Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 381
Filtrar
1.
J Phys Chem B ; 128(16): 3807-3823, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38605466

RESUMO

The origin of highly efficient asymmetric aminohydroxylation of styrene catalyzed by engineered cytochrome c is investigated by the developed Atom-Bond Electronegativity Equalization Method polarizable force field (ABEEM PFF), which is a combined outcome of electronic and steric effects. Model molecules were used to establish the charge parameters of the ABEEM PFF, for which the bond-stretching and angle-bending parameters were obtained by using a combination of modified Seminario and scan methods. The interactions between carbon-radical Fe-porphyrin (FePP) and waters are simulated by molecular dynamics, which shows a clear preference for the pre-R over the pre-S. This preference is attributed to the hydrogen-bond between the mutated 100S and 101P residues as well as van der Waals interactions, enforcing a specific conformation of the carbon-radical FePP complex within the binding pocket. Meanwhile, the hydrogen-bond between water and the nitrogen atom in the active intermediate dictates the stereochemical outcome. Quantum mechanics/molecular mechanics (QM/MM (ABEEM PFF)) and free-energy perturbation calculations elucidate that the 3RTS is characterized by sandwich-like structure among adjacent amino acid residues, which exhibits greater stability than crowed arrangement in 3STS and enables the R enantiomer to form more favorably. Thus, this study provides mechanistic insight into the catalytic reaction of hemoproteins.


Assuntos
Citocromos c , Simulação de Dinâmica Molecular , Teoria Quântica , Estereoisomerismo , Citocromos c/química , Citocromos c/metabolismo , Hidrólise , Carbono/química , Engenharia de Proteínas , Ligação de Hidrogênio , Biocatálise , Metaloporfirinas/química , Metaloporfirinas/metabolismo
2.
Int J Mol Sci ; 24(7)2023 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-37047131

RESUMO

Myocardial ischemia-reperfusion injury (I/R) causes damage to cardiomyocytes through oxidative stress and apoptosis. We investigated the cardioprotective effects of MnTnBuOE-2-PyP5+ (BMX-001), a superoxide dismutase mimic, in an in vitro model of I/R injury in H9c2 cardiomyocytes. We found that BMX-001 protected against hypoxia/reoxygenation (H/R)-induced oxidative stress, as evident by a significant reduction in intracellular and mitochondrial superoxide levels. BMX-001 pre-treatment also reduced H/R-induced cardiomyocyte apoptosis, as marked by a reduction in TUNEL-positive cells. We further demonstrated that BMX-001 pre-treatment significantly improved mitochondrial function, particularly O2 consumption, in mouse adult cardiomyocytes subjected to H/R. BMX-001 treatment also attenuated cardiolipin peroxidation, 4-hydroxynonenal (4-HNE) level, and 4-HNE adducted proteins following H/R injury. Finally, the pre-treatment with BMX-001 improved cell viability and lactate dehydrogenase (LDH) activity in H9c2 cells following H/R injury. Our findings suggest that BMX-001 has therapeutic potential as a cardioprotective agent against oxidative stress-induced H/R damage in H9c2 cardiomyocytes.


Assuntos
Metaloporfirinas , Mimetismo Molecular , Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos , Estresse Oxidativo , Superóxido Dismutase , Superóxido Dismutase/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Metaloporfirinas/metabolismo , Metaloporfirinas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Lactato Desidrogenases/metabolismo , Linhagem Celular , Animais , Ratos , Cardiolipinas/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Metabolismo Energético/efeitos dos fármacos , Apoptose/efeitos dos fármacos
3.
Planta Med ; 89(7): 700-708, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36889328

RESUMO

Licarin A, a dihydrobenzofuranic neolignan presents in several medicinal plants and seeds of nutmeg, exhibits strong activity against protozoans responsible for Chagas disease and leishmaniasis. From biomimetic reactions by metalloporphyrin and Jacobsen catalysts, seven products were determined: four isomeric products yielded by epoxidation from licarin A, besides a new product yielded by a vicinal diol, a benzylic aldehyde, and an unsaturated aldehyde in the structure of the licarin A. The incubation with rat and human liver microsomes partially reproduced the biomimetic reactions by the production of the same epoxidized product of m/z 343 [M + H]+. In vivo acute toxicity assays of licarin A suggested liver toxicity based on biomarker enzymatic changes. However, microscopic analysis of tissues sections did not show any tissue damage as indicative of toxicity after 14 days of exposure. New metabolic pathways of the licarin A were identified after in vitro biomimetic oxidation reaction and in vitro metabolism by rat or human liver microsomes.


Assuntos
Lignanas , Metaloporfirinas , Ratos , Humanos , Animais , Biomimética , Oxirredução , Lignanas/toxicidade , Metaloporfirinas/metabolismo , Microssomos Hepáticos/metabolismo
4.
ACS Appl Mater Interfaces ; 13(49): 58508-58521, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34871496

RESUMO

An extracellular electron transfer (EET) process between an electroactive biofilm and an electrode is a crucial step for the performance of microbial fuel cells (MFCs), which is highly related to the enrichment of exoelectrogens and the electrocatalytic activity of the electrode. Herein, an efficient N- and Fe-abundant carbon cloth (CC) electrode with the comodification of iron porphyrin (FePor) and polyquaternium-7 (PQ) was synthesized using a facile solvent evaporation and immersion method and developed as an anode (named FePor-PQ) in MFCs. The surface structural characterizations confirmed the successful introduction of N and Fe atoms, whereas FePor-PQ achieved the N content of 9.59 at %, which may offer various active sites for EET. The introduction of PQ contributed to improving the surface hydrophilicity, providing the composite electrode good biocompatibility for bacterial attachment and colonization as well as substrate diffusion. Based on the advantages, the MFC with the FePor-PQ anode produced a maximum power density of 2165.7 mW m-2, strikingly higher than those of CC (1124.0 mW m-2), PQ (1668.8 mW m-2), and FePor (1978.9 mW m-2). Furthermore, with the EET mediated by the binding of flavins and c-type cytochromes on the outer membrane was enhanced prominently, the typical exoelectrogen Geobacter was enriched up to 55.84% in the FePor-PQ anode biofilm. This work reveals a synergistic effect from heteroatom coating and surface properties tailoring to boost both the EET efficiency and exoelectrogen enrichment for enhancing MFC performance, which also provides valuable insights for designing electrodes in other bio-electrochemical systems.


Assuntos
Bactérias/química , Fontes de Energia Bioelétrica , Acrilamidas/síntese química , Acrilamidas/química , Acrilamidas/metabolismo , Cloreto de Amônio/síntese química , Cloreto de Amônio/química , Cloreto de Amônio/metabolismo , Bactérias/citologia , Bactérias/metabolismo , Aderência Bacteriana , Materiais Biocompatíveis , Carbono/química , Eletrodos , Transporte de Elétrons , Elétrons , Teste de Materiais , Metaloporfirinas/síntese química , Metaloporfirinas/química , Metaloporfirinas/metabolismo , Tamanho da Partícula , Propriedades de Superfície
5.
J Inorg Biochem ; 223: 111551, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34340058

RESUMO

G-quadruplex nucleic acids (G4s) are RNA and DNA secondary structures involved in the regulation of multiple key biological processes. They can be found in telomeres, oncogene promoters, RNAs, but also in viral genomes. Due to their unique structural features, very distinct from the canonical duplexes or single-strands, G4s represent promising pharmacological targets for small molecules, namely G4-ligands. Gold(III) penta-cationic porphyrins, as specific G4 ligands, are able to inhibit HIV-1 infectivity and their antiviral activity correlates with their affinity for G4s. Up to now, one of the best antiviral compounds is meso-5,10,15,20-tetrakis[4-(N-methyl-pyridinium-2-yl)phenyl]porphyrinato gold(III) (1). Starting from this compound, we report a structure/affinity relationship study of gold(III) cationic porphyrins to find out the best porphyrin candidate for functionalization, in order to study the antiviral mechanism of action of these gold(III) porphyrins.


Assuntos
Fármacos Anti-HIV/metabolismo , DNA/metabolismo , Quadruplex G , Metaloporfirinas/metabolismo , Fármacos Anti-HIV/síntese química , DNA/genética , Ouro/química , HIV-1/química , Metaloporfirinas/síntese química , Simulação de Acoplamento Molecular , Estrutura Molecular , Relação Estrutura-Atividade
6.
J Inorg Biochem ; 219: 111431, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33798828

RESUMO

Oxidative stress that results from an imbalance between the concentrations of reactive species (RS) and antioxidant defenses is associated with many pathologies. Superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase are among the key enzymes that maintain the low nanomolar physiological concentrations of superoxide and hydrogen peroxide. The increase in the levels of these species and their progeny could have deleterious effects. In this context, chemists have developed SOD and CAT mimics to supplement them when cells are overwhelmed with oxidative stress. However, the beneficial activity of such molecules in cells depends not only on their intrinsic catalytic activities but also on their stability in biological context, their cell penetration and their cellular localization. We have employed cellular assays to characterize several compounds that possess SOD and CAT activities and have been frequently used in cellular and animal models. We used cellular assays that address SOD and CAT activities of the compounds. Finally, we determined the effect of compounds on the suppression of the inflammation in HT29-MD2 cells challenged by lipopolysaccharide. When the assay requires penetration inside cells, the SOD mimics Mn(III) meso-tetrakis(N-(2'-n-butoxyethyl)pyridinium-2-yl)porphyrin (MnTnBuOE-2-PyP5+) and Mn(II) dichloro[(4aR,13aR,17aR,21aR)-1,2,3,4,4a,5,6,12,13,13a,14,15,16,17,17a,18,19,20,21,21a-eicosahydro-11,7-nitrilo-7Hdibenzo[b,h] [1,4, 7,10] tetraazacycloheptadecine-κN5,κN13,κN18,κN21,κN22] (Imisopasem manganese, M40403, CG4419) were found efficacious at 10 µM, while Mn(II) chloro N-(phenolato)-N,N'-bis[2-(N-methyl-imidazolyl)methyl]-ethane-1,2-diamine (Mn1) requires an incubation at 100 µM. This study thus demonstrates that MnTnBuOE-2-PyP5+, M40403 and Mn1 were efficacious in suppressing inflammatory response in HT29-MD2 cells and such action appears to be related to their ability to enter the cells and modulate reactive oxygen species (ROS) levels.


Assuntos
Catalase/metabolismo , Manganês/metabolismo , Compostos Organometálicos/metabolismo , Superóxido Dismutase/metabolismo , Animais , Antioxidantes/metabolismo , Linhagem Celular , Glutationa Peroxidase/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Metaloporfirinas/metabolismo , Mimetismo Molecular , Oxirredução , Estresse Oxidativo , Porfirinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo
7.
Redox Rep ; 26(1): 85-93, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33902399

RESUMO

OBJECTIVE: The aim of this study was to investigate how modifications at the periphery of the porphyrin ring affect the anticancer activity of Mn porphyrins (MnPs)-based SOD mimics. METHODS: Six compounds: MnTE-2-PyP with a short ethyl chain on the pyridyl ring; MnTnHexOE-2-PyP and MnTnOct-2-PyP with linear 8-atom alkyl chains, but the former with an oxygen atom within the alkyl chain; MnTE-2-PyPhP and MnTPhE-2-PyP with pyridyl and phenyl substituents, were investigated. Cytotoxicity was studied using pII and MDA-MB-231 cancer cell lines. Viability was assessed by the MTT (3-[4,5-dimethylthiazol-2-yl)]-2,5-diphenyltetrazolium bromide) assay and cell proliferation was determined by the sulforhodamine B assay. RESULTS: Cellular uptake was increased with the increase of the lipophilicity of the compounds, whereas reduction potential (E½) of the Mn(III)/Mn(II) redox couple shifted away from the optimal value for efficient redox cycling with ascorbate, necessary for ROS production. Amphiphilic MnPs, however, exerted anticancer activity by a mechanism not involving ROS. CONCLUSION: Two different processes account for MnPs cytotoxicity. MnPs with appropriate E½ act via a ROS-dependent mechanism. Amphiphilic MnPs with suitable structure damage sensitive cellular constituents, leading to the suppression of proliferation and loss of viability. Design of compounds interacting directly with sensitive cellular targets is highly promising in the development of anticancer drugs with high selectivity and specificity.


Assuntos
Metaloporfirinas , Porfirinas , Antioxidantes , Ácido Ascórbico/farmacologia , Metaloporfirinas/metabolismo , Metaloporfirinas/farmacologia , Oxirredução , Porfirinas/farmacologia , Superóxido Dismutase/metabolismo
8.
Bioorg Med Chem Lett ; 40: 127931, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33705911

RESUMO

Green photosynthetic bacteria with an efficient light-harvesting system contain special chlorophyll molecules, called bacteriochlorophylls c, d, e, in their main antennae. In the biosynthetic pathway, a BciC enzyme is proposed to catalyze the hydrolysis of the C132-methoxycarbonyl group of chlorophyllide a, but the resulting C132-carboxy group has not been detected yet because it is spontaneously removed due to the instability of the ß-keto-carboxylic acid. In this study, the in vitro BciC enzymatic reactions of zinc methyl (131R/S)-hydroxy-mesochlorophyllides a were examined and a carboxylic acid possessing the C132S-OH was first observed as the hydrolyzed product of the C132-COOCH3.


Assuntos
Hidrolases de Éster Carboxílico/metabolismo , Clorofilídeos/metabolismo , Metaloporfirinas/metabolismo , Proteínas de Bactérias/metabolismo , Chlorobi/enzimologia , Clorofilídeos/química , Hidrólise , Metaloporfirinas/química , Estrutura Molecular , Zinco/química
9.
Metallomics ; 13(3)2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33576808

RESUMO

Understanding the toxicological properties of MnIII-porphyrins (MnTPPS, MnTMPyP, or MnTBAP) can provide important biochemical rationales in developing them as the therapeutic drugs against protein tyrosine nitration-induced inflammation diseases. Here, we present a comprehensive understanding of the pH-dependent redox behaviors of these MnIII-porphyrins and their structural effects on catalyzing bovine serum albumin (BSA) nitration in the presence of H2O2 and NO2-. It was found that both MnTPPS and MnTBAP stand out in catalyzing BSA nitration at physiologically close condition (pH 8), yet they are less effective at pH 6 and 10. MnTMPyP was shown to have no ability to catalyze BSA nitration under all tested pHs (pH 6, 8, and 10). The kinetics and active intermediate determination through electrochemistry method revealed that both the pH-dependent redox behavior of the central metal cation and the antioxidant capability of porphin derivative contribute to the catalytic activities of three MnIII-porphyrins in BSA nitration in the presence of H2O2/NO2-. These comprehensive studies on the oxidative reactivity of MnIII-porphyrins toward BSA nitration may provide new clues for searching the manganese-based therapeutic drugs against the inflammation-related diseases.


Assuntos
Peróxido de Hidrogênio/química , Metaloporfirinas/química , Nitratos/química , Nitritos/química , Estresse Oxidativo , Soroalbumina Bovina/química , Tirosina/química , Animais , Bovinos , Peróxido de Hidrogênio/metabolismo , Concentração de Íons de Hidrogênio , Metaloporfirinas/metabolismo , Nitritos/metabolismo
10.
J Am Chem Soc ; 143(1): 252-259, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33373215

RESUMO

De novo protein design offers the opportunity to test our understanding of how metalloproteins perform difficult transformations. Attaining high-resolution structural information is critical to understanding how such designs function. There have been many successes in the design of porphyrin-binding proteins; however, crystallographic characterization has been elusive, limiting what can be learned from such studies as well as the extension to new functions. Moreover, formation of highly oxidizing high-valent intermediates poses design challenges that have not been previously implemented: (1) purposeful design of substrate/oxidant access to the binding site and (2) limiting deleterious oxidation of the protein scaffold. Here we report the first crystallographically characterized porphyrin-binding protein that was programmed to not only bind a synthetic Mn-porphyrin but also maintain binding site access to form high-valent oxidation states. We explicitly designed a binding site with accessibility to dioxygen units in the open coordination site of the Mn center. In solution, the protein is capable of accessing a high-valent Mn(V)-oxo species which can transfer an O atom to a thioether substrate. The crystallographic structure is within 0.6 Å of the design and indeed contained an aquo ligand with a second water molecule stabilized by hydrogen bonding to a Gln side chain in the active site, offering a structural explanation for the observed reactivity.


Assuntos
Hemeproteínas/química , Manganês/química , Metaloporfirinas/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Hemeproteínas/genética , Hemeproteínas/metabolismo , Oxirredução , Ligação Proteica , Engenharia de Proteínas , Sulfetos/metabolismo
11.
Chem Commun (Camb) ; 57(4): 476-479, 2021 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-33326521

RESUMO

Replacing coenzyme F430, an Ni(i) F430-like cofactor derived from vitamin B12 (F430-B12) is revealed by DFT calculations to be able to catalyze methane formation in methyl-coenzyme M reductase with a barrier of 13.3 kcal mol-1, demonstrating the correctness of the route starting from vitamin B12. The structure-activity relationships of F430 and F430-B12 (especially the roles of the F ring) are discovered and several sources of inspiration promoting the application of F430-B12 are also obtained, coming closer to using F430 chemistry in man-made catalysis.


Assuntos
Metaloporfirinas/química , Oxirredutases/química , Vitamina B 12/análogos & derivados , Proteínas Arqueais/química , Proteínas Arqueais/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Teoria da Densidade Funcional , Metaloporfirinas/metabolismo , Metano/biossíntese , Methanobacteriaceae/enzimologia , Modelos Químicos , Estrutura Molecular , Níquel/química , Oxirredutases/metabolismo , Ligação Proteica , Relação Estrutura-Atividade , Termodinâmica , Vitamina B 12/metabolismo
12.
J Med Chem ; 63(13): 7268-7292, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32462865

RESUMO

An experimental approach is described for late-stage lead diversification of frontrunner drug candidates using nanomole-scale amounts of lead compounds for structure-activity relationship development. The process utilizes C-H bond activation methods to explore chemical space by transforming candidates into newly functionalized leads. A key to success is the utilization of microcryoprobe nuclear magnetic resonance (NMR) spectroscopy, which permits the use of low amounts of lead compounds (1-5 µmol). The approach delivers multiple analogues from a single lead at nanomole-scale amounts as DMSO-d6 stock solutions with a known structure and concentration for in vitro pharmacology and absorption, distribution, metabolism, and excretion testing. To demonstrate the feasibility of this approach, we have used the antihistamine agent loratadine (1). Twenty-six analogues of loratadine were isolated and fully characterized by NMR. Informative SAR analogues were identified, which display potent affinity for the human histamine H1 receptor and improved metabolic stability.


Assuntos
Loratadina/análogos & derivados , Loratadina/farmacocinética , Relação Estrutura-Atividade , Animais , Cromatografia Líquida de Alta Pressão , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Dimetil Sulfóxido/química , Cães , Descoberta de Drogas/métodos , Antagonistas não Sedativos dos Receptores H1 da Histamina/química , Antagonistas não Sedativos dos Receptores H1 da Histamina/farmacologia , Humanos , Ligação de Hidrogênio , Inativação Metabólica , Loratadina/química , Espectroscopia de Ressonância Magnética , Metaloporfirinas/química , Metaloporfirinas/metabolismo , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometria de Massas em Tandem , Distribuição Tecidual
13.
J Inorg Biochem ; 206: 111049, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32171934

RESUMO

The potential energy surfaces of the H2S binding to iron-porphyrin (FeP) with the imidazole (Im) ligand via intersystem crossings are investigated by using density functional theory. The minimum energy intersystem crossing point (MEISCP) between the quintet and triplet states (MEISCPTQ) for the Fe(II)P(Im)-H2S complex is located at a Fe-S distance of 3.39 Šwith only 1.1 kcal/mol above the quintet state minimum. The second spin-crossover point, where a change from the triplet to the singlet state occurs, comes at a much shorter Fe-S distance of 2.79 Å, and the MEISCPST is located at 3.7 kcal/mol above the triplet state minimum. The nature of the chemical bonding along the Fe-S reaction coordinate from the ground state singlet to the quintet state along the path to the separated species is analyzed. An inspection of the vibrational modes reveals that the largest contribution to the triplet-quintet transition around the quintet and triplet state minimum comes from the symmetric shrinking of the pyrrole units of the porphyrin ring, indicating that the related reaction coordinate plays a main role in the intersystem crossing. The fully optimized structures of the Fe(II)P(Im)-HS- complex corresponding to three different spin multiplicities (M = 1, 3, 5) are characterized by a bent Fe-H-S conformation. The binding of the hydrosulfide anion to Fe(II)P(Im) in the quintet state induces a 0.2 Šdisplacement of the Fe atom out of the nitrogen porphyrin (Npyr) plane. The fully optimized structure of the ground state of Fe(II)P(Im)-HS- agrees well with experimental data for the corresponding heme models.


Assuntos
Heme/metabolismo , Sulfeto de Hidrogênio/metabolismo , Imidazóis/metabolismo , Ferro/metabolismo , Metaloporfirinas/metabolismo , Teoria Quântica , Elétrons , Heme/química , Sulfeto de Hidrogênio/química , Imidazóis/química , Ferro/química , Metaloporfirinas/química , Modelos Moleculares , Conformação Molecular , Estrutura Molecular
14.
Chem Commun (Camb) ; 56(20): 3089-3092, 2020 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-32052805

RESUMO

Reaction of FeIII(O2˙-)(TPP) with 2,3-dimethylindole at -40 °C gives the ring-opened, dioxygenated N-(2-acetyl-phenyl)-acetamide product. The reaction was monitored in situ by low-temperature UV-vis and 1H NMR spectroscopies. This work demonstrates that a discrete iron(iii)(superoxo) porphyrin is competent to carry out indole oxidation, as proposed for the tryptophan and indoleamine 2,3-dioxygenases.


Assuntos
Compostos Férricos/química , Indolamina-Pirrol 2,3,-Dioxigenase/química , Indóis/química , Metaloporfirinas/química , Superóxidos/química , Triptofano Oxigenase/química , Compostos Férricos/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Indóis/metabolismo , Metaloporfirinas/metabolismo , Estrutura Molecular , Oxirredução , Superóxidos/metabolismo , Triptofano Oxigenase/metabolismo
15.
J Photochem Photobiol B ; 201: 111640, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31734545

RESUMO

Fluorescence image guided surgical resection (FIGR) of high grade gliomas (HGGs) takes advantage of the accumulation of the tracer protoporphyrin IX (PpIX) in glioma cells following administration of 5-aminolevulinic acid (5-ALA). Occasionally, PpIX fluorescence intensity may be insufficient, thus compromising the efficacy and precision of the surgical intervention. The cause for the signal variation is unclear and strategies to improve the intensity of PpIX fluorescence are considered necessary. We have previously shown that differential expression of the epidermal growth factor receptor in glioblastoma cells affects PpIX fluorescence. Herein, we investigated other factors impairing PpIX accumulation and pharmacological treatments able to enhance PpIX fluorescence in glioblastoma cells displaying lower signal. In the present study we demonstrate that presence of serum in cell culture medium and differences in cellular confluence can negatively influence PpIX accumulation in U87 cell lines. We hypothesized that PpIX fluorescence intensity results from the interplay between the metabolic clearance of PpIX mediated by ferrochelatase and heme oxygenase-1 and the cellular efflux of PpIX through the ATP-binding cassette subfamily G member 2 (ABCG2). Based on the availability of compounds targeting these proteins and inhibiting them, in this study we used modulators such as genistein, an isoflavone able to inhibit ABCG2; deferoxamine, which chelate iron ions impairing FECH activity and tin protoporphyrin IX (SnPP), the specific HO-1 inhibitor. Finally, we showed the efficacy of a precisely tuned pharmacological treatment in increasing PpIX accumulation and consequently fluorescence in glioblastoma cells. This strategy may translate in more sensitive tracing of tumor cells in-vivo and improved FIGR of HGGs and possibly low grade gliomas (LGGs).


Assuntos
Corantes Fluorescentes/química , Microscopia Confocal , Protoporfirinas/química , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Ácido Aminolevulínico/química , Ácido Aminolevulínico/metabolismo , Ácido Aminolevulínico/farmacologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/metabolismo , Corantes Fluorescentes/metabolismo , Genisteína/metabolismo , Genisteína/farmacologia , Glioblastoma/patologia , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/metabolismo , Humanos , Metaloporfirinas/química , Metaloporfirinas/metabolismo , Metaloporfirinas/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Protoporfirinas/metabolismo , Protoporfirinas/farmacologia
16.
J Am Chem Soc ; 141(22): 9087-9095, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31079455

RESUMO

The cavities of artificial receptors are defined by how their components fit together. The encapsulation of specific molecules can thus be engineered by considering geometric principles; however, intermolecular interactions and steric fit scale with receptor size, such that the ability to bind multiple guests from a specific class of compounds remains a current challenge. By employing metal-organic self-assembly, we have prepared a triangular prism from two different ligands that is capable of binding more than 20 different natural products, drugs, and steroid derivatives within its prolate cavity. Encapsulation inflates the host, enhancing its ability to bind other guests in peripheral pockets and thus enabling our system to bind combinations of different drug and natural product cargoes in different locations simultaneously. This new mode of entropically favorable self-assembly thus enables central encapsulation to amplify guest-binding events around the periphery of an artificial receptor.


Assuntos
Alcaloides Indólicos/metabolismo , Metaloporfirinas/metabolismo , Derivados da Morfina/metabolismo , Receptores Artificiais/metabolismo , Esteroides/metabolismo , Sítios de Ligação , Entropia , Metaloporfirinas/síntese química , Metaloporfirinas/química , Receptores Artificiais/síntese química , Receptores Artificiais/química , Zinco/química
17.
Int J Biol Macromol ; 134: 445-457, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31078597

RESUMO

The evidence that Human Serum Albumin (HSA) binds metal ions and organometallic compounds has generated interest in its physiological role as a metalloprotein and as a vehicle for synthetic biology applications (e.g., synthetic blood and solar energy conversion). HSA has been shown to bind metallo-porphyrins, however, the structural details of such interactions are available only for the HSA:heme complex. A typical challenge for studying the interaction of proteins with metalloporphyrins is the poor solubility of the ligands that affect the characterization the complexes. The manuscript shows that a combination of dialysis and centrifugation yields aqueous solutions that contain >90% HSA:porphyrin complexes and virtually eliminate aggregated ligands. The removal of aggregates increases the quality of the optical spectroscopy data which, in turn, yield more accurate binding constants (~0.1 and 2.1 × 106 M-1) and reveal FRET between Trp214 and the porphyrins. The Trp-porphyrin distance was estimated to be within the 28-34 Šrange and was used to guide the search of binding sites through a novel feedback approach with docking simulations. Results suggest while some protoporphyrins (metal-free, Fe(III)PPIX and Mg(II)PPIX) bind HSA at the heme site, others (Zn(II)PPIX, Mn(III)PPIX and Sn(IV)PPIX) are more likely to bind the Cys34.


Assuntos
Metaloporfirinas/química , Modelos Moleculares , Protoporfirinas/química , Albumina Sérica Humana/química , Albumina Sérica Humana/metabolismo , Sítios de Ligação , Complexos de Coordenação/química , Complexos de Coordenação/isolamento & purificação , Humanos , Ligantes , Metaloporfirinas/metabolismo , Conformação Molecular , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Protoporfirinas/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Análise Espectral , Relação Estrutura-Atividade
18.
Oxid Med Cell Longev ; 2019: 8639791, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30931081

RESUMO

BACKGROUND: As a key step in enhancing cancer cell invasion and metastasis, epithelial-mesenchymal transition (EMT) plays an important role in colorectal cancer progression. EMT is triggered by a variety of signaling pathways, among which the transforming growth factor ß (TGF-ß) signaling pathway has been implicated as a primary inducer. Accumulating evidence demonstrates that MnTE-2-PyP (chemical name: manganese(III) meso-tetrakis-(N-ethylpyridinium-2-yl), a superoxide dismutase (SOD) mimetic, inhibits TGF-ß signaling; however, its ability to inhibit TGF-ß-induced EMT in colorectal cancer has not yet been explored. METHODS: To verify our hypothesis that MnTE-2-PyP attenuates TGF-ß-induced EMT, human colorectal cancer cells were treated with TGF-ß in the presence or absence of MnTE-2-PyP. Cells were analyzed by several techniques including western blotting, real-time quantitative PCR, transwell assay, and wound healing assay. RESULTS: MnTE-2-PyP reverses cell phenotypes induced by TGF-ß in colon cancer cells. MnTE-2-PyP treatment significantly reduced the expression of mesenchymal markers but maintained epithelial marker expression. Mechanistically, MnTE-2-PyP suppressed the phosphorylated Smad2/3 protein levels induced by TGF-ß in SW480 cells, but MnTE-2-PyP failed to suppress TGF-ß-induced Slug and Snail expression in colorectal cells. Furthermore, MnTE-2-PyP effectively suppressed TGF-ß-mediated cell migration and invasion and the expression of matrix metalloproteinase 2 (MMP-2) and matrix metalloproteinase 9 (MMP-9) in colorectal cells. CONCLUSION: Taken together, we provide an in-depth mechanism by which MnTE-2-PyP inhibits colorectal cancer progression, supporting an important role for MnTE-2-PyP as an effective and innovative antitumor agent to enhance treatment outcomes in colorectal cancer.


Assuntos
Neoplasias Colorretais/genética , Metaloporfirinas/metabolismo , Proteína Smad2/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Transição Epitelial-Mesenquimal , Humanos , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo
19.
J Inorg Biochem ; 195: 61-70, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30925402

RESUMO

Coproheme decarboxylase (ChdC) catalyzes the oxidative decarboxylation of coproheme to heme b, i.e. the last step in the recently described coproporphyrin-dependent pathway. Coproheme decarboxylation from Listeria monocytogenes is a robust enzymatic reaction of low catalytic efficiency. Coproheme acts as both substrate and redox cofactor activated by H2O2. It fully depends on the catalytic Y147 close to the propionyl group at position 2. In the present study we have investigated the effect of disruption of the comprehensive and conserved hydrogen bonding network between the four propionates and heme cavity residues on (i) the conformational stability of the heme cavity, (ii) the electronic configuration of the ferric redox cofactor/substrate, (iii) the binding of carbon monoxide and, (iv) the decarboxylation reaction mediated by addition of H2O2. Nine single, double and triple mutants of ChdC from Listeria monocytogenes were produced in E. coli. The respective coproheme- and heme b-complexed proteins were studied by UV-Vis, resonance Raman, circular dichroism spectroscopy, and mass spectrometry. Interactions of propionates 2 and 4 with residues in the hydrophobic cavity are crucial for maintenance of the heme cavity architecture, for the mobile distal glutamine to interact with carbon monoxide, and to keep the heme cavity in a closed conformation during turnover. By contrast, the impact of substitution of residues interacting with solvent exposed propionates 6 and 7 was negligible. Except for Y147A and K151A all mutant ChdCs exhibited a wild-type-like catalytic activity. The findings are discussed with respect to the structure-function relationships of ChdCs.


Assuntos
Carboxiliases/metabolismo , Listeria monocytogenes/enzimologia , Metaloporfirinas/metabolismo , Monóxido de Carbono/metabolismo , Carboxiliases/química , Carboxiliases/genética , Catálise , Domínio Catalítico , Ligação de Hidrogênio , Peróxido de Hidrogênio/química , Metaloporfirinas/química , Mutagênese Sítio-Dirigida , Mutação , Ligação Proteica , Conformação Proteica
20.
Inorg Chem ; 58(5): 2954-2964, 2019 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-30735039

RESUMO

Emulating enzymatic reactivity using small molecules has been a long-time challenging pursuit of the scientific community. Peroxidases, ubiquitous heme enzymes that are involved in hormone synthesis and the immune system, have been a prime target of such efforts due to their tremendous potential in the chemical industry as well as in wastewater treatment. Here it is demonstrated that inclusion of a second sphere guanidine moiety in an iron porphyrin not only makes this small molecule a veritable peroxidase catalyst but also offers an auxiliary binding site for organic substrates, facilitating their rapid oxidation with a green oxidant like H2O2. This small molecule analogue exhibits a "ping-pong" mechanism and Michaelis-Menten type kinetics, which is generally typical of metallo-enzymes and follows a mechanism of the natural enzyme in its entirety, including the formation of compound I as the primary oxidant.


Assuntos
Ferro/metabolismo , Metaloporfirinas/metabolismo , Peroxidases/metabolismo , Sítios de Ligação , Biocatálise , Cristalografia por Raios X , Guanidina/química , Guanidina/metabolismo , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Ferro/química , Cinética , Metaloporfirinas/síntese química , Metaloporfirinas/química , Modelos Moleculares , Estrutura Molecular , Oxirredução , Peroxidases/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...