that protects cells from Cu-mediated toxicity, such as uncontrolled formation of ROS. This Memo1 functionality may be a safety mechanism to cope with the increased demand of Cu ions in cancer cells.


Asunto(s)
Proteínas Transportadoras de Cobre , Cobre , Péptidos y Proteínas de Señalización Intracelular , Metalochaperonas , Chaperonas Moleculares , Línea Celular Tumoral , Cobre/metabolismo , Proteínas Transportadoras de Cobre/genética , Proteínas Transportadoras de Cobre/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Iones/metabolismo , Metalochaperonas/genética , Metalochaperonas/metabolismo , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Oxidación-Reducción , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo
4.
J Inorg Biochem ; 234: 111870, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35660722

RESUMEN

Binuclear Ru(II) polypyridyl complexes [Ru2(NN)4(BIPMB)]4+ (1-4), where N-N = 2,2'-bipyridine (bpy), 1,10-phenanthroline (phen), dipyrido [3,2-d:2',3'-f] quinoxaline (dpq), and dipyrido[3,2-a:2',3'-c]phenazine (dppz), have been synthesized using suitable precursors and bridging ligand (BIPMB), where BIPMB = 3,3'-bis-{(imidazol-1-yl)-[4,5-f]-1,10-phenanthroline) methyl}-1,1'-biphenyl. The binding mode and affinity of complexes 1-4 with Calf Thymus DNA (CT-DNA) were determined by absorption and steady-state fluorescence spectroscopy. The decrease in viscosity of CT-DNA on sequential addition of these complexes indicated DNA condensation and the result was corroborated by circular dichroism (CD). The nanosized globular aggregates of CT-DNA induced by complexes 1-4 were observed by dynamic light scattering (DLS) and transmission electron microscopy (TEM) measurements. The gel electrophoretic mobility studies revealed the small orderly particles of supercoiled plasmid pBR322 DNA induced by these complexes. Additionally, the morphology and size of compact plasmid DNA condensates were studied by DLS and atomic force microscopy (AFM). The complexes were moderately cytotoxic against MCF-7 cells.


Asunto(s)
Compuestos Organometálicos , Rutenio , Dicroismo Circular , ADN/química , División del ADN , Compuestos Organometálicos/química , Fenantrolinas/química , Rutenio/química
5.
Biophys J ; 121(7): 1194-1204, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35202609

RESUMEN

Abnormal cellular copper levels have been clearly implicated in genetic diseases, cancer, and neurodegeneration. Ctr1, a high-affinity copper transporter, is a homotrimeric integral membrane protein that provides the main route for cellular copper uptake. Together with a sophisticated copper transport system, Ctr1 regulates Cu(I) metabolism in eukaryotes. Despite its pivotal role in normal cell function, the molecular mechanism of copper uptake and transport via Ctr1 remains elusive. In this study, electron paramagnetic resonance (EPR), UV-visible spectroscopy, and all-atom simulations were employed to explore Cu(I) binding to full-length human Ctr1 (hCtr1), thereby elucidating how metal binding at multiple distinct sites affects the hCtr1 conformational dynamics. We demonstrate that each hCtr1 monomer binds up to five Cu(I) ions and that progressive Cu(I) binding triggers a marked structural rearrangement in the hCtr1 C-terminal region. The observed Cu(I)-induced conformational remodeling suggests that the C-terminal region may play a dual role, serving both as a channel gate and as a shuttle mediating the delivery of copper ions from the extracellular hCtr1 selectivity filter to intracellular metallochaperones. Our findings thus contribute to a more complete understanding of the mechanism of hCtr1-mediated Cu(I) uptake and provide a conceptual basis for developing mechanism-based therapeutics for treating pathological conditions linked to de-regulated copper metabolism.


Asunto(s)
Proteínas de Transporte de Catión , Proteínas Transportadoras de Cobre , Transportador de Cobre 1 , Cobre , Cobre/química , Cobre/metabolismo , Proteínas Transportadoras de Cobre/química , Proteínas Transportadoras de Cobre/metabolismo , Transportador de Cobre 1/química , Transportador de Cobre 1/metabolismo , Humanos , Iones/química , Iones/metabolismo
6.
ChemistryOpen ; 10(4): 486-492, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33908707

RESUMEN

The Cu(II)-diacetyl-bis (N4-methylthiosemicarbazone) complex (ATSM-Cu(II)) has been suggested as a promising positron emission tomography (PET) agent for hypoxia imaging. Several in-vivo studies have shown its potential to detect hypoxic tumors. However, its uptake mechanism and its specificity to various cancer cell lines have been less studied. Herein, we tested ATSM-Cu(II) toxicity, uptake, and reduction, using four different cell types: (1) mouse breast cancer cells (DA-3), (2) human embryonic kidney cells (HEK-293), (3) breast cancer cells (MCF-7), and (4) cervical cancer cells (Hela) under normoxic and hypoxic conditions. We showed that ATSM-Cu(II) is toxic to breast cancer cells under normoxic and hypoxic conditions; however, it is not toxic to normal HEK-293 non-cancer cells. We showed that the Cu(I) content in breast cancer cell after treatment with ATSM-Cu(II) under hypoxic conditions is higher than in normal cells, despite that the uptake of ATSM-Cu(II) is a bit higher in normal cells than in breast cancer cells. This study suggests that the redox potential of ATSM-Cu(II) is higher in breast cancer cells than in normal cells; thus, its toxicity to cancer cells is increased.


