Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.450
Filter
1.
Sci Rep ; 14(1): 18443, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117886

ABSTRACT

Hypoxia is an important feature of the tumor microenvironment (TME) of most solid tumors, and it is closely linked to cancer cell proliferation, therapy resistance, and the tumor immune response. Herein, we describe a method for hypoxia-induced heterogeneous oxygen distribution in xenograft tumors based on phosphorescence imaging microscopy (PLIM) using intravascular and intracellular oxygen probes. We synthesized Ir(III) complexes with polyethylene glycol (PEG) units of different molecular weights into the ligand as intravascular oxygen probes, BTP-PEGm (m = 2000, 5000, 10000, 20000). BTP-PEGm showed red emission with relatively high emission quantum yield and high oxygen sensitivity in saline. Cellular and in vivo experiments using these complexes revealed that BTP-PEG10000 was the most suitable probe in terms of blood retention and ease of intravenous administration in mice. PLIM measurements of xenograft tumors in mice treated with BTP-PEG10000 allowed simultaneous imaging of the tumor microvasculature and quantification of oxygen partial pressures. From lifetime images using the red-emitting intracellular oxygen probe BTPDM1 and the green-emitting intravascular fluorescent probe FITC-dextran, we demonstrated hypoxic heterogeneity in the TME with a sparse vascular network and showed that the oxygen levels of tumor cells gradually decreased with vascular distance.


Subject(s)
Microscopy, Confocal , Oxygen , Animals , Oxygen/metabolism , Mice , Humans , Microscopy, Confocal/methods , Cell Line, Tumor , Iridium/chemistry , Tumor Microenvironment , Polyethylene Glycols/chemistry , Mice, Nude , Fluorescent Dyes/chemistry , Neoplasms/diagnostic imaging , Neoplasms/pathology
2.
Sci Data ; 11(1): 870, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127782

ABSTRACT

Iridium(III) complexes nowadays became rising stars in various health-related applications. Thus, there is a necessity to assess cytotoxicity of the synthesized molecules against cancer/normal cell lines. In this report, we present a dataset of 2694 experimental cytotoxicity values of 803 iridium complexes against 127 different cell lines. We specify the experimental conditions and provide representation of the complexes molecules in machine-readable format. The dataset provides a starting point for exploration of new iridium-based cellular probes and opens new possibilities for predictions of toxicities and data-driven generation of new organometallic anticancer agents.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Iridium , Iridium/chemistry , Humans , Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Cell Line , Cell Line, Tumor , Drug Screening Assays, Antitumor
3.
Carbohydr Polym ; 342: 122325, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39048214

ABSTRACT

Hemostasis, infection, oxidative stress, and inflammation still severely impede the wound repair process. It is significant to develop multifunctional wound dressings that can function as needed in various stages of wound healing. In this study, iridium nanoparticles (IrNPs) with multi-enzyme mimetic activity were complexed with chitosan (CS) and fucoidan (FD) for the first time to make a multifunctional CS/FD/IrNPs hydrogel with excellent antioxidant effect. The hydrogel has excellent physicochemical properties. In particular, the incorporation of IrNPs imparts excellent antioxidant properties to the hydrogel, which could scavenge reactive oxygen species (ROS). In addition, the hydrogel shows excellent hemostatic and antibacterial properties. The CS/FD/IrNPs hydrogel performs fast and efficient hemostasis in 21 s. Moreover, the blood loss of the CS/FD/IrNPs hydrogel group was approximately 10% of that in the control group and the antibacterial rate of CS/FD/IrNPs hydrogel against E. coli and S. aureus was up to 95 %. In vivo results demonstrate that CS/FD/IrNPs hydrogel promotes wound healing by attenuating ROS levels, reducing oxidative damage, mitigating inflammation, and accelerating angiogenesis. To summarize, the CS/FD/IrNPs hydrogel system, with hemostatic, antibacterial, antioxidant, anti-inflammatory and pro-healing activities, can be a promising and effective strategy for the treatment of clinically difficult-to-heal wounds.


Subject(s)
Anti-Bacterial Agents , Antioxidants , Chitosan , Escherichia coli , Hydrogels , Iridium , Polysaccharides , Staphylococcus aureus , Wound Healing , Chitosan/chemistry , Chitosan/pharmacology , Wound Healing/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Iridium/chemistry , Iridium/pharmacology , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Polysaccharides/chemistry , Polysaccharides/pharmacology , Antioxidants/pharmacology , Antioxidants/chemistry , Skin/drug effects , Reactive Oxygen Species/metabolism , Mice , Male , Hemostatics/chemistry , Hemostatics/pharmacology , Oxidative Stress/drug effects , Nanoparticles/chemistry , Metal Nanoparticles/chemistry , Rats , Humans
4.
Metallomics ; 16(7)2024 07 01.
Article in English | MEDLINE | ID: mdl-38955388

ABSTRACT

Both 8-hydroxyquinoline compounds and iridium (Ir) complexes have emerged as potential novel agents for tumor therapy. In this study, we synthesized and characterized two new Ir(III) complexes, [Ir(L1)(bppy)2] (Br-Ir) and [Ir(L2)(bppy)2] (Cl-Ir), with 5,7-dibromo-2-methyl-8-hydroxyquinoline (HL-1) or 5,7-dichloro-2-methyl-8-hydroxyquinoline as the primary ligand. Complexes Br-Ir and Cl-Ir successfully inhibited antitumor activity in Hep-G2 cells. In addition, complexes Br-Ir and Cl-Ir were localized in the mitochondrial membrane and caused mitochondrial damage, autophagy, and cellular immunity in Hep-G2 cells. We tested the proteins related to mitochondrial and mitophagy by western blot analysis, which showed that they triggered mitophagy-mediated apoptotic cell death. Remarkably, complex Br-Ir showed high in vivo antitumor activity, and the tumor growth inhibition rate was 63.0% (P < 0.05). In summary, our study on complex Br-Ir revealed promising results in in vitro and in vivo antitumor activity assays.


Subject(s)
Antineoplastic Agents , Iridium , Mitochondria , Humans , Iridium/chemistry , Iridium/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Animals , Hep G2 Cells , Mice , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Apoptosis/drug effects , Oxyquinoline/pharmacology , Oxyquinoline/chemistry , Oxyquinoline/analogs & derivatives , Mice, Inbred BALB C , Mitophagy/drug effects , Mice, Nude
5.
Eur J Med Chem ; 276: 116648, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38968786

ABSTRACT

Medical applications of iridium (III) complexes include their use as state-of-the-art theranostic agents - molecules that combine therapeutic and diagnostic functions into a single entity. These complexes offer a promising avenue in medical diagnostics, precision imaging at single-cell resolution and targeted anticancer therapy due to their unique properties. In this review we report a short summary of their application in medical diagnostics, imaging at single-cell level and targeted anticancer therapy. The exceptional photophysical properties of Iridium (III) complexes, including their brightness and photostability, make them excellent candidates for bioimaging. They can be used to image cellular processes and the microenvironment within single cells with unprecedented clarity, aiding in the understanding of disease mechanisms at the molecular level. Moreover the iridium (III) complexes can be designed to selectively target cancer cells,. Upon targeting, these complexes can act as photosensitizers for photodynamic therapy (PDT), generating reactive oxygen species (ROS) upon light activation to induce cell death. The integration of diagnostic and therapeutic capabilities in Iridium (III) complexes offers the potential for a holistic approach to cancer treatment, enabling not only the precise eradication of cancer cells but also the real-time monitoring of treatment efficacy and disease progression. This aligns with the goals of personalized medicine, offering hope for more effective and less invasive cancer treatment strategies.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Iridium , Neoplasms , Humans , Iridium/chemistry , Iridium/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Neoplasms/pathology , Neoplasms/diagnosis , Photochemotherapy , Theranostic Nanomedicine , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Animals , Precision Medicine , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Single-Cell Analysis
6.
Eur J Med Chem ; 276: 116618, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38972079

ABSTRACT

Ir(III) and Ru(II) polypyridyl complexes are promising photosensitizers (PSs) for photodynamic therapy (PDT) due to their outstanding photophysical properties. Herein, one series of cyclometallated Ir(III) complexes and two series of Ru(II) polypyridyl derivatives bearing three different thiazolyl-ß-carboline N^N' ligands have been synthesized, aiming to evaluate the impact of the different metal fragments ([Ir(C^N)2]+ or [Ru(N^N)2]2+) and N^N' ligands on the photophysical and biological properties. All the compounds exhibit remarkable photostability under blue-light irradiation and are emissive (605 < λem < 720 nm), with the Ru(II) derivatives displaying higher photoluminescence quantum yields and longer excited state lifetimes. The Ir PSs display pKa values between 5.9 and 7.9, whereas their Ru counterparts are less acidic (pKa > 9.3). The presence of the deprotonated form in the Ir-PSs favours the generation of reactive oxygen species (ROS) since, according to theoretical calculations, it features a low-lying ligand-centered triplet excited state (T1 = 3LC) with a long lifetime. All compounds have demonstrated anticancer activity. Ir(III) complexes 1-3 exhibit the highest cytotoxicity in dark conditions, comparable to cisplatin. Their activity is notably enhanced by blue-light irradiation, resulting in nanomolar IC50 values and phototoxicity indexes (PIs) between 70 and 201 in different cancer cell lines. The Ir(III) PSs are also activated by green (with PI between 16 and 19.2) and red light in the case of complex 3 (PI = 8.5). Their antitumor efficacy is confirmed by clonogenic assays and using spheroid models. The Ir(III) complexes rapidly enter cells, accumulating in mitochondria and lysosomes. Upon photoactivation, they generate ROS, leading to mitochondrial dysfunction and lysosomal damage and ultimately cell apoptosis. Additionally, they inhibit cancer cell migration, a crucial step in metastasis. In contrast, Ru(II) complex 6 exhibits moderate mitochondrial activity. Overall, Ir(III) complexes 1-3 show potential for selective light-controlled cancer treatment, providing an alternative mechanism to chemotherapy and the ability to inhibit lethal cancer cell dissemination.


Subject(s)
Antineoplastic Agents , Carbolines , Coordination Complexes , Drug Screening Assays, Antitumor , Iridium , Photochemotherapy , Photosensitizing Agents , Ruthenium , Humans , Iridium/chemistry , Iridium/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Ligands , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/chemical synthesis , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Carbolines/chemistry , Carbolines/pharmacology , Carbolines/chemical synthesis , Molecular Structure , Structure-Activity Relationship , Cell Proliferation/drug effects , Pyridines/chemistry , Pyridines/pharmacology , Pyridines/chemical synthesis , Reactive Oxygen Species/metabolism , Dose-Response Relationship, Drug , Cell Line, Tumor , Cell Survival/drug effects , Apoptosis/drug effects
7.
Chem Commun (Camb) ; 60(59): 7622-7625, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-38957144

ABSTRACT

Pyridyloxy-directed Ir(III)-catalyzed diacylmethylation of protected tyrosines was achieved with alkyl and (hetero)aryl sulfoxonium ylides, furnishing tyrosine-based unnatural amino acids in good yields. Furthermore, the late stage exemplification of the strategy was successfully accomplished in tyrosine-containing dipeptides, tripeptides and tetrapeptides in moderate yields. This methodology is distinguished by its site-selectivity, tolerance of sensitive functional groups, scalability, and retention of the chiral configuration for tyrosine motifs.


Subject(s)
Iridium , Peptides , Tyrosine , Iridium/chemistry , Catalysis , Tyrosine/chemistry , Methylation , Peptides/chemistry , Sulfonium Compounds/chemistry , Molecular Structure
8.
Dalton Trans ; 53(28): 11836-11849, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38949269

ABSTRACT

A novel lysosome-targeted photosensitizer/photoredox catalyst based on cyclometalated Ir(III) complex IrL has been designed and synthesized, which exhibited excellent phosphorescence properties and the ability to generate single oxygen (1O2) and photocatalytically oxidize 1,4-dihydronicotinamide adenine dinucleotide (NADH) under light irradiation. Most importantly, the aforementioned activities are significantly enhanced due to protonation under acidic conditions, which makes them highly attractive in light-activated tumor therapy, especially for acidic lysosomes and tumor microenvironments. The photocytotoxicity of IrL and the mechanism of cell death have been investigated. Additionally, the tumor-killing ability of IrL under light irradiation was evaluated using a 4T1 tumor-bearing mouse model. This work provides a strategy for the development of lysosome-targeted photosensitizers/photoredox catalysts to overcome hypoxic tumors.


Subject(s)
Coordination Complexes , Iridium , Lysosomes , Oxidation-Reduction , Photosensitizing Agents , Lysosomes/metabolism , Iridium/chemistry , Iridium/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Animals , Catalysis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Mice , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Humans , Photochemotherapy , Cell Line, Tumor , Light , Mice, Inbred BALB C
9.
Inorg Chem ; 63(31): 14641-14655, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39053139

ABSTRACT

Organotin(IV) and iridium(III) complexes have shown good application potential in the field of anticancer; however, the aggregation-caused quenching (ACQ) effect induced by high concentration or dose has limited the research on their targeting and anticancer mechanism. Then, a series of aggregation-induced emission (AIE)-activated butyltin(IV)-iridium(III) imidazole-phenanthroline complexes were prepared in this study. Complexes exhibited significant fluorescence improvement in the aggregated state because of the restricted intramolecular rotation (RIR), accompanied by an absolute fluorescence quantum yield of up to 29.2% (IrSn9). Complexes demonstrated potential in vitro antiproliferative and antimigration activity against A549 cells, following a lysosomal-mitochondrial apoptotic pathway. Nude mouse models further confirmed that complexes had favorable in vivo antitumor and antimigration activity in comparison to cisplatin. Therefore, butyltin(IV)-iridium(III) imidazole-phenanthroline complexes possess the potential as potential substitutes for platinum-based drugs.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Coordination Complexes , Drug Screening Assays, Antitumor , Imidazoles , Iridium , Phenanthrolines , Phenanthrolines/chemistry , Phenanthrolines/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Humans , Animals , Mice , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Cell Proliferation/drug effects , Imidazoles/chemistry , Imidazoles/pharmacology , Iridium/chemistry , Iridium/pharmacology , Mice, Nude , Apoptosis/drug effects , Organotin Compounds/chemistry , Organotin Compounds/pharmacology , Organotin Compounds/chemical synthesis , Molecular Structure , A549 Cells
10.
ACS Appl Mater Interfaces ; 16(30): 38916-38930, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39041453

ABSTRACT

Despite the potential of photodynamic therapy (PDT) in cancer treatment, the development of efficient and photostable photosensitizing molecules that operate at long wavelengths of light has become a major hurdle. Here, we report for the first time an Ir(III)-phthalocyanine conjugate (Ir-ZnPc) as a novel photosensitizer for high-efficiency synergistic PDT treatment that takes advantage of the long-wavelength excitation and near infrared (NIR) emission of the phthalocyanine scaffold and the known photostability and high phototoxicity of cyclometalated Ir(III) complexes. In order to increase water solubility and cell membrane permeability, the conjugate and parent zinc phthalocyanine (ZnPc) were encapsulated in amphoteric redox-responsive polyurethane-polyurea hybrid nanocapsules (Ir-ZnPc-NCs and ZnPc-NCs, respectively). Photobiological evaluations revealed that the encapsulated Ir-ZnPc conjugate achieved high photocytotoxicity in both normoxic and hypoxic conditions under 630 nm light irradiation, which can be attributed to dual Type I and Type II reactive oxygen species (ROS) photogeneration. Interestingly, PDT treatments with Ir-ZnPc-NCs and ZnPc-NCs significantly inhibited the growth of three-dimensional (3D) multicellular tumor spheroids. Overall, the nanoencapsulation of Zn phthalocyanines conjugated to cyclometalated Ir(III) complexes provides a new strategy for obtaining photostable and biocompatible red-light-activated nano-PDT agents with efficient performance under challenging hypoxic environments, thus offering new therapeutic opportunities for cancer treatment.


Subject(s)
Antineoplastic Agents , Indoles , Isoindoles , Photochemotherapy , Photosensitizing Agents , Humans , Indoles/chemistry , Indoles/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Iridium/chemistry , Iridium/pharmacology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Zinc Compounds/chemistry , Reactive Oxygen Species/metabolism , Nanocapsules/chemistry , Cell Line, Tumor , Cell Survival/drug effects
11.
Dalton Trans ; 53(31): 12917-12926, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39028267

ABSTRACT

The fast-moving field of photodynamic therapy (PDT) has provided fresh opportunities to expand the potential of metallodrugs to combat cancers in a light-controlled manner. As such, in the present study, a series of cyclometalated Ir(III) complexes modified with a tetrazine functional group (namely, Ir-ppy-Tz, Ir-pbt-Tz, and Ir-dfppy-Tz) are developed as potential two-photon photodynamic anticancer agents. These complexes target mitochondria but exhibit low toxicity towards HLF primary lung fibroblast normal cells in the dark. When receiving a low-dose one- or two-photon PDT, they become highly potent towards A549 lung cancer cells (with IC50 values ranging from 24.0 nM to 96.0 nM) through the generation of reactive oxygen species (ROS) to induce mitochondrial damage and subsequent apoptosis. Our results indicated that the incorporation of tetrazine with cyclometalated Ir(III) matrices would increase the singlet oxygen (1O2) quantum yield (ΦΔ) and, meanwhile, enable a type I PDT mechanism. Ir-pbt-Tz, with the largest two-photon absorption (TPA) cross-section (σ2 = 102 GM), shows great promise in serving as a two-photon PDT agent for phototherapy.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Iridium , Mitochondria , Photochemotherapy , Photosensitizing Agents , Iridium/chemistry , Iridium/pharmacology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Photons , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , A549 Cells , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Molecular Structure
12.
Anal Biochem ; 693: 115597, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38969155

ABSTRACT

Vibrio parahaemolyticus (V. parahaemolyticus) is a major foodborne pathogen, which can cause serious foodborne illnesses like diarrhoea. Rapid on-site detection of foodborne pathogens is an ideal way to respond to foodborne illnesses. Herein, we provide an electrochemical sensor for rapid on-site detection. This sensor utilized a pH-sensitive metal-oxide material for the concurrent isothermal amplification and label-free detection of nucleic acids. Based on a pH-sensitive hydrated iridium oxide oxyhydroxide film (HIROF), the electrode transforms the hydrogen ion compound generated during nucleic acid amplification into potential, so as to achieve a real-time detection. The results can be transmitted to a smartphone via Bluetooth. Moreover, HIROF was applied in nucleic acid device detection, with a super-Nernst sensitivity of 77.6 mV/pH in the pH range of 6.0-8.5, and the sensitivity showed the best results so far. Detection of V. parahaemolyticus by this novel method showed a detection limit of 1.0 × 103 CFU/mL, while the time consumption was only 30 min, outperforming real-time fluorescence loop-mediated isothermal amplification (LAMP). Therefore, the characteristics of compact, portable, and fast make the sensor more widely used in on-site detection.


Subject(s)
Electrochemical Techniques , Iridium , Vibrio parahaemolyticus , Vibrio parahaemolyticus/isolation & purification , Vibrio parahaemolyticus/genetics , Hydrogen-Ion Concentration , Electrochemical Techniques/methods , Iridium/chemistry , Nucleic Acid Amplification Techniques/methods , Biosensing Techniques/methods , Limit of Detection , Electrodes
13.
Anal Sci ; 40(8): 1459-1473, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38900232

ABSTRACT