Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
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
2.
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
3.
J Biol Chem ; 293(43): 16623-16634, 2018 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-30217815

RESUMO

Human cytochrome P450 enzymes are membrane-bound heme-containing monooxygenases. As is the case for many heme-containing enzymes, substitution of the metal in the center of the heme can be useful for mechanistic and structural studies of P450 enzymes. For many heme proteins, the iron protoporphyrin prosthetic group can be extracted and replaced with protoporphyrin containing another metal, but human membrane P450 enzymes are not stable enough for this approach. The method reported herein was developed to endogenously produce human membrane P450 proteins with a nonnative metal in the heme. This approach involved coexpression of the P450 of interest, a heme uptake system, and a chaperone in Escherichia coli growing in iron-depleted minimal medium supplemented with the desired trans-metallated protoporphyrin. Using the steroidogenic P450 enzymes CYP17A1 and CYP21A2 and the drug-metabolizing CYP3A4, we demonstrate that this approach can be used with several human P450 enzymes and several different metals, resulting in fully folded proteins appropriate for mechanistic, functional, and structural studies including solution NMR.


Assuntos
Citocromo P-450 CYP3A/metabolismo , Metaloporfirinas/metabolismo , Metais/metabolismo , Protoporfirinas/metabolismo , Esteroide 17-alfa-Hidroxilase/metabolismo , Esteroide 21-Hidroxilase/metabolismo , Citocromo P-450 CYP3A/química , Humanos , Metaloporfirinas/química , Dobramento de Proteína , Protoporfirinas/química , Esteroide 17-alfa-Hidroxilase/química , Esteroide 21-Hidroxilase/química
4.
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
5.
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
6.
J Am Chem Soc ; 138(38): 12451-8, 2016 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-27575374

RESUMO

We introduce a strategy that expands the functionality of hemoproteins through orthogonal enzyme/heme pairs. By exploiting the ability of a natural heme transport protein, ChuA, to promiscuously import heme derivatives, we have evolved a cytochrome P450 (P450BM3) that selectively incorporates a nonproteinogenic cofactor, iron deuteroporphyrin IX (Fe-DPIX), even in the presence of endogenous heme. Crystal structures show that selectivity gains are due to mutations that introduce steric clash with the heme vinyl groups while providing a complementary binding surface for the smaller Fe-DPIX cofactor. Furthermore, the evolved orthogonal enzyme/cofactor pair is active in non-natural carbenoid-mediated olefin cyclopropanation. This methodology for the generation of orthogonal enzyme/cofactor pairs promises to expand cofactor diversity in artificial metalloenzymes.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sistema Enzimático do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/metabolismo , Ferro/química , Metaloporfirinas/química , NADPH-Ferri-Hemoproteína Redutase/química , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Domínio Catalítico , Coenzimas , Evolução Molecular Direcionada , Metaloporfirinas/metabolismo , Modelos Moleculares , Estrutura Molecular , Mutação , Oxirredução
7.
Inorg Chem ; 54(12): 5634-45, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26000802

RESUMO

Binding of nitric oxide (NO) to metalloporphyrins and heme groups is important in biochemistry while challenging to describe accurately by density functional theory (DFT) calculations. Here, the structural and thermochemical aspect of NO binding to Co(II) and Mn(II) porphyrins is investigated by DFT and DFT-D (dispersion-corrected) calculations, supported by reliable coupled-cluster methodology (CCSD(T)), and critically correlated with the experimental data. It is argued that whereas the bonding of NO to Co(II) porphyrin is a simple radical recombination, the bonding of NO to Mn(II) porphyrin is accompanied by a crossing of spin states. For this reason, the spin-state conversion energy contributes to the Mn-NO bond energy, and the paradigmatic correlation between bond length and bond energy is violated for the considered nitrosyl complexes: the Mn-NO bond is (structurally) shorter by ∼0.2 Å, albeit (energetically) weaker by a few kcal/mol, compared with the Co-NO bond. Moreover, none of the many tested DFT methods can reproduce the Co-NO and Mn-NO bond energies simultaneously, except for calculations with B3LYP*-D3, TPSSh-D3, and M06-D3 methods supplemented with the proposed spin-state energy correction (to compensate for an error on the calculated spin-state conversion energy). The results of this study are important to appreciate the role of spin-state changes in ligand binding properties of heme-related models. They also highlight the need for accurate calculations for correct interpretation of experimental data, including the qualitative structure-energy relationship.


Assuntos
Cobalto/química , Manganês/química , Metaloporfirinas/metabolismo , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Metaloporfirinas/química , Modelos Moleculares
8.
PLoS One ; 9(12): e115362, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25506911

RESUMO

High temperature requirement protein A1 (HtrA1), a secreted serine protease of the HtrA family, is associated with a multitude of human diseases. However, the exact functions of HtrA1 in these diseases remain poorly understood. We seek to unravel the mechanisms of HtrA1 by elucidating its interactions with chemical or biological modulators. To this end, we screened a small molecule library of 500 bioactive compounds to identify those that alter the formation of extracellular HtrA1 complexes in the cell culture medium. An initial characterization of two novel hits from this screen showed that protoporphyrin IX (PPP-IX), a precursor in the heme biosynthetic pathway, and its metalloporphyrin (MPP) derivatives fostered the oligomerization of HtrA1 by binding to the protease domain. As a result of the interaction with MPPs, the proteolytic activity of HtrA1 against Fibulin-5, a specific HtrA1 substrate in age-related macular degeneration (AMD), was increased. This physical interaction could be abolished by the missense mutations of HtrA1 found in patients with cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL). Furthermore, knockdown of HtrA1 attenuated apoptosis induced by PPP-IX. These results suggest that PPP-IX, or its derivatives, and HtrA1 may function as co-factors whereby porphyrins enhance oligomerization and the protease activity of HtrA1, while active HtrA1 elevates the pro-apoptotic actions of porphyrin derivatives. Further analysis of this interplay may shed insights into the pathogenesis of diseases such as AMD, CARASIL and protoporphyria, as well as effective therapeutic development.


Assuntos
Protoporfirinas/farmacologia , Serina Endopeptidases/efeitos dos fármacos , Apoptose , Avaliação Pré-Clínica de Medicamentos , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Humanos , Metaloporfirinas/metabolismo , Metaloporfirinas/farmacologia , Ligação Proteica , Estrutura Quaternária de Proteína , Protoporfirinas/metabolismo , Serina Endopeptidases/metabolismo
9.
J Am Chem Soc ; 136(52): 18087-99, 2014 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-25475739

RESUMO

Axial Cu-S(Met) bonds in electron transfer (ET) active sites are generally found to lower their reduction potentials. An axial S(Met) bond is also present in cytochrome c (cyt c) and is generally thought to increase the reduction potential. The highly covalent nature of the porphyrin environment in heme proteins precludes using many spectroscopic approaches to directly study the Fe site to experimentally quantify this bond. Alternatively, L-edge X-ray absorption spectroscopy (XAS) enables one to directly focus on the 3d-orbitals in a highly covalent environment and has previously been successfully applied to porphyrin model complexes. However, this technique cannot be extended to metalloproteins in solution. Here, we use metal K-edge XAS to obtain L-edge like data through 1s2p resonance inelastic X-ray scattering (RIXS). It has been applied here to a bis-imidazole porphyrin model complex and cyt c. The RIXS data on the model complex are directly correlated to L-edge XAS data to develop the complementary nature of these two spectroscopic methods. Comparison between the bis-imidazole model complex and cyt c in ferrous and ferric oxidation states show quantitative differences that reflect differences in axial ligand covalency. The data reveal an increased covalency for the S(Met) relative to N(His) axial ligand and a higher degree of covalency for the ferric states relative to the ferrous states. These results are reproduced by DFT calculations, which are used to evaluate the thermodynamics of the Fe-S(Met) bond and its dependence on redox state. These results provide insight into a number of previous chemical and physical results on cyt c.


Assuntos
Citocromos c/química , Imidazóis/química , Ferro/química , Metaloporfirinas/química , Metionina/química , Difração de Raios X , Citocromos c/metabolismo , Transporte de Elétrons , Metaloporfirinas/metabolismo , Teoria Quântica
10.
J Biol Phys ; 40(3): 275-83, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24824526

RESUMO

The c-MYC gene plays an important role in the regulation of cell proliferation and growth and it is overexpressed in a wide variety of human cancers. Around 90% of c-MYC transcription is controlled by the nuclease-hypersensitive element III1 (NHE III1), whose 27-nt purine-rich strand has the ability to form a G-quadruplex structure under physiological conditions. Therefore, c-MYC DNA is an attractive target for drug design, especially for cancer chemotherapy. Here, the interaction of water-soluble tetrapyridinoporphyrazinatozinc(II) with 27-nt G-rich strand (G/c-MYC), its equimolar mixture with the complementary sequence (GC/c-MYC) and related C-rich oligonucleotide (C/c-MYC) is investigated. Circular dichroism (CD) measurements of the G-rich 27-mer oligonucleotide in 150 mM KCl, pH 7 demonstrate a spectral signature consistent with parallel G-quadruplex DNA. Furthermore, the CD spectrum of the GC rich oligonucleotide shows characteristics of both duplex and quadruplex structures. Absorption spectroscopy implies that the complex binding of G/c-MYC and GC/c-MYC is a two-step process; in the first step, a very small red shift and hypochromicity and in the second step, a large red shift and hyperchromicity are observed in the Q band. Emission spectra of zinc porphyrazine are quenched upon addition of three types of DNA. According to the results of spectroscopy, it can be concluded the dominant binding mode is probably, outside binding and end stacking.


Assuntos
DNA/química , Genes myc/genética , Metaloporfirinas/química , Análise Espectral , Animais , Sequência de Bases , Bovinos , DNA/genética , DNA/metabolismo , Sequência Rica em GC , Metaloporfirinas/metabolismo , Solubilidade , Água/química
11.
Arch Biochem Biophys ; 529(1): 45-54, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23142682

RESUMO

Peroxynitrite (ONOO(-)) formation in mitochondria may be favored due to the constant supply of superoxide radical (O(2)(∙-)) by the electron transport chain plus the facile diffusion of nitric oxide ((∙)NO) to this organelle. Herein, a model system of submitochondrial particles (SMP) in the presence of succinate plus the respiratory inhibitor antimycin A (to increase O(2)(∙-) rates) and the (∙)NO-donor NOC-7 was studied to directly establish and quantitate peroxynitrite by a multiplicity of methods including chemiluminescence, fluorescence and immunochemical analysis. While all the tested probes revealed peroxynitrite at near stoichiometric levels with respect to its precursor radicals, coumarin boronic acid (a probe that directly reacts with peroxynitrite) had the more straightforward oxidation profile from O(2)(∙-)-forming SMP as a function of the (∙)NO flux. Interestingly, immunospintrapping studies verified protein radical generation in SMP by peroxynitrite. Substrate-supplemented SMP also reduced Mn(III)porphyrins (MnP) to Mn(II)P under physiologically-relevant oxygen levels (3-30 µM); then, Mn(II)P were capable to reduce peroxynitrite and protect SMP from the inhibition of complex I-dependent oxygen consumption and protein radical formation and nitration of membranes. The data directly support the formation of peroxynitrite in mitochondria and demonstrate that MnP can undergo a catalytic redox cycle to neutralize peroxynitrite-dependent mitochondrial oxidative damage.