Asunto(s)
Hipoxia/metabolismo , Compuestos Organometálicos/metabolismo , Tiosemicarbazonas/metabolismo , Animales , Línea Celular Tumoral , Complejos de Coordinación , Radioisótopos de Cobre/química , Radioisótopos de Cobre/metabolismo , Transportador de Cobre 1/metabolismo , Células HEK293 , Humanos , Ratones , Compuestos Organometálicos/química , Compuestos Organometálicos/toxicidad , Oxidación-Reducción , Tiosemicarbazonas/química , Tiosemicarbazonas/toxicidad
7.
ACS Omega ; 4(7): 12278-12285, 2019 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-31460344

RESUMEN

Hypoxia is commonly encountered in the tumor microenvironment and drives proliferation, angiogenesis, and resistance to therapy. Imaging of hypoxia is important in many disease states in oncology, cardiology, and neurology. Finding clinically approved imaging biomarkers for hypoxia has proved challenging. Candidate biomarkers have shown low uptake into tumors and low signal to background ratios that adversely affect imaging quality. Copper complexes have been identified as potential biomarkers for hypoxia owing to their redox ability. Active uptake of copper complexes into cells could ensure selectivity and high sensitivity. We explored the reactivity and selectivity of the ATSM-Cu(II) biomarker to proteins that are involved in the copper cycle using electron paramagnetic resonance (EPR) spectroscopy and UV-vis measurements. We show that the affinity of the ATSM-Cu(II) complex to proteins in the copper cycle is low and the cell probably does not actively uptake ATSM-Cu(II).

8.
PLoS One ; 14(8): e0219337, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31465444

RESUMEN

The dissemination of resistant pathogenic microbes has become one of the most challenging problems that modern medicine has faced. Developing novel drugs based on new molecular targets that previously were not targeted, is therefore the highest priority in antibiotics research. One approach that has been recently suggested is to inhibit copper transporters in prokaryotic systems. Copper is required for many biological pathways, but sometimes it can harm the cell. Pathogenic systems have a highly sophisticated copper-regulation network; therefore, a better understanding of how this network operates at the molecular level should assist in developing the next generation of antibiotics. The CusB protein is part of the CusCBA periplasmic Cu(I) efflux system in Gram-negative bacteria, and was recently reported to play a key role in the functioning of the whole CusCBA system, in which conformational changes as well as the assembly/disassembly process control the opening of the transporter. More knowledge of the underlying mechanism is needed to attain a full understanding of CusB functioning, which is associated with targeting specific and crucial residues in CusB. Here, we combine in-vitro structural measurements, which use EPR spectroscopy and UV-Vis measurements, with cell experiments to explore the role of the various methionine residues in CusB. We targeted two methionine residues (M227 and M241) that are essential for the proper functioning of CusB.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Metionina/metabolismo , Supervivencia Celular/efectos de los fármacos , Cobre/metabolismo , Cobre/farmacología , Escherichia coli/citología , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Proteínas de Escherichia coli/genética , Proteínas de Transporte de Membrana/genética , Modelos Moleculares , Mutación , Dominios Proteicos , Estrés Fisiológico/efectos de los fármacos
9.
Chem Commun (Camb) ; 54(84): 11945-11948, 2018 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-30288543

RESUMEN

The catalytic redox activity of Cu(ii) bound to the motif NH2-Xxx-Zzz-His (ATCUN) with ascorbate and H2O2/O2 is very low and can be stopped via Cu(i)-chelation. This impacts its application as an artificial Cu-enzyme to degrade biomolecules via production of reactive oxygen species in a Cu(i)-chelator rich environment like the cytosol.

10.
Inorg Chem ; 57(13): 7524-7535, 2018 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-29893118

RESUMEN

Two heteronuclear ruthenium(II)-platinum(II) complexes [Ru(bpy)2(BPIMBp)PtCl2]2+ (3) and [Ru(phen)2(BPIMBp)PtCl2]2+ (4), where bpy = 2,2'-bipyridine, phen = 1,10-phenanthroline, and BPIMBp = 1,4'-bis[(2-pyridin-2-yl)-1H-imidazol-1-ylmethyl]-1,1'-biphenyl, have been designed and synthesized from their mononuclear precursors [Ru(bpy)2(BPIMBp)]2+ (1) and [Ru(phen)2(BPIMBp)]2+ (2) as multitarget molecules for Alzheimer's disease (AD). The inclusion of the cis-PtCl2 moiety facilitates the covalent interaction of Ru(II) polypyridyl complexes with amyloid ß (Aß) peptide. These multifunctional complexes act as inhibitors of acetylcholinesterase (AChE), Aß aggregation, and Cu-induced oxidative stress and protect neuronal cells against Aß-toxicity. The study highlights the design of metal based anti-Alzheimer's disease (AD) systems.

11.
Inorg Chem ; 56(7): 3729-3732, 2017 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-28318262

RESUMEN

A variety of fluorescent probes are proposed to monitor the intracellular copper content. So far, none of the probes have been evaluated for their potential to inhibit copper-associated intracellular oxidative stress. Herein, we studied the ability of a fluorescent copper probe, OBEP-CS1, to inhibit intracellular oxidative stress associated with an amyloid ß (Aß) peptide-copper complex. The data showed that OBEP-CS1 completely inhibits the copper-catalyzed oxidation as well as decarboxylation/deamination of Aß1-16. Moreover, the cell imaging experiments confirmed that OBEP-CS1 can inhibit Aß-CuII-catalyzed reactive oxygen species production in SH-SY5Y cells. We also demonstrated that Aß1-16 peptide can bind intracellular copper and thereby exert oxidative stress.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Quelantes/farmacología , Cobre/metabolismo , Colorantes Fluorescentes/farmacología , Fragmentos de Péptidos/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Péptidos beta-Amiloides/química , Catálisis , Línea Celular Tumoral , Quelantes/química , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Cobre/química , Colorantes Fluorescentes/química , Humanos , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/química , Especies Reactivas de Oxígeno/química , Especies Reactivas de Oxígeno/metabolismo
12.
Free Radic Res ; 50(4): 405-13, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26690929

RESUMEN

The binding of metal ions to Aß peptide plays an important role in the etiology of AD. Copper coordinates chiefly to His residues and produces reactive oxygen species (ROS) upon redox cycling. ROS builds enormous burden on the normal functioning of neuronal cells and results into deleterious effects. Recently, two structurally distinct copper binding sites with contrasting redox properties were characterized. Here, we demonstrate for the first time the effect of binding of two equivalents of Cu(2+) on redox properties and cytotoxicity of Aß peptide. Our electrochemical data and ascorbate consumption assay suggest that in the presence of two equivalents of copper; Aß peptide has higher propensity of H2O2 generation. The oxidation of Aß1-16 peptide due to both gamma radiolysis and metal catalyzed oxidation in the presence of two equivalents of copper is inhibited confirming the binding of both equivalents of copper to peptide. The electrochemical and cytotoxicity study shows that negative shift in the reduction potential is reflected as slightly higher cytotoxicity in SH-SY5Y cell lines for Aß1-16-Cu(2+) (1:2) complex.


Asunto(s)
Péptidos beta-Amiloides/química , Complejos de Coordinación/farmacología , Cobre/química , Citotoxinas/farmacología , Histidina/química , Peróxido de Hidrógeno/química , Fragmentos de Péptidos/química , Secuencia de Aminoácidos , Ácido Ascórbico/farmacología , Línea Celular Tumoral , Complejos de Coordinación/química , Citotoxinas/química , Técnicas Electroquímicas , Rayos gamma , Humanos , Hidrólisis , Neuronas/química , Neuronas/citología , Neuronas/efectos de los fármacos , Oxidación-Reducción , Unión Proteica
13.
Inorg Chem ; 53(19): 10003-5, 2014 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-25237806

RESUMEN

Cisplatin was studied for its effect on the copper-catalyzed oxidation of amyloid ß (Aß) peptide. The interaction of cisplatin with Aß1-16 in the presence of Cu(II) was investigated using cyclic voltammetry and mass spectrometry. The positive shift in the E1/2 value of Aß1-16-Cu(II) suggests that the interaction of cisplatin alters the copper-binding properties of Aß1-16. The mass spectrometry data show complete inhibition of copper-catalyzed decarboxylation/deamination of the Asp1 residue of Aß1-16, while there is a significant decrease in copper-catalyzed oxidation of Aß1-16 in the presence of cisplatin. Overall, our results provide a novel mode by which cisplatin inhibits copper-catalyzed oxidation of Aß. These findings may lead to the design of better platinum complexes to treat oxidative stress in Alzheimer's disease and other related neurological disorders.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Cisplatino/farmacología , Cobre/química , Catálisis , Cisplatino/química , Humanos , Estructura Molecular , Oxidación-Reducción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA