Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 659
Filter
1.
J Biol Inorg Chem ; 29(2): 217-241, 2024 03.
Article in English | MEDLINE | ID: mdl-38369679

ABSTRACT

We previously reported that the bismuth(III) dithiocarbamate derivative, bismuth diethyldithiocarbamate (1) exhibited greater cytotoxicity while inducing apoptosis via the intrinsic pathway in MCF-7 cells. We further evaluated the other bismuth(III) dithiocarbamate derivatives, Bi[S2CNR]3, with R = (CH2CH2OH)(iPr), (CH2)4, and (CH2CH2OH)(CH3), denoted as 2, 3, and 4, respectively, in the same MCF-7 cell line. 2-4 were found to exhibit IC50 values of 10.33 ± 0.06 µM, 1.07 ± 0.01 µM and 25.37 ± 0.12 µM, respectively, compared to that of cisplatin at 30.53 ± 0.23 µM. Apoptotic promotion via the mitochondrial-dependent pathway was due to the elevation of intracellular reactive oxygen species (ROS), promotion of caspases, release of cytochrome c, fragmentation of DNA, and results of staining assay observed in all compound-treated cells. 2-4 are also capable of suppressing MCF-7 cell invasion and modulate Lys-48 also Lys-63 linked polyubiquitination, leading to proteasomal degradation. Analysis of gene expression via qRT-PCR revealed their modulation, which supported all activities conducted upon treatment with 2-4. Altogether, bismuth dithiocarbamate derivatives, with bismuth(III) as the metal center bound to ligands, isopropyl ethanol, pyrrolidine, and methyl ethanol dithiocarbamate, are potential anti-breast cancer agents that induce apoptosis and suppress metastasis. Further studies using other breast cancer cell lines and in vivo studies are recommended to clarify the anticancer effects of these compounds.


Subject(s)
Antineoplastic Agents , Apoptosis , Bismuth , Breast Neoplasms , Mitochondria , Thiocarbamates , Humans , Bismuth/chemistry , Bismuth/pharmacology , Apoptosis/drug effects , Thiocarbamates/pharmacology , Thiocarbamates/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , MCF-7 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Reactive Oxygen Species/metabolism , Female , Neoplasm Invasiveness , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects
2.
Ecotoxicol Environ Saf ; 283: 116852, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39142049

ABSTRACT

Dithiocarbamates have been widely used in various industrial applications, such as insecticides (ferbam) or drug (disulfiram). This study explored the inhibitory effects of dithiocarbamates on human and rat gonadal 3ß-hydroxysteroid dehydrogenases (3ß-HSD) and investigated the structure-activity relationship and mechanistic insights. The inhibitory activity of six dithiocarbamates and thiourea on the conversion of pregnenolone to progesterone was evaluated using human KGN cell and rat testicular microsomes, with subsequent progesterone measurement using HPLC-MS/MS. The study found that among the tested compounds disulfiram, ferbam, and thiram exhibited significant inhibitory activity against human 3ß-HSD2 and rat 3ß-HSD1, with ferbam demonstrating the highest potency. The mode of action for these compounds was characterized, showing mixed inhibition for human 3ß-HSD2 and mixed/noncompetitive inhibition for rat 3ß-HSD1. Additionally, it was observed that dithiothreitol dose-dependently reversed the inhibitory effects of dithiocarbamates on both human and rat gonadal 3ß-HSD enzymes. The study also delved into the penetration of these dithiocarbamates through the human KGN cell membrane and their impact on progesterone production, highlighting their potency in inhibiting human 3ß-HSD2. Furthermore, bivariate correlation analysis revealed a positive correlation of LogP (lipophilicity) with IC50 values for both enzymes. Docking analysis indicated that dithiocarbamates bind to NAD+ and steroid-binding sites, with some interactions with cysteine residues. In conclusion, this study provides valuable insights into the structure-activity relationship and mechanistic aspects of dithiocarbamates as inhibitors of human and rat gonadal 3ß-HSDs, suggesting that these compounds likely exert their inhibitory effects through binding to cysteine residues.