Assuntos
Antioxidantes/química , Manganês/química , Metaloporfirinas/química , Óxido Nítrico/química , Ácido Peroxinitroso/metabolismo , Partículas Submitocôndricas/metabolismo , Animais , Antimicina A/farmacologia , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Catálise , Transporte de Elétrons/efeitos dos fármacos , Hidrazinas/farmacologia , Medições Luminescentes , Manganês/metabolismo , Metaloporfirinas/metabolismo , Metaloporfirinas/farmacologia , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , NAD/metabolismo , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Oxirredução , Oxigênio/química , Oxigênio/metabolismo , Ácido Peroxinitroso/antagonistas & inibidores , Ratos , Espectrometria de Fluorescência , Partículas Submitocôndricas/efeitos dos fármacos , Ácido Succínico/metabolismo , Superóxidos/antagonistas & inibidores , Superóxidos/metabolismo
12.
Food Chem Toxicol ; 49(5): 1047-55, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21272610

RESUMO

In this study, we analyzed the anti-inflammatory effects of Angelica dahurica Bentham et Hooker ethanolic extract (ADEE) on RAW264.7 cells, to understand the mechanism underlying its observed effects. ADEE inhibited cyclooxygenase 2 (COX-2) and inducible nitric oxide synthase (iNOS) expression, leading to the suppression of COX-2-derived prostaglandin E(2) and iNOS-derived production in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. These inhibitory effects of ADEE were accompanied by the reduced production of tumor necrosis factor α and interleukin (IL)-6. ADEE also inhibited nuclear factor κB (NFκB) translocation to the nucleus by interrupting inhibitor kappa Bα (IκBα) degradation. ADEE upregulated heme oxygenase 1 expression, and treatment with tin protoporphyrin IX (SnPP), a selective inhibitor of HO-1, reversed the LPS-induced generation of proinflammatory cytokines. ADEE also induced IL-4 and IL-5 expression in concanavalin-A-stimulated splenocytes. These results suggest that ADEE has anti-inflammatory activity, which acts via the suppression of the NF-κB pathway.


Assuntos
Angelica/química , Anti-Inflamatórios/farmacologia , Heme Oxigenase-1/metabolismo , Proteínas de Membrana/metabolismo , Extratos Vegetais/farmacologia , Animais , Western Blotting , Linhagem Celular , Sobrevivência Celular , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Proteínas I-kappa B/metabolismo , Interleucina-4/metabolismo , Interleucina-5/metabolismo , Interleucina-6/antagonistas & inibidores , Interleucina-6/metabolismo , Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Metaloporfirinas/metabolismo , Camundongos , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Protoporfirinas/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
13.
J Am Chem Soc ; 132(11): 3997-4005, 2010 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-20192195

RESUMO

The first example of a computationally de novo designed protein that binds an emissive abiological chromophore is presented, in which a sophisticated level of cofactor discrimination is pre-engineered. This heterotetrameric, C(2)-symmetric bundle, A(His):B(Thr), uniquely binds (5,15-di[(4-carboxymethyleneoxy)phenyl]porphinato)zinc [(DPP)Zn] via histidine coordination and complementary noncovalent interactions. The A(2)B(2) heterotetrameric protein reflects ligand-directed elements of both positive and negative design, including hydrogen bonds to second-shell ligands. Experimental support for the appropriate formulation of [(DPP)Zn:A(His):B(Thr)](2) is provided by UV/visible and circular dichroism spectroscopies, size exclusion chromatography, and analytical ultracentrifugation. Time-resolved transient absorption and fluorescence spectroscopic data reveal classic excited-state singlet and triplet PZn photophysics for the A(His):B(Thr):(DPP)Zn protein (k(fluorescence) = 4 x 10(8) s(-1); tau(triplet) = 5 ms). The A(2)B(2) apoprotein has immeasurably low binding affinities for related [porphinato]metal chromophores that include a (DPP)Fe(III) cofactor and the zinc metal ion hemin derivative [(PPIX)Zn], underscoring the exquisite active-site binding discrimination realized in this computationally designed protein. Importantly, elements of design in the A(His):B(Thr) protein ensure that interactions within the tetra-alpha-helical bundle are such that only the heterotetramer is stable in solution; corresponding homomeric bundles present unfavorable ligand-binding environments and thus preclude protein structural rearrangements that could lead to binding of (porphinato)iron cofactors.


Assuntos
Desenho de Fármacos , Metaloporfirinas/química , Metaloporfirinas/metabolismo , Modelos Moleculares , Proteínas/química , Proteínas/metabolismo , Zinco/química , Absorção , Sequência de Aminoácidos , Dicroísmo Circular , Simulação por Computador , Elétrons , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Especificidade por Substrato , Ultracentrifugação
14.
J Neurochem ; 98(4): 1324-42, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16895586

RESUMO

We evaluated whether both inert and catalytically active metalloporphyrin antioxidants, meso-substituted with either phenyl-based or N-alkylpyridinium-based groups, suppress Ca(2+)-dependent neurotoxicity in cell culture models of relevance to cerebral ischemia. Representatives from both metalloporphyrin classes, regardless of antioxidant strength, protected cultured cortical neurons or PC-12 cultures against the Ca(2+) ionophores ionomycin or A23187, by suppressing neurotoxic Ca(2+) influx. Some metalloporphyrins suppressed excitotoxic Ca(2+) influx indirectly induced by the Ca(2+) ionophores in cortical neurons. Metalloporphyrins did not quench intracellular fluorescence, suggesting localization to the plasma membrane interface and/or interference with Ca(2+) ionophores. Metalloporphyrins suppressed ionomycin-induced Mn(2+) influx, but did not protect cortical neurons against pyrithione, a Zn(2+) ionophore. In other Ca(2+)-dependent paradigms, Ca(2+) influx via plasma membrane depolarization, but not through reversal of plasmalemmal Na(+)/Ca(2+) exchangers, was modestly suppressed by Mn(III)meso-tetrakis(4-benzoic acid)porphyrin (Mn(III)TBAP) or by an inert analog, Zn(II)TBAP. Mn(III)TBAP and Zn(II)TBAP potently protected cortical neurons against long-duration oxygen-glucose deprivation (OGD), performed in the presence of antagonists of NMDA, alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate and L-type voltage-gated Ca(2+) channels, raising the possibility of an unconventional mode of blockade of transient receptor protein melastatin 7 channels by a metalloTBAP family of metalloporphyrins. The present study extends the range of Ca(2+)-dependent insults for which metalloporphyrins demonstrate unconventional neuroprotection. MetalloTBAPs appear capable of targeting an OGD temporal continuum.


Assuntos
Antioxidantes/farmacologia , Sinalização do Cálcio/fisiologia , Metaloporfirinas/metabolismo , Fármacos Neuroprotetores , Animais , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Cálcio/metabolismo , Catálise , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Fluoresceínas/metabolismo , Ácido Glutâmico/metabolismo , N-Metilaspartato/farmacologia , Neurônios/patologia , Síndromes Neurotóxicas/patologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Canais de Cátion TRPM/metabolismo
15.
Comb Chem High Throughput Screen ; 8(4): 347-52, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16101011

RESUMO

Metalloporphyrin catalyzed biomimetic oxidation was used for the identification of nitric oxide (NO) donors with diverse chemical structure. Methodology was validated by testing known NO donors. Efficient automation of the test allowed us to investigate a subset of our corporate library. Several hits identified in this campaign were validated in both the chemical and also microsomal model that revealed all hits to be active in the biological system, as well. One of the hits showed comparable activity to V-PYRRO/NO, the prototypic liver selective NO donor.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Doadores de Óxido Nítrico/farmacologia , Animais , Biomimética , Técnicas de Química Combinatória , Peróxido de Hidrogênio/metabolismo , Masculino , Metaloporfirinas/química , Metaloporfirinas/metabolismo , Metaloporfirinas/farmacologia , Microssomos Hepáticos , Ratos , Ratos Wistar , Reprodutibilidade dos Testes
16.
J Control Release ; 93(2): 141-50, 2003 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-14636720

RESUMO

Photosensitizers play a crucial role in the photodynamic therapy (PDT) of cancer. In this study, a third-generation aryl ether dendrimer porphyrin with 32 primary amine groups on the periphery, [NH2CH2CH2NHCO]32DPZn, and pH-sensitive, polyion complex micelles (PIC) composed of the porphyrin dendrimer and PEG-b-poly(aspartic acid), were evaluated as new photosensitizers (PSs) for PDT in the Lewis Lung Carcinoma (LLC) cell line. The preliminary photophysical characteristics of [NH2CH2CH2NHCO]32DPZn and the corresponding micelles were investigated. Electrostatic assembly resulted in a red-shift of the Soret peak of the porphyrin core and the enhanced fluorescence. Compared to the dendrimer porphyrin [NH2CH2CH2NHCO]32DPZn, relatively low cellular uptake of dendrimer porphyrin [NH2CH2CH2NHCO]32DPZn incorporated in the PIC micelle was observed, yet the latter exhibited enhanced photodynamic efficacy on the LLC cell line. Importantly, the use of PIC micelles as a delivery system reduced the dark toxicity of the cationic dendrimer porphyrin, probably due to the biocompatible PEG shell of the micelles.


Assuntos
Portadores de Fármacos/farmacologia , Metaloporfirinas/farmacologia , Micelas , Neoplasias/terapia , Éteres Fenílicos/farmacologia , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/química , Animais , Ácido Aspártico/química , Carcinoma Pulmonar de Lewis/patologia , Previsões , Substâncias Macromoleculares , Metaloporfirinas/síntese química , Metaloporfirinas/metabolismo , Nanotecnologia , Éteres Fenílicos/síntese química , Éteres Fenílicos/metabolismo , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/farmacologia , Fototerapia , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Polímeros , Proteínas/química , Proteínas/farmacologia , Espectrometria de Fluorescência , Células Tumorais Cultivadas
17.
FEMS Microbiol Lett ; 208(1): 77-81, 2002 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-11934497

RESUMO

Vibrio vulnificus strain L-180, a clinical isolate, can obtain iron from a synthetic heme, iron-tetra(4-sulfonatophenyl)porphyrin (Fe-TPPS), as well as from a natural heme, protoheme. This assimilation of iron bound to TPPS was demonstrated to be a common property of V. vulnificus by testing a total of 27 strains isolated from both clinical and environmental sources. Strain L-180 could also utilize Fe-TCPP, but not Fe-TMPyP, as a sole iron source. TPPS or its complex with a metal ion reduced bacterial multiplication in the broth containing a minimum dose of Fe-TPPS. When inoculated into human serum supplemented with Fe-TCPP, L-180 could grow only in the presence of a protease from the same bacterium. In both TPPS and TCPP, each side chain of a porphyrin ring has a negative charge. Therefore, this negative charge may be important for interaction with an outer membrane receptor involving in a heme-assimilating system of V. vulnificus.


Assuntos
Heme/metabolismo , Ferro/metabolismo , Metaloporfirinas/metabolismo , Vibrio/metabolismo , Animais , Sangue/microbiologia , Meios de Cultura , Microbiologia Ambiental , Heme/química , Humanos , Camundongos , Especificidade por Substrato , Vibrio/crescimento & desenvolvimento , Vibrioses/microbiologia
18.
J Clin Laser Med Surg ; 14(5): 343-8, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9612202

RESUMO

Cancer and cardiovascular disease are the leading causes of death in the western world. Photodynamic therapy (PDT) has demonstrated activity in the treatment of superficial cancerous lesions and as an intraoperative adjunct during surgical debulking. Texaphyrins are pure, synthetic water-soluble macrocycles that localize in both cancerous lesions and atheromatous plaque. Lutetium texaphyrin (PCI-0123) is activated by tissue-penetrating far red light (720-760 nm). Patient diagnosis and treatment planning is possible via magnetic resonance imaging (MRI) with the paramagnetic gadolinium texaphyrin (PCI-0120) or via fluorescence imaging using the diamagnetic PCI-0123. In this study it is shown that texaphyrins localize selectively in cancer and atheromatous plaque. PDT with PCI-0123 is found to cause selective photodamage to the diseased tissue. Specifically, PCI-0123 acts to eradicate the SMT-F murine mammary tumors and diet-induced atheromatous plaque in rabbits.


Assuntos
Antineoplásicos/uso terapêutico , Arteriosclerose/tratamento farmacológico , Metaloporfirinas/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Animais , Antineoplásicos/química , Argônio , Colesterol/metabolismo , Gadolínio , Terapia a Laser , Lutécio , Masculino , Metaloporfirinas/química , Metaloporfirinas/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/metabolismo , Coelhos , Análise de Sobrevida , Células Tumorais Cultivadas
19.
Can J Physiol Pharmacol ; 74(3): 278-85, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8773407

RESUMO

Synthetic metalloporphyrin derivatives of heme have been identified as competitive inhibitors of heme oxygenase, the rate-limiting enzyme in the heme degradation pathway that produces equimolar quantities of carbon monoxide, biliverdin, and bilirubin. Therefore, administration of metalloporphyrin has been proposed as one means to prevent excessive hyperbilirubinemia in human neonates. Tin proto- and meso-porphyrin have been studied for the suppression of neonatal jaundice. However, these compounds are also photosensitizers. This property may limit the clinical use of these compounds, particularly in infants being exposed simultaneously to phototherapy. Via measurements of carbon monoxide, we have studied the photooxidative, metabolic, and inhibitory characteristics of metalloporphyrins that have not yet been studied for this purpose: deuteroporphyrin bisglycol (DBG), iron deuteroporphyrin bisglycol (FeBG), tin deuteroporphyrin bisglycol (SnBG),tin deuteroporphyrin (SnDP), chromium deuteroporphyrin (CrDP), and zinc deuteroporphyrin (ZnDP). Of the six compounds, SnDP, SnBG, and DBG were significantly photoreactive in vitro. Furthermore, in vitro measurements of brain, liver, and spleen heme oxygenase activity inhibition indicated that, of the nonphotoreactive compounds, CrDP and ZnDP were the most potent inhibitors. Only FeBG served as a substrate for the heme oxygenase reaction. The concentration for 50% inhibition with the three tissues ranged from 0.6 to 1.3 microM for CrDP and from 11.0 to 13.5 microM for ZnDP. When 25 mumol CrDP and 50 mumol ZnDP per kilogram body weight were administered to rats given a heme load of 30 mumol/kg body weight, the total body carbon monoxide excretion due to the administered heme load was reduced by 46 and 32%, respectively, at t = 7.5 h, when the experiment was terminated in order to measure heme oxygenase activity. Brain heme oxygenase activity was not affected by the metalloporphyrin treatment. However, both CrDP and ZnDP inhibited liver tissue heme oxygenase activity to 5 and 20%, respectively, whereas only CrDP inhibited spleen tissue heme oxygenase, to 7% of untreated control. Thus, CrDP appears to be the most attractive of the six compounds and deserves further study.


Assuntos
Inibidores Enzimáticos/farmacologia , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Metaloporfirinas/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Inibidores Enzimáticos/metabolismo , Hemólise/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Metaloporfirinas/metabolismo , Estrutura Molecular , Ratos , Ratos Wistar , Baço/efeitos dos fármacos , Baço/enzimologia
20.
Nucleic Acids Res ; 19(22): 6283-8, 1991 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-1720241

RESUMO

Cationic manganese-porphyrin complexes, free or targetted with an intercalating agent, are able to cleave DNA using oxygen atom donors like potassium monopersulfate or magnesium monoperphthalate as coreactants. Detailed studies of the cleavage of calf thymus DNA, before and after a heating step, show that free bases and 5-methylene-2-furanone are the main reaction products, indicating that hydroxylation at the 1'-carbon atom is the main target of these chemical agents. These data confirm that metalloporphyrin derivatives interact with the minor groove of double-stranded DNA. Hydroxylation of one of the two C-H bonds at position-5' is another initial DNA damage, characterized by the formation of furfural as sugar degradation product. Besides these two main initial damage sites, a low contribution of a hydroxylation reaction at C4' can not be definitively discounted, while an hydroperoxidation route at C4' can be excluded.


Assuntos
Dano ao DNA , DNA/metabolismo , Metaloporfirinas/metabolismo , Animais , Bleomicina/toxicidade , Cátions , Bovinos , DNA/efeitos dos fármacos , Furanos/metabolismo , Hidroxilação , Cinética , Oxirredução , Extratos Vegetais/metabolismo , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA