Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 188: 114523, 2021 06.
Article in English | MEDLINE | ID: mdl-33741331

ABSTRACT

Triple-negative breast cancer (TNBC) shares the molecular features facilitating epithelial-to-mesenchymal transition (EMT), which contributed to tumor invasion and metastasis. A platinum(IV) conjugate ketoplatin deriving from FDA-approved drugs cisplatin and ketoprofen was designed and prepared to enhance antitumor activity and suppress EMT in TNBC via positive impact on inflammatory microenvironment by modulating COX-2 signal. As a prodrug, ketoplatin afforded 50.26-fold higher cytotoxicity than cisplatin against TNBC mesenchymal-stem cell-like MDA-MB-231 cells, partly attributing to its dramatic increase of cellular uptake and DNA damage. More importantly, EMT progress in MDA-MB-231 was markedly restrained by ketoplatin, resulting from the suppression of vimentin and N-cadherin mediated by down-regulated COX-2. Further in vivo investigation exhibited that ketoplatin effectively inhibited tumor growth and reduced systemic toxicity compared to cisplatin. Overall, ketoplatin possessed high antitumor activity and low toxicity against TNBC MDA-MB-231 in vitro and in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/analogs & derivatives , Ketoprofen/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment/drug effects , A549 Cells , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antineoplastic Agents/chemistry , Cisplatin/administration & dosage , Cisplatin/chemistry , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Female , HeLa Cells , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Ketoprofen/analogs & derivatives , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Treatment Outcome , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/physiology , Xenograft Model Antitumor Assays/methods
2.
J Med Chem ; 63(11): 6096-6106, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32401032

ABSTRACT

Multitargeted therapy could rectify various oncogenic pathways to block tumorigenesis and progression. The combination of endocrine-, immune-, and chemotherapy might exert a highly synergistic effect against certain tumors. Herein, a series of smart Pt(IV) prodrugs 3-6, named Melatplatin, were rationally designed not only to multitarget DNA, MT1, and estrogen receptor (ER) but also to activate immune response. Melatplatin, conjugating first-line chemotherapeutic Pt drugs with human endogenous melatonin (MT), significantly enhanced drug efficacy especially in ER high-expression (ER+) cells, among which 3 presented the most potent cytotoxicity toward ER+ MCF-7 with nanomolar IC50 values 100-fold lower than cisplatin. Melatplatin could bind well to melatonin receptor (MT1) according to molecular docking. Besides, 3 evidently increased intracellular accumulation and DNA damage, upregulated γH2AX and P53, and silenced NF-κB to induce massive apoptosis. Most strikingly, 3 effectively inhibited tumor growth and attenuated systemic toxicity compared to cisplatin in vivo, promoting lymphocyte proliferation in spleen to achieve immune modulation.


Subject(s)
Antineoplastic Agents/chemistry , Platinum/chemistry , Prodrugs/chemistry , Receptor, Melatonin, MT1/metabolism , Receptors, Estrogen/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Binding Sites , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Humans , Immune System/drug effects , Immune System/metabolism , Mice , Mice, Nude , Molecular Docking Simulation , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Prodrugs/pharmacology , Prodrugs/therapeutic use , Protein Structure, Tertiary , Receptor, Melatonin, MT1/chemistry , Receptors, Estrogen/chemistry
3.
J Med Chem ; 62(9): 4543-4554, 2019 05 09.
Article in English | MEDLINE | ID: mdl-31002510

ABSTRACT

As FDA-approved chemotherapeutic agents, cisplatin, oxaliplatin, and 5-fluorouracil are widely used in clinic but limited by severe side-effects. To ameliorate their respective defects, a series of "dual-prodrug" by linking oxoplatin and 5-FU were designed and synthesized. The assembled compounds 10-17, named Fuplatin, exhibited much higher cytotoxicity against the tested cancer cells while lower cytotoxicity toward the human normal lung cells than free drugs or their combinations. Among them, 14 enhanced cellular accumulation with 62- and 825-fold amount of oxaliplatin and 8 at 9 h, respectively, significantly induced DNA damage and cell apoptosis, and inhibited migration and invasion in HCT-116 cells. Compound 14 arrested the cell cycle at S and G2 phases and up-regulated thymidylate synthase and p53, consistent with the results of the combination, suggesting 14 adopted a collaborative mode of 5-FU and oxaliplatin to kill cancer cells. In vivo, compound 14 showed high antitumor effect and no observable toxicity in NOD/SCID mice bearing HCT-116 tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/analogs & derivatives , Fluorouracil/analogs & derivatives , Fluorouracil/therapeutic use , Prodrugs/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cisplatin/chemical synthesis , Cisplatin/pharmacology , Cisplatin/therapeutic use , DNA Damage/drug effects , Drug Synergism , Fluorouracil/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Mice, Inbred NOD , Mice, SCID , Prodrugs/chemical synthesis , Prodrugs/pharmacology , S Phase Cell Cycle Checkpoints/drug effects , Thymidylate Synthase/metabolism , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
4.
Eur J Med Chem ; 167: 377-387, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30784875

ABSTRACT

The great interest in epithelial-to-mesenchymal transition (EMT) programme lies in its association with process of metastasis and invasion, which is a crucial cause of cancer-related death. Herein, we designed and reported three new NSAID-Pt(IV) prodrugs, taking Non-Steroid Anti-Inflammatory Drugs (NSAIDs) to disrupt EMT programme and assist genotoxic platinum-based drugs as a cytotoxicity booster, to offer a class of potential anticarcinogens with a multi-functional action mechanism. The NSAID-Pt(IV) prodrugs, especially Eto-Pt(IV), highly enhanced cellular uptake with amount up to 42-fold at 3 h compared with CDDP, and greatly increased DNA damage and cell apoptosis, showing much higher cytotoxicity than cisplatin in the tested cancer cells even in A549/cis cells. Among of them, Eto-Pt(IV) and Car-Pt(IV) exhibited more excellent activity than Sul-Pt(IV), arising from their reduction-labile and favorable lipophilicity. Most strikingly, Eto-Pt(IV) markedly inhibited metastasis and invasion of MCF-7 cells, owing to its COX-2 suppression that down-regulated active MMP-2, vimentin protein and up-regulated E-cadherin. In vivo, Eto-Pt(IV) displayed potent antitumor activity and no observable toxicity in BALB/c nude mice bearing MCF-7 tumors.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antineoplastic Agents/chemistry , Drug Design , Platinum Compounds/chemistry , Prodrugs/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage/drug effects , Heterografts , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis/drug therapy , Neoplasms/drug therapy , Neoplasms/pathology , Prodrugs/pharmacology
5.
Eur J Med Chem ; 157: 1292-1299, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30195239

ABSTRACT

Modification of platinum (II) into lipophilic platinum (IV) compounds by introducing biologically active molecules were widely employed to develop new platinum-based prodrugs in the past decade. In this paper, two chlorambucil platinum (IV) complexes, CLB-Pt and CLB-Pt-CLB, were synthesized and displayed very potent antiproliferative activity against all the tested cancer cell lines, such as A549, HeLa and MCF-7, especially to treat the well-known refractory triple-negative breast cancer. CLB-Pt-CLB significantly improved cell-killing effect in triple-negative subtype MDA-MB-231 cells, and showed much stronger cytotoxicity than either monotherapy or combination of cisplatin and chlorambucil. CLB-Pt-CLB prodrug entered cells in dramatically increased amount compared with cisplatin and enhanced DNA damage, inducing cancer cell apoptosis. It exhibited high anticancer activity and no observable toxicity in BALB/c nude mice bearing MDA-MB-231 tumors. The chlorambucil moiety not only greatly assisted the passive diffusion of CLB-Pt-CLB into cells, but also produced the synergism with cisplatin in targeting DNA.


Subject(s)
Antineoplastic Agents/pharmacology , Chlorambucil/pharmacology , Organoplatinum Compounds/pharmacology , Prodrugs/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Chlorambucil/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Nude , Molecular Structure , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Prodrugs/chemical synthesis , Prodrugs/chemistry , Structure-Activity Relationship , Triple Negative Breast Neoplasms/pathology , Wound Healing/drug effects
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 195: 157-164, 2018 Apr 15.
Article in English | MEDLINE | ID: mdl-29414573

ABSTRACT

A new aluminum ion fluorescent probe (4-(diethylamino)-2-hydroxybenzylidene)isoquinoline-1-carbohydrazide (HL1) has been conveniently synthesized and characterized. HL1 exhibited a highly selective and pronounced enhancement for Al3+ in the fluorescence emission over other common cations by forming a 2:1 complex, with a recognition mechanism based on excited-state intramolecular proton transfer (ESIPT) and intramolecular charge transfer (ICT). The strong fluorescent emission can be observed even at ppm level concentration of the probe in the presence of Al3+ with 41 fold intensity enhancement at 545 nm. HL1 displays good linear relationship with Al3+ in the low concentration and the limit of detection is 8.08 × 10-8 mol/L. Similar molecules with different substituents on salicylaldehyde phenyl ring were synthesized for studying the structure-activity relationship. Density-functional theory (DFT) calculations are in agreement with the proposed mechanism. It is confirmed that HL1 could be used to detect Al3+ ions in real sample by fluorescence spectrometry and Al3+ ions in cells by bioimaging.

7.
Am J Cancer Res ; 6(6): 1271-86, 2016.
Article in English | MEDLINE | ID: mdl-27429843

ABSTRACT

In this study, a novel adamantyl nitroxide derivative was synthesized and its antitumor activities in vitro and in vivo were investigated. The adamantyl nitroxide derivative 4 displayed a potent anticancer activity against all the tested human hepatoma cells, especially with IC50 of 68.1 µM in Bel-7404 cells, compared to the positive control 5-FU (IC50=607.7 µM). The significant inhibition of cell growth was also observed in xenograft mouse model, with low toxicity. Compound 4 suppressed the cell migration and invasion, induced the G2/M phase arrest. Further mechanistic studies revealed that compound 4 induced cell death, which was accompanied with damaging mitochondria, increasing the generation of intracellular reactive oxygen species, cleavages of caspase-9 and caspase-3, as well as activations of Bax and Bcl-2. These results confirmed that adamantyl nitroxide derivative exhibited selective antitumor activities via mitochondrial apoptosis pathway in Bel-7404 cells, and would be a potential anticancer agent for liver cancer.

8.
J Inorg Biochem ; 146: 52-60, 2015 May.
Article in English | MEDLINE | ID: mdl-25771239

ABSTRACT

Three novel structurally associated copper(II) complexes [Cu(II)(SalCl-Gly)(H2O)2] (1), [Cu(II)(SalCl-Ala)(H2O)] (2) and [Cu(II)(SalCl-Gly)(bipy)]·0.5H2O (3) (SalCl-Gly=5-chloro-2-hydroxybenzylidene-glycine, SalCl-Ala=5-chloro-2-hydroxybenzylidene-alanine, bipy=2,2'-bipyridine) have been synthesized and characterized by X-ray crystallography, elemental analysis, IR and fluorescence spectroscopy. Single-crystal diffraction reveals that complex 1 is an infinite 1D zigzag chain in which SalCl-Gly serves as both a chelating and a bridging ligand, while complexes 2 and 3 are mononuclear. Cu(II) ions in complexes 1-3 exhibit distorted quasi-hexacoordinated octahedral, tetracoordinated square planar, and pentacoordinated square pyramid geometry, respectively. Their interactions with calf thymus DNA (CT-DNA) have been investigated by viscosity measurements and fluorescence spectroscopy. The apparent binding constant (Kapp) values for 1-3 are 1.02×10(5), 0.98×10(5) and 1.57×10(5)M(-1), respectively. All complexes displayed efficient oxidative cleavage of supercoiled DNA in the presence of H2O2. Complex 2, whose ligand can be regarded as a methyl-modification of SalCl-Gly of 1, showed a reduced DNA cleavage activity and a little-changed DNA-binding ability compared with 1. While attaching a 2,2'-bipyridine group to 1, the resulting complex 3 was conferred an enhanced intercalation into DNA. Moreover, cytotoxicity studies of three complexes against HepG-2 (human liver hepatocellular carcinoma) and NCI-H460 (human large-cell lung carcinoma) cells indicated that, thereto, complex 3 possessed the highest inhibition on viability of tested cells.


Subject(s)
Aldehydes/chemistry , Amino Acids/chemistry , Copper/chemistry , Organometallic Compounds/chemical synthesis , Schiff Bases/chemistry , DNA/chemistry , Hep G2 Cells , Humans , Organometallic Compounds/toxicity
9.
J Inorg Biochem ; 136: 13-23, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24690556

ABSTRACT

Four novel thiosemicarbazone metal complexes, [Cu(Am4M)(OAc)]·H2O (1), [Zn(HAm4M)Cl2] (2), [Zn2(Am4M)2Br2] (3) and [Zn2(Am4M)2(OAc)2]·2MeOH (4) [HAm4M=(Z)-2-(amino(pyridin-2-yl)methylene)-N-methylhydrazinecarbothioamide], have been synthesized and characterized by X-ray crystallography, elemental analysis, ESI-MS and IR. X-ray analysis revealed that complexes 1 and 2 are mononuclear, which possess residual coordination sites for Cu(II) ion in 1 and good leaving groups (Cl(-)) for Zn(II) ion in 2. Both 3 and 4 displayed dinuclear units, in which the metal atoms are doubly bridged by S atoms of two Am4M(-) ligands in 3 and by two acetate ions in bi- and mono-dentate forms, respectively, in 4. Their antiproliferative activities on human epithelial cervical cancer cell line (HeLa), human liver hepatocellular carcinoma cell line (HepG-2) and human gastric cancer cell line (SGC-7901) were screened. Inspiringly, IC50 value (11.2±0.9 µM) of complex 1 against HepG-2 cells was nearly 0.5 fold of that against human hepatic cell lines LO2, showing a lower toxicity to human liver cells. Additionally, it displayed a stronger inhibition on the viability of HepG-2 cells than cisplatin (IC50=25±3.1 µM), suggesting complex 1 might be a potential high efficient antitumor agent. Furthermore, fluorescence microscopic observation and flow cytometric analysis revealed that complex 1 could significantly suppress HepG-2 cell viability and induce apoptosis. Several indexes, such as DNA cleavage, reactive oxygen species (ROS) generation, comet assay and cell cycle analysis indicated that the antitumor mechanism of complex 1 on HepG-2 cells might be via ROS-triggered apoptosis pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Coordination Complexes/pharmacology , Copper/chemistry , Thiosemicarbazones/chemistry , Zinc/chemistry , Antineoplastic Agents/chemical synthesis , Cell Cycle/drug effects , Cell Proliferation , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Crystallography, X-Ray , DNA Cleavage , Drug Screening Assays, Antitumor , HeLa Cells , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Plasmids/chemistry , Reactive Oxygen Species
10.
Biometals ; 27(1): 155-72, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24368745

ABSTRACT

Exploring novel chemotherapeutic agents is a great challenge in cancer medicine. To that end, 2-substituted benzimidazole copper(II) complex, [Cu(BMA)Cl2]·(CH3OH) (1) [BMA = N,N'-bis(benzimidazol-2-yl-methyl)amine], was synthesized and its cytotoxicity was characterized. The interaction between complex 1 and calf thymus DNA was detected by spectroscopy methods. The binding constant (K b = 1.24 × 10(4 )M(-1)) and the apparent binding constant (K app = 6.67 × 10(6 )M(-1)) of 1 indicated its moderate DNA affinity. Complex 1 induced single strand breaks of pUC19 plasmid DNA in the presence of H2O2 through an oxidative pathway. Cytotoxicity studies proved that complex 1 could inhibit the proliferation of human cervical carcinoma cell line HeLa in both time- and dose-dependent manners. The results of nuclei staining by Hoechst 33342 and alkaline single-cell gel electrophoresis proved that complex 1 caused cellular DNA damage in HeLa cells. Furthermore, treatment of HeLa cells with 1 resulted in S-phase arrest, loss of mitochondrial potential, and up-regulation of caspase-3 and -9 in HeLa cells, suggesting that complex 1 was capable of inducing apoptosis in cancer cells through the intrinsic mitochondrial pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzimidazoles/chemistry , Copper/chemistry , DNA Damage , DNA/drug effects , Organometallic Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cattle , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , HeLa Cells , Humans , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Structure-Activity Relationship , Time Factors , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
11.
J Inorg Biochem ; 117: 1-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23073509

ABSTRACT

In this study, we investigated the newly synthesized Schiff base copper(II) complex, [Cu(II)(5-Cl-pap)(OAc)(H(2)O)]·2H(2)O (1) (5-Cl-pap=N-2-pyridiylmethylidene-2-hydroxy-5-chloro-phenylamine), inducing growth inhibition and apoptosis in human breast cancer cell line MCF-7 and its potential antitumor mechanism. The results of cytotoxicity research, fluorescence microscopic observation and flow cytometric analysis revealed that complex 1 could significantly suppress MCF-7 cell viability and induce apoptosis. Comet assay indicated that severe DNA fragmentation in MCF-7 cells was induced after treatment with complex 1. Flow cytometric analysis showed that the antitumor effect of complex 1 on MCF-7 cells was associated with the cell cycle arrest. In addition, atomic absorption analyses displayed that complex 1 caused a rapid increase of intracellular copper uptake in MCF-7 cells in a time-dependent manner. The present work suggested that the antitumor mechanism of complex 1 on MCF-7 cells might be via the mitochondrial pathway, based on the up-regulated expression of Bax and activation of caspase-9 and caspase-3.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Coordination Complexes/pharmacology , Copper , Mitochondria/metabolism , Schiff Bases/pharmacology , Antineoplastic Agents/chemistry , Breast Neoplasms , Caspase 3/metabolism , Caspase 9/metabolism , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Female , Humans , MCF-7 Cells , Mitochondria/drug effects , Schiff Bases/chemistry , Up-Regulation
12.
Dalton Trans ; 41(39): 12220-32, 2012 Oct 21.
Article in English | MEDLINE | ID: mdl-22930131

ABSTRACT

Three new zinc(II) complexes: [Zn(2)(L(1))(2)Cl(2)](ClO(4))(2)·C(2)H(5)OH (1) and [ZnL(2)X(4)]·2CH(3)CN (X = Br for 2, Cl for 3), utilizing two new and interrelated di-nucleating polypyridyl ligands (L(1), L(2)), have been synthesized and characterized by using various physico-chemical techniques. The interactions of three complexes with CT-DNA have been explored by using absorption, emission and CD spectral methods, which reveal that three complexes bind to CT-DNA by partial intercalation binding modes. Notably, in the presence of H(2)O(2) as a revulsant or an activator, the cleavage abilities of all complexes are obviously enhanced. The hydrolytic mechanism was demonstrated by adding standard radical scavengers and anaerobic reaction. Further, the protein binding ability has been monitored by quenching of tryptophan emission in the presence of complexes using BSA as a model protein. The quenching mechanisms of BSA by the complexes are static procedures. In addition, the in vitro cytotoxicity of the complexes on three human tumor cells lines (HeLa, MCF-7 and RL952) and the apoptosis-inducing activity of were assessed by MTT, Clonogenic assay, Hoechst 33342 staining, Cell cycle and Annexin V binding experiments.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Zinc/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemistry , Crystallography, X-Ray , DNA/metabolism , DNA Cleavage , Humans , Hydrogen Peroxide/pharmacology , Ligands , Molecular Structure , Protein Binding , Serum Albumin, Bovine/metabolism , Zinc/chemistry
13.
J Inorg Biochem ; 105(5): 728-37, 2011 May.
Article in English | MEDLINE | ID: mdl-21463568

ABSTRACT

A new cytotoxic copper(II) complex with Schiff base ligand [Cu(II)(5-Cl-pap)(OAc)(H(2)O)]·2H(2)O (1) (5-Cl-pap=N-2-pyridiylmethylidene-2-hydroxy-5-chloro-phenylamine), was synthesized and structurally characterized by X-ray diffraction. Single-crystal analysis revealed that the copper atom shows a 4+1 pyramidal coordination, a water oxygen appears in the apical position, and three of the basal positions are occupied by the NNO tridentate ligand and the fourth by an acetate oxygen. The interaction of Schiff base copper(II) complex 1 with DNA was investigated by UV-visible spectra, fluorescence spectra and agarose gel electrophoresis. The apparent binding constant (K(app)) value of 6.40×10(5) M(-1) for 1 with DNA suggests moderate intercalative binding mode. This copper(II) complex displayed efficient oxidative cleavage of supercoiled DNA, which might indicate that the underlying mechanism involve hydroxyl radical, singlet oxygen-like species, and hydrogen peroxide as reactive oxygen species. In addition, our present work showed the antitumor effect of 1 on cell cycle and apoptosis. Flow cytometric analysis revealed that HeLa cells were arrested in the S phase after treatment with 1. Fluorescence microscopic observation indicated that complex 1 can induce apoptosis of HeLa cells, whose process was mediated by intrinsic mitochondrial apoptotic pathway owing to the activation of caspase-9 and caspase-3.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Apoptosis , Coordination Complexes/chemistry , Coordination Complexes/toxicity , DNA/metabolism , Antineoplastic Agents/chemical synthesis , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Coordination Complexes/chemical synthesis , Copper/chemistry , Crystallography, X-Ray , DNA/chemistry , HeLa Cells , Humans , Ligands , Models, Molecular , Schiff Bases/chemistry , Spectrometry, Fluorescence
14.
Dalton Trans ; 39(30): 7159-66, 2010 Aug 14.
Article in English | MEDLINE | ID: mdl-20601983

ABSTRACT

The directed assembly of N,N-bis(benzimidazol-2-yl-methyl)amine (BMA) with Cu(II), Mn(II), Ni(II) and Zn(II) salts based on dicyanamide (mu(1,5)-dca) and terephthalate (mu-ta) linear bridging ligands, respectively, leads to four novel compounds: [Cu(BMA)(mu(1,5)-dca)(ClO(4))](n) (1), {[Mn(BMA)(mu(1,5)-dca)(CH(3)OH)] x ClO(4) x C(10)H(9)N(3)O x CH(3)OH}(n) (2), {[Ni(2)(BMA)(2)(mu-ta)(mu(1,5)-dca)] x ClO(4) x CH(3)OH x H(2)O}(n) (3), and {[Zn(2)(BMA)(2)(mu-ta)(mu(1,5)-dca)] x ClO(4) x CH(3)OH}(n) (4), which were characterized by single-crystal X-ray diffraction, elemental analysis, IR, fluorescence spectroscopy, and magnetic measurement. X-Ray analysis revealed that 1 and 2 are two infinite 1D coordination polymers, in which dca units serving as an end-to-end out-of-plane bridge bring about linear chains for 1 and zigzag chains for 2. Complexes 3 and 4 are similar, in which the metal atoms are bridged alternately by terephthalate and mu(1,5)-dicyanamide ligands into 1D zigzag chains. In all these complexes, each BMA ligand adopts a tridentate chelating mode to coordinate with a transition metal forming a [M(BMA)](2+) node. Different rigidity bridging ligands together with the stereochemistry and supramolecular effects of benzimidazol planes may result in the dramatic structural changes from 1D to multidimensional networks for all 1-4. Fluorescent measurements established that, in solution, complex 3 displays weak blue luminescence which originates from the BMA but is significantly red-shifted and has a much lower emission intensity, compared to the free BMA ligand. Complex 4 shows stronger luminescence than 3 and still reduces luminescence efficiency compared to the free BMA ligand. The variable-temperature magnetic susceptibility measurements (2-300 K) of 1 and 3 show the dominant weak ferromagnetic interactions between the copper(II) centers with J = 3.02 cm(-1), zJ' = -2.70 cm(-1) for 1, and the nickel(II) centers with J = 1.94 cm(-1), J'; = -0.38 cm(-1) for 3, while weak antiferromagnetic interactions between the Mn(II) centers for 2 with J = -0.27 cm(-1).


Subject(s)
Benzimidazoles/chemistry , Cyanamide/chemistry , Organometallic Compounds/chemical synthesis , Phthalic Acids/chemistry , Polymers/chemical synthesis , Copper/chemistry , Crystallography, X-Ray , Ligands , Manganese/chemistry , Models, Molecular , Nickel/chemistry , Organometallic Compounds/chemistry , Polymers/chemistry , Temperature , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...