Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters











Publication year range
1.
JACS Au ; 4(8): 2746-2766, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39211583

ABSTRACT

Photocatalysis is a versatile and rapidly developing field with applications spanning artificial photosynthesis, photo-biocatalysis, photoredox catalysis in solution or supramolecular structures, utilization of abundant metals and organocatalysts, sustainable synthesis, and plastic degradation. In this Perspective, we summarize conclusions from an interdisciplinary workshop of young principal investigators held at the Lorentz Center in Leiden in March 2023. We explore how diverse fields within photocatalysis can benefit from one another. We delve into the intricate interplay between these subdisciplines, by highlighting the unique challenges and opportunities presented by each field and how a multidisciplinary approach can drive innovation and lead to sustainable solutions for the future. Advanced collaboration and knowledge exchange across these domains can further enhance the potential of photocatalysis. Artificial photosynthesis has become a promising technology for solar fuel generation, for instance, via water splitting or CO2 reduction, while photocatalysis has revolutionized the way we think about assembling molecular building blocks. Merging such powerful disciplines may give rise to efficient and sustainable protocols across different technologies. While photocatalysis has matured and can be applied in industrial processes, a deeper understanding of complex mechanisms is of great importance to improve reaction quantum yields and to sustain continuous development. Photocatalysis is in the perfect position to play an important role in the synthesis, deconstruction, and reuse of molecules and materials impacting a sustainable future. To exploit the full potential of photocatalysis, a fundamental understanding of underlying processes within different subfields is necessary to close the cycle of use and reuse most efficiently. Following the initial interactions at the Lorentz Center Workshop in 2023, we aim to stimulate discussions and interdisciplinary approaches to tackle these challenges in diverse future teams.

2.
J Phys Chem Lett ; 15(29): 7430-7435, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39007727

ABSTRACT

Triplet-triplet annihilation upconversion is a bimolecular process converting low-energy photons into high-energy photons. Here, we report a calcium-sensing system working via triplet-triplet annihilation (TTA) upconverted emission. The probe itself was obtained by covalent conjugation of a blue emitter, perylene, with a calcium-chelating moiety, and it was sensitized by the red-light-absorbing photosensitizer palladium(II) tetraphenyltetrabenzoporphyrin (PdTPTBP). Sensing was selective for Ca2+ and occurred in the micromolar domain. In deoxygenated conditions, the TTA upconverted luminescence gradually appeared upon adding an increasing concentration of calcium ions, to reach a maximum upconversion quantum yield of 0.0020.

3.
J Med Chem ; 67(13): 11086-11102, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38924492

ABSTRACT

Photoactivated chemotherapy agents form a new branch of physically targeted anticancer agents with potentially lower systemic side effects for patients. On the other hand, limited information exists on the intracellular interactions between the photoreleased metal cage and the photoreleased anticancer inhibitor. In this work, we report a new biological study of the known photoactivated compound Ru-STF31 in the glioblastoma cancer cell line, U87MG. Ru-STF31 targets nicotinamide phosphoribosyltransferase (NAMPT), an enzyme overexpressed in U87MG. Ru-STF31 is activated by red light irradiation and releases two photoproducts: the ruthenium cage and the cytotoxic inhibitor STF31. This study shows that Ru-STF31 can significantly decrease intracellular NAD+ levels in both normoxic (21% O2) and hypoxic (1% O2) U87MG cells. Strikingly, NAD+ depletion by light activation of Ru-STF31 in hypoxic U87MG cells could not be rescued by the addition of extracellular NAD+. Our data suggest an oxygen-dependent active role of the ruthenium photocage released by light activation.


Subject(s)
Antineoplastic Agents , NAD , Nicotinamide Phosphoribosyltransferase , Oxygen , Ruthenium , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Humans , Ruthenium/chemistry , Ruthenium/pharmacology , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Oxygen/metabolism , NAD/metabolism , Cytokines/metabolism , Light , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis
4.
Planta Med ; 90(7-08): 588-594, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38843798

ABSTRACT

Antimicrobial photodynamic therapy (aPDT) is an evolving treatment strategy against human pathogenic microbes such as the Candida species, including the emerging pathogen C. auris. Using a modified EUCAST protocol, the light-enhanced antifungal activity of the natural compound parietin was explored. The photoactivity was evaluated against three separate strains of five yeasts, and its molecular mode of action was analysed via several techniques, i.e., cellular uptake, reactive electrophilic species (RES), and singlet oxygen yield. Under experimental conditions (λ = 428 nm, H = 30 J/cm2, PI = 30 min), microbial growth was inhibited by more than 90% at parietin concentrations as low as c = 0.156 mg/L (0.55 µM) for C. tropicalis and Cryptococcus neoformans, c = 0.313 mg/L (1.10 µM) for C. auris, c = 0.625 mg/L (2.20 µM) for C. glabrata, and c = 1.250 mg/L (4.40 µM) for C. albicans. Mode-of-action analysis demonstrated fungicidal activity. Parietin targets the cell membrane and induces cell death via ROS-mediated lipid peroxidation after light irradiation. In summary, parietin exhibits light-enhanced fungicidal activity against all Candida species tested (including C. auris) and Cryptococcus neoformans, covering three of the four critical threats on the WHO's most recent fungal priority list.


Subject(s)
Antifungal Agents , Cryptococcus neoformans , Microbial Sensitivity Tests , Antifungal Agents/pharmacology , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/radiation effects , Candida auris/drug effects , Light , Candida/drug effects , Reactive Oxygen Species/metabolism , Photochemotherapy/methods , Anthraquinones/pharmacology , Photosensitizing Agents/pharmacology
5.
Chem Commun (Camb) ; 60(49): 6308-6311, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38818705

ABSTRACT

The famous ''light-switch'' ruthenium complex [Ru(bpy)2(dppz)](PF6)2 (1) has been long known for its DNA binding properties in vitro. However, the biological utility of this compound has been hampered by its poor cellular uptake in living cells. Here we report a bioimaging application of 1 as cell viability probe in both 2D cells monolayer and 3D multi-cellular tumor spheroids of various human cancer cell lines (U87, HepG2, A549). When compared to propidium iodide, a routinely used cell viability probe, 1 was found to enhance the staining of dead cells in particular in tumor spheroids. 1 has high photostability, longer Stokes shift, and displays lower cytotoxicity compared to propidium iodide, which is a known carcinogenic. Finally, 1 was also found to displace the classical DNA binding dye Hoechst in dead cells, which makes it a promising dye for time-dependent imaging of dead cells in cell cultures, including multi-cellular tumor spheroids.


Subject(s)
Cell Survival , Coordination Complexes , DNA , Ruthenium , Spheroids, Cellular , Humans , Cell Survival/drug effects , Spheroids, Cellular/metabolism , Ruthenium/chemistry , DNA/chemistry , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Cell Line, Tumor , Light , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Optical Imaging , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology
6.
Chem Sci ; 15(10): 3596-3609, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38455019

ABSTRACT

In nature, light-driven water oxidation (WO) catalysis is performed by photosystem II via the delicate interplay of different cofactors positioned in its protein scaffold. Artificial systems for homogeneous photocatalytic WO are based on small molecules that often have limited solubility in aqueous solutions. In this work, we alleviated this issue and present a cobalt-based WO-catalyst containing artificial metalloenzyme (ArM) that is active in light-driven, homogeneous WO catalysis in neutral-pH aqueous solutions. A haem-containing electron transfer protein, cytochrome B5 (CB5), served to host a first-row transition-metal-based WO catalyst, CoSalen (CoIISalen, where H2Salen = N,N'-bis(salicylidene)ethylenediamine), thus producing an ArM capable of driving photocatalytic WO. The CoSalen ArM formed a water-soluble pre-catalyst in the presence of [Ru(bpy)3](ClO4)2 as photosensitizer and Na2S2O8 as the sacrificial electron acceptor, with photocatalytic activity similar to that of free CoSalen. During photocatalysis, the CoSalen-protein interactions were destabilized, and the protein partially unfolded. Rather than forming tens of nanometer sized CoOx nanoparticles as free CoSalen does under photocatalytic WO conditions, the CB5 : CoSalen ArM showed limited protein cross-linking and remained soluble. We conclude that a weak, dynamic interaction between a soluble cobalt species and apoCB5 was formed, which generated a catalytically active adduct during photocatalysis. A detailed analysis was performed on protein stability and decomposition processes during the harsh oxidizing reaction conditions of WO, which will serve for the future design of WO ArMs with improved activity and stability.

7.
Phys Rev Lett ; 132(2): 028201, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38277585

ABSTRACT

We show here that soap films-typically expected to host symmetric molecular arrangements-can be constructed with differing opposite surfaces, breaking their symmetry, and making them reminiscent of functional biological motifs found in nature. Using fluorescent molecular probes as dopants on different sides of the film, resonance energy transfer could be employed to confirm the lack of symmetry, which was found to persist on timescales of several minutes. Further, a theoretical analysis of the main transport phenomena involved yielded good agreement with the experimental observations.

8.
Angew Chem Int Ed Engl ; 63(5): e202316425, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38061013

ABSTRACT

Photoactivated chemotherapy (PACT) is a promising cancer treatment modality that kills cancer cells via photochemical uncaging of a cytotoxic drug. Most ruthenium-based photocages used for PACT are activated with blue or green light, which penetrates sub-optimally into tumor tissues. Here, we report amide functionalization as a tool to fine-tune the toxicity and excited states of a terpyridine-based ruthenium photocage. Due to conjugation of the amide group with the terpyridine π system in the excited state, the absorption of red light (630 nm) increased 8-fold, and the photosubstitution rate rose 5-fold. In vitro, red light activation triggered inhibition of tubulin polymerization, which led to apoptotic cell death both in normoxic (21 % O2 ) and hypoxic (1 % O2 ) cancer cells. In vivo, red light irradiation of tumor-bearing mice demonstrated significant tumor volume reduction (45 %) with improved biosafety, thereby demonstrating the clinical potential of this compound.


Subject(s)
Antineoplastic Agents , Neoplasms , Ruthenium , Animals , Mice , Ruthenium/pharmacology , Ruthenium/chemistry , Polymerization , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , Microtubules
9.
J Chem Inf Model ; 63(24): 7816-7825, 2023 Dec 25.
Article in English | MEDLINE | ID: mdl-38048559

ABSTRACT

Despite the proven potential of metal complexes as therapeutics, the lack of computational tools available for the high-throughput screening of their interactions with proteins is a limiting factor toward clinical developments. To address this challenge, we introduce MetalDock, an easy-to-use, open access docking software for docking metal complexes to proteins. Our tool integrates the AutoDock docking engine with three well-known quantum software packages to automate the docking of metal-organic complexes to proteins. We used a Monte Carlo sampling scheme to obtain the missing Lennard-Jones parameters for 12 metal atom types and demonstrated that these parameters generalize exceptionally well. Our results show that the poses obtained by MetalDock are highly accurate, as they predict the binding geometries experimentally determined by crystal structures with high spatial reproducibility. Three different case studies are presented that demonstrate the versatility of MetalDock for the docking of diverse metal-organic compounds to different biomacromolecules, including nucleic acids.


Subject(s)
Coordination Complexes , Access to Information , Reproducibility of Results , Ligands , Proteins/chemistry , Software , Molecular Docking Simulation , Protein Binding
10.
J Am Chem Soc ; 145(43): 23397-23415, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37846939

ABSTRACT

Ruthenium(II) polypyridyl complexes form a vast family of molecules characterized by their finely tuned photochemical and photophysical properties. Their ability to undergo excited-state deactivation via photosubstitution reactions makes them quite unique in inorganic photochemistry. As a consequence, they have been used, in general, for building dynamic molecular systems responsive to light but, more particularly, in the field of oncology, as prodrugs for a new cancer treatment modality called photoactivated chemotherapy (PACT). Indeed, the ability of a coordination bond to be selectively broken under visible light irradiation offers fascinating perspectives in oncology: it is possible to make poorly toxic agents in the dark that become activated toward cancer cell killing by simple visible light irradiation of the compound inside a tumor. In this Perspective, we review the most important concepts behind the PACT idea, the relationship between ruthenium compounds used for PACT and those used for a related phototherapeutic approach called photodynamic therapy (PDT), and we discuss important questions about real-life applications of PACT in the clinic. We conclude this Perspective with important challenges in the field and an outlook.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Photochemotherapy , Ruthenium , Humans , Ruthenium/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Coordination Complexes/chemistry , Light , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/chemistry
11.
J Nat Prod ; 86(10): 2247-2257, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37708055

ABSTRACT

The photoantimicrobial potential of four mushroom species (i.e., Cortinarius cinnabarinus, C. holoxanthus, C. malicorius, and C. sanguineus) was explored by studying the minimal inhibitory concentrations (MIC) via a light-modified broth microdilution assay based on the recommended protocols of the European Committee on Antimicrobial Susceptibility Testing (EUCAST). The extracts were tested against Candida albicans, Escherichia coli, and Staphylococcus aureus under blue (λ = 428 and 478 nm, H = 30 J/cm2) and green light (λ = 528 nm, H = 30 J/cm2) irradiation. Three extracts showed significant photoantimicrobial effects at concentrations below 25 µg/mL. Targeted isolation of the major pigments from C. sanguineus led to the identification of two new potent photoantimicrobials, one of them (i.e., dermocybin) being active against S. aureus and C. albicans under green light irradiation [PhotoMIC530 = 39.5 µM (12.5 µg/mL) and 2.4 µM (0.75 µg/mL), respectively] and the other one (i.e., emodin) being in addition active against E. coli in a low micromolar range [PhotoMIC428 = 11.1 µM (3 µg/mL)]. Intriguingly, dermocybin was not (photo)cytotoxic against the three tested cell lines, adding an additional level of selectivity. Since both photoantimicrobials are not charged, this discovery shifts the paradigm of cationic photosensitizers.


Subject(s)
Antifungal Agents , Photosensitizing Agents , Antifungal Agents/pharmacology , Photosensitizing Agents/pharmacology , Escherichia coli , Staphylococcus aureus , Candida albicans , Anthraquinones/pharmacology , Microbial Sensitivity Tests
12.
Commun Chem ; 6(1): 179, 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37644120

ABSTRACT

Cardiovascular diseases are the leading cause of death worldwide and are not typically diagnosed until the disease has manifested. Endothelial dysfunction is an early, reversible precursor in the irreversible development of cardiovascular diseases and is characterized by a decrease in nitric oxide production. We believe that more reliable and reproducible methods are necessary for the detection of endothelial dysfunction. Both nitric oxide and calcium play important roles in the endothelial function. Here we review different types of molecular sensors used in biological settings. Next, we review the current nitric oxide and calcium sensors available. Finally, we review methods for using both sensors for the detection of endothelial dysfunction.

13.
J Am Chem Soc ; 145(24): 13420-13434, 2023 Jun 21.
Article in English | MEDLINE | ID: mdl-37294954

ABSTRACT

While photosubstitution reactions in metal complexes are usually thought of as dissociative processes poorly dependent on the environment, they are, in fact, very sensitive to solvent effects. Therefore, it is crucial to explicitly consider solvent molecules in theoretical models of these reactions. Here, we experimentally and computationally investigated the selectivity of the photosubstitution of diimine chelates in a series of sterically strained ruthenium(II) polypyridyl complexes in water and acetonitrile. The complexes differ essentially by the rigidity of the chelates, which strongly influenced the observed selectivity of the photosubstitution. As the ratio between the different photoproducts was also influenced by the solvent, we developed a full density functional theory modeling of the reaction mechanism that included explicit solvent molecules. Three reaction pathways leading to photodissociation were identified on the triplet hypersurface, each characterized by either one or two energy barriers. Photodissociation in water was promoted by a proton transfer in the triplet state, which was facilitated by the dissociated pyridine ring acting as a pendent base. We show that the temperature variation of the photosubstitution quantum yield is an excellent tool to compare theory with experiments. An unusual phenomenon was observed for one of the compounds in acetonitrile, for which an increase in temperature led to a surprising decrease in the photosubstitution reaction rate. We interpret this experimental observation based on complete mapping of the triplet hypersurface of this complex, revealing thermal deactivation to the singlet ground state through intersystem crossing.

14.
J Am Chem Soc ; 145(27): 14963-14980, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37379365

ABSTRACT

To investigate the potential of tumor-targeting photoactivated chemotherapy, a chiral ruthenium-based anticancer warhead, Λ/Δ-[Ru(Ph2phen)2(OH2)2]2+, was conjugated to the RGD-containing Ac-MRGDH-NH2 peptide by direct coordination of the M and H residues to the metal. This design afforded two diastereoisomers of a cyclic metallopeptide, Λ-[1]Cl2 and Δ-[1]Cl2. In the dark, the ruthenium-chelating peptide had a triple action. First, it prevented other biomolecules from coordinating with the metal center. Second, its hydrophilicity made [1]Cl2 amphiphilic so that it self-assembled in culture medium into nanoparticles. Third, it acted as a tumor-targeting motif by strongly binding to the integrin (Kd = 0.061 µM for the binding of Λ-[1]Cl2 to αIIbß3), which resulted in the receptor-mediated uptake of the conjugate in vitro. Phototoxicity studies in two-dimensional (2D) monolayers of A549, U87MG, and PC-3 human cancer cell lines and U87MG three-dimensional (3D) tumor spheroids showed that the two isomers of [1]Cl2 were strongly phototoxic, with photoindexes up to 17. Mechanistic studies indicated that such phototoxicity was due to a combination of photodynamic therapy (PDT) and photoactivated chemotherapy (PACT) effects, resulting from both reactive oxygen species generation and peptide photosubstitution. Finally, in vivo studies in a subcutaneous U87MG glioblastoma mice model showed that [1]Cl2 efficiently accumulated in the tumor 12 h after injection, where green light irradiation generated a stronger tumoricidal effect than a nontargeted analogue ruthenium complex [2]Cl2. Considering the absence of systemic toxicity for the treated mice, these results demonstrate the high potential of light-sensitive integrin-targeted ruthenium-based anticancer compounds for the treatment of brain cancer in vivo.


Subject(s)
Antineoplastic Agents , Brain Neoplasms , Coordination Complexes , Prodrugs , Ruthenium , Animals , Humans , Mice , Ruthenium/pharmacology , Ruthenium/chemistry , Prodrugs/pharmacology , Prodrugs/therapeutic use , Prodrugs/chemistry , Integrins , Peptides, Cyclic , Peptides , Brain Neoplasms/drug therapy , Cell Line, Tumor , Coordination Complexes/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry
15.
Nat Chem ; 15(7): 980-987, 2023 07.
Article in English | MEDLINE | ID: mdl-37169984

ABSTRACT

Self-assembling molecular drugs combine the easy preparation typical of small-molecule chemotherapy and the tumour-targeting properties of drug-nanoparticle conjugates. However, they require a supramolecular interaction that survives the complex environment of a living animal. Here we report that the metallophilic interaction between cyclometalated palladium complexes generates supramolecular nanostructures in living mice that have a long circulation time (over 12 h) and efficient tumour accumulation rate (up to 10.2% of the injected dose per gram) in a skin melanoma tumour model. Green light activation leads to efficient tumour destruction due to the type I photodynamic effect generated by the self-assembled palladium complexes, as demonstrated in vitro by an up to 96-fold cytotoxicity increase upon irradiation. This work demonstrates that metallophilic interactions are well suited to generating stable supramolecular nanotherapeutics in vivo with exceptional tumour-targeting properties.


Subject(s)
Antineoplastic Agents , Nanoparticles , Nanostructures , Skin Neoplasms , Animals , Mice , Palladium , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Nanoparticles/chemistry
16.
Dalton Trans ; 52(3): 598-608, 2023 Jan 17.
Article in English | MEDLINE | ID: mdl-36562298

ABSTRACT

Metal compounds form an attractive class of ligands for a variety of nucleic acids. Five metal complexes bearing aminopyridyl-2,2'-bipyridine tetradentate ligands and possessing a quasi-planar geometry were challenged toward different types of nucleic acid molecules including RNA polynucleotides in the duplex or triplex form, an RNA Holliday four-way junction, natural double helix DNA and a DNA G-quadruplex. The binding process was monitored comparatively using different spectroscopic and melting methods. The binding preferences that emerge from our analysis are discussed in relation to the structural features of the metal complexes.


Subject(s)
Coordination Complexes , Platinum , Platinum/chemistry , Coordination Complexes/chemistry , 2,2'-Dipyridyl , Palladium/chemistry , Gold , Ligands , DNA/chemistry , RNA
17.
Photochem Photobiol ; 99(2): 777-786, 2023 03.
Article in English | MEDLINE | ID: mdl-36315051

ABSTRACT

Photosubstitutionally active ruthenium complexes show high potential as prodrugs for the photoactivated chemotherapy (PACT) treatment of tumors. One of the problems in PACT is that the localization of the ruthenium compound is hard to trace. Here, a ruthenium PACT prodrug, [Ru(3)(biq)(STF-31)](PF6 )2 (where 3 = 3-(([2,2':6',2″-ter- pyridin]-4'-yloxy)propyl-4-(pyren-1-yl)butanoate) and biq = 2,2'-biquinoline), has been prepared, in which a pyrene tracker is attached via an ester bond. The proximity between the fluorophore and the ruthenium center leads to fluorescence quenching. Upon intracellular hydrolysis of the ester linkage, however, the fluorescence of the pyrene moiety is recovered, thus demonstrating prodrug cellular uptake. Further light irradiation of this molecule liberates by photosubstitution STF-31, a known cytotoxic nicotinamide phosphoribosyltransferase (NAMPT) inhibitor, as well as singlet oxygen via excitation of the free pyrene chromophore. The dark and light cytotoxicity of the prodrug, embedded in liposomes, as well as the appearance of blue emission upon uptake, were evaluated in A375 human skin melanoma cells. The cytotoxicity of the liposome-embedded prodrug was indeed increased by light irradiation. This work realizes an in vitro proof-of-concept of the lock-and-kill principle, which may ultimately be used to design strategies aimed at knowing where and when light irradiation should be realized in vivo.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Prodrugs , Ruthenium , Humans , Coordination Complexes/chemistry , Ruthenium/chemistry , Prodrugs/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
18.
J Am Chem Soc ; 144(42): 19353-19364, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36250745

ABSTRACT

Photosystem II, the natural water-oxidizing system, is a large protein complex embedded in a phospholipid membrane. A much simpler system for photocatalytic water oxidation consists of liposomes functionalized with amphiphilic ruthenium(II)-tris-bipyridine photosensitizer (PS) and 6,6'-dicarboxylato-2,2'-bipyridine-ruthenium(II) catalysts (Cat) with a water-soluble sacrificial electron acceptor (Na2S2O8). However, the effect of embedding this photocatalytic system in liposome membranes on the mechanism of photocatalytic water oxidation was not well understood. Here, several phenomena have been identified by spectroscopic tools, which explain the drastically different kinetics of water photo-oxidizing liposomes, compared with analogous homogeneous systems. First, the oxidative quenching of photoexcited PS* by S2O82- at the liposome surface occurs solely via static quenching, while dynamic quenching is observed for the homogeneous system. Moreover, the charge separation efficiency after the quenching reaction is much smaller than unity, in contrast to the quantitative generation of PS+ in homogeneous solution. In parallel, the high local concentration of the membrane-bound PS induces self-quenching at 10:1-40:1 molar lipid-PS ratios. Finally, while the hole transfer from PS+ to catalyst is rather fast in homogeneous solution (kobs > 1 × 104 s-1 at [catalyst] > 50 µM), in liposomes at pH = 4, the reaction is rather slow (kobs ≈ 17 s-1 for 5 µM catalyst in 100 µM DMPC lipid). Overall, the better understanding of these productive and unproductive pathways explains what limits the rate of photocatalytic water oxidation in liposomal vs homogeneous systems, which is required for future optimization of light-driven catalysis within self-assembled lipid interfaces.


Subject(s)
Ruthenium , Water , Water/chemistry , Lipid Bilayers , Ruthenium/chemistry , Liposomes , Photosensitizing Agents/chemistry , 2,2'-Dipyridyl , Photosystem II Protein Complex , Dimyristoylphosphatidylcholine , Oxidation-Reduction
19.
Inorg Chem ; 61(40): 16045-16054, 2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36171738

ABSTRACT

Upon coordination to metal centers, tetradentate ligands based on the 6,6'-bis(2″-aminopyridyl)-2,2'-bipyridine (bapbpy) structure form helical chiral complexes due to the steric clash between the terminal pyridines of the ligand. For octahedral ruthenium(II) complexes, the two additional axial ligands bound to the metal center, when different, generate diastereotopic aromatic protons that can be distinguished by NMR. Based on these geometrical features, the inversion barrier of helical [RuII(L)(RR'SO)Cl]+ complexes, where L is a sterically hindered bapbpy derivative and RR'SO is a chiral or achiral sulfoxide ligand, was studied by variable-temperature 1H NMR. The coalescence energies for the inversion of the helical chirality of [Ru(bapbpy)(DMSO)(Cl)]Cl and [Ru(bapbpy)(MTSO)(Cl)]Cl (where MTSO is (R)-methyl p-tolylsulfoxide) were found to be 43 and 44 kJ/mol, respectively. By contrast, in [Ru(biqbpy)(DMSO)(Cl)]Cl (biqbpy = 6,6'-bis(aminoquinolyl)-2,2'-bipyridine), increased strain caused by the larger terminal quinoline groups resulted in a coalescence temperature higher than 376 K, which pointed to an absence of helical chirality inversion at room temperature. Further increasing the steric strain by introducing methoxy groups ortho to the nitrogen atoms of the terminal pyridyl groups in bapbpy resulted in the serendipitous discovery of a ring-closing reaction that took place upon trying to make [Ru(OMe-bapbpy)(DMSO)Cl]+ (OMe-bapbpy = 6,6'-bis(6-methoxy-aminopyridyl)-2,2'-bipyridine). This reaction generated, in excellent yields, a chiral complex [Ru(L″)(DMSO)Cl]Cl, where L″ is an asymmetric tetrapyridyl macrocycle. This unexpected transformation appears to be specific to ruthenium(II) as macrocyclization did not occur upon coordination of the same ligand to palladium(II) or rhodium(III).

20.
Chem Sci ; 13(23): 6899-6919, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35774173

ABSTRACT

In vivo data are rare but essential for establishing the clinical potential of ruthenium-based photoactivated chemotherapy (PACT) compounds, a new family of phototherapeutic drugs that are activated via ligand photosubstitution. Here a novel trisheteroleptic ruthenium complex [Ru(dpp)(bpy)(mtmp)](PF6)2 ([2](PF6)2, dpp = 4,7-diphenyl-1,10-phenanthroline, bpy = 2,2'-bipyridine, mtmp = 2-methylthiomethylpyridine) was synthesized and its light-activated anticancer properties were validated in cancer cell monolayers, 3D tumor spheroids, and in embryonic zebrafish cancer models. Upon green light irradiation, the non-toxic mtmp ligand is selectively cleaved off, thereby releasing a phototoxic ruthenium-based photoproduct capable notably of binding to nuclear DNA and triggering DNA damage and apoptosis within 24-48 h. In vitro, fifteen minutes of green light irradiation (21 mW cm-2, 19 J cm-2, 520 nm) were sufficient to generate high phototherapeutic indexes (PI) for this compound in a range of cancer cell lines including lung (A549), prostate (PC3Pro4), conjunctival melanoma (CRMM1, CRMM2, CM2005.1) and uveal melanoma (OMM1, OMM2.5, Mel270) cancer cell lines. The therapeutic potential of [2](PF6)2 was further evaluated in zebrafish embryo ectopic (PC3Pro4) or orthotopic (CRMM1, CRMM2) tumour models. The ectopic model consisted of red fluorescent PC3Pro4-mCherry cells injected intravenously (IV) into zebrafish, that formed perivascular metastatic lesions at the posterior ventral end of caudal hematopoietic tissue (CHT). By contrast, in the orthotopic model, CRMM1- and CRMM2-mCherry cells were injected behind the eye where they developed primary lesions. The maximally-tolerated dose (MTD) of [2](PF6)2 was first determined for three different modes of compound administration: (i) incubating the fish in prodrug-containing water (WA); (ii) injecting the prodrug intravenously (IV) into the fish; or (iii) injecting the prodrug retro-orbitally (RO) into the fish. To test the anticancer efficiency of [2](PF6)2, the embryos were treated 24 h after engraftment at the MTD. Optimally, four consecutive PACT treatments were performed on engrafted embryos using 60 min drug-to-light intervals and 90 min green light irradiation (21 mW cm-2, 114 J cm-2, 520 nm). Most importantly, this PACT protocol was not toxic to the zebrafish. In the ectopic prostate tumour models, where [2](PF6)2 showed the highest photoindex in vitro (PI > 31), the PACT treatment did not significantly diminish the growth of primary lesions, while in both conjunctival melanoma orthotopic tumour models, where [2](PF6)2 showed more modest photoindexes (PI ∼ 9), retro-orbitally administered PACT treatment significantly inhibited growth of the engrafted tumors. Overall, this study represents the first demonstration in zebrafish cancer models of the clinical potential of ruthenium-based PACT, here against conjunctival melanoma.

SELECTION OF CITATIONS
SEARCH DETAIL