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.
Biochem Pharmacol ; 95(3): 133-44, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25888926

RESUMO

Substitutionally inert Pt(IV) prodrugs, combining bioactive axial ligands with Pt(IV) derivatives of antitumor Pt(II) compounds, represent a new generation of anticancer drugs. The rationale behind these prodrugs is to release, by reductive elimination inside the cancer cell, an active Pt(II) drug which binds nuclear DNA as well as bioactive ligands that may potentiate toxic effects of the Pt(II) drugs by an independent pathway. Platinum prodrugs, such as Pt(IV) derivatives of cisplatin containing axial valproic acid (VPA) ligands, destroy cancer cells with greater efficacy than conventional cisplatin. These axial ligands were chosen because VPA inhibits histone deacetylase (HDAC) activity, thereby decondensing chromatin and subsequently increasing the accessibility of DNA within chromatin to DNA-binding agents. We examined the mechanism of cytotoxic activity of Pt(IV) derivatives of cisplatin with VPA axial ligands. Particular attention was paid to the role of the VPA ligand in these Pt(IV) prodrugs in the mechanism underlying their toxic effects in human ovarian tumor cells. We demonstrate that (i) treatment of the cells with these prodrugs resulted in enhanced histone H3 acetylation and decondensation of heterochromatin markedly more effectively than free VPA; (ii) of the total Pt inside the cells, a considerably higher fraction of Pt from the Pt(IV)-VPA conjugates is bound to DNA than from the conjugates with biologically inactive ligands. The results indicate that the enhanced cytotoxicity of the Pt(IV)-VPA conjugates is a consequence of several processes involving enhanced cellular accumulation, downregulation of HDACs and yet other biochemical processes (not involving HDACs) which may potentiate antitumor effects.


Assuntos
Epigênese Genética , Compostos Organoplatínicos/metabolismo , Neoplasias Ovarianas/metabolismo , Ácido Valproico/metabolismo , Acetilação , Linhagem Celular Tumoral , Feminino , Glutationa/metabolismo , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Compostos Organoplatínicos/química , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Ácido Valproico/química
2.
J Inorg Biochem ; 140: 72-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25063910

RESUMO

We report new anticancer prodrugs, platinum(IV) derivatives of oxaliplatin conjugated with valproic acid (VPA), a well-known drug having histone deacetylase inhibitory activity. Like most platinum(IV) derivatives, the cytotoxicity of the conjugates was lower in cell culture than that of oxaliplatin, but greater than those of its Pt(IV) derivative containing biologically inactive axial ligands in several cancer cell lines. Notably, these conjugates display activity in both cisplatin sensitive- and resistant tumor cells capable of both markedly enhanced accumulation in tumor cells and acting in a dual threat manner, concurrently targeting histone deacetylase and genomic DNA. These results demonstrate the dual targeting strategy to be a valuable route to pursue in the design of platinum agents which may be more effective in cancer types that are typically resistant to therapy by conventional cisplatin. Moreover, platinum(IV) derivatives containing VPA axial ligands seem to be promising dual-targeting candidates for additional preclinical studies.


Assuntos
Antineoplásicos/química , Compostos Organoplatínicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Compostos Organoplatínicos/farmacologia , Oxaliplatina
3.
Metallomics ; 6(8): 1491-501, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24828756

RESUMO

The cellular mechanism of action of an iridium(III) half-sandwich complex [(η(5)-C5Me4C6H4C6H5)Ir(phen)Cl]PF6 (phen = phenanthroline) (1) is reported. Complex 1 was used to treat several cell lines, including cisplatin-sensitive, cisplatin-resistant (with intrinsic and acquired resistance) carcinoma cells with wild type p53 status as well as the cells with no intact p53 gene, and nontumorigenic cells. Complex 1 preferentially kills cancer cells over nontumorigenic cells and exhibits no cross-resistance with cisplatin. It appears to retain significant activity in human tumor cell lines that are refractory or poorly responsive to cisplatin, and in contrast to cisplatin it displays a high activity in human tumor cell lines that are characterized by both wild type and mutant p53 gene. The mechanism of cell killing was established through detailed cell-based assays. Complex 1 exhibits dual effects in killing cancer cells causing nuclear DNA damage and mitochondrial dysfunction involving ROS production simultaneously. Flow cytometric studies and impedance-based monitoring of cellular responses to 1 demonstrated that 1 acts more quickly than cisplatin to induce cell death and that 1 is a more effective apoptosis inducer than cisplatin in particular in early stages of treatment, when the apoptotic effects predominate over necrosis. Overall, our findings confirm that 1 and its iridium derivatives represent promising candidates for further pre-clinical studies and new additions to the growing family of nonplatinum metal-based anticancer complexes.


Assuntos
Antineoplásicos/farmacologia , Irídio/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Citometria de Fluxo , Humanos , Espécies Reativas de Oxigênio/metabolismo
4.
Stem Cells Dev ; 23(20): 2443-54, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24836366

RESUMO

The genomic destabilization associated with the adaptation of human embryonic stem cells (hESCs) to culture conditions or the reprogramming of induced pluripotent stem cells (iPSCs) increases the risk of tumorigenesis upon the clinical use of these cells and decreases their value as a model for cell biology studies. Base excision repair (BER), a major genomic integrity maintenance mechanism, has been shown to fail during hESC adaptation. Here, we show that the increase in the mutation frequency (MF) caused by the inhibition of BER was similar to that caused by the hESC adaptation process. The increase in MF reflected the failure of DNA maintenance mechanisms and the subsequent increase in MF rather than being due solely to the accumulation of mutants over a prolonged period, as was previously suggested. The increase in the ionizing-radiation-induced MF in adapted hESCs exceeded the induced MF in nonadapted hESCs and differentiated cells. Unlike hESCs, the overall DNA maintenance in iPSCs, which was reflected by the MF, was similar to that in differentiated cells regardless of the time spent in culture and despite the upregulation of several genes responsible for genome maintenance during the reprogramming process. Taken together, our results suggest that the changes in BER activity during the long-term cultivation of hESCs increase the mutagenic burden, whereas neither reprogramming nor long-term propagation in culture changes the MF in iPSCs.


Assuntos
Loci Gênicos , Hipoxantina Fosforribosiltransferase/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Taxa de Mutação , Diferenciação Celular/efeitos da radiação , Linhagem Celular , Raios gama , Humanos , Hipoxantina Fosforribosiltransferase/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia
5.
Chemistry ; 18(35): 10983-90, 2012 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-22806942

RESUMO

The interaction with native DNA of a 2,7-diazapyrenium-based ligand 1 and its Pt(II) rectangular metallacycle 2 is explored through circular and linear dichroism and fluorescence spectroscopies. The metal-free ligand 1 binds through intercalation, with a binding constant of approximately 5×10(5) M(-1), whereas the metallacycle 2 binds and bends the DNA with a binding constant of 7×10(6) M(-1). PCR assays show that metallo-supramolecular box 2 interferes with DNA transactions in vitro whereas the intercalator 1 does not. The metallacycle is active against four human cancer cell lines, with IC(50) values ranging between 3.1 and 19.2 µM and shows similar levels of efficacy, but a different spectrum of activity, to cisplatin.


Assuntos
Antineoplásicos/química , DNA de Neoplasias/química , Substâncias Intercalantes/química , Compostos Organoplatínicos/química , Fenantrolinas/química , Platina/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dicroísmo Circular , Cisplatino/química , DNA de Neoplasias/antagonistas & inibidores , Humanos , Concentração Inibidora 50 , Substâncias Intercalantes/farmacologia , Cinética , Ligantes , Conformação Molecular , Desnaturação de Ácido Nucleico/efeitos dos fármacos , Compostos Organoplatínicos/farmacologia , Espectrometria de Fluorescência , Termodinâmica
6.
Mol Pharm ; 9(7): 1990-9, 2012 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-22591133

RESUMO

cis-[Pt(II)(NH3)2(malSAHA-2H)], a cisplatin adduct conjugated to a potent histone deacetylase inhibitor (HDACi), suberoylanilide hydroxamic acid (SAHA), was previously developed as a potential anticancer agent. This Pt-HDACi conjugate was demonstrated to have comparable cytotoxicity to cisplatin against A2780 ovarian cancer cells but significantly reduced cytotoxicity against a representative normal cell line, NHDF. Thus, with a view to (i) understanding more deeply the effects that may play an important role in the biological (pharmacological) properties of this new conjugate against cancer cells and (ii) developing the next generation of Pt-HDACi conjugates, the cytotoxicity, DNA binding, cellular accumulation and HDAC inhibitory activity of cis-[Pt(II)(NH3)2(malSAHA-2H)] were investigated and are reported herein. cis-[Pt(II)(NH3)2(malSAHA-2H)] was found to have marginally lower cytotoxicity against a panel of cancer cell lines as compared to cisplatin and SAHA. cis-[Pt(II)(NH3)2(malSAHA-2H)] was also found to accumulate better in cancer cells but bind DNA less readily as compared to cisplatin. DNA binding experiments indicated that cis-[Pt(II)(NH3)2(malSAHA-2H)] bound DNA more effectively in cellulo as compared to in cell-free media. Activation of the Pt-HDACi conjugate was therefore investigated. The binding of cis-[Pt(II)(NH3)2(malSAHA-2H)] to DNA was found to be enhanced by the presence of thiol-containing molecules such as glutathione and thiourea, and activation occurred in cytosolic but not nuclear extract of human cancer cells. The activity of cis-[Pt(NH3)2(malSAHA-2H)] as a HDAC inhibitor was also examined; the conjugate exhibited no inhibition of HDAC activity in CH1 cells. In light of these results, novel Pt-HDACi conjugates are currently being developed, with particular emphasis, through subtle structural modifications, on enhancing the rate of DNA binding and enhancing HDAC inhibitory activity.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/intoxicação , Compostos Organoplatínicos/farmacologia , Linhagem Celular Tumoral , DNA/metabolismo , Humanos , Vorinostat
7.
Chem Res Toxicol ; 25(5): 1099-111, 2012 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-22420335

RESUMO

The Pt(IV) diazido complex trans,trans,trans-[Pt(N(3))(2)(OH)(2)(pyridine)(2)] (1) is unreactive in the dark but is cytotoxic when photoactivated by UVA and visible light. We have shown that 1 when photoactivated accumulates in tumor cells and binds strongly to nuclear DNA under conditions in which it is toxic to tumor cells. The nature of the DNA adducts, including conformational alterations, induced by photoactivated 1 are distinctly different from those produced in DNA by conventional cisplatin or transplatin. In addition, the observation that major DNA adducts of photoactivated 1 are able to efficiently stall RNA polymerase II more efficiently than cisplatin suggests that transcription inhibition may contribute to the cytotoxicity levels observed for photoactivated 1. Hence, DNA adducts of 1 could trigger a number of downstream cellular effects different from those triggered in cancer cells by DNA adducts of cisplatin. This might lead to the therapeutic effects that could radically improve chemotherapy by platinum complexes. The findings of the present work help to explain the different cytotoxic effects of photoactivated 1 and conventional cisplatin and thereby provide new insights into mechanisms associated with the antitumor effects of platinum complexes photoactivated by UVA and visible light.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Adutos de DNA/metabolismo , Compostos Organoplatínicos/química , Compostos Organoplatínicos/farmacologia , Piridinas/química , Piridinas/farmacologia , Animais , Azidas/química , Azidas/farmacologia , Bovinos , Linhagem Celular Tumoral , DNA/metabolismo , Adutos de DNA/química , Feminino , Humanos , Luz , Neoplasias Ovarianas/tratamento farmacológico , Tioureia/metabolismo , Raios Ultravioleta
8.
Chem Res Toxicol ; 25(2): 500-9, 2012 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-22250642

RESUMO

The cellular and molecular pharmacology of the new class of anticancer drugs, in which the CDK inhibitor bohemine and its analogues are coordinated to Pt(II) to form cisplatin derivatives, was investigated. The results revealed the unique anticancer profile of a cisplatin-derived platinum(II) dichlorido complex involving N(7)-coordinated bohemine (C1). Although the IC(50) values were ∼6-fold higher for C1 than for cisplatin in cisplatin-sensitive tumor cells, the tumor cells in which C1 was also active are those which acquired resistance to cisplatin. In addition, among the novel conjugates of bohemine and its analogues with cisplatin, marked selectivity of C1 for tumor cells relative to the nontumorigenic, normal cells was observed. However, coordination of bohemine to platinum in C1 considerably reduced one of the dual functionalities anticipated to be effective after C1 reaches the nucleus. Further studies performed in the cells with wt p53 status show differences between cisplatin and C1 at the level of cell cycle regulation. Impedance-based real-time monitoring of the effects of C1 and cisplatin on cell growth supported the thesis that critical differences exist in the rate and mechanisms of cell kill caused by the two agents and that C1 was a more potent inducer of apoptosis and/or necrosis than cisplatin. The results also showed that the distinct differences in cell killing observed for C1 and cisplatin might be associated with processes at the DNA level. The DNA binding experiments carried out in a cell-free medium demonstrated that modification reactions resulting in the irreversible coordination of C1 to DNA were slower than that of cisplatin. Transcription mapping experiments and determination of interstrand cross-linking efficiency of C1 suggested that several aspects of DNA binding mode of C1 and cisplatin were similar. It was concluded that C1 remains a promising prototype of compounds for the generation of novel drug candidates with cytotoxicity profiles different from those of the platinum drugs currently in use.


Assuntos
Antineoplásicos/farmacologia , Proteína Quinase CDC2/antagonistas & inibidores , Cisplatino/análogos & derivados , Cisplatino/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Animais , Células CHO , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , DNA/metabolismo , Humanos
9.
Mol Pharm ; 8(3): 949-57, 2011 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-21480648

RESUMO

The new monofunctional Ru(II)-arene complex [(η6-arene)Ru(II)(en)Cl]+, where en = 1,2-diaminoethane and the arene is para-terphenyl (complex 1) exhibits promising cytotoxic effects in human tumor cells including those resistant to conventional cisplatin (J. Med. Chem.2008, 51, 5310). The present study is focused on the cellular pharmacology of 1 to elucidate more deeply the mechanisms underlying its antitumor effects. We have identified several cellular mechanisms induced by 1 in human ovarian carcinoma cells, including inhibition of DNA synthesis, overexpression and activation of p53, expression of proapoptotic proteins p21(WAF1) and Bax, G0/G1 arrest, and nuclear fragmentation as a result of apoptotic, and, to a much lower extent, also necrotic processes. Thus, 1 inhibits growth of the cancer cells through induction of apoptotic cell death and G0/G1 cell cycle arrest. Further investigations have shown that 1 induces apoptosis by regulating the expression of Bcl-2 family proteins. There were significant differences in cellular responses to the treatment with 1 and with conventional cisplatin, particularly in the kinetics and the extent of these responses. In addition, the distinct p53 activation profile of 1 compared with cisplatin provides an explanation for the activity of this ruthenium drug against cisplatin-resistant cells. Hence complex 1 may provide an alternative therapy in patients with acquired cisplatin resistance, particularly with respect to its very low mutagenicity and different mode of action compared to platinum antitumor drugs in clinical use.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Cisplatino/química , Cisplatino/farmacologia , Rutênio/química , Rutênio/farmacologia , Compostos de Terfenil/química , Compostos de Terfenil/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Proteína Supressora de Tumor p53/metabolismo
10.
J Inorg Biochem ; 105(5): 652-62, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21450268

RESUMO

The photocytotoxicity and photobiochemical properties of the new complex trans,trans,trans-[Pt(N(3))(2)(OH)(2)(NH(3))(piperidine)] (5) are compared with its analogue containing the less basic and less lipophilic ligand pyridine (4). The log P (n-octanol/water) values were of -1.16 and -1.84 for the piperidine and pyridine complexes, respectively, confirmed that piperidine increases the hydrophobicity of the complex. Density Functional Theory (DFT) and time-dependent density functional theory (TDDFT) calculations indicate that 5 has accessible singlet and triplet states which can promote ligand dissociation when populated by both UVA and visible white light. When activated by UVA or white light, both compounds showed similar cytotoxic potencies in various human cancer cell lines although their selectivity was different. The time needed to reach similar antiproliferative activity was noticeably decreased by introducing the piperidine ligand. Neither compound showed cross-resistance in three oxoplatin-resistant cell lines. Furthermore, both compounds showed similar anticlonogenic activity when activated by UVA radiation. Interactions of the light-activated complexes with DNA showed similar kinetics and levels of DNA platination and similar levels of DNA interstrand cross-linking (ca. 5%). Also the ability to unwind double stranded DNA were comparable for the piperidine analogue (24°, respectively), while the piperidine complex showed higher potency in changing the conformation of DNA, as measured in an ethidium bromide binding assay. These results indicate that the nature of the heterocyclic nitrogen ligand can have subtle influences on both the phototoxicity and photobiochemistry of this class of photochemotherapeutic agents.


Assuntos
Reagentes de Ligações Cruzadas/química , DNA/química , Compostos Organoplatínicos/química , Piperidinas/química , Piridinas/química , 1-Octanol/química , 1-Octanol/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Cisplatino/química , Cisplatino/metabolismo , Reagentes de Ligações Cruzadas/metabolismo , DNA/metabolismo , Adutos de DNA , Etídio/química , Etídio/metabolismo , Humanos , Ligantes , Luz , Compostos Organoplatínicos/metabolismo , Compostos Organoplatínicos/efeitos da radiação , Compostos Organoplatínicos/toxicidade
11.
Biochem Pharmacol ; 80(3): 344-51, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20399754

RESUMO

The primary objective was to understand more deeply the molecular mechanism underlying different antitumor effects of dinuclear Pt(II) complexes containing aromatic linkers of different length, {[cis-Pt(NH(3))(2)Cl](2)(4,4'-methylenedianiline)}(2+) (1) and {[cis-Pt(NH(3))(2)Cl](2)(alpha,alpha'-diamino-p-xylene)}(2+) (2). These complexes belong to a new generation of promising polynuclear platinum drugs resistant to decomposition by sulfur nucleophiles which hampers clinical use of bifunctional polynuclear trans Pt(II) complexes hitherto tested. Results obtained with the aid of methods of molecular biophysics and pharmacology reveal differences and new details of DNA modifications by 1 and 2 and recognition of these modifications by cellular components. The results indicate that the unique properties of DNA interstrand cross-links of this class of polynuclear platinum complexes and recognition of these cross-links may play a prevalent role in antitumor effects of these metallodrugs. Moreover, the results show for the first time a strong specific recognition and binding of high-mobility-group-domain proteins, which are known to modulate antitumor effects of clinically used platinum drugs, to DNA modified by a polynuclear platinum complex.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Cisplatino/química , Cisplatino/farmacologia , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/farmacologia , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Cisplatino/metabolismo , Reagentes de Ligações Cruzadas/metabolismo , Adutos de DNA/química , Adutos de DNA/metabolismo , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Células HeLa , Humanos
12.
Biochem Pharmacol ; 79(2): 112-21, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19682435

RESUMO

Reported herein is a detailed biochemical and molecular biophysics study of the molecular mechanism of action of antitumor dinuclear Pt(II) complex [{PtCl(DACH)}(2)-mu-Y](4+) [DACH=1,2-diaminocyclohexane, Y=H(2)N(CH(2))(6)NH(2)(CH(2))(2)NH(2)(CH(2))(6)NH(2)] (complex 1). This new, long-chain bifunctional dinuclear Pt(II) complex is resistant to metabolic decomposition by sulfur-containing nucleophiles. The results show that DNA adducts of 1 can largely escape repair and yet inhibit very effectively transcription so that they should persist longer than those of conventional cisplatin. Hence, they could trigger a number of downstream cellular effects different from those triggered in cancer cells by DNA adducts of cisplatin. This might lead to the therapeutic effects that could radically improve chemotherapy by platinum complexes. In addition, the findings of the present work make new insights into mechanisms associated with antitumor effects of dinuclear/trinuclear Pt(II) complexes possible.


Assuntos
Antineoplásicos/farmacologia , DNA/química , Conformação de Ácido Nucleico , Compostos Organoplatínicos/farmacologia , Enxofre/química , Antineoplásicos/química , Sequência de Bases , Sistema Livre de Células , Reparo do DNA , Fluorescência , Glutationa/química , Dados de Sequência Molecular , Compostos Organoplatínicos/química
13.
Biochem Pharmacol ; 79(4): 552-64, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19782655

RESUMO

Earlier studies have described promising antitumor activity of a large-ring chelate complex [PtCl(2)(cis-1,4-DACH)] (DACH=diaminocyclohexane). Encouraging antitumor activity of this analogue of cisplatin prompted us to perform studies focused on the mechanistic basis of pharmacological effects of this complex. Four early steps in the mechanism of biological activity of cisplatin have been delineated: cell entry, reactions with sulfur-containing compounds, platinum-DNA binding along with processing platinated DNA by proteins (enzymes) and DNA repair. Here, we describe comparative experiments (involving also cisplatin) revealing: (i) improved cytotoxicity (3.4-5.4-fold) of [PtCl(2)(cis-1,4-DACH)] in human tumor ovarian cell lines; (ii) enhanced cellular uptake (approximately 1.5-fold) of [PtCl(2)(cis-1,4-DACH)]; (iii) somewhat enhanced rate of reactions of [PtCl(2)(cis-1,4-DACH)] with glutathione (approximately 1.5-fold), but a similar rate of reactions with metallothionenin-2; (iv) enhanced rate of DNA binding of [PtCl(2)(cis-1,4-DACH)] in cell-free media (approximately 2-fold); (v) similar sequence preference of DNA binding of [PtCl(2)(cis-1,4-DACH)] in cell-free media; (vi) identical DNA interstrand cross-linking efficiency (6%); (vii) similar bending (32 degrees) and enhanced local unwinding (approximately 1.5-fold) induced in DNA by the major 1,2-GG-intrastrand cross-link; (viii) markedly enhanced inhibiting effects of DNA adducts of [PtCl(2)(cis-1,4-DACH)] on processivity of DNA polymerase; and (ix) a slightly lower efficiency of DNA repair systems to remove the adducts of [PtCl(2)(cis-1,4-DACH)] from DNA.


Assuntos
Antineoplásicos/metabolismo , Cicloexilaminas/metabolismo , Reparo do DNA/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Glutationa/metabolismo , Compostos Organoplatínicos/metabolismo , Compostos Organoplatínicos/toxicidade , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Bovinos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cicloexilaminas/química , Cicloexilaminas/farmacologia , Reparo do DNA/fisiologia , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/toxicidade , Células HeLa , Humanos , Ligantes , Compostos de Platina/química , Compostos de Platina/metabolismo , Compostos de Platina/toxicidade , Coelhos
14.
J Med Chem ; 51(17): 5310-9, 2008 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-18702458

RESUMO

We have compared the cancer cell cytotoxicity, cell uptake, and DNA binding properties of the isomeric terphenyl complexes [(eta(6)-arene)Ru(en)Cl](+), where the arene is ortho- (2), meta- (3), or para-terphenyl (1) (o-, m-, or p-terp). Complex 1, the X-ray crystal structure of which confirms that it has the classical "piano-stool" geometry, has a similar potency to cisplatin but is not cross-resistant and has a much higher activity than 2 or 3. The extent of Ru uptake into A2780 or A2780cis cells does not correlate with potency. Complex 1 binds to DNA rapidly and quantitatively, preferentially to guanine residues, and causes significant DNA unwinding. Circular and linear dichroism, competitive binding experiments with ethidium bromide, DNA melting, and surface-enhanced Raman spectroscopic data are consistent with combined intercalative and monofunctional (coordination) binding mode of complex 1. This unusual DNA binding mode may therefore make a major contribution to the high potency of complex 1.


Assuntos
Antineoplásicos/química , DNA/efeitos dos fármacos , Compostos Organometálicos/farmacocinética , Rutênio , Compostos de Terfenil/farmacologia , Animais , Antineoplásicos/farmacologia , Sítios de Ligação , Linhagem Celular Tumoral , Cristalografia por Raios X , DNA/química , DNA/metabolismo , DNA Super-Helicoidal , Guanina/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Desnaturação de Ácido Nucleico , Compostos Organometálicos/metabolismo , Compostos Organometálicos/farmacologia , Análise Espectral , Relação Estrutura-Atividade , Compostos de Terfenil/química
15.
Sensors (Basel) ; 8(1): 429-444, 2008 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-27879715

RESUMO

Using a paraffin impregnated graphite electrode (PIGE) and mercury-modifiedpyrolytic graphite electrode with basal orientation (Hg-PGEb) copper(II) and Cu(II)-DNApurine base solutions have been studied by cyclic (CV) and linear sweep voltammetry(LSV) in connection with elimination voltammetry with linear scan (EVLS). In chlorideand bromide solutions (pH 6), the redox process of Cu(II) proceeded on PIGE with twocathodic and two anodic potentially separated signals. According to the eliminationfunction E4, the first cathodic peak corresponds to the reduction Cu(II) e⁻ → Cu(I) withthe possibility of fast disproportionation 2Cu(I) → Cu(II) Cu(0). The E4 of the secondcathodic peak signalized an electrode process controlled by a surface reaction. Theelectrode system of Cu(II) on Hg-PGEb in borate buffer (pH 9.2) was characterized by onecathodic and one anodic peak. Anodic stripping voltammetry (ASV) on PIGE and cathodicstripping voltammetry (CSV) on Hg-PGEb were carried out at potentials where thereduction of copper ions took place and Cu(I)-purine complexes were formed. By usingASV and CSV in combination with EVLS, the sensitivity of Cu(I)-purine complexdetection was enhanced relative to either ASV or CSV alone, resulting in higher peakcurrents of more than one order of magnitude. The statistical treatment of CE data wasused to determine the reproducibility of measurements. Our results show that EVLS inconnection with the stripping procedure is useful for both qualitative and quantitativemicroanalysis of purine derivatives and can also reveal details of studied electrodeprocesses.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...