Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 107
1.
Dalton Trans ; 53(18): 7866-7879, 2024 May 07.
Article En | MEDLINE | ID: mdl-38632950

Rhenium(I) tricarbonyl complexes are widely studied for their cell imaging properties and anti-cancer and anti-microbial activities, but the complexes with S-donor ligands remain relatively unexplored. A series of six fac-[Re(NN)(CO)3(SR)] complexes, where (NN) is 2,2'-bipyridyl (bipy) or 1,10-phenanthroline (phen), and RSH is a series of thiocarboxylic acid methyl esters, have been synthesized and characterized. Cellular uptake and anti-proliferative activities of these complexes in human breast cancer cell lines (MDA-MB-231 and MCF-7) were generally lower than those of the previously described fac-[Re(NN)(CO)3(OH2)]+ complexes; however, one of the complexes, fac-[Re(CO)3(phen)(SC(Ph)CH2C(O)OMe)] (3b), was active (IC50 ∼ 10 µM at 72 h treatment) in thiol-depleted MDA-MB-231 cells. Moreover, unlike fac-[Re(CO)3(phen)(OH2)]+, this complex did not lose activity in the presence of extracellular glutathione. Taken together these properties show promise for further development of 3b and its analogues as potential anti-cancer drugs for co-treatment with thiol-depleting agents. Conversely, the stable and non-toxic complex, fac-[Re(bipy)(CO)3(SC(Me)C(O)OMe)] (1a), predominantly localized in the lysosomes of MDA-MB-231 cells, as shown by live cell confocal microscopy (λex = 405 nm, λem = 470-570 nm). It is strongly localized in a subset of lysosomes (25 µM Re, 4 h treatment), as shown by co-localization with a Lysotracker dye. Longer treatment times with 1a (25 µM Re for 48 h) resulted in partial migration of the probe into the mitochondria, as shown by co-localization with a Mitotracker dye. These properties make complex 1a an attractive target for further development as an organelle probe for multimodal imaging, including phosphorescence, carbonyl tag for vibrational spectroscopy, and Re tag for X-ray fluorescence microscopy.


Antineoplastic Agents , Cell Proliferation , Coordination Complexes , Rhenium , Sulfur , Humans , Rhenium/chemistry , Rhenium/pharmacology , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Ligands , Sulfur/chemistry , Sulfur/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Molecular Structure
2.
Inorg Chem ; 62(43): 17804-17817, 2023 Oct 30.
Article En | MEDLINE | ID: mdl-37858311

Limited stability of most transition-metal complexes in biological media has hampered their medicinal applications but also created a potential for novel cancer treatments, such as intratumoral injections of cytotoxic but short-lived anticancer drugs. Two related V(V) complexes, [VO(Hshed)(dtb)] (1) and [VO(Hshed)(cat)] (2), where H2shed = N-(salicylideneaminato)-N'-(2-hydroxyethyl)-1,2-ethanediamine, H2dtb = 3,5-di-tert-butylcatechol, and H2cat = 1,2-catechol, decomposed within minutes in cell culture medium at 310 K (t1/2 = 43 and 9 s for 1 and 2, respectively). Despite this, both complexes showed high antiproliferative activities in triple-negative human breast cancer (MDA-MB-231) cells, but the mechanisms of their activities were radically different. Complex 1 formed noncovalent adducts with human serum albumin, rapidly entered cells via passive diffusion, and was nearly as active in a short-term treatment (IC50 = 1.9 ± 0.2 µM at 30 min) compared with a long-term treatment (IC50 = 1.3 ± 0.2 µM at 72 h). The activity of 1 decreased about 20-fold after its decomposition in cell culture medium for 30 min at 310 K. Complex 2 showed similar activities (IC50 ≈ 12 µM at 72 h) in both fresh and decomposed solutions and was inactive in a short-term treatment. The activity of 2 was mainly due to the reactions among V(V) decomposition products, free catechol, and O2 in cell culture medium. As a result, the activity of 1 was less sensitive than that of 2 to the effects of hypoxic conditions that are characteristic of solid tumors and to the presence of apo-transferrin that acts as a scavenger of V(V/IV) decomposition products in blood serum. In summary, complex 1, but not 2, is a suitable candidate for further development as an anticancer drug delivered via intratumoral injections. These results demonstrate the importance of fine-tuning the ligand properties for the optimization of biological activities of metal complexes.


Coordination Complexes , Organometallic Compounds , Humans , Coordination Complexes/pharmacology , Vanadium/pharmacology , Organometallic Compounds/pharmacology , Transferrin , Albumins , Hypoxia , Catechols/pharmacology
3.
Chemistry ; 29(68): e202302271, 2023 Dec 06.
Article En | MEDLINE | ID: mdl-37581946

Two new series of complexes with pyridine-containing Schiff bases, [VV O(SALIEP)L] and [VV O(Cl-SALIEP)L] (SALIEP=N-(salicylideneaminato)-2-(2-aminoethylpyridine; Cl-SALIEP=N-(5-chlorosalicylideneaminato)-2-(2-aminoethyl)pyridine, L=catecholato(2-) ligand) have been synthesized. Characterization by 1 H and 51 V NMR and UV-Vis spectroscopies confirmed that: 1) most complexes form two major geometric isomers in solution, and [VV O(SALIEP)(DTB)] (DTB=3,5-di-tert-butylcatecholato(2-)) forms two isomers that equilibrate in solution; and 2) tert-butyl substituents were necessary to stabilize the reduced VIV species (EPR spectroscopy and cyclic voltammetry). The pyridine moiety within the Schiff base ligands significantly changed their chemical properties with unsubstituted catecholate ligands compared with the parent HSHED (N-(salicylideneaminato)-N'-(2-hydroxyethyl)-1,2-ethanediamine) Schiff base complexes. Immediate reduction to VIV occurred for the unsubstituted-catecholato VV complexes on dissolution in DMSO. By contrast, the pyridine moiety within the Schiff base significantly improved the hydrolytic stability of [VV O(SALIEP)(DTB)] compared with [VV O(HSHED)(DTB)]. [VV O(SALIEP)(DTB)] had moderate stability in cell culture media. There was significant cellular uptake of the intact complex by T98G (human glioblastoma) cells and very good anti-proliferative activity (IC50 6.7±0.9 µM, 72 h), which was approximately five times higher than for the non-cancerous human cell line, HFF-1 (IC50 34±10 µM). This made [VV O(SALIEP)(DTB)] a potential drug candidate for the treatment of advanced gliomas by intracranial injection.


Antineoplastic Agents , Coordination Complexes , Glioblastoma , Organometallic Compounds , Humans , Vanadium/chemistry , Schiff Bases/chemistry , Organometallic Compounds/chemistry , Glioblastoma/drug therapy , Antineoplastic Agents/chemistry , Pyridines/chemistry , Electron Spin Resonance Spectroscopy , Oxidation-Reduction , Ligands , Coordination Complexes/pharmacology
4.
Chemistry ; 29(54): e202203323, 2023 Sep 26.
Article En | MEDLINE | ID: mdl-37385951

As shown by IncuCyte Zoom imaging proliferation assays, invasive triple-negative human breast MDA-MB-231 cancer cells treated with sub-toxic doses (5.0-20 µM, 72 h) of [GaQ3 ] (Q=8-hydroxyquinolinato) caused profound morphological changes and inhibition of cell migration, which were likely due to terminal cell differentiation or similar phenotypical change. This is the first demonstration of potential use of a metal complex in differentiation anti-cancer therapy. Additionally, a trace amount of Cu(II) (0.20 µM) added to the medium dramatically increased [GaQ3 ] cytotoxicity (IC50 ~2 µM, 72 h) due to its partial dissociation and the action of the HQ ligand as a Cu(II) ionophore, as shown with electrospray mass spectrometry and fluorescence spectroscopy assays in the medium. Hence, cytotoxicity of [GaQ3 ] is strongly linked to ligand binding of essential metal ions in the medium, for example, Cu(II). Appropriate delivery mechanisms of such complexes and their ligands could enable a powerful new triple therapeutic approach for cancer chemotherapy, including cytotoxicity against primary tumour, arrest of metastases, and activation of innate and adaptive immune responses.


Antineoplastic Agents , Coordination Complexes , Humans , Copper/chemistry , Ligands , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Metals/pharmacology , Cell Proliferation , Cell Line, Tumor
5.
Chem Commun (Camb) ; 59(45): 6877-6880, 2023 Jun 01.
Article En | MEDLINE | ID: mdl-37195631

A new photoluminescent polypyridylruthenium(II) stain for extracellular vesicles (EVs) released from lipopolysaccharide-stimulated THP-1 monocytes enabled important new insights into how the bacteria-induced immune system affects the blood-brain barrier (BBB). These included previously unknown aspects of EV interactions with BBB microvascular endothelial cells and the extracellular matrix relevant to human brain diseases.


Endothelial Cells , Extracellular Vesicles , Humans , Endothelium , Brain , Blood-Brain Barrier
6.
Chemosphere ; 329: 138673, 2023 Jul.
Article En | MEDLINE | ID: mdl-37054846

The regular incremental secretion of enamel and dentine can be interrupted during periods of stress resulting in accentuated growth lines. These accentuated lines, visible under light microscopy, provide a chronology of an individual's stress exposure. Previously, we showed that small biochemical changes along accentuated growth lines detected by Raman spectroscopy, coincided with the timing of medical history events and disruptions of weight trajectory in teeth from captive macaques. Here, we translate those techniques to study biochemical changes related to illness and prolonged medical treatment during early infancy in humans. Chemometric analysis revealed biochemical changes related to known stress-induced changes in circulating phenylalanine as well as other biomolecules. Changes in phenylalanine are also known to affect biomineralization which is reflected in changes in the wavenumbers of hydroxyapatite phosphate bands associated with stress in the crystal lattice. Raman spectroscopy mapping of teeth is an objective, minimally-destructive technique that can aid in the reconstruction of an individual's stress response history and provide important information on the mixture of circulating biochemicals associated with medical conditions, as applied in epidemiological and clinical samples.


Tooth , Humans , Tooth/chemistry , Microscopy , Spectrum Analysis, Raman
7.
Dalton Trans ; 52(15): 4835-4848, 2023 Apr 11.
Article En | MEDLINE | ID: mdl-36939381

Twelve Re(I) tricarbonyl diimine (2,2'-bipyridine and 1,10-phenanthroline) complexes with thiotetrazolato ligands have been synthesised and fully characterised. Structural characterisation revealed the capacity of the tetrazolato ligand to bind to the Re(I) centre through either the S atom or the N atom with crystallography revealing most complexes being bound to the N atom. However, an example where the Re(I) centre is linked via the S atom has been identified. In solution, the complexes exist as an equilibrating mixture of linkage isomers, as suggested by comparison of their NMR spectra at room temperature and 373 K, as well as 2D exchange spectroscopy. The complexes are photoluminescent in fluid solution at room temperature, with emission either at 625 or 640 nm from the metal-to-ligand charge transfer excited states of triplet multiplicity, which seems to be exclusively dependent on the nature of the diimine ligand. The oxygen-sensitive excited state lifetime decay ranges between 12.5 and 27.5 ns for the complexes bound to 2,2'-bipyrdine, or between 130.6 and 155.2 ns for those bound to 1.10-phenanthroline. Quantum yields were measured within 0.4 and 1.5%. The complexes were incubated with human lung (A549), brain (T98g), and breast (MDA-MB-231) cancer cells, as well as with normal human skin fibroblasts (HFF-1), revealing low to moderate cytotoxicity, which for some compounds exceeded that of a standard anti-cancer drug, cisplatin. Low cytotoxicity combined with significant cellular uptake and photoluminescence properties provides potential for their use as cellular imaging agents. Furthermore, the complexes were assessed in disc diffusion and broth microdilution assays against methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus (VRE), Escherichia coli (E. coli), and Pseudomonas aeruginosa (P. aeruginosa) bacterial strains, which revealed negligible antibacterial activity in the dark or after irradiation.


Antineoplastic Agents , Coordination Complexes , Methicillin-Resistant Staphylococcus aureus , Humans , Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Escherichia coli , Ligands , Rhenium
8.
J Biol Inorg Chem ; 28(1): 43-55, 2023 02.
Article En | MEDLINE | ID: mdl-36469143

Imbalances in metal homeostasis have been implicated in the progression and drug response of cancer cells. Understanding these changes will enable identification of new treatment regimes and precision medicine approaches to cancer treatment. In particular, there has been considerable interest in the interplay between copper homeostasis and response to platinum-based chemotherapeutic agents. Here, we have studied differences in the Cu uptake and distributions in the ovarian cancer cell line, A2780, and its cisplatin resistant form, A2780.CisR, by measuring total Cu content and the bioavailable Cu pool. Atomic absorption spectroscopy (AAS) revealed a lower total Cu uptake in A2780.CisR compared to A2780 cells. Conversely, live-cell confocal microscopy studies with the ratiometric Cu(I)-sensitive fluorescent dye, InCCu1, revealed higher relative cellular content of labile Cu in A2780.CisR cells compared with A2780 cells. These results demonstrate that Cu trafficking, homeostasis and speciation are different in the Pt-sensitive and resistant cells and may be associated with the predominance of different phenotypes for A2780 (epithelial) and A2780.CisR (mesenchymal) cells.


Antineoplastic Agents , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Copper/pharmacology , Fluorescent Dyes , Cell Line, Tumor , Drug Resistance, Neoplasm , Organoplatinum Compounds/metabolism , Cisplatin/pharmacology
9.
J Inorg Biochem ; 239: 112082, 2023 02.
Article En | MEDLINE | ID: mdl-36459780

Tumour cell heterogeneity affects cisplatin but not doxorubicin cytotoxicity in two phenotypes of the triple-negative breast cancer (TNBC) cell line, MDA-MB-231. A mesenchymal MDA-MB-231 phenotype was three times less sensitive to cisplatin than an epithelial-type phenotype from the same cell line, which has important implications in the success of TNBC chemotherapies.


Cisplatin , Triple Negative Breast Neoplasms , Humans , Cisplatin/pharmacology , Cisplatin/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Cell Proliferation , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use
10.
Inorg Chem ; 61(51): 20757-20773, 2022 Dec 26.
Article En | MEDLINE | ID: mdl-36519680

A hydrophobic Schiff base catecholate vanadium complex was recently discovered to have anticancer properties superior to cisplatin and suited for intratumoral administration. This [VO(HSHED)(DTB)] complex, where HSHED is N-(salicylideneaminato)-N'-(2-hydroxyethyl)-1,2-ethanediamine and the non-innocent catecholato ligand is di-t-butylcatecholato (DTB), has higher stability compared to simpler catecholato complexes. Three new chloro-substituted Schiff base complexes of vanadium(V) with substituted catecholates as co-ligands were synthesized for comparison with their non-chlorinated Schiff base vanadium complexes, and their properties were characterized. Up to four geometric isomers for each complex were identified in organic solvents using 51V and 1H NMR spectroscopies. Spectroscopy was used to characterize the structure of the major isomer in solution and to demonstrate that the observed isomers are exchanged in solution. All three chloro-substituted Schiff base vanadium(V) complexes with substituted catecholates were also characterized by UV-vis spectroscopy, mass spectrometry, and electrochemistry. Upon testing in human glioblastoma multiforme (T98g) cells as an in vitro model of brain gliomas, the most sterically hindered, hydrophobic, and stable compound [t1/2 (298 K) = 15 min in cell medium] was better than the two other complexes (IC50 = 4.1 ± 0.5 µM DTB, 34 ± 7 µM 3-MeCat, and 19 ± 2 µM Cat). Furthermore, upon aging, the complexes formed less toxic decomposition products (IC50 = 9 ± 1 µM DTB, 18 ± 3 µM 3-MeCat, and 8.1 ± 0.6 µM Cat). The vanadium complexes with the chloro-substituted Schiff base were more hydrophobic, more hydrolytically stable, more easily reduced compared to their corresponding parent counterparts, and the most sterically hindered complex of this series is only the second non-innocent vanadium Schiff base complex with a potent in vitro anticancer activity that is an order of magnitude more potent than cisplatin under the same conditions.


Coordination Complexes , Vanadium , Humans , Vanadium/pharmacology , Vanadium/chemistry , Cisplatin , Schiff Bases/pharmacology , Schiff Bases/chemistry , Water , Magnetic Resonance Spectroscopy , Coordination Complexes/pharmacology , Ligands
11.
Biomolecules ; 12(9)2022 09 18.
Article En | MEDLINE | ID: mdl-36139158

Ruthenium complexes are at the forefront of developments in metal-based anticancer drugs, but many questions remain open regarding their reactivity in biological media, including the role of transferrin (Tf) in their transport and cellular uptake. A well-known anticancer drug, KP1019 ((IndH)[RuIIICl4(Ind)2], where Ind = indazole) and a reference complex, [RuIII(nta)2]3- (nta = nitrilotriacetato(3-)) interacted differently with human apoTf, monoFeTf, or Fe2Tf. These reactions were studied by biolayer interferometry (BLI) measurements of Ru-Fe-Tf binding to recombinant human transferrin receptor 1 (TfR1) in conjunction with UV-vis spectroscopy and particle size analysis. Cellular Ru uptake in human hepatoma (HepG2) cells was measured under the conditions of the BLI assays. The mode of Tf binding and cellular Ru uptake were critically dependent on the nature of Ru complex, availability of Fe(III) binding sites of Tf, and the presence of proteins that competed for metal binding, particularly serum albumin. Cellular uptake of KP1019 was not Tf-mediated and occurred mostly by passive diffusion, which may also be suitable for treatments of inoperable cancers by intratumoral injections. High cellular Ru uptake from a combination of [RuIII(nta)2]3- and Fe2Tf in the absence of significant Ru-Tf binding was likely to be due to trapping of Ru(III) species into the endosome during TfR1-mediated endocytosis of Fe2Tf.


Antineoplastic Agents , Organometallic Compounds , Ruthenium , Antineoplastic Agents/chemistry , Ferric Compounds/metabolism , Humans , Indazoles/chemistry , Organometallic Compounds/chemistry , Receptors, Transferrin/metabolism , Ruthenium/chemistry , Ruthenium/pharmacology , Ruthenium Compounds , Serum Albumin , Transferrin/metabolism
12.
Pharmaceutics ; 14(4)2022 Apr 04.
Article En | MEDLINE | ID: mdl-35456624

Injections of highly cytotoxic or immunomodulating drugs directly into the inoperable tumor is a procedure that is increasingly applied in the clinic and uses established Pt-based drugs. It is advantageous for less stable anticancer metal complexes that fail administration by the standard intravenous route. Such hydrophobic metal-containing complexes are rapidly taken up into cancer cells and cause cell death, while the release of their relatively non-toxic decomposition products into the blood has low systemic toxicity and, in some cases, may even be beneficial. This concept was recently proposed for V(V) complexes with hydrophobic organic ligands, but it can potentially be applied to other metal complexes, such as Ti(IV), Ga(III) and Ru(III) complexes, some of which were previously unsuccessful in human clinical trials when administered via intravenous injections. The potential beneficial effects include antidiabetic, neuroprotective and tissue-regenerating activities for V(V/IV); antimicrobial activities for Ga(III); and antimetastatic and potentially immunogenic activities for Ru(III). Utilizing organic ligands with limited stability under biological conditions, such as Schiff bases, further enhances the tuning of the reactivities of the metal complexes under the conditions of intratumoral injections. However, nanocarrier formulations are likely to be required for the delivery of unstable metal complexes into the tumor.

13.
Cells ; 10(7)2021 07 05.
Article En | MEDLINE | ID: mdl-34359866

Biochemical changes in specific organelles underpin cellular function, and studying these changes is crucial to understand health and disease. Fluorescent probes have become important biosensing and imaging tools as they can be targeted to specific organelles and can detect changes in their chemical environment. However, the sensing capacity of fluorescent probes is highly specific and is often limited to a single analyte of interest. A novel approach to imaging organelles is to combine fluorescent sensors with vibrational spectroscopic imaging techniques; the latter provides a comprehensive map of the relative biochemical distributions throughout the cell to gain a more complete picture of the biochemistry of organelles. We have developed NpCN1, a bimodal fluorescence-Raman probe targeted to the lipid droplets, incorporating a nitrile as a Raman tag. NpCN1 was successfully used to image lipid droplets in 3T3-L1 cells in both fluorescence and Raman modalities, reporting on the chemical composition and distribution of the lipid droplets in the cells.


Fluorescent Dyes/chemistry , Optical Imaging , Spectrum Analysis, Raman , 3T3-L1 Cells , Animals , Cell Survival , Fluorescence , Mice , Naphthalimides/chemistry , Principal Component Analysis
14.
Gels ; 8(1)2021 Dec 27.
Article En | MEDLINE | ID: mdl-35049554

Transferrin (Tf) is a crucial transporter protein for Fe(III), but its biological role in binding other metal ions and their delivery into cells remain highly controversial. The first systematic exploration of the effect of non-Fe(III) metal ion binding on Tf conformation has been performed by urea-polyacrylamide gel electrophoresis (urea-PAGE), which is commonly used for nucleic acids but rarely for proteins. Closed Tf conformation, similar to that caused by Fe(III)-Tf binding, was formed for In(III), V(III) or Cr(III) binding to Tf. In all these cases, metal distribution between Tf lobes and/or the rate of metal release under acidic conditions differed from that of Fe(III)-Tf. By contrast, Ga(III) and V(IV) did not form closed Tf conformation under urea-PAGE conditions. Apart from Fe(III), only In(III) was able to increase the proportion of closed Tf conformation in whole serum. These results suggest that Tf is unlikely to act as a natural carrier of any metal ion, except Fe(III), into cells but can reduce toxicity of exogenous metal ions by binding them in serum and preventing their entry into cells.

15.
Inorg Chem ; 59(22): 16143-16153, 2020 Nov 16.
Article En | MEDLINE | ID: mdl-32578416

The role of vanadium binding to transferrin (Tf) in the biological activities of vanadium-based drugs is a matter of considerable debate. In order to determine whether V(V) and/or V(IV) binding to Tf (in apo, monoferric(III), and diferric(III) forms) enhances or inhibits biological activities, cellular V uptake and in vitro antiproliferative activity were examined in the presence and absence of different forms of Tf and other biomolecules under normoxic conditions. These data were combined with studies on V-Tf binding in cell culture medium and its role in Tf interactions with transferrin receptor 1 (TfR1), using the biolayer interferometry (BLI) model of the Tf cycle that was developed in our group. The results showed that both V(V) and V(IV) oxidation states efficiently bind to vacant Fe(III) binding sites of Tf even in the presence of a 20-fold molar excess of albumin, although V does not displace Tf-bound Fe(III) under these conditions. Binding of V(V) or V(IV) to Tf in cell culture medium drastically reduced its cellular uptake and antiproliferative activity in the A549 (human lung cancer) cell line that expresses TfR1. BLI and gel electrophoresis studies showed that V(V/IV) binding to partially Fe(III) saturated Tf did not enhance the affinity of Tf binding to TfR1 at pH 7.4 but did disrupt Tf conformational changes under endosome-mimicking conditions (pH 5.6, 0.10 mM citrate). Hence, it is postulated that the absence of a significant cellular uptake of Tf-bound V(V/IV) is likely to be due to the return of undissociated V(V/IV)-Tf adducts to the cell surface after the endosomal step. Collectively, these data show that the biotransformation of V-based drugs leads to V(V/IV)-Tf binding in the blood serum and inhibits, rather than enhances, the biological activity of such drugs under aerobic conditions. These results indicate that the design of V-based drugs that are stable enough to survive in the blood, enter cells intact, and release the active components intracellularly is likely to be required for their clinical success.


Antineoplastic Agents/pharmacology , Transferrin/pharmacology , Vanadium/pharmacology , A549 Cells , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Molecular Conformation , Transferrin/chemistry , Transferrin/metabolism , Tumor Cells, Cultured , Vanadium/chemistry , Vanadium/metabolism
16.
Angew Chem Int Ed Engl ; 59(37): 15834-15838, 2020 09 07.
Article En | MEDLINE | ID: mdl-32598089

The chemistry and short lifetimes of metal-based anti-cancer drugs can be turned into an advantage for direct injections into tumors, which then allow the use of highly cytotoxic drugs. The release of their less toxic decomposition products into the blood will lead to decreased toxicity and can even have beneficial effects. We present a ternary VV complex, 1 ([VOL1 L2 ], where L1 is N-(salicylideneaminato)-N'-(2-hydroxyethyl)ethane-1,2-diamine and L2 is 3,5-di-tert-butylcatechol), which enters cells intact to induce high cytotoxicity in a range of human cancer cells, including T98g (glioma multiforme), while its decomposition products in cell culture medium were ≈8-fold less toxic. 1 was 12-fold more toxic than cisplatin in T98g cells and 6-fold more toxic in T98g cells than in a non-cancer human cell line, HFF-1. Its high toxicity in T98g cells was retained in the presence of physiological concentrations of the two main metal-binding serum proteins, albumin and transferrin. These properties favor further development of 1 for brain cancer treatment by intratumoral injections.


Antineoplastic Agents/chemistry , Brain Neoplasms/drug therapy , Coordination Complexes/chemistry , Vanadium Compounds/chemistry , Antineoplastic Agents/therapeutic use , Brain Neoplasms/pathology , Cell Line, Tumor , Coordination Complexes/therapeutic use , Culture Media , Drug Discovery , Drug Screening Assays, Antitumor , Humans , Proton Magnetic Resonance Spectroscopy , Vanadium Compounds/therapeutic use
17.
PLoS One ; 15(5): e0233300, 2020.
Article En | MEDLINE | ID: mdl-32428015

OBJECTIVE: Damage to locus ceruleus neurons could play a part in the pathogenesis of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis because of impairment of the blood-brain barrier and enhanced neuroinflammation. The locus ceruleus has connections throughout the brain and spinal cord, so the characteristic widespread multifocal pathology in these disorders could be due to damage to different subsets of locus ceruleus neurons. Previous studies have shown that only certain locus ceruleus neurons accumulate the neurotoxic metal mercury. To find out if concentrations of other toxic metals or of essential trace elements also vary between individual locus ceruleus neurons, we used synchrotron X-ray fluorescence microscopy on frozen sections of locus ceruleus neurons taken from people with multiple sclerosis, in whom the locus ceruleus is structurally intact. MATERIALS AND METHODS: Paraffin embedded sections containing the locus ceruleus from seven people with multiple sclerosis were stained with autometallography that demonstrates accumulations of mercury, silver and bismuth. These were compared to maps of multiple elements obtained from frozen sections of locus ceruleus neurons from the same people using X-ray fluorescence microscopy. Neurons in the anterior pons from three of these donors were used as internal controls. RESULTS: Autometallography staining was observed in scattered locus ceruleus neurons from three of the seven donors. X-ray fluorescence microscopy showed variations among individual locus ceruleus neurons in levels of mercury, selenium, iron, copper, lead, bromine, and rubidium. Variations between donors of locus ceruleus neuronal average levels of mercury, iron, copper, and bromine were also detected. Anterior pons neurons contained no mercury, had varied levels of iron, and had lower copper levels than locus ceruleus neurons. CONCLUSIONS: Individual human locus ceruleus neurons contain varying levels of toxic metals and essential trace elements. In contrast, most toxic metals are absent or at low levels in nearby anterior pons neurons. The locus ceruleus plays a role in numerous central nervous system functions, including maintaining the blood-brain-barrier and limiting neuroinflammation. Toxic metals, or alterations in essential trace metals within individual locus ceruleus neurons, could be one factor determining the non-random destruction of locus ceruleus neurons in normal aging and neurodegenerative diseases, and subsequently the sites of the widespread multifocal central nervous system pathology in these disorders.


Locus Coeruleus/metabolism , Metals, Heavy/analysis , Neurons/metabolism , Trace Elements/analysis , Aged , Autopsy , Female , Heavy Metal Poisoning , Humans , Locus Coeruleus/physiology , Middle Aged , Motor Neurons/metabolism , Multiple Sclerosis/metabolism , Spectrometry, X-Ray Emission/methods , Spinal Cord
18.
Chembiochem ; 21(8): 1188-1200, 2020 04 17.
Article En | MEDLINE | ID: mdl-31701616

RuII -arene complexes provide a versatile scaffold for novel anticancer drugs. Seven new RuII -arene-thiocarboxylato dimers were synthesized and characterized. Three of the complexes (2 a, b and 5) showed promising antiproliferative activities in MDA-MB-231 (human invasive breast cancer) cells, and were further tested in a panel of fifteen cancerous and noncancerous cell lines. Complex 5 showed moderate but remarkably selective activity in MDA-MB-231 cells (IC50 =39±4 µm Ru). Real-time proliferation studies showed that 5 induced apoptosis in MDA-MB-231 cells but had no effect in A549 (human lung cancer, epithelial) cells. By contrast, 2 a and b showed moderate antiproliferative activity, but no apoptosis, in either cell line. Selective cytotoxicity of 5 in aggressive, mesenchymal-like MDA-MB-231 cells over many common epithelial cancer cell lines (including noninvasive breast cancer MCF-7) makes it an attractive lead compound for the development of specifically antimetastatic Ru complexes with low systemic toxicity.


Antineoplastic Agents/pharmacology , Apoptosis , Breast Neoplasms/pathology , Carboxylic Acids/chemistry , Coordination Complexes/pharmacology , Ruthenium/chemistry , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Cell Proliferation , Coordination Complexes/chemistry , Drug Screening Assays, Antitumor , Female , Humans , Molecular Structure , Structure-Activity Relationship , Thiophenes/chemistry , Tumor Cells, Cultured
19.
J Inorg Biochem ; 201: 110815, 2019 12.
Article En | MEDLINE | ID: mdl-31520878

Vanadium complexes are intensively tested for anti-cancer activities, particularly for the novel treatment protocols involving injections of cytotoxic compounds directly into the tumor. This approach is increasingly applied to difficult-to-treat cancers, such as pancreatic cancer. The first study of in-vitro anti-cancer properties of a rare stable non-oxido V(V) complex, (NH4)[V(dtbc)3], where dtbcH2 is 3,5-di-tert-butylcatechol, was performed by a combination of end-point viability assays and real-time (IncuCyte) proliferation and cytotoxicity assays in human pancreatic cancer (PANC-1) cells. An improved synthetic procedure led to a nearly quantitative yield of the complex under ambient conditions. Reactions of (NH4)[V(dtbc)3] either in polar organic solvents or in neutral aqueous media led to the formation of V(V)-oxido-catecholato intermediates (characterized by electrospray mass spectrometry) that were responsible for its anti-proliferative and cytotoxic (apoptotic or necrotic) activity (IC50, 3.5-18 µM V in 72 h assays). These results demonstrate the link between solution speciation and biological activity of V complexes. Reaction of (NH4)[V(dtbc)3] with human serum albumin (HSA) in aqueous media led to the formation of protein-bound V(V) oxido-catecholato species that showed high anti-proliferative activity (IC50 ~10 µM V) combined with low cytotoxicity. Formation of HSA adducts of hydrophobic V complexes, such as (NH4)[V(dtbc)3], is a promising way to achieve their sustained delivery to cancer tumors.


Antineoplastic Agents/chemical synthesis , Organometallic Compounds/chemical synthesis , Vanadium Compounds/chemical synthesis , 1-Butanol/chemistry , Antineoplastic Agents/pharmacology , Catechols/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Organometallic Compounds/pharmacology , Serum Albumin/chemistry , Vanadium Compounds/pharmacology
20.
ACS Chem Biol ; 14(5): 893-900, 2019 05 17.
Article En | MEDLINE | ID: mdl-30973710

Fe(III) delivery from blood plasma to cells via the transferrin (Tf) cycle was studied intensively due to its crucial role in Fe homeostasis. Tf-cycle disruptions are linked to anemia, infections, immunodeficiency, and neurodegeneration. Biolayer interferometry (BLI) enabled direct kinetic and thermodynamic measurements for all Tf-cycle steps in a single in vitro experiment using Tf within blood serum or released into the medium by cultured liver cells. In these media, known Tf cycle features were reproduced, and unprecedented insights were gained into conditions of rapid endosomal (pH 5.6) Fe(III) release from the Tf-Tf receptor 1 (TfR1) adduct. This release occurred via synergistic citrate and ascorbate effects, which pointed to respective roles as the likely elusive Fe chelator and reductant within the Tf cycle. These results explain enhanced cellular Fe uptake by ascorbate, the clinical efficacy of anemia treatment with Fe citrate and ascorbate, and dietary effects associated with loss of Fe homeostasis, including the large health burden of infections and neurodegeneration.


Ascorbic Acid/metabolism , Citric Acid/metabolism , Ferric Compounds/metabolism , Transferrin/metabolism , Endosomes/metabolism , Kinetics
...