Subject(s)
Fungicides, Industrial , Animals , Humans , Fungicides, Industrial/toxicity , Rats , Male , Cysteine , Structure-Activity Relationship , Thiocarbamates/pharmacology , Thiocarbamates/chemistry , Testis/drug effects , Testis/enzymology , Molecular Docking Simulation , 3-Hydroxysteroid Dehydrogenases/metabolism , Microsomes/drug effects , Microsomes/enzymology
3.
Molecules ; 29(10)2024 May 19.
Article in English | MEDLINE | ID: mdl-38792249

ABSTRACT

Indole phytoalexins, found in economically significant Cruciferae family plants, are synthesized in response to pathogen attacks or stress, serving as crucial components of plant defense mechanisms against bacterial and fungal infections. Furthermore, recent research indicates that these compounds hold promise for improving human health, particularly in terms of potential anticancer effects that have been observed in various studies. Since our last comprehensive overview in 2016 focusing on the antiproliferative effects of these substances, brassinin and camalexin have been the most extensively studied. This review analyses the multifaceted pharmacological effects of brassinin and camalexin, highlighting their anticancer potential. In this article, we also provide an overview of the antiproliferative activity of new synthetic analogs of indole phytoalexins, which were synthesized and tested at our university with the aim of enhancing efficacy compared to the parent compound.


Subject(s)
Indoles , Phytoalexins , Sesquiterpenes , Indoles/chemistry , Indoles/pharmacology , Humans , Sesquiterpenes/pharmacology , Sesquiterpenes/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Thiazoles/pharmacology , Thiazoles/chemistry , Cell Proliferation/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Thiocarbamates/pharmacology , Thiocarbamates/chemistry
4.
Inorg Chem ; 62(16): 6332-6338, 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37026841

ABSTRACT

Formate dehydrogenase (FDH) enzymes catalyze redox interconversion of CO2 and HCO2-, with a key mechanistic step being the transfer of H- from HCO2- to an oxidized active site featuring a [MVI≡S] group in a sulfur-rich environment (M = Mo or W). Here, we report reactivity studies with HCO2- and other reducing agents of a synthetic [WVI≡S] model complex ligated by dithiocarbamate (dtc) ligands. Reactions of [WVIS(dtc)3][BF4] (1) conducted in MeOH solvent generated [WVIS(S2)(dtc)2] (2) and [WVS(µ-S)(dtc)]2 (3) products by a solvolysis pathway that was accelerated by the presence of [Me4N][HCO2] but did not require it. Under MeOH-free conditions, the reaction of 1 with [Et4N][HCO2] produced some [WIV(µ-S)(µ-dtc)(dtc)]2 (4), but predominantly [WV(dtc)4]+ (5), along with stoichiometric CO2 detected by headspace gas chromatography (GC) analysis. Stronger hydride sources such as K-selectride generated the more reduced analogue, 4, exclusively. The reaction of 1 with the electron donor, CoCp2, also produced 4 and 5 in varying amounts depending on reaction conditions. These results indicate that formates and borohydrides act as electron donors rather than hydride donors toward 1, an outcome that diverges from the behavior of FDHs. The difference is ascribed to the more oxidizing potential of [WVI≡S] complex 1 when supported by monoanionic dtc ligands that allows electron transfer to outcompete hydride transfer, as compared to the more reduced [MVI≡S] active sites supported by dianionic pyranopterindithiolate ligands in FDHs.


Subject(s)
Carbon Dioxide , Formate Dehydrogenases , Catalytic Domain , Formate Dehydrogenases/chemistry , Oxidation-Reduction , Tungsten Compounds/chemistry , Thiocarbamates/chemistry
5.
Bioorg Chem ; 118: 105474, 2022 01.
Article in English | MEDLINE | ID: mdl-34794102

ABSTRACT

The worldwide prevalence of NDM-1-producing Gram-negative pathogens has drastically undermined the clinical efficacy of carbapenems, prompting a need to devise an effective strategy to preserve their clinical value. Here we constructed a focused compound library of dithiocarbamates and systematically evaluated their potential synergistic antibacterial activities combined with copper. SA09-Cu exhibited excellent inhibition against a series of clinical NDM-1-producing carbapenem-resistant Enterobacteriaceae (CRE) in restoring meropenem effect, and slowed down the development of carbapenem resistance. Enzymatic kinetic and isothermal titration calorimetry studies demonstrated that SA09-Cu was a noncompetitive NDM-1 inhibitor. The electron paramagnetic resonance (EPR) and X-ray photoelectron spectroscopy (XPS) revealed a novel inhibition mechanism, which is that SA09-Cu could convert NDM-1 into an inactive state by oxidizing the Zn(II)-thiolate site of the enzyme. Importantly, SA09-Cu showed a unique redox tuning ability, and avoided to be reduced by intracellular thiols of bacteria. In vivo experiments indicated that SA09 combined with CuGlu could effectively potentiate MER's effect against NDM-1-producing E. coli (EC23) in the murine infection model. This study provides a highly promising scaffold in developing novel inhibitors to combat NDM-1-producing CREs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbapenem-Resistant Enterobacteriaceae/drug effects , Coordination Complexes/pharmacology , Copper/pharmacology , Enzyme Inhibitors/pharmacology , Thiocarbamates/pharmacology , beta-Lactamases/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Carbapenem-Resistant Enterobacteriaceae/enzymology , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Copper/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli/drug effects , Escherichia coli/enzymology , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Thiocarbamates/chemistry
6.
J Enzyme Inhib Med Chem ; 37(1): 1-8, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34894954

ABSTRACT

Recently, inorganic anions and sulphonamides, two of the main classes of zinc-binding carbonic anhydrase inhibitors (CAIs), were investigated for inhibition of the α-class carbonic anhydrase (CA, EC 4.2.1.1) from Neisseria gonorrhoeae, NgCA. As an extension to our previous studies, we report that dithiocarbamates (DTCs) derived from primary or secondary amines constitute a class of efficient inhibitors of NgCA. KIs ranging between 83.7 and 827 nM were measured for a series of 31 DTCs that incorporated various aliphatic, aromatic, and heterocyclic scaffolds. A subset of DTCs were selected for antimicrobial testing against N. gonorrhoeae, and three molecules displayed minimum inhibitory concentration (MIC) values less than or equal to 8 µg/mL. As NgCA was recently validated as an antibacterial drug target, the DTCs may lead to development of novel antigonococcal agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Neisseria gonorrhoeae/drug effects , Thiocarbamates/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Neisseria gonorrhoeae/enzymology , Structure-Activity Relationship , Thiocarbamates/chemical synthesis , Thiocarbamates/chemistry
7.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35163241

ABSTRACT

Dithiocarbamate ligands have the ability to form stable complexes with transition metals, and this chelating ability has been utilized in numerous applications. The complexes have also been used to synthesize other useful compounds. Here, the up-to-date applications of dithiocarbamate ligands and complexes are extensively discussed. Some of these are their use as enzyme inhibitor and treatment of HIV and other diseases. The application as anticancer, antimicrobial, medical imaging and anti-inflammatory agents is examined. Moreover, the application in the industry as vulcanization accelerator, froth flotation collector, antifouling, coatings, lubricant additives and sensors is discussed. The various ways in which they have been employed in synthesis of other compounds are highlighted. Finally, the agricultural uses and remediation of heavy metals via dithiocarbamate compounds are comprehensively discussed.


Subject(s)
Thiocarbamates/chemistry , Thiocarbamates/chemical synthesis , Transition Elements/chemistry , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors , Humans , Ligands , Metals, Heavy , Thiocarbamates/pharmacology
8.
Bioorg Med Chem Lett ; 43: 128102, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33984471

ABSTRACT

In order to find a 99mTc-labeled deferoxamine radiotracer for bacterial infection imaging, deferoxamine dithiocarbamate (DFODTC) was successfully synthesized and it was radiolabeled with [99mTcN]2+ core to prepare the 99mTcN(DFODTC)2 complex. 99mTcN(DFODTC)2 was obtained with high radiochemical purity without further purification. The complex was lipophilic and exhibited good in vitro stability. According to the result of bacterial binding study, the binding of 99mTcN(DFODTC)2 to bacteria was specific. Biodistribution in mice study indicated that 99mTcN(DFODTC)2 had a higher uptake in bacterial infection tissues than in turpentine-induced abscesses at 120 min after injection, which showed that the radiotracer could differentiate between bacterial infection and sterile inflammation. SPECT/CT images showed that there was a clear accumulation in infection sites, suggesting that 99mTcN(DFODTC)2 could be a potential bacterial infection imaging radiotracer.


Subject(s)
Bacterial Infections/diagnostic imaging , Deferoxamine/chemistry , Organotechnetium Compounds/chemistry , Radiopharmaceuticals/chemistry , Thiocarbamates/chemistry , Animals , Inflammation/diagnostic imaging , Mice , Molecular Structure , Organotechnetium Compounds/chemical synthesis , Radiopharmaceuticals/chemical synthesis
9.
Bioorg Med Chem ; 47: 116373, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34467870

ABSTRACT

A series of "2 + 1" mixed ligand tricarbonyl complexes of the general formula fac-[Re/99mTc/186Re(CO)3(DDTC)(L)] containing diethyldithiocarbamate (DDTC) as a monoanionic bidentate ligand and a series of monodentate ligands L was synthesized, characterized and evaluated. The impact of ligand L on the radiochemical yield (RCY) and biodistribution of the final compounds was also investigated. DDTC and the appropriate L ligand [cyclohexyl isocyanide (cisc), tert-butyl isocyanide (tbi), triphenylphosphine (PPh3), methyldiphenylphosphine (PPh2Me), triphenylarsine (AsPh3), imidazole (im), and 4-aminopyridine (4AP)] readily reacted in equimolar amounts with the [Et4N]2[Re(CO)3Br3] precursor to afford fac-[Re(CO)3(DDTC)(cisc)], Re1, fac-[Re(CO)3(DDTC)(tbi)], Re2, fac-[Re(CO)3(DDTC)(PPh3)], Re3, fac-[Re(CO)3(DDTC)(PPh2Me)], Re4, fac-[Re(CO)3(DDTC)(AsPh3)], Re5, fac-[Re(CO)3(DDTC)(im)], Re6 and fac-[Re(CO)3(DDTC)(4AP)], Re7, complexes in high yields (>80%). All Re complexes were fully characterized by IR, NMR, and in addition Re4, Re5, and Re7 with X-ray crystallography. Analogous reactions as performed with Re were subsequently explored on the 99mTc and 186Re-tracer levels using the corresponding fac-[99mTc/186Re(CO)3(H2O)3]+ precursor. Complexes 99mTc1 - 99mTc5, 186Re1 and 186Re3 were obtained in high radiochemical yield (>91%), while the complexes 99mTc6, 99mTc7 and 186Re7 formed with radiochemical yields of 55%, 28%, and 75%, respectively. The 99mTc and 186Re-complexes were characterized by comparative HPLC analysis using the analogous Re complexes. During histidine and cysteine challenge experiments at 37 °C through 6 h, complexes 99mTc1 - 99mTc5 remained > 92% stable, while complexes 99mTc6 and 99mTc7 remained only 8% stable through 3 h. Similar studies for 186Re-complexes showed that 186Re1 and 186Re3 remained > 95% stable for up to 48 h, while 186Re7 had decreased to 7% after 3 h. LogD7.4 data of 99mTc1 - 99mTc5, 186Re1, and 186Re3 complexes, which ranged from 2.59 to 3.39, suggested high lipophilicity. Biodistribution studies in healthy Swiss albino mice showed hepatobiliary excretion for 99mTc1, 99mTc2, and 99mTc4, fast blood clearance for 99mTc4, while high liver uptake and retention for 99mTc3 and 99mTc5 were measured. Moreover, 99mTc2 showed high accumulation in the lungs with sustained retention (52.80% ID/g at 4 h p.i.) and significant brain uptake at 2 min p.i. (1.89% ID/g). The study showed the great influence of monodentate ligand in the synthesis and biodistribution of the mixed ligand complexes.


Subject(s)
Radiopharmaceuticals/pharmacokinetics , Rhenium/pharmacokinetics , Technetium/pharmacokinetics , Thiocarbamates/pharmacokinetics , Animals , Ligands , Male , Mice , Molecular Structure , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Rhenium/chemistry , Technetium/chemistry , Thiocarbamates/chemistry , Tissue Distribution
10.
Bioorg Chem ; 115: 105203, 2021 10.
Article in English | MEDLINE | ID: mdl-34371375

ABSTRACT

A novel class of potential MAO-B inhibitors was designed and synthesized in good yield by combining the pyridazinone moiety with the dithiocarbamate framework, two relevant pharmacophores for drug discovery. The biological results obtained for the different pyridazinone/dithiocarbamate hybrids (compounds 8-14) indicated that most of them reversibly and selectively inhibit the hMAO-B in vitro with IC50 values in the µM range and exhibit not significant cellular toxicity. The analogues 9a1, 11a1, 12a2, 12b1 and 12b2, which present the dithiocarbamate fragment derivatized with a piperidin-1-yl or pyrrolidin-1-yl group and placed at C3 or C4 of the diazine ring, were the most attractive compounds of these series. Molecular modeling studies were performed to analyze the binding mode to the enzyme and the structure activity relationships of the titled compounds, as well as to predict their drug-like properties.


Subject(s)
Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Pyridazines/pharmacology , Thiocarbamates/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Pyridazines/chemical synthesis , Pyridazines/chemistry , Structure-Activity Relationship , Thiocarbamates/chemistry
11.
J Enzyme Inhib Med Chem ; 36(1): 593-604, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33557648

ABSTRACT

Colorectal cancer (CRC) is a common malignant tumour of human digestive tract. The high mortality rate of CRC is closely related to the limitations of existing treatments. Thus, there is an urgent need to search for new anti-CRC agents. In this work, twenty novel coumarin-dithiocarbamate derivatives (IDs) were designed, synthesized and evaluated in vitro. The results suggest that the most active compound ID-11 effectively inhibited the proliferation of CRC cell lines while shown little impact on normal colon epithelial cells. Mechanism studies revealed that ID-11 displayed bromodomain-containing protein 4 inhibitory activity, and induced G2/M phase arrest, apoptosis as well as decreased the expression levels of the key genes such as c-Myc and Bcl-2 in CRC cell lines. Moreover, the ADMET properties prediction results shown that ID-11 possess well metabolic characteristics without obvious toxicities. Our data demonstrated that compound ID-11 may be a promising anti-CRC agent and deserved for further development.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Coumarins/pharmacology , Drug Design , Thiocarbamates/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Coumarins/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Thiocarbamates/chemistry , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
12.
Molecules ; 26(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209921

ABSTRACT

Three novel gold(III) complexes (1-3) of general composition [Au(Bipydc)(S2CNR2)]Cl2 (Bipydc = 2,2'-bipyridine-3,3'-dicarboxylic acid and R = methyl for dimethyldithiocarbamate (DMDTC), ethyl for diethyldithiocarbamate (DEDTC), and benzyl for dibenzyldithiocarbamate (DBDTC)) have been synthesized and characterized by elemental analysis, FTIR and NMR spectroscopic techniques. The spectral results confirmed the presence of both the Bipydc and dithiocarbamate ligands in the complexes. The in vitro cytotoxic studies demonstrated that compounds 1-3 were highly cytotoxic to A549, HeLa, MDA-231, and MCF-7 cancer cells with activities much higher (about 25-fold) than cisplatin. In order to know the possible mode of cell death complex 2, [Au(Bipydc)(DEDTC)]Cl2 was further tested for induction of apoptosis towards the MCF-7 cells. The results indicated that complex 2 induces cell death through apoptosis.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Gold/chemistry , Pyridines/chemistry , Thiocarbamates/chemistry , A549 Cells , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , HeLa Cells , Humans , MCF-7 Cells
13.
Molecules ; 26(13)2021 Jul 03.
Article in English | MEDLINE | ID: mdl-34279414

ABSTRACT

Malignant tumors have affected the human being since the pharaoh period, but in the last century the incidence of this disease has increased due to a large number of risk factors, including deleterious lifestyle habits (i.e., smoking) and the higher longevity. Many efforts have been spent in the last decades on achieving an early stage diagnosis of cancer, and more effective cures, leading to a decline in age-standardized cancer mortality rates. In the last years, our research groups have developed new metal-based complexes, with the aim to obtain a better selectivity for cancer cells and less side effects than the clinically established reference drug cisplatin. This work is focused on four novel Au(III) and Ru(III) complexes that share the piperidine dithiocarbamato (pipe-DTC) as the ligand, in a different molar ratio. The compounds [AuCl2(pipeDTC)], [Au(pipeDTC)2]Cl, [Ru(pipeDTC)3] and ß-[Ru2(pipeDTC)5] have been synthesized and fully characterized by several chemical analyses. We have then investigated their biological properties in two different cell lines, namely, AGS (gastric adenocarcinoma) and HCT116 (colon carcinomas), showing significant differences among the four compounds. First, the two gold-based compounds and ß-[Ru2(pipeDTC)5] display IC50 in the µM range, significantly lower than cisplatin. Second, we showed that [AuCl2(pipeDTC)] and ß-[Ru2(pipeDTC)5]Cl drive different molecular mechanisms. The first was able to induce the protein level of the DNA damage response factor p53 and the autophagy protein p62, in contrast to the second that induced the ATF4 protein level, but repressed p62 expression. This study highlights that the biological activity of different complexes bringing the same organic ligand depends on the electronic and structural properties of the metal, which are able to fine tune the biological properties, giving us precious information that can help to design more selective anticancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation , Coordination Complexes/pharmacology , Gold/chemistry , Ruthenium/chemistry , Stomach Neoplasms/drug therapy , Thiocarbamates/chemistry , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Humans , Stomach Neoplasms/pathology , Tumor Cells, Cultured
14.
Molecules ; 26(5)2021 Mar 07.
Article in English | MEDLINE | ID: mdl-33800091

ABSTRACT

The use of cisplatin is severely limited by its toxic side-effects, which has spurred chemists to employ different strategies in the development of new metal-based anticancer agents. Here, three novel dehydroabietyl piperazine dithiocarbamate ruthenium (II) polypyridyl complexes (6a-6c) were synthesized as antitumor agents. Compounds 6a and 6c exhibited better in vitro antiproliferative activity against seven tumor cell lines than cisplatin, they displayed no evident resistance in the cisplatin-resistant cell line A549/DPP. Importantly, 6a effectively inhibited tumor growth in the T-24 xenograft mouse model in comparison with cisplatin. Gel electrophoresis assay indicated that DNA was the potential targets of 6a and 6c, and the upregulation of p-H2AX confirmed this result. Cell cycle arrest studies demonstrated that 6a and 6c arrested the cell cycle at G1 phase, accompanied by the upregulation of the expression levels of the antioncogene p27 and the down-regulation of the expression levels of cyclin E. In addition, 6a and 6c caused the apoptosis of tumor cells along with the upregulation of the expression of Bax, caspase-9, cytochrome c, intracellular Ca2+ release, reactive oxygen species (ROS) generation and the downregulation of Bcl-2. These mechanistic study results suggested that 6a and 6c exerted their antitumor activity by inducing DNA damage, and consequently causing G1 stage arrest and the induction of apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Ruthenium/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Coordination Complexes/chemical synthesis , DNA Damage , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Humans , Magnetic Resonance Spectroscopy , Mice , Piperazines/chemistry , Thiocarbamates/chemistry , Xenograft Model Antitumor Assays
15.
Acc Chem Res ; 52(9): 2723-2731, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31390174

ABSTRACT

In addition to nitric oxide and carbon monoxide, hydrogen sulfide (H2S) has been recently recognized as an important biological signaling molecule with implications in a wide variety of processes, including vasodilation, cytoprotection, and neuromodulation. In parallel to the growing number of reports highlighting the biological impact of H2S, interest in developing H2S donors as both research tools and potential therapeutics has led to the growth of different H2S-releasing strategies. Many H2S investigations in model systems use direct inhalation of H2S gas or aqueous solutions of NaSH or Na2S; however, such systems do not mimic endogenous H2S production. This stark contrast drives the need to develop better sources of caged H2S. To address these limitations, different small organosulfur donor compounds have been prepared that release H2S in the presence of specific activators or triggers. Such compounds, however, often lack suitable  control compounds, which limits the use of these compounds in probing the effects of H2S directly. To address these needs, our group has pioneered the development of carbonyl sulfide (COS) releasing compounds as a new class of H2S donor motifs. Inspired by a commonly used carbamate prodrug scaffold, our approach utilizes self-immolative thiocarbamates to access controlled release of COS, which is rapidly converted to H2S by the ubiquitous enzyme carbonic anhydrase (CA). In addition, this design enables access to key control compounds that release CO2/H2O rather than COS/H2S, which enables delineation of the effects of COS/H2S from the organic donor byproducts. In this Account, we highlight a library of first-generation COS/H2S donors based on self-immolative thiocarbamates developed in our lab and also highlight challenges related to H2S donor development. We showcase the release of COS in the presence of specific triggers and activators, including biological thiols and bio-orthogonal reactants for targeted applications. We also demonstrate the design and development of a series of H2O2/reactive oxygen species (ROS)-triggered donors and show that such compounds can be activated by endogenous levels of ROS production. Utilizing approaches in bio-orthogonal activation, we establish that donors functionalized with an o-nitrobenzyl photocage can enable access to light-activated donors. Similar to endogenous production by cysteine catabolism, we also prepared a cysteine-selective COS donor activated by a Strongin ligation mechanism. In efforts to help delineate potential differences in the chemical biology of COS and H2S, we also report a simple esterase-activated donor, which demonstrated fast COS-releasing kinetics and inhibition of mitochondrial respiration in BEAS-2B cells. Additional investigations revealed that COS release rates and cytotoxicity correlated directly within this series of compounds with different ester motifs. In more recent and applied applications of this H2S donation strategy, we also highlight the development of donors that generate either a colorimetric or fluorescent optical response upon COS release. Overall, the work described in this Account outlines the development and initial application of a new class of H2S donors, which we anticipate will help to advance our understanding of the rapidly emerging chemical biology of H2S and COS.


Subject(s)
Carbonic Anhydrases/metabolism , Hydrogen Sulfide/metabolism , Sulfur Oxides/metabolism , Animals , Carbonic Anhydrases/chemistry , Cell Survival/drug effects , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Hydrogen Sulfide/chemistry , Mice , Molecular Structure , RAW 264.7 Cells , Sulfur Oxides/chemical synthesis , Sulfur Oxides/chemistry , Thiocarbamates/chemistry , Thiocarbamates/metabolism , Thiocarbamates/pharmacology
16.
Anticancer Drugs ; 31(7): 672-683, 2020 08.
Article in English | MEDLINE | ID: mdl-32282370

ABSTRACT

The anticancer drug candidates 1,3-dibenzyl-4,5-diphenyl-imidazol-2-ylidene gold(I) dimethylamino dithiocarbamate and 2,3,4,6-tetra-O-acetyl-α-D-glucopyranosyl-1-thiolate derivative exhibited nanomolar in-vitro activity against prostate cancer cells advanced prostate cancer (PC3) and micromolar inhibition of mammalian thioredoxin reductase. Encouraging maximum tolerable dose experiments led to human prostate cancer subcutaneous xenograft experiments; 1,3-dibenzyl-4,5-diphenyl-imidazol-2-ylidene gold(I) dimethylamino dithiocarbamate and 2,3,4,6-tetra-O-acetyl-α-D-glucopyranosyl-1-thiolate derivative were applied twelve times at two doses in groups of n = 5 PC3 to tumor-bearing NMRI:nu/nu mice. 1,3-dibenzyl-4,5-diphenyl-imidazol-2-ylidene gold(I) dimethylamino dithiocarbamate and 2,3,4,6-tetra-O-acetyl-α-D-glucopyranosyl-1-thiolate derivative at the dose of 10 and 20 mg/kg showed good tolerability, while no significant body weight loss was seen in both groups. In particular, for the drug 1,3-dibenzyl-4,5-diphenyl-imidazol-2-ylidene gold(I) dimethylamino dithiocarbamate the tumor growth inhibition suggested to be dose dependent, reflected by the respective optimal T/C values of 0.45 at the dose of 10 mg/kg and of 0.31 at the dose of 20 mg/kg. By contrast, the 2,3,4,6-tetra-O-acetyl-α-D-glucopyranosyl-1-thiolate derivative treated groups showed no indication for dose-dependent antitumoral activity, as reflected by the optimal T/C values of 0.44 for the 10 mg/kg and for the 20 mg/kg treated mice. Immunohistochemical experiments involving Ki67 staining of tumor tissue showed that both compounds reduced PC3 cell proliferation against the difficult to treat advanced human prostate tumors derived from PC3.


Subject(s)
Gold/pharmacology , Organometallic Compounds/pharmacology , Prostatic Neoplasms/drug therapy , Thiocarbamates/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Growth Processes/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/metabolism , Gold/chemistry , Humans , Male , Mice , Mice, Nude , Molecular Docking Simulation , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , PC-3 Cells , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Random Allocation , Sulfhydryl Compounds/chemical synthesis , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology , Thiocarbamates/chemical synthesis , Thiocarbamates/chemistry , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Xenograft Model Antitumor Assays
17.
Bioorg Med Chem Lett ; 30(11): 127148, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32253061

ABSTRACT

Disulfiram in conjunction with copper has been shown to be a potent anticancer agent. However, disulfiram's therapeutic potential in prostate cancer is hindered by off-target effects due to its reactive and nucleophilic thiol-containing component, diethyldithiocarbamate (DTC). To minimize undesirable reactivity, we have strategically blocked the thiol moiety in DTC with a cleavable p-aminobenzyl (pAB) group linked to peptide substrates recognized by prostate specific antigen (PSA). Here we report the synthesis and evaluation in cancer cell models of two PSA-activatable prodrugs: HPD (Ac-HSSKLQL-pAB-DTC and RPD (RSSYYSL-pAB-DTC). In vitro exposure to PSA was found to trigger activation of HPD and RPD to release diethyldithiocarbamate, and both prodrugs were found to induce toxicity in prostate cancer cells, with HPD showing the most promising selectivity. With copper supplementation, the IC50 of HPD was 1.4 µM in PSA-expressing LNCaP cells, and 11 µM in PC3 cells that do not express PSA. These studies demonstrate the utility of using peptide recognition handles to direct the activity of dithiocarbamate prodrugs for selective cytotoxicity of cancer cells.


Subject(s)
Prodrugs/chemistry , Prostate-Specific Antigen/chemistry , Thiocarbamates/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Copper/chemistry , Humans , Male , Prodrugs/metabolism , Prodrugs/pharmacology , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/pathology , Thiocarbamates/metabolism , Thiocarbamates/pharmacology
18.
Org Biomol Chem ; 18(7): 1369-1376, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31996878

ABSTRACT

This paper discloses a transition metal-free selective C-H dithiocarbamation of drug skeletons using disulfiram (DSF) in the presence of KI/K2S2O8 in DMF/H2O. Drug skeletons, including 5-aminopyrazoles, indoles, pyrroloquinoline, and Julolidine, underwent C-H dithiocarbamation smoothly to afford a variety of drug-like molecules in moderate to good yields. It was found that the in situ formed 5-aminopyrazole iodide is the key intermediate for the dithiocarbamation. Bioassay results show that some of these N-heterocyclic dithiocarbamate derivatives exhibit good antifungal activity against Colletotrichum gloeosprioides and Fusarium oxysporum, F. proliferatum, Fusarium solani, Geotrichum candidum, Penicillium digitatum, Penicillium italicum, Phyricularia grisea.


Subject(s)
Antifungal Agents/pharmacology , Colletotrichum/drug effects , Fusarium/drug effects , Geotrichum/drug effects , Penicillium/drug effects , Thiocarbamates/pharmacology , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Benzene Derivatives/chemistry , Microbial Sensitivity Tests , Molecular Structure , Stereoisomerism , Thiocarbamates/chemical synthesis , Thiocarbamates/chemistry
19.
J Biochem Mol Toxicol ; 34(11): e22581, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32662241

ABSTRACT

Consecutive substitution reactions of arsenic(III)chloride with sodium salts of various oximes and morpholinedithiocarbamate (morphdtc) were carried out in 1:2:1 stoichiometry to obtain six new arsenic(III) mixed derivatives of the type: [(R)(R1 )C = NO]2 Sb[S2 CN(CH2 CH2 )2 O] [where R is -C6 H5 , R1 = -CH3 (1); R = -C6 H4 CH3 , R1 = -CH3 (2); R = -C6 H4 Cl, R1 = -CH3 (3); R = -C6 H4 Br, R1 = -CH3 (4); R = -C6 H4 OH, R1 = -H (5); R(R1 )C = (6)]. These derivatives are characterized by elemental and physicochemical analyses and the tentative distorted trigonalbipyramidal geometry around arsenic assigned using spectral data of infrared (1 H, 13 C) nuclear magnetic resonance and liquid chromatography-mass. Powder X-ray diffraction study revealed their nanoranged particle size to be approximately 40 nm and crystalline nature. These derivatives examined against microbes and results revealed that these derivatives expressed more antifungal potential than antibacterial. The antioxidant activity was carried out by ferric reducing ability of plasma assay, and the cytotoxic study was performed in 3T3 fibroblast cell lines by tetrazolium-based colorimetric assay.


Subject(s)
Anti-Infective Agents/pharmacology , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Arsenicals/pharmacology , Morpholines/pharmacology , Oximes/pharmacology , Thiocarbamates/pharmacology , Microbial Sensitivity Tests , Morpholines/chemistry , Spectrum Analysis/methods , Thiocarbamates/chemistry
20.
Bioorg Chem ; 104: 104174, 2020 11.
Article in English | MEDLINE | ID: mdl-32932119

ABSTRACT

Cathepsins have emerged as important targets in various tissues degenerative disorders due to their involvement in degradation of extracellular matrices and endogenous protein turnover. Elevated cathepsins levels vis-à-vis decreased concentration of endogenous inhibitors has been reported at different diseased sites. The design and synthesis of specific potential anti-cathepsin agents is therefore of great significance. Most of potential anti-cathepsin agents developed have peptide based structures with an active warhead. Due to oral instability and immunogenic problems related to peptidyl inhibitors drift the synthesis and evaluation of non-peptide cathepsin inhibitors in last two decades. The present work provides a detailed structure activity relationship for developing potential non-peptide anticathepsin agents based on in-vitro inhibition studies of a library of synthesized thiocarbamoyl- non-peptide inhibitors.


Subject(s)
Cathepsin B/antagonists & inhibitors , Cathepsin H/antagonists & inhibitors , Cathepsin L/antagonists & inhibitors , Protease Inhibitors/pharmacology , Thiocarbamates/pharmacology , Cathepsin B/isolation & purification , Cathepsin B/metabolism , Cathepsin H/isolation & purification , Cathepsin H/metabolism , Cathepsin L/isolation & purification , Cathepsin L/metabolism , Dose-Response Relationship, Drug , Humans , Molecular Structure , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Structure-Activity Relationship , Thiocarbamates/chemical synthesis , Thiocarbamates/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL