Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Methods Mol Biol ; 1876: 25-35, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30317472

RESUMO

Enzymes with homology to nitrogenase are essential for the reduction of chemically stable double bonds within the biosynthetic pathways of bacteriochlorophyll and coenzyme F430. These tetrapyrrole-based compounds are crucial for bacterial photosynthesis and the biogenesis of methane in methanogenic archaea. Formation of bacteriochlorophyll requires the unique ATP-dependent enzyme chlorophyllide oxidoreductase (COR) for the two-electron reduction of chlorophyllide to bacteriochlorophyllide. COR catalysis is based on the homodimeric protein subunit BchX2, which facilitates the transfer of electrons to the corresponding heterotetrameric catalytic subunit (BchY/BchZ)2. By analogy to the nitrogenase system, the dynamic switch protein BchX2 contains a [4Fe-4S] cluster that triggers the ATP-driven transfer of electrons onto a second [4Fe-4S] cluster located in (BchY/BchZ)2. The subsequent substrate reduction and protonation is unrelated to nitrogenase catalysis, with no further involvement of a molybdenum-containing cofactor. The biosynthesis of the nickel-containing coenzyme F430 includes the six-electron reduction of the tetrapyrrole macrocycle of Ni2+-sirohydrochlorin a,c-diamide to Ni2+-hexahydrosirohydrochlorin a,c-diamide catalyzed by CfbC/D. The homodimeric CfbC2 subunit carrying a [4Fe-4S] cluster shows close homology to BchX2. Accordingly, parallelism for the initial ATP-driven electron transfer steps of CfbC/D was proposed. Electrons are received by the dimeric catalytic subunit CfbD2, which contains a second [4Fe-4S] cluster and carries out the saturation of an overall of three double bonds in a highly orchestrated spatial and regioselective process. Following a short introduction to nitrogenase catalysis, this chapter will focus on the recent progress toward the understanding of the nitrogenase-like enzymes COR and CfbC/D, with special emphasis on the underlying enzymatic mechanism(s).


Assuntos
Oxirredutases do Álcool/metabolismo , Archaea/enzimologia , Bacterioclorofilas/biossíntese , Metaloporfirinas/metabolismo , Nitrogenase/química , Trifosfato de Adenosina/metabolismo , Oxirredutases do Álcool/química , Archaea/química , Proteínas Arqueais/química , Proteínas Arqueais/metabolismo , Catálise , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Níquel , Homologia de Sequência de Aminoácidos
8.
Chem Commun (Camb) ; 54(95): 13423-13426, 2018 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-30427327

RESUMO

Metal-containing enzyme cofactors achieve their unusual reactivity by stabilizing uncommon metal oxidation states with structurally complex ligands. In particular, the specific cofactor promoting both methanogenesis and anaerobic methane oxidation is a porphyrinoid chelated to a nickel(i) atom via a multi-step biosynthetic path, where nickel reduction is achieved through extensive molecular hydrogenation. Here, we demonstrate an alternative route to porphyrin reduction by charge transfer from a selected copper substrate to commercially available 5,10,15,20-tetraphenyl-porphyrin nickel(ii). X-ray absorption measurements at the Ni L3-edge unequivocally show that NiTPP species adsorbed on Cu(100) are stabilized in the highly reactive Ni(i) oxidation state by electron transfer to the molecular orbitals. Our approach highlights how some fundamental properties of synthetically inaccessible biological cofactors may be reproduced by hybridization of simple metalloporphyrins with metal surfaces, with implications towards novel approaches to heterogenous catalysis.


Assuntos
Coenzimas/metabolismo , Metaloporfirinas/metabolismo , Adsorção , Catálise , Coenzimas/química , Cobre/química , Metaloporfirinas/química , Estrutura Molecular , Tamanho da Partícula , Propriedades de Superfície
9.
Planta Med ; 84(15): 1141-1148, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29763945

RESUMO

Piperlongumine is an amide alkaloid found in Piperaceae species that shows a broad spectrum of biological properties, including antitumor and antiparasitic activities. Herein, the leishmanicidal effect of piperlongumine and its derivatives produced by a biomimetic model using metalloporphyrins was investigated. The results showed that IC50 values of piperlongumine in promastigote forms of Leishmania infantum and Leishmania amazonensis were 7.9 and 3.3 µM, respectively. The IC50 value of piperlongumine in the intracellular amastigote form of L. amazonensis was 0.4 µM, with a selectivity index of 25. The piperlongumine biomimetic derivatives, Ma and Mb, also showed leishmanicidal effects. We also carried out an in vitro metabolic degradation study showing that Ma is the most stable piperlongumine derivative in rat liver microsome incubations. The results presented here indicate that piperlongumine is a potential leishmanicidal candidate and support the biomimetic approach for development of new antileishmanial derivatives.


Assuntos
Anti-Helmínticos/farmacologia , Antiprotozoários/farmacologia , Dioxolanos/farmacologia , Leishmania infantum/efeitos dos fármacos , Piperaceae/química , Piperidonas/farmacologia , Animais , Anti-Helmínticos/química , Antiprotozoários/química , Biomimética , Dioxolanos/química , Feminino , Concentração Inibidora 50 , Fígado/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Metaloporfirinas/metabolismo , Camundongos Endogâmicos BALB C , Microssomos , Piperidonas/química , Ratos
10.
Biochemistry ; 57(3): 334-343, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29211462

RESUMO

The enzymes hydroxylamine oxidoreductase and cytochrome (cyt) P460 contain related unconventional "heme P460" cofactors. These cofactors are unusual in their inclusion of nonstandard cross-links between amino acid side chains and the heme macrocycle. Mutagenesis studies performed on the Nitrosomonas europaea cyt P460 that remove its lysine-heme cross-link show that the cross-link is key to defining the spectroscopic properties and kinetic competence of the enzyme. However, exactly how this cross-link confers these features remains unclear. Here we report the 1.45 Å crystal structure of cyt P460 from Nitrosomonas sp. AL212 and conclude that the cross-link does not lead to a change in hybridization of the heme carbon participating in the cross-link but rather enforces structural distortions to the macrocycle away from planarity. Time-dependent density functional theory coupled to experimental structural and spectroscopic analysis suggest that this geometric distortion is sufficient to define the spectroscopic properties of the heme P460 cofactor and provide clues toward establishing a relationship between heme P460 electronic structure and function.


Assuntos
Amônia/metabolismo , Proteínas de Bactérias/metabolismo , Coenzimas/metabolismo , Citocromos/metabolismo , Ferro/química , Compostos Macrocíclicos/metabolismo , Metaloporfirinas/metabolismo , Nitrosomonas europaea/enzimologia , Proteínas de Bactérias/química , Coenzimas/química , Cristalografia por Raios X , Citocromos/química , Espectroscopia de Ressonância de Spin Eletrônica , Compostos Macrocíclicos/química , Metaloporfirinas/química , Estrutura Molecular , Oxirredução , Espectrofotometria Ultravioleta
11.
Angew Chem Int Ed Engl ; 56(44): 13752-13756, 2017 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-28856780

RESUMO

Photodynamic therapy (PDT) has emerged as an important minimally invasive tumor treatment technology. The search for an effective photosensitizer to realize selective cancer treatment has become one of the major foci in recent developments of PDT technology. Controllable singlet-oxygen release based on specific cancer-associated events, as another major layer of selectivity mode, has attracted great attention in recent years. Here, for the first time, we demonstrated that a novel mixed-metal metal-organic framework nanoparticle (MOF NP) photosensitizer can be activated by a hydrogen sulfide (H2 S) signaling molecule in a specific tumor microenvironment for PDT against cancer with controllable singlet-oxygen release in living cells. The effective removal of tumors in vivo further confirmed the satisfactory treatment effect of the MOF NP photosensitizer.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Metaloporfirinas/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Oxigênio Singlete/metabolismo , Animais , Linhagem Celular Tumoral , Células HCT116 , Células Hep G2 , Humanos , Metaloporfirinas/administração & dosagem , Metaloporfirinas/metabolismo , Camundongos Nus , Nanopartículas/administração & dosagem , Nanopartículas/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Fotoquimioterapia , Fármacos Fotossensibilizantes/administração & dosagem , Fármacos Fotossensibilizantes/metabolismo , Microambiente Tumoral/efeitos dos fármacos
12.
J Biol Chem ; 292(32): 13205-13229, 2017 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-28655775

RESUMO

Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Moreover, intestinal iron is incorporated as ferrous iron, which is transported via the iron importer, divalent metal transporter 1 (DMT1). Recently, we demonstrated that the iron chaperone poly(rC)-binding protein 2 (PCBP2) can directly receive ferrous iron from DMT1 or transfer iron to the iron exporter, ferroportin 1. To promote intracellular iron flux, an iron chaperone may be essential for receiving iron generated by heme catabolism, but this hypothesis is untested so far. Herein, we demonstrate that HO1 binds to PCBP2, but not to other PCBP family members, namely PCBP1, PCBP3, or PCBP4. Interestingly, HO1 formed a complex with either CPR or PCBP2, and it was demonstrated that PCBP2 competes with CPR for HO1 binding. Using PCBP2-deletion mutants, we demonstrated that the PCBP2 K homology 3 domain is important for the HO1/PCBP2 interaction. In heme-loaded cells, heme prompted HO1-CPR complex formation and decreased the HO1/PCBP2 interaction. Furthermore, in vitro reconstitution experiments with purified recombinant proteins indicated that HO1 could bind to PCBP2 in the presence of heme, whereas loading of PCBP2 with ferrous iron caused PCBP2 to lose its affinity for HO1. These results indicate that ferrous iron released from heme can be bound by PCBP2 and suggest a model for an integrated heme catabolism and iron transport metabolon.


Assuntos
Heme Oxigenase-1/metabolismo , Heme/metabolismo , Ferro/metabolismo , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Proteínas de Ligação a RNA/metabolismo , Substituição de Aminoácidos , Sítios de Ligação , Ligação Competitiva , Transporte Biológico , Linhagem Celular , Deleção de Genes , Heme Oxigenase (Desciclizante)/química , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/química , Heme Oxigenase-1/genética , Humanos , Metaloporfirinas/metabolismo , Mutação , NADPH-Ferri-Hemoproteína Redutase/antagonistas & inibidores , NADPH-Ferri-Hemoproteína Redutase/química , NADPH-Ferri-Hemoproteína Redutase/genética , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Transporte Proteico , Interferência de RNA , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Homologia Estrutural de Proteína , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
Molecules ; 22(7)2017 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-28661455

RESUMO

Two new water-soluble metal carboxyl porphyrins, manganese (III) meso-tetrakis (carboxyl) porphyrin and iron (III) meso-tetrakis (carboxyl) porphyrin, were synthesized and characterized. Their interactions with ct-DNA were investigated by UV-Vis titration, fluorescence spectra, viscosity measurement and CD spectra. The results showed they can strongly bind to ct-DNA via outside binding mode. Electrophoresis experiments revealed that both complexes can cleave pBR322 DNA efficiently in the presence of hydrogen peroxide, albeit 2-Mn exhibited a little higher efficiency. The inhibitor tests suggest the oxidative DNA cleavage by these two complexes may involve hydroxyl radical active intermediates. Notably, 2-Mn exhibited considerable photocytotoxicity against Hep G2 cell via triggering a significant generation of ROS and causing disruption of MMP after irradiation.


Assuntos
Ferro/química , Manganês/química , Metaloporfirinas/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , DNA/química , DNA/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/farmacologia , Humanos , Metaloporfirinas/metabolismo , Metaloporfirinas/farmacologia , Oxirredução/efeitos dos fármacos , Estresse Oxidativo , Ligação Proteica , Proteólise , Espécies Reativas de Oxigênio , Solubilidade , Análise Espectral , Água/química
14.
Chem Pharm Bull (Tokyo) ; 65(4): 336-340, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28381673

RESUMO

The specific intermolecular interaction between an anionic tetraarylporphyrin and per-O-methylated ß-cyclodextrin (TMe-ß-CD) paved the way to produce a functional supramolecule that works as a strong carbon monoxide (CO)-depleting agent in living organisms. The supramolecular complex, hemoCD, that is composed of meso-tetrakis(4-sulfonatophenyl)porphinatoiron(II) and a TMe-ß-CD dimer linked by a pyridine linker, captured internal CO from carboxyhemoglobin during its circulation in the blood of animals. HemoCD thus produced the pseudo-knockdown (loss-of-functional) state of endogenous CO in the animals. This unique property led us to investigate the biological function of endogenous CO as a gaseous signal mediator in living systems. In this paper, we introduce our recent study on the hemoCD complex as a biological CO-depleting agent.


Assuntos
Monóxido de Carbono/metabolismo , Ciclodextrinas/metabolismo , Compostos Ferrosos/metabolismo , Metaloporfirinas/metabolismo , Animais , Monóxido de Carbono/química , Ciclodextrinas/química , Compostos Ferrosos/química , Humanos , Substâncias Macromoleculares/síntese química , Substâncias Macromoleculares/química , Substâncias Macromoleculares/metabolismo , Metaloporfirinas/química , Estrutura Molecular
16.
Nucleosides Nucleotides Nucleic Acids ; 36(2): 122-138, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27831822

RESUMO

The interaction of nickel (II) phthalocyanine tetrasulfonic acid tetrasodium salt with bovine serum albumin (BSA) has been investigated by combination of fluorescence, UV-vis absorption, Fourier transform infrared (FT-IR), and circular dichorism (CD) spectroscopies as well as through molecular docking. Fluorescence quenching and absorption spectra were investigated as a mean for estimating the binding parameters. Analysis of fluorescence quenching data at different temperatures was performed in order to specify the thermodynamics parameters for interactions of phthalocyanine complex with BSA. According to experimental data it was suggested that phthalocyanine had a significant binding affinity to BSA and the process was entropy driven. Based on the results of molecular docking it was indicated that the main active binding site for this phthalocyanine complex is site I in subdomain IIA of BSA. The results provide useful information for understanding the binding mechanism of anticancer drug-albumin and gives insight into the biological activity and metabolism of the drug in blood.


Assuntos
Metaloporfirinas/química , Soroalbumina Bovina/química , Sítios de Ligação , Dicroísmo Circular , Fluorescência , Indóis/química , Isoindóis , Metaloporfirinas/metabolismo , Simulação de Acoplamento Molecular , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/metabolismo , Soroalbumina Bovina/metabolismo , Espectrofotometria Ultravioleta , Espectroscopia de Infravermelho com Transformada de Fourier , Termodinâmica
17.
Rapid Commun Mass Spectrom ; 29(6): 521-32, 2015 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-26160418

RESUMO

RATIONALE: Metalloporphyrins can be useful in the production of drug metabolites, as they enable easier production of oxidative metabolites usually produced by the cytochrome P450 enzymes. Our aim was to test metalloporphyrin-based biomimetic oxidation (BMO) methods for production and S-glutathione trapping of reactive drug metabolites in addition to phase I metabolites. METHODS: Clozapine, ticlopidine and citalopram were selected as model compounds. These were incubated with the BMO assay and the incubations were analyzed with high-resolution liquid chromatography/mass spectrometry (LC/MS) and tandem mass spectrometry (LC/MS/MS). Additionally, incubations with human liver S9 fraction were performed to compare the results with the BMO assay. RESULTS: Six glutathione conjugates were identified for clozapine from the S9 incubation, while the BMO assay produced four of these. Four out of the five phase I metabolites produced by S9 were detected using the BMO assay. For ticlopidine, four glutathione conjugates were detected from the S9 incubation, but none of these were observed using the BMO assay. Eight of the nine phase I metabolites produced by S9 incubation were detected in the BMO assay. As expected, no glutathione conjugates were detected for citalopram, and the same three phase I metabolites were detected in both S9 and BMO incubations. CONLUSIONS: Differences in formation of GSH-trapped reactive metabolites by BMO assay between clozapine and ticlopidine are probably due to different reactive intermediates and reaction mechanisms. The reactive intermediate of clozapine, the nitrenium ion was generated, but the reactive intermediates of ticlopidine, S-oxide and epoxide, were not detected from the incubations. However, the results show that for selected cases the use of biomimetic assays can be used to produce high amounts of S-glutathione conjugates identical to those from liver subfraction incubations, on a scale that is relevant for purification and subsequent identification by NMR spectroscopy; which is often difficult using incubations with liver subfractions.


Assuntos
Citalopram/metabolismo , Clozapina/metabolismo , Glutationa/metabolismo , Metaloporfirinas/química , Ticlopidina/metabolismo , Citalopram/química , Clozapina/química , Glutationa/química , Humanos , Metaloporfirinas/metabolismo , Microssomos Hepáticos/química , Microssomos Hepáticos/metabolismo , Oxirredução , Espectrometria de Massas em Tandem , Ticlopidina/química
18.
J Am Chem Soc ; 137(24): 7560-3, 2015 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-26067900

RESUMO

Iron porphyrin carbenes (IPCs) are important intermediates in various chemical reactions catalyzed by iron porphyrins and engineered heme proteins, as well as in the metabolism of various xenobiotics by cytochrome P450. However, there are no prior theoretical reports to help understand their formation mechanisms and identify key information governing the binding mode, formation feasibility, and stability/reactivity. A systematic quantum chemical study was performed to investigate the effects of carbene substituent, porphyrin substituent, and axial ligand on IPC formation pathways. Results not only are consistent with available experimental data but also provide a number of unprecedented insights into electronic, steric, and H-bonding effects of various structural factors on IPC formation mechanisms. These results shall facilitate research on IPC and related systems for sustainable chemical catalysis and biocatalysis.


Assuntos
Compostos de Ferro/química , Metaloporfirinas/química , Metano/análogos & derivados , Biocatálise , Catálise , Sistema Enzimático do Citocromo P-450/metabolismo , Hemeproteínas/metabolismo , Compostos de Ferro/metabolismo , Ligantes , Metaloporfirinas/metabolismo , Metano/química , Metano/metabolismo , Modelos Moleculares , Termodinâmica
19.
Vascul Pharmacol ; 72: 163-71, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26015376

RESUMO

AIMS: Angiogenesis is defined as the sprouting of capillaries from pre-existing vasculature. It is a complex process that includes endothelial proliferation, migration, and tube formation. Previous data have demonstrated a high expression level of manganese-superoxide dismutase (MnSOD) in endothelial cells and suggested an important role of MnSOD in several cardiovascular diseases. In addition, manganese (III) tetrakis (4-benzoic acid) porphyrin (MnTBAP) has been shown to mimic some of the effects of MnSOD in various tissues. However, its effect on the vasculature remains unknown. METHODS AND RESULTS: HUVECs were treated with MnTBAP. Migration, tube formation, and capillary sprouting assays were performed to evaluate the pro-angiogenic effect in vitro. Matrigel plug assay was performed to assess capillary ingrowth in vivo. Compared to control, treatment with MnTBAP revealed increased cell migration, tube formation and capillary sprouting along with more capillary ingrowth in the Matrigel plug assay. This effect was mediated through a mitofusin (Mfn)-1-dependent pathway. Expression of Tie-2, Ang-2 and VEGF mRNA was increased in muscle tissues after ligation in MnTBAP treated mice. However, revascularization in the hindlimb ischemia model was not statistically significant at day 10 in MnTBAP treated mice. CONCLUSION: In summary, our data demonstrate a strong pro-angiogenic, but less pro-arteriogenic effect of MnTBAP in HUVECs mediated by Mfn-1.


Assuntos
Células Endoteliais/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Metaloporfirinas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Neovascularização Patológica/metabolismo , Animais , Movimento Celular/fisiologia , Células Cultivadas , Colágeno/metabolismo , Combinação de Medicamentos , Células Endoteliais da Veia Umbilical Humana , Humanos , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteoglicanas/metabolismo , RNA Mensageiro/metabolismo , Receptor TIE-2/metabolismo , Fator A de Crescimento do Endotélio Vascular , Proteínas de Transporte Vesicular/metabolismo
20.
J Biol Chem ; 290(16): 10176-90, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25733668

RESUMO

Iron is an essential metal cofactor that is required for many biological processes. Eukaryotic cells have consequently developed different strategies for its acquisition. Until now, Schizosaccharomyces pombe was known to use reductive iron uptake and siderophore-bound iron transport to scavenge iron from the environment. Here, we report the identification of a gene designated shu1(+) that encodes a protein that enables S. pombe to take up extracellular heme for cell growth. When iron levels are low, the transcription of shu1(+) is induced, although its expression is repressed when iron levels rise. The iron-dependent down-regulation of shu1(+) requires the GATA-type transcriptional repressor Fep1, which strongly associates with a proximal promoter region of shu1(+) in vivo in response to iron repletion. HA4-tagged Shu1 localizes to the plasma membrane in cells expressing a functional shu1(+)-HA4 allele. When heme biosynthesis is selectively blocked in mutated S. pombe cells, their ability to acquire exogenous hemin or the fluorescent heme analog zinc mesoporphyrin IX is dependent on the expression of Shu1. Further analysis by absorbance spectroscopy and hemin-agarose pulldown assays showed that Shu1 interacts with hemin, with a KD of ∼2.2 µm. Taken together, results reported here revealed that S. pombe possesses an unexpected pathway for heme assimilation, which may also serve as a source of iron for cell growth.


Assuntos
Regulação Fúngica da Expressão Gênica , Heme/metabolismo , Ferro/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/genética , Membrana Celular/metabolismo , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/metabolismo , Hemina/metabolismo , Transporte de Íons , Proteínas de Membrana Transportadoras/metabolismo , Metaloporfirinas/metabolismo , Oxirredução , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA