Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ChemMedChem ; 17(10): e202200139, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35385211

RESUMO

In silico adsorption of eight antimalarials that inhibit ß-hematin (synthetic hemozoin) formation identified a primary binding site on the (001) face, which accommodates inhibitors via formation of predominantly π-π interactions. A good correlation (r2 =0.64, P=0.017) between adsorption energies and the logarithm of ß-hematin inhibitory activity was found for this face. Of 53 monocyclic, bicyclic and tricyclic scaffolds, the latter yielded the most favorable adsorption energies. Five new amino-phenoxazine compounds were pursued as ß-hematin inhibitors based on adsorption behaviour. The 2-substituted phenoxazines show good to moderate ß-hematin inhibitory activity (<100 µM) and Plasmodium falciparum blood stage activity against the 3D7 strain. N1 ,N1 -diethyl-N4 -(10H-phenoxazin-2-yl)pentane-1,4-diamine (P2a) is the most promising hit with IC50 values of 4.7±0.6 and 0.64±0.05 µM, respectively. Adsorption energies are predictive of ß-hematin inhibitory activity, and thus the in silico approach is a beneficial tool for structure-based development of new non-quinoline inhibitors.


Assuntos
Antimaláricos , Hemeproteínas , Adsorção , Hemeproteínas/química , Plasmodium falciparum
2.
Acc Chem Res ; 54(11): 2649-2659, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33982570

RESUMO

Over the last century, malaria deaths have decreased by more than 85%. Nonetheless, there were 405 000 deaths in 2018, mostly resulting from Plasmodium falciparum infection. In the 21st century, much of the advance has arisen from the deployment of insecticide-treated bed nets and artemisinin combination therapy. However, over the past few decades parasites with a delayed artemisinin clearance phenotype have appeared in Southeast Asia, threatening further gains. The effort to find new drugs is thus urgent. A prominent process in blood stage malaria parasites, which we contend remains a viable drug target, is hemozoin formation. This crystalline material consisting of heme can be readily seen when parasites are viewed microscopically. The process of its formation in the parasite, however, is still not fully understood.In early work, we recognized hemozoin formation as a biomineralization process. We have subsequently investigated the kinetics of synthetic hemozoin (ß-hematin) crystallization catalyzed at lipid-aqueous interfaces under biomimetic conditions. This led us to the use of neutral detergent-based high-throughput screening (HTS) for inhibitors of ß-hematin formation. A good hit rate against malaria parasites was obtained. Simultaneously, we developed a pyridine-based assay which proved successful in measuring the concentrations of hematin not converted to ß-hematin.The pyridine assay was adapted to determine the effects of chloroquine and other clinical antimalarials on hemozoin formation in the cell. This permitted the determination of the dose-dependent amounts of exchangeable heme and hemozoin in P. falciparum for the first time. These studies have shown that hemozoin inhibitors cause a dose-dependent increase in exchangeable heme, correlated with decreased parasite survival. Electron spectroscopic imaging (ESI) showed a relocation of heme iron into the parasite cytoplasm, while electron microscopy provided evidence of the disruption of hemozoin crystals. This cellular assay was subsequently extended to top-ranked hits from a wide range of scaffolds found by HTS. Intriguingly, the amounts of exchangeable heme at the parasite growth IC50 values of these scaffolds showed substantial variation. The amount of exchangeable heme was found to be correlated with the amount of inhibitor accumulated in the parasitized red blood cell. This suggests that heme-inhibitor complexes, rather than free heme, lead to parasite death. This was supported by ESI using a Br-containing compound which showed the colocalization of Fe and Br as well as by confocal Raman microscopy which confirmed the presence of a complex in the parasite. Current evidence indicates that inhibitors block hemozoin formation by surface adsorption. Indeed, we have successfully introduced molecular docking with hemozoin to find new inhibitors. It follows that the resulting increase in free heme leads to the formation of the parasiticidal heme-inhibitor complex. We have reported crystal structures of heme-drug complexes for several aryl methanol antimalarials in nonaqueous media. These form coordination complexes but most other inhibitors interact noncovalently, and the determination of their structures remains a major challenge.It is our view that key future developments will include improved assays to measure cellular heme levels, better in silico approaches for predicting ß-hematin inhibition, and a concerted effort to determine the structure and properties of heme-inhibitor complexes.


Assuntos
Antimaláricos/farmacologia , Desenvolvimento de Medicamentos , Heme/metabolismo , Malária/tratamento farmacológico , Animais , Antimaláricos/síntese química , Antimaláricos/química , Humanos , Malária/metabolismo , Malária/parasitologia , Testes de Sensibilidade Parasitária , Plasmodium falciparum/efeitos dos fármacos
3.
RSC Med Chem ; 11(1): 85-91, 2020 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33479606

RESUMO

In this study, we investigated a series of triarylimidazoles, in an effort to elucidate critical SAR information pertaining to their anti-plasmodial and ß-hematin inhibitory activity. Our results showed that in addition to the positional effects of ring substitution, subtle changes to lipophilicity and imidazole ionisability were important factors in SAR interpretation. Finally, in silico adsorption analysis indicated that these compounds exert their effect by inhibiting ß-hematin crystal growth at the fast growing 001 face.

4.
Eur J Med Chem ; 159: 243-254, 2018 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-30296683

RESUMO

The 2-phenylbenzimidazole scaffold has recently been discovered to inhibit ß-hematin (synthetic hemozoin) formation by high throughput screening. Here, a library of 325,728 N-4-(1H-benzo[d]imidazol-2-yl)aryl)benzamides was enumerated, and Bayesian statistics used to predict ß-hematin and Plasmodium falciparum growth inhibition. Filtering predicted inactives and compounds with negligible aqueous solubility reduced the library to 35,124. Further narrowing to compounds with terminal aryl ring substituents only, reduced the library to 18, 83% of which were found to inhibit ß-hematin formation <100 µM and 50% parasite growth <2 µM. Four compounds showed nanomolar parasite growth inhibition activities, no cross-resistance in a chloroquine resistant strain and low cytotoxicity. QSAR analysis showed a strong association of parasite growth inhibition with inhibition of ß-hematin formation and the most active compound inhibited hemozoin formation in P. falciparum, with consequent increasing exchangeable heme. Pioneering use of molecular docking for this system demonstrated predictive ability and could rationalize observed structure activity trends.


Assuntos
Antimaláricos/farmacologia , Benzimidazóis/farmacologia , Hemeproteínas/antagonistas & inibidores , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Antimaláricos/síntese química , Antimaláricos/química , Benzimidazóis/síntese química , Benzimidazóis/química , Relação Dose-Resposta a Droga , Simulação de Acoplamento Molecular , Estrutura Molecular , Testes de Sensibilidade Parasitária , Relação Estrutura-Atividade
5.
ChemMedChem ; 10(12): 2099-110, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26447782

RESUMO

7-Chloroquinoline-based antimalarial drugs are effective in the inhibition of hemozoin formation in the food vacuole of the Plasmodium parasite, the causative agent of malaria. We synthesized five series of ferroquine (FQ) and phenylequine (PQ) derivatives, which display good in vitro efficacy toward both the chloroquine-sensitive (CQS) NF54 (IC50 : 4.2 nm) and chloroquine-resistant (CQR) Dd2 (IC50 : 33.7 nm) strains of P. falciparum. Several compounds were found to have good inhibitory activity against ß-hematin formation in an NP-40 detergent assay, with IC50 values ranging between 10.4 and 19.2 µm.


Assuntos
Aminoquinolinas/química , Antimaláricos/síntese química , Cloroquina/química , Desenho de Fármacos , Compostos Ferrosos/química , Aminoquinolinas/farmacologia , Antimaláricos/química , Antimaláricos/farmacologia , Cloroquina/síntese química , Cloroquina/farmacologia , Cristalografia por Raios X , Resistência a Medicamentos/efeitos dos fármacos , Compostos Ferrosos/farmacologia , Hemeproteínas/antagonistas & inibidores , Hemeproteínas/metabolismo , Concentração Inibidora 50 , Metalocenos , Testes de Sensibilidade Microbiana , Conformação Molecular , Octoxinol , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/metabolismo , Polietilenoglicóis/química , Ligação Proteica , Relação Estrutura-Atividade
6.
Dalton Trans ; 44(38): 16767-77, 2015 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-26335948

RESUMO

The quinoline methanol antimalarial drug mefloquine is a structural analogue of the Cinchona alkaloids, quinine and quinidine. We have elucidated the single crystal X-ray diffraction structure of the complexes formed between racemic erythro mefloquine and ferriprotoporphyrin IX (Fe(iii)PPIX) and show that alkoxide coordination is a key interaction in the solid-state. Mass spectrometry confirms the existence of coordination complexes of quinine, quinidine and mefloquine to Fe(iii)PPIX in acetonitrile. The length of the iron(iii)-O bond in the quinine and quinidine complexes as determined by Extended X-ray Absorption Fine Structure (EXAFS) spectroscopy unequivocally confirms that coordination of the quinoline methanol compounds to Fe(iii)PPIX occurs in non-aqueous aprotic solution via their benzylic alkoxide functional group. UV-visible spectrophotometric titrations of the low-spin bis-pyridyl-Fe(iii)PPIX complex with each of the quinoline methanol compounds results in the displacement of a single pyridine molecule and subsequent formation of a six-coordinate pyridine-Fe(iii)PPIX-drug complex. We propose that formation of the drug-Fe(iii)PPIX coordination complexes is favoured in a non-aqueous environment, such as that found in lipid bodies or membranes in the malaria parasite, and that their existence may contribute to the mechanism of haemozoin inhibition or other toxicity effects that lead ultimately to parasite death. In either case, coordination is a key interaction to be considered in the design of novel antimalarial drug candidates.


Assuntos
Acetonitrilas/química , Antimaláricos/química , Complexos de Coordenação/química , Compostos Férricos/química , Mefloquina/química , Protoporfirinas/química , Quinidina/química , Quinina/química , Antimaláricos/metabolismo , Alcaloides de Cinchona/química , Complexos de Coordenação/metabolismo , Compostos Férricos/metabolismo , Mefloquina/metabolismo , Protoporfirinas/metabolismo , Quinidina/metabolismo , Espectrofotometria , Termodinâmica , Difração de Raios X
7.
J Am Chem Soc ; 135(3): 1037-47, 2013 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-23253048

RESUMO

Single crystals of solvated ß-hematin were grown from a DMSO solution containing the antimalarial drug chloroquine, a known inhibitor of ß-hematin formation. In addition, a kinetics study employing biomimetic lipid-water emulsion conditions was undertaken to further investigate the effect of chloroquine and quinidine on the formation of ß-hematin. Scanning electron microscopy shows that the external morphology of the ß-hematin DMSO solvate crystals is almost indistinguishable from that of malaria pigment (hemozoin), and single crystal X-ray diffraction confirms the presence of µ-propionato coordination dimers of iron(III) protoporphyrin IX. The free propionic acid functional groups of adjacent dimers hydrogen bond to included DMSO molecules, rather than forming carboxylic acid dimers. The observed exponential kinetics were modeled using the Avrami equation, with an Avrami constant equal to 1. The decreased rate of ß-hematin formation observed at low concentrations of both drugs could be accounted for by assuming a mechanism of drug adsorption to sites on the fastest growing face of ß-hematin. This behavior was modeled using the Langmuir isotherm. Higher concentrations of drug resulted in decreased final yields of ß-hematin, and an irreversible drug-induced precipitation of iron(III) protoporphyrin IX was postulated to account for this. The model permits determination of the equilibrium adsorption constant (K(ads)). The values for chloroquine (log K(ads) = 5.55 ± 0.03) and quinidine (log K(ads) = 4.92 ± 0.01) suggest that the approach may be useful as a relative probe of the mechanism of action of novel antimalarial compounds.


Assuntos
Cloroquina/química , Dimetil Sulfóxido/química , Hemeproteínas/química , Animais , Cloroquina/farmacologia , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Hemeproteínas/antagonistas & inibidores , Hemeproteínas/síntese química , Cinética , Modelos Moleculares , Estrutura Molecular , Soluções , Relação Estrutura-Atividade , Suínos
8.
ACS Chem Biol ; 7(4): 666-71, 2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-22276975

RESUMO

The antimalarial properties of the Cinchona alkaloids quinine and quinidine have been known for decades. Surprisingly, 9-epiquinine and 9-epiquinidine are almost inactive. A lack of definitive structural information has precluded a clear understanding of the relationship between molecular structure and biological activity. In the current study, we have determined by single crystal X-ray diffraction the structures of the complexes formed between quinine and quinidine and iron(III) protoporphyrin IX (Fe(III)PPIX). Coordination of the alkaloid to the Fe(III) center is a key feature of both complexes, and further stability is provided by an intramolecular hydrogen bond formed between a propionate side chain of Fe(III)PPIX and the protonated quinuclidine nitrogen atom of either alkaloid. These interactions are believed to be responsible for inhibiting the incorporation of Fe(III)PPIX into crystalline hemozoin during its in vivo detoxification. It is also possible to rationalize the greater activity of quinidine compared to that of quinine.


Assuntos
Antimaláricos/química , Alcaloides de Cinchona/química , Ferro/química , Protoporfirinas/química , Modelos Moleculares , Conformação Molecular , Quinidina , Quinina , Relação Estrutura-Atividade , Difração de Raios X
9.
Dalton Trans ; 39(5): 1235-44, 2010 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-20104349

RESUMO

The mechanism of formation of haemozoin, a detoxification by-product of several blood-feeding organisms including malaria parasites, has been a subject of debate; however, recent studies suggest that neutral lipids may serve as a catalyst. In this study, a model system consisting of an emulsion of neutral lipid particles was employed to investigate the formation of beta-haematin, the synthetic counterpart of haemozoin, at the lipid-water interface. A solution of monoglyceride, either monostearoylglycerol (MSG) or monopalmitoylglycerol (MPG), dissolved in acetone and methanol was introduced to an aqueous surface. Fluorescence, confocal and transmission electron microscopic (TEM) imaging and dynamic light scattering analysis of samples obtained from beneath the surface confirmed the presence of homogeneous lipid particles existing in two major populations: one in the low micrometre size range and the other in the hundred nanometre range. The introduction of haem (Fe(iii)PPIX) to this lipid particle system under biomimetic conditions (37 degrees C, pH 4.8) produced beta-haematin with apparent first-order kinetics and an average half life of 0.5 min. TEM of monoglycerides (MSG or MPG) extruded through a 200 nm filter with haem produced beta-haematin crystals aligned and parallel to the lipid-water interface. These TEM data, together with a model system replacing the lipid with an aqueous organic solvent interface using either methyl laurate or docosane demonstrated that the OH and C[double bond, length as m-dash]O groups are apparently necessary for efficient nucleation. This suggests that beta-haematin crystallizes via epitaxial nucleation at the lipid-water interface through interaction of Fe(iii)PPIX with the polar head group. Once nucleated, the crystal grows parallel to the interface until growth is terminated by the curvature of the lipid particle. The hydrophobic nature of the mature crystal favours an interior transport resulting in crystals aligned parallel to the lipid-water interface and each other, strikingly similar to that seen in malaria parasites.


Assuntos
Hemeproteínas/química , Lipídeos/química , 1,2-Dipalmitoilfosfatidilcolina/química , Cristalização , Heme/química , Cinética , Microscopia Eletrônica de Transmissão , Espectrofotometria Infravermelho , Difração de Raios X
10.
Molecules ; 14(8): 2868-87, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19701131

RESUMO

Haem is believed to be the target of some of the historically most important antimalarial drugs, most notably chloroquine. This target is almost ideal as haem is host-derived and the process targeted, haemozoin formation, is a physico-chemical process with no equivalent in the host. The result is that the target remains viable despite resistance to current drugs, which arises from mutations in parasite membrane transport proteins. Recent advances in high-throughput screening methods, together with a better understanding of the interaction of existing drugs with this target, have created new prospects for discovering novel haem-targeting chemotypes and for target-based structural design of new drugs. Finally, the discovery that Schistosoma mansoni also produces haemozoin suggests that new drugs of this type may be chemotherapeutic not only for malaria, but also for schistosomiasis. These recent developments in the literature are reviewed.


Assuntos
Antimaláricos/química , Antimaláricos/uso terapêutico , Malária/tratamento farmacológico , Esquistossomose/tratamento farmacológico , Esquistossomicidas/química , Esquistossomicidas/uso terapêutico , Desenho de Fármacos , Humanos , Estrutura Molecular , Relação Estrutura-Atividade
11.
Inorg Chem ; 48(16): 7994-8003, 2009 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-19572726

RESUMO

The speciation of ferriprotoporphyrin IX (Fe(III)PPIX) in aqueous and mixed aqueous-organic solvents has been investigated by UV-vis, (1)H NMR, magnetic, and diffusion measurements. Fe(III)PPIX has been found to form monomers, pi-pi dimers, mu-oxo dimers, and pi-stacked aggregates of mu-oxo dimers depending on concentration, pH, the presence of salts, temperature, and solvent identity. This highlights the complexity of the behavior of Fe(III)PPIX in solution. However, the presence or absence of the mu-oxo dimer is clearly dependent on solvent, with a series of aprotic solvents (5.64 M DMSO, acetone, DMF, THF, 2,6-lutidine) all promoting mu-oxo dimer formation at pH 10. By contrast, protic solvents (methanol, ethanol, propanol, ethylene glycol, diethylene glycol, and formamide) at the same concentration and under the same conditions give rise only to the pi-pi dimer variously mixed with monomer depending on solvent polarity. The pi-pi dimer has previously been shown to be present in purely aqueous solution. In the presence of 4.25 M NaCl in aqueous solution, on the other hand, both UV-vis spectra and diffusion measurements suggest the presence of large pi-stacked aggregates of mu-oxo dimers at pH 10. In aqueous DMSO at least, the temperature dependence of the dimerization constant shows that the process of mu-oxo dimer formation is endothermic and hence entirely entropy driven. This strongly suggests that formation of the mu-oxo dimer is driven by desolvation, with solvents that can act as both hydrogen bond donors and acceptors to the axial water/hydroxide ligand of Fe(III)PPIX preventing formation of this dimer species, while those that cannot act as hydrogen bond donors facilitate it. The findings permit prediction of the Fe(III)PPIX species present in different mixed solvent systems and in the case of aqueous DMSO at any given pH, concentration, and temperature.


Assuntos
Hemina/química , Cloreto de Sódio/química , Solventes/química , Água/química , Dimerização , Dimetil Sulfóxido/química , Concentração de Íons de Hidrogênio , Magnetismo , Metanol/química , Soluções , Análise Espectral , Termodinâmica
12.
Anal Chem ; 81(7): 2516-24, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19278236

RESUMO

Synchrotron Fourier transform infrared (FT-IR) spectra of fixed single erythrocytes infected with Plasmodium falciparum at different stages of the intraerythrocytic cycle are presented for the first time. Bands assigned to the hemozoin moiety at 1712, 1664, and 1209 cm(-1) are observed in FT-IR difference spectra between uninfected erythrocytes and infected trophozoites. These bands are also found to be important contributors in separating the trophozoite spectra from the uninfected cell spectra in principal components analysis. All stages of the intraerythrocytic lifecycle of the malarial parasite, including the ring and schizont stage, can be differentiated by visual inspection of the C-H stretching region (3100-2800 cm(-1)) and by using principal components analysis. Bands at 2922, 2852, and 1738 cm(-1) assigned to the nu(asym)(CH(2) acyl chain lipids), nu(sym)(CH(2) acyl chain lipids), and the ester carbonyl band, respectively, increase as the parasite matures from its early ring stage to the trophozoite and finally to the schizont stage. Training of an artificial neural network showed that excellent automated spectroscopic discrimination between P. falciparum-infected cells and the control cells is possible. FT-IR difference spectra indicate a change in the production of unsaturated fatty acids as the parasite matures. The ring stage spectrum shows bands associated with cis unsaturated fatty acids. The schizont stage spectrum displays no evidence of cis bands and suggests an increase in saturated fatty acids. These results demonstrate that different phases of the P. falciparum intraerthyrocytic life cycle are characterized by different lipid compositions giving rise to distinct spectral profiles in the C-H stretching region. This insight paves the way for an automated infrared-based technology capable of diagnosing malaria at all intraerythrocytic stages of the parasite's life cycle.


Assuntos
Eritrócitos/parasitologia , Estágios do Ciclo de Vida , Malária/patologia , Malária/parasitologia , Redes Neurais de Computação , Plasmodium falciparum/crescimento & desenvolvimento , Síncrotrons , Animais , Biomarcadores/análise , Biomarcadores/química , Biomarcadores/metabolismo , Eritrócitos/química , Eritrócitos/metabolismo , Hemeproteínas/análise , Hemeproteínas/química , Hemeproteínas/metabolismo , Humanos , Metabolismo dos Lipídeos , Lipídeos/química , Malária/diagnóstico , Plasmodium falciparum/fisiologia , Análise de Componente Principal , Espectroscopia de Infravermelho com Transformada de Fourier
13.
J Inorg Biochem ; 102(8): 1660-7, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18508124

RESUMO

The crystal structure of the complex formed between the antimalarial drug halofantrine and ferriprotoporphyrin IX (Fe(III)PPIX) has been determined by single crystal X-ray diffraction. The structure shows that halofantrine coordinates to the Fe(III) center through its alcohol functionality in addition to pi-stacking of the phenanthrene ring over the porphyrin. The length of the Fe(III)-O bond is consistent with an alkoxide and not an alcohol coordinating group. The iron porphyrin is five coordinate and monomeric. Changes in the electronic spectrum of Fe(III)PPIX upon addition of halofantrine base in acetonitrile solution are almost identical to those observed upon addition of quinidine free base in the same solvent. This suggests homologous binding. Molecular mechanics modeling of Fe(III)PPIX complexes of quinidine, quinine, 9-epiquinine and 9-epiquinidine based on this homology suggests that the antimalarially active quinidine and quinine can readily adopt conformations that permit formation of an intramolecular salt bridge between the protonated quinuclidine tertiary amino group and unprotonated heme propionate group, while the inactive epimers 9-epiquinidine and 9-epiquinine have to adopt high energy conformations in order to accommodate such salt bridge formation. We propose that salt bridge formation may interrupt formation of the hemozoin precursor dimer formed during the heme detoxification pathway and so account for the strong activity of the two active isomers.


Assuntos
Hemina/química , Fenantrenos/química , Álcoois , Antimaláricos/química , Sítios de Ligação , Cristalografia por Raios X , Estrutura Molecular
14.
J Biol Inorg Chem ; 12(1): 101-17, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16972088

RESUMO

Changes in epsilon (393) (the Soret band) of aqueous ferriprotoporphyrin IX [Fe(III)PPIX] with concentration indicate that it dimerizes, but does not form higher aggregates. Diffusion measurements support this observation. The diffusion coefficient of aqueous Fe(III)PPIX is half that of the hydrated monomeric dicyano complex. Much of the apparent instability of aqueous Fe(III)PPIX solutions could be attributed to adsorption onto glass and plastic surfaces. However, epsilon (347) was found to be independent of the aggregation state of the porphyrin and was used to correct for the effects of adsorption. The UV-vis spectrum of the aqueous dimer is not consistent with that expected for a mu-oxo dimer and the (1)H NMR spectrum is characteristic of five-coordinate, high-spin Fe(III)PPIX. Magnetic susceptibility measurements using the Evans method showed that there is no antiferromagnetic coupling in the dimer. By contrast, when the mu-oxo dimer is induced in 10% aqueous pyridine, characteristic UV-vis and (1)H NMR spectra of this species are observed and the magnetic moment is consistent with strong antiferromagnetic coupling. We propose a model in which the spontaneously formed aqueous Fe(III)PPIX dimer involves noncovalent interaction of the unligated faces of two five-coordinate H(2)O/HO-Fe(III)PPIX molecules, with the axial H(2)O/OH(-) ligands directed outwards. This arrangement is consistent with the crystal structures of related five-coordinate iron(III) porphyrins and accounts for the observed pH dependence of the dimerization constant and the spectra of the monomer and dimer. Structures for the aqueous dimer are proposed on the basis of molecular dynamics/simulated annealing calculations using a force field previously developed for modeling metalloporphyrins.


Assuntos
Hemina/química , Adsorção , Fenômenos Químicos , Físico-Química , Cristalografia por Raios X , Difusão , Dimerização , Concentração de Íons de Hidrogênio , Ligantes , Espectroscopia de Ressonância Magnética/métodos , Espectroscopia de Ressonância Magnética/normas , Magnetismo , Modelos Moleculares , Padrões de Referência , Sensibilidade e Especificidade , Soluções/química , Espectrofotometria Ultravioleta/métodos , Propriedades de Superfície , Água/química
15.
FEBS Lett ; 580(21): 5105-10, 2006 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-16956610

RESUMO

Several blood-feeding organisms, including the malaria parasite detoxify haem released from host haemoglobin by conversion to the insoluble crystalline ferriprotoporphyrin IX dimer known as haemozoin. To date the mechanism of haemozoin formation has remained unknown, although lipids or proteins have been suggested to catalyse its formation. We have found that beta-haematin (synthetic haemozoin) forms rapidly under physiologically realistic conditions near octanol/water, pentanol/water and lipid/water interfaces. Molecular dynamics simulations show that a precursor of the haemozoin dimer forms spontaneously in the absence of the competing hydrogen bonds of water, demonstrating that this substance probably self-assembles near a lipid/water interface in vivo.


Assuntos
Hemeproteínas/química , Hemeproteínas/metabolismo , Lipídeos/química , Água/química , Animais , Hemeproteínas/análise , Hemeproteínas/ultraestrutura , Cinética , Modelos Moleculares , Plasmodium falciparum/química , Espectrofotometria Infravermelho , Análise Espectral Raman , Difração de Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...