Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 84
1.
Eur J Pharmacol ; 949: 175728, 2023 Jun 15.
Article En | MEDLINE | ID: mdl-37062501

GLUT5, a key protein encoded by the SLC2A5 gene, is involved in the uptake of fructose from the intestine. Currently, with the increased consumption of this sugar and the associated increased incidence of obesity, diabetes and cancer, GLUT5 may represent an important molecular target in the prevention and treatment of these diseases. Here, we demonstrate that overexpression of the SNAI1 and SNAI2 transcription factors in cells expressing high levels of SLC2A5 mRNA reduced SLC2A5 gene expression. Furthermore, a histone deacetylase inhibitor, trichostatin A, which induces SNAI1 and SNAI2 expression, inhibits SLC2A5/GLUT5 expression and sensitizes colon cancer cells to cisplatin and oxaliplatin. This finding might have potential relevance for the development of therapeutic treatments aimed at modulating fructose transport or genes involved in this process for use with certain cancers.


Colonic Neoplasms , Transcription Factors , Humans , Transcription Factors/metabolism , Platinum Compounds/metabolism , Fructose , Colonic Neoplasms/genetics , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Glucose Transporter Type 5
2.
Cancer Sci ; 112(11): 4655-4668, 2021 Nov.
Article En | MEDLINE | ID: mdl-34533854

Platinum-based regimens are the most widely used chemotherapy regimens, but cancer cells often develop resistance, which impedes therapy outcome for patients. Previous studies have shown that fibroblast growth factor 13 (FGF13) is associated with resistance to platinum drugs in HeLa cells. However, the mechanism and universality of this effect have not been clarified. Here, we found that FGF13 was associated with poor platinum-based chemotherapy outcomes in a variety of cancers, such as lung, endometrial, and cervical cancers, through bioinformatics analysis. We then found that FGF13 simultaneously regulates the expression and distribution of hCTR1 and ATP7A in cancer cells, causes reduced platinum influx, and promotes platinum sequestration and efflux upon cisplatin exposure. We subsequently observed that FGF13-mediated platinum resistance requires the microtubule-stabilizing effect of FGF13. Only overexpression of FGF13 with the -SMIYRQQQ- tubulin-binding domain could induce the platinum resistance effect. This phenomenon was also observed in SK-MES-1 cells, KLE cells, and 5637 cells. Our research reveals the mechanism of FGF13-induced platinum drug resistance and suggests that FGF13 can be a sensibilization target and prognostic biomarker for chemotherapy.


Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Copper Transporter 1/metabolism , Copper-Transporting ATPases/metabolism , Drug Resistance, Neoplasm , Fibroblast Growth Factors/physiology , A549 Cells , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cisplatin/metabolism , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Microtubules/drug effects , Platinum Compounds/metabolism , Platinum Compounds/pharmacology , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism
3.
Angew Chem Int Ed Engl ; 59(43): 19229-19236, 2020 10 19.
Article En | MEDLINE | ID: mdl-32662563

It is of great significance to track the platinum drugs in real time with super-resolution to elucidate their mechanism of action, such as their behavior and distribution in live cells. Such information is required for further drug development. However, it is always challenging to design platinum complexes suitable for such research. Herein, we design a luminescent building block (L) for metal complexes and a dinuclear platinum complex (Pt2 L) for super-resolution imaging. Because of its super-large Stokes shift and excellent photophysical properties, Pt2 L is capable of serving as an ideal candidate for super-resolution imaging with extremely low luminescence background and high photobleaching resistance. Moreover, upon light stimulation, a matter flux of Pt2 L escaping from autolysosomes to nucleus was observed, which represents a new transportation path. Utilizing the photoactivated escape properties, we can regulate the nuclear accessibility of Pt2 L form autolysosomes with photo-selectivity, which provides a new way to improve the targeting of platinum drugs.


Color , Lysosomes/metabolism , Platinum Compounds/chemistry , A549 Cells , Biological Transport , Cell Nucleus/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence/methods , Mitochondria/metabolism , Platinum Compounds/metabolism
4.
Hum Cell ; 33(4): 1294-1301, 2020 Oct.
Article En | MEDLINE | ID: mdl-32710259

Carcinostatic effects of combined use of hydrogen nano-bubbles (nano-H) and platinum-povidone (PVP--Pt) were examined. Hydrogen-dissolved medium was prepared by hydrogen-gas bubbling with a microporous gas-emittance-terminal into a medium in the absence or presence of PVP-Pt (nano-H, nano-H/PVP-Pt). Human esophagus-derived carcinoma cells KYSE70 were repressed for cell proliferation with nano-H/PVP-Pt more markedly than with nano-H, indicating the hydrogen-intensification for PVP-Pt-alone-carcinostasis. However, the intensified carcinostasis required co-administration of nano-H and PVP-Pt, and no intensified carcinostasis was shown in two-step separate administration of nano-H and PVP-Pt. Furthermore, hydrogen bubbling into PVP-Pt-containing medium achieved more appreciable carcinostasis than mere addition of PVP-Pt into nano-H-containing medium, indicating the potent interaction of hydrogen and PVP-Pt. The nano-H/PVP-Pt-administered human tongue-derived carcinoma cells HSC-4 were repressed for cell proliferation more markedly than pre-malignant human tongue-derived epitheliocytes DOK, concurrently with more abundant intracellular Pt-intake into HSC-4 cells than DOK as analyzed by ICP-MS. Thus, PVP-Pt is able to adsorb hydrogen nano-bubbles on Pt and applicable for cancer therapy by diminishing the side-effects to normal cells.


Antineoplastic Agents , Carcinoma/pathology , Cell Proliferation/drug effects , Drug Interactions , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Hydrogen/pharmacology , Nanoparticles , Platinum Compounds/adverse effects , Platinum Compounds/metabolism , Platinum Compounds/pharmacology , Povidone/adverse effects , Povidone/pharmacology , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology , Carcinoma/metabolism , Cell Line, Tumor , Colloids , Culture Media , Drug Combinations , Esophageal Neoplasms/drug therapy , Gases , Humans , Hydrogen/administration & dosage , Povidone/metabolism , Tongue Neoplasms/drug therapy
5.
Small ; 16(26): e2002044, 2020 07.
Article En | MEDLINE | ID: mdl-32500659

Platinum (Pt) drugs are widely used in anti-cancer treatment although many reports advocated that tumor cells could inactivate Pt drugs via glutathione-Pt (GSH-Pt) adducts formation. To date, GSH chelated Pt molecules have not been assessed in cancer treatment because GSH-Pt adducts are not capable of killing cancer cells, which is widely accepted and well followed. In this report, endogenous biothiol is utilized to precisely synthesize a GSH chelated Pt molecule (Pt6 GS4 ). This Pt6 GS4 molecule can be well taken up by aggressive triple negative breast cancer (TNBC) cells. Subsequently, its metabolites could enter nuclei to interact with DNA, finally the DNA-Pt complex triggers TNBC cell apoptosis via the p53 pathway. Impressively, high efficacy for anti-cancer treatment is achieved by Pt6 GS4 both in vitro and in vivo when compared with traditional first-line carboplatin in the same dosage. Compared with carboplatin, Pt6 GS4 keeps tumor bearing mice alive for a longer time and is non-toxic for the liver and kidneys. This work opens a route to explore polynuclear Pt compound with accurate architecture for enhancing therapeutic effects and reducing systemic toxicity.


Antineoplastic Agents , Platinum Compounds , Triple Negative Breast Neoplasms , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Glutathione/chemistry , Humans , Mice , Platinum/chemistry , Platinum Compounds/metabolism , Platinum Compounds/pharmacology , Survival Analysis , Triple Negative Breast Neoplasms/drug therapy
6.
Theranostics ; 9(8): 2158-2166, 2019.
Article En | MEDLINE | ID: mdl-31149035

Platinum complexes have been used for anti-cancer propose for decades, however, their high side effects resulting from damage to healthy cells cannot be neglected and prevent further clinical utilisation. Here, we designed a cyclometalated platinum (II) complex that can bind the endogenous nuclear factor-κB (NF-κB) protein. Employing detailed colocalization studies in co-culture cell line models, we show that by binding to NF-κB, the platinum (II) complex is capable of upregulated nuclear translocation specifically in cancer but not normal cells, thereby impairing cancer proliferation without disturbing healthy cells. In a murine tumour model, the platinum (II) complex prevents tumour growth to a greater extent than cisplatin and with considerably lower side-effects and kidney damage. Considering its weak damage to normal cells combined with high toxicity to cancer cells, this NF-κB-binding platinum complex is a potential anti-cancer candidate and acts to verify the strategy of hijacking endogenous trans-nuclear proteins to achieve cancer-cell specificity and enhance therapeutic indices.


Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , NF-kappa B p50 Subunit/metabolism , Neoplasms/therapy , Platinum Compounds/metabolism , Platinum Compounds/pharmacology , Theranostic Nanomedicine/methods , Active Transport, Cell Nucleus , Animals , Antineoplastic Agents/administration & dosage , Cell Survival/drug effects , Disease Models, Animal , Mice , Models, Biological , Neoplasm Transplantation , Platinum Compounds/administration & dosage , Protein Binding , Transplantation, Heterologous , Treatment Outcome
7.
Metallomics ; 10(7): 1003-1015, 2018 07 18.
Article En | MEDLINE | ID: mdl-29978878

The present paper deals with investigation of the interaction between selected simple structure Au(iii) ([AuCl4]-, [AuCl2(dmso)2]+, [AuCl2(bipy)]+) and Pt(ii) ([PtCl2(dmso)2]) complexes with Na/K-ATPase as the target enzyme, using an experimental and theoretical approach. Reaction stoichiometries and binding constants for these enzyme/complex systems were determined, while kinetic measurements were used in order to reveal the type of inhibition. Based on the results obtained by quantum mechanical calculations (electrostatic surface potential (ESP), volume and surface of the complexes) the nature of the investigated complexes was characterized. By using the solvent accessible surface area (SASA) applied on specific inhibitory sites (ion channel and intracellular domains) the nature of these sites was described. Docking studies were used to determine the theoretical probability of the non-covalent metal binding site positions. Inhibition studies implied that all the investigated complexes decreased the activity of the enzyme while the kinetic analysis indicated an uncompetitive mode of inhibition for the selected complexes. Docking results suggested that the main inhibitory site of all these complexes is located in the ion translocation pathway on the extracellular side in the E2P enzyme conformation, similar to the case of cardiac glycosides, specific Na/K-ATPase inhibitors. Also, based on our knowledge, the hydrolyzed forms of [AuCl4]- and [PtCl2(dmso)2] complexes were investigated for the first time by theoretical calculations in this paper. Thereby, a new inhibitory site situated between the M2 and M4 helices was revealed. Binding in this site induces conformational changes in the enzyme domains and perturbs the E1-E2P conformational equilibrium, causing enzyme inhibition.


Coordination Complexes/metabolism , Gold Compounds/metabolism , Models, Theoretical , Platinum Compounds/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Binding Sites , Coordination Complexes/chemistry , Gold Compounds/chemistry , Humans , Kinetics , Models, Molecular , Molecular Docking Simulation , Platinum Compounds/chemistry , Protein Conformation , Sodium-Potassium-Exchanging ATPase/chemistry
8.
Met Ions Life Sci ; 182018 02 05.
Article En | MEDLINE | ID: mdl-29394021

Polynuclear platinum complexes (PPCs) represent a discrete structural class of DNA-binding agents with excellent antitumor properties. The use of at least two platinum coordinating units automatically means that multifunctional DNA binding modes are possible. The structural variability inherent in a polynuclear platinum structure can be harnessed to produce discrete modes of DNA binding, with conformational changes distinct from and indeed inaccessible to, the mononuclear agents such as cisplatin. Since our original contributions in this field a wide variety of dinuclear complexes especially have been prepared, their DNA binding studied, and potential relevance to cytotoxicity examined. This chapter focuses on how DNA structure and reactivity is modulated through interactions with PPCs with emphasis on novel aspects of such structure and reactivity. How these major changes are further reflected in damaged DNA-protein binding and cellular effects are reviewed. We further review, for the first time, the great structural diversity achieved in PPC complex design and summarize their major DNA binding effects.


Antineoplastic Agents/therapeutic use , DNA, Neoplasm/metabolism , Neoplasms/drug therapy , Organometallic Compounds/therapeutic use , Platinum Compounds/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Coordination Complexes , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , G-Quadruplexes , Humans , Models, Molecular , Molecular Structure , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Platinum Compounds/chemistry , Platinum Compounds/metabolism , Protein Binding , Structure-Activity Relationship
9.
Met Ions Life Sci ; 182018 02 05.
Article En | MEDLINE | ID: mdl-29394023

Glycosaminoglycans (GAGs) such as heparin and heparan sulfate (HS) are large complex carbohydrate molecules that bind to a wide variety of proteins and exercise important physiological and pathological processes. This chapter focuses on the concept of metalloglycomics and reviews the structure and conformation of GAGs and the role of various metal ions during the interaction of GAGs with their biological partners such as proteins and enzymes. The use of metal complexes in heparin analysis is discussed. Cleavage of heparan sulfate proteoglycans (HSPGs) by the enzyme heparanase modulates tumor-related events including angiogenesis, cell invasion, metastasis, and inflammation. HS is identified as a ligand receptor for polynuclear platinum complexes (PPCs) defining a new mechanism of cellular accumulation for platinum drugs with implications for tumor selectivity. The covalent and noncovalent interaction of PPCs with GAGs and the functional consequences of strong binding with HS are explained in detail. Sulfate cluster anchoring shields the sulfates from recognition by charged protein residues preventing the exercise of the HS-enzyme/protein function, such as growth factor recognition and the activity of heparanase on HS. The cellular consequences are inhibition of invasion and angiogenesis. Metalloglycomics is a potentially rich new area of endeavor for bioinorganic chemists to study the relevance of intrinsic metal ions in heparin/ HS-protein interactions and for development of new compounds for therapeutic, analytical, and imaging applications.


Antineoplastic Agents/chemistry , Glycomics/methods , Heparan Sulfate Proteoglycans/chemistry , Heparin/chemistry , Organometallic Compounds/chemistry , Platinum Compounds/chemistry , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Binding Sites , Carbohydrate Conformation , Coordination Complexes , Heparan Sulfate Proteoglycans/metabolism , Heparin/metabolism , Humans , Models, Molecular , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Organometallic Compounds/metabolism , Organometallic Compounds/therapeutic use , Platinum Compounds/metabolism , Platinum Compounds/therapeutic use , Protein Binding , Protein Conformation , Structure-Activity Relationship
10.
Sci Rep ; 8(1): 767, 2018 01 15.
Article En | MEDLINE | ID: mdl-29335501

G-quadruplexes (GQ) folded by the oncogenic G-rich sequences are the promising targets for developing anticancer therapeutic molecules. However, the current drug development mainly focused on non-covalent dynamic binders to stabilize GQ structures, while the covalent targeting from inorganic complexes via chelating principles, as a potent therapeutic strategy was surprisingly lack of exploration. Herein, a series of dinuclear platinum complexes, [(Pt(Dip)Cl)2(µ-diamine)](NO3)2 (Dip: 4,7-diphenyl-1,10-phenanthroline), were designed to contain two dual-functional Pt cores connected by an alkyl linkage. Pt3 with nonanediamine linkage optimized the specific binding towards c-myc G-quadruplex via dual functional clamp on GQ as 1) non-covalently π-stacking of aromatic ligands, and 2) two Pt(II) cores covalently chelated to guanines at both 3'- and 5'-ends.


DNA/metabolism , G-Quadruplexes , Genes, myc , Platinum Compounds/metabolism , Chemical Phenomena , Molecular Docking Simulation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
Biotechnol Bioeng ; 113(2): 301-10, 2016 Feb.
Article En | MEDLINE | ID: mdl-26192088

PCR cannot distinguish live microorganisms from dead ones. To circumvent this disadvantage, ethidium/propidium-monoazide (EMA/PMA) and psoralen to discriminate live from dead bacteria have been used for 2 decades. These methods require the use of numerous laborious procedures. We introduce an innovative method that uses platinum compounds, which are primarily used as catalysts in organic chemistry and partly used as anti-cancer drugs. Microorganisms are briefly exposed to platinum compounds in vivo, and these compounds penetrate dead (compromised) microorganisms but not live ones and are chelated by chromosomal DNA. The use of platinum compounds permits clear discrimination between live and dead microorganisms in water and milk (including Cronobacter sakazakii and Escherichia coli) via PCR compared with typically used PMA. This platinum-PCR method could enable the specific detection of viable coliforms in milk at a concentration of 5-10 CFU mL(-1) specified by EU/USA regulations after a 4-h process. For sample components, environmental water contains lower levels of PCR inhibitors than milk does, and milk is similar to infant formula, skim milk and blood; thus, the use of the platinum-PCR method could also prevent food poisoning due to the presence of C. sakazakii in dairy products. This method could provide outstanding rapidity for use in environmental/food/clinical tests. Platinum-PCR could also be a substitute for the typical culture-based methods currently used.


Microbial Viability , Platinum Compounds/metabolism , Polymerase Chain Reaction/methods , Animals , Cronobacter sakazakii/drug effects , Cronobacter sakazakii/genetics , DNA, Bacterial/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Milk/microbiology , Sensitivity and Specificity , Time Factors , Water Microbiology
12.
Biochem J ; 472(2): 217-23, 2015 Dec 01.
Article En | MEDLINE | ID: mdl-26399480

The copper chaperone Cox17 (cytochrome c oxidase copper chaperone) has been shown to facilitate the delivery of cisplatin to mitochondria, which contributes to the overall cytotoxicity of the drug [Zhao et al. (2014) Chem. Commun. 50: , 2667-2669]. Kinetic data indicate that Cox17 has reactivity similar to glutathione (GSH), the most abundant thiol-rich molecule in the cytoplasm. In the present study, we found that GSH significantly modulates the reaction of platinum complexes with Cox17. GSH enhances the reactivity of three anti-cancer drugs (cisplatin, carboplatin and oxaliplatin) to Cox17, but suppresses the reaction of transplatin. Surprisingly, the pre-formed cisplatin-GSH adducts are highly reactive to Cox17; over 90% platinum transfers from GSH to Cox17. On the other hand, transplatin-GSH adducts are inert to Cox17. These different effects are consistent with the drug activity of these platinum complexes. In addition, GSH attenuates the protein aggregation of Cox17 induced by platination. These results indicate that the platinum-protein interactions could be substantially influenced by the cellular environment.


Antineoplastic Agents/metabolism , Carrier Proteins/metabolism , Copper/metabolism , Glutathione/metabolism , Organoplatinum Compounds/metabolism , Platinum Compounds/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoproteins/genetics , Apoproteins/metabolism , Binding, Competitive , Carrier Proteins/chemistry , Carrier Proteins/genetics , Copper Transport Proteins , Humans , Kinetics , Ligands , Organoplatinum Compounds/agonists , Organoplatinum Compounds/antagonists & inhibitors , Organoplatinum Compounds/pharmacology , Oxidation-Reduction , Platinum Compounds/agonists , Platinum Compounds/antagonists & inhibitors , Platinum Compounds/pharmacology , Protein Aggregates/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solubility
13.
Article En | MEDLINE | ID: mdl-25004912

BACKGROUND: We studied the interaction of oxaliplatin derivatives involving cytotoxic adenine-based cyclin-dependent kinase inhibitors, with human liver microsomal cytochrome P450. METHODS AND RESULTS: The activities of 9 human liver microsomal CYP forms (CYPs 1A2, 7-ethoxyresorufin O-deethylation; 2A6, coumarin 7-hydroxylation; 2B6, 7-ethoxy-4-(trifluoromethyl) coumarin O-deethylation; 2C8, luciferin-6´ methyl ether demethylation; 2C9, diclofenac 4´-hydroxylation, 6´-deoxyluciferin hydroxylation; 2C19, (S)-mephenytoin 4´-hydroxylation; 2D6, bufuralol 1´-hydroxylation, 2E1, chlorzoxazone 6-hydroxylation; 3A4, testosterone 6ß-hydroxylation, luciferin-6´ benzyl ether debenzylation) were tested using HPLC, fluorescence and luminescence product detection. At 100 µM platinum(II) oxalato complex concentration, CYP inhibition was in general 25%-50%, except for the CYP3A4 form which showed roughly twice the inhibition (72%-95%). At low complex concentration (10 µM), the difference in inhibition of CYP3A4 and other forms was even more pronounced. Dixon and Lineweaver-Burk plots indicated a partially noncompetitive mechanism of CYP3A4 inhibition. CONCLUSIONS: The tested complexes significantly inhibit human liver microsomal CYP3A4 activity even at clinically relevant concentrations. This could be a serious drawback for the use of these compounds in clinical practice.


Cytochrome P-450 CYP3A/metabolism , Microsomes, Liver/metabolism , Platinum Compounds/metabolism , Humans , Hydroxylation , Ligands , Oxidation-Reduction
14.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 9): 1132-4, 2014 Sep.
Article En | MEDLINE | ID: mdl-25195880

This study examines the binding and chemical stability of the platinum hexahalides K2PtCl6, K2PtBr6 and K2PtI6 when soaked into pre-grown hen egg-white lysozyme (HEWL) crystals as the protein host. Direct comparison of the iodo complex with the chloro and bromo complexes shows that the iodo complex is partly chemically transformed to a square-planar PtI3 complex bound to the N(δ) atom of His15, a chemical behaviour that is not exhibited by the chloro or bromo complexes. Each complex does, however, bind to HEWL in its octahedral form either at one site (PtI6) or at two sites (PtBr6 and PtCl6). As heavy-atom derivatives of a protein, the octahedral shape of the hexahalides could be helpful in cases of difficult-to-interpret electron-density maps as they would be recognisable 'objects'.


Halogens/chemistry , Histidine/metabolism , Muramidase/metabolism , Platinum Compounds/metabolism , Crystallography, X-Ray , Platinum Compounds/chemistry
15.
J Inorg Biochem ; 137: 40-5, 2014 08.
Article En | MEDLINE | ID: mdl-24803025

Binding to plasma proteins is one of the major metabolic pathways of metallodrugs. In the case of platinum-based anticancer drugs, it is the interaction with serum albumin that affects most strongly their in vivo behavior. Since both the configuration, i.e. cis-trans-isomerism, and the nature of leaving groups have an effect on the reactivity of Pt(II) coordination compounds toward biomolecules, a set of cis- and trans-configured complexes with halide leaving groups (Cl(-), Br(-), and I(-)) and 2-propanone oxime as carrier ligands was chosen for this study. Binding experiments were performed both with albumin and human serum and the Pt content in ultrafiltrates was quantified using inductively coupled plasma mass spectrometry. In order to shed light on the binding mechanism, the albumin binding constant (KHSA) and the octanol-water partition coefficient (P) were experimentally determined and relationships between log KHSA and log P were explored. The correlation was found significant only for cis-configured platinum complexes (R(2)=0.997 and standard deviation=0.02), indicating a certain contribution of the nonspecific binding which is largely dominated by the lipophilicity of compounds. In contrast, for trans-complexes a specific molecular recognition element plays a significant role. The participation of albumin in drug distribution in blood serum was assessed using an equilibrium distribution model and by comparing the percentage binding in the albumin and serum-protein fractions. Irrespective of the compound polarity, albumin contributes from 85 to 100% to the overall binding in serum.


Antineoplastic Agents/chemistry , Blood Proteins/chemistry , Platinum Compounds/chemistry , Serum Albumin/chemistry , Antineoplastic Agents/administration & dosage , Blood Proteins/metabolism , Drug Design , Humans , Isomerism , Ligands , Mass Spectrometry , Platinum/blood , Platinum/chemistry , Platinum Compounds/blood , Platinum Compounds/metabolism , Protein Binding , Serum Albumin/metabolism
16.
Expert Opin Ther Targets ; 18(4): 403-14, 2014 Apr.
Article En | MEDLINE | ID: mdl-24479491

INTRODUCTION: Platinum drugs are widely used for the treatment of testicular, bladder, ovarian, colorectal, lung and prostate cancers. With regard to ovarian cancer in particular, the prognosis is poor for tumours that are (or have become) platinum-resistant. Determining the mechanism underlying platinum resistance may aid in the identification of therapeutic targets for the treatment of platinum-resistant tumours. AREAS COVERED: This review gives an overview of the characteristics and functions of Annexin (Anx) A4, the mechanism of Anx A4-induced platinum resistance, the association between platinum resistance and platinum transporters, recent reports that Anx A4 overexpression promotes the efflux of platinum drugs via platinum transporters and the association between other Anxs and chemoresistance. The reader will gain an understanding of recent studies on the mechanism of Anx A4-induced chemoresistance. Anx A4 represents a therapeutic target for the treatment of Anx A4-overexpressing platinum-resistant tumours. EXPERT OPINION: Anx A4 is overexpressed in ovarian clear cell carcinoma (CCC), and enhanced Anx A4 expression induces platinum resistance. Recent studies showed that Anx A4 is also associated with platinum resistance in cancers other than ovarian CCC. Furthermore, other Anxs are reportedly associated with chemoresistance, suggesting a relationship between the Anx family and chemoresistance.


Annexin A4/metabolism , Antineoplastic Agents/metabolism , Biomarkers, Tumor/metabolism , Drug Delivery Systems/methods , Drug Resistance, Neoplasm/physiology , Platinum Compounds/metabolism , Animals , Annexin A4/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm/drug effects , Humans , Platinum Compounds/administration & dosage , Protein Binding/physiology , Treatment Outcome
17.
Sci Rep ; 3: 2060, 2013.
Article En | MEDLINE | ID: mdl-23792883

A quaternized trigeminal ligand, 4-[4,6-di(4-pyridyl)-1,3,5-(2-triazinyl)]-1-methylpyridine-1-ium hexafluorophosphate (dptmp·PF6), and two derivative V-shaped dinuclear Pt(II) complexes, {[Pt(dien)]2(dptmp)}(PF6)5 (1) and {[Pt(dpa)]2(dptmp)}(PF6)5 (2), were synthesized, characterized and applied to a series of biochemical studies. FRET and SPR analyses showed these compounds, especially Pt(II) complexes, bound more strongly to human telomeric (hTel) G-quadruplex than to promoters (such as c-myc and bcl2) or to the duplex DNA. PCR-stop assays revealed that the Pt(II) complexes could bind to and stabilize G-quadruplex far more effectively than corresponding ligand. CD analyses further indicated the three compounds likely stabilized the formation of mixed-type parallel/antiparallel G-quadruplex structures. Their efficacy as telomerase inhibitors and potential anticancer drugs was explored via TRAP. The IC50 value was determined to be 0.113 ± 0.019 µM for 1, indicating that it is one of the strongest known telomerase inhibitors. These results confirm that both V-shaped dinuclear Pt(II) complexes act as selective G-quadruplex binders and significant telomerase inhibitors.


G-Quadruplexes , Platinum Compounds/metabolism , Telomerase/antagonists & inhibitors , Telomere , Humans , Nucleic Acid Conformation
18.
J Toxicol Environ Health A ; 76(6): 345-53, 2013.
Article En | MEDLINE | ID: mdl-23557233

The chemotherapeutic agent cisplatin (cDDP) is widely used to treat a variety of solid and hematological tumors. However, cDDP exerts severe side effects, such as nephrotoxicity, neurotoxicity, and bone-marrow suppression. The use of some dietary compounds to protect organs that are not targets in association with chemotherapy has been encouraged. This study evaluated the protective effects of chlorophyll b (CLb) on DNA damage induced by cDDP by use of single-cell gel electrophoresis (SCGE) assays. Further, this investigation also determined platinum (Pt) and magnesium (Mg) bioaccumulation in mice tissues after treatment with CLb alone and/or in association of cDDP (simultaneous treatment) by inductively coupled plasma-mass spectroscopy (ICP-MS). All parameters were studied in peripheral blood cells (PBC), kidneys, and liver of mice after administration of CLb (0.2 or 0.5 mg/kg of body weight [b.w.]), cDDP (6 mg/kg b.w.), and the combination CLb 0.2 plus cDDP or CLb 0.5 plus cDDP. Pt accumulation in liver and kidneys was higher than that found in PBC, while DNA damage was higher in kidneys and liver than in PBC. Further, treatment with CLb alone did not induce DNA damage. Evidence indicates that genotoxic effects produced by cDDP may not be related to Pt accumulation and distribution. In combined treatments, CLb decreased DNA damage in tissues, but the PT contents did not change and these treatments also showed that CLb may be an important source of Mg. Thus, our results indicate that consumption of CLb-rich foods may diminish the adverse health effects induced by cDDP exposure.


Antimutagenic Agents/pharmacology , Antineoplastic Agents/toxicity , Chlorophyll/pharmacology , Cisplatin/toxicity , DNA Damage/drug effects , Animals , Antineoplastic Agents/pharmacokinetics , Cisplatin/pharmacokinetics , Comet Assay , Female , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Magnesium Compounds/metabolism , Male , Mice , Platinum Compounds/metabolism
19.
Dalton Trans ; 42(23): 8426-36, 2013 Jun 21.
Article En | MEDLINE | ID: mdl-23620195

The kinetics of chloride substitution from a series of square-planar platinum(II) complexes, viz. [Pt(terpyridine)Cl](+), (Pt1), [Pt{2-(2'-pyridyl)-1,10-phenanthroline}Cl]Cl, (Pt2), [Pt{4'-(2'''-CH3-phenyl)-2,2':6',2''-terpyridine}Cl]CF3SO3 (Pt3) and [Pt{4'-(2'''-CH3-phenyl)-6-(3''-isoquinoyl)-2,2'bipyridine}Cl]SbF6 (Pt4) were studied using a series of five-membered heterocyclic neutral nitrogen donor nucleophiles, viz. pyrazole (Pz), triazole (Tz), imidazole (Im), 1-methylimidazole (MIm) and 1,2-dimethylimidazole (DMIm) under pseudo-first-order conditions in methanol using UV/Visible spectrophotometry and conventional stopped-flow techniques. The observed second-order rate constants, k2, followed a two term rate law k(obs) = k2[nucleophile] + k(s) except for DMIm with Pt1, Pt3, Pt4 and Pz, Tz with Pt1. Increasing the π-conjugation in the cis position decreases the rate of chloride substitution by decreasing the π-acceptor property of the terpy moiety. However, increasing the π-conjugation in the cis/trans position increases the rate of substitution by enhancing the π-acceptor property within the ligand framework whereby increasing the reactivity of the metal centre. The observed trend for the reactivity was Pt2 > Pt1 > Pt3 > Pt4. The substitution kinetics was influenced by the basicity of the incoming nucleophiles except for the sterically hindered nucleophile, DMIm. The general trend observed for the reactivity of the nucleophiles is MIm > Im > DMIm > Pz > Tz.


Azoles/chemistry , Platinum Compounds/chemistry , Pyridines/chemistry , Azoles/metabolism , Crystallography, X-Ray , Ligands , Platinum Compounds/metabolism , Pyridines/metabolism
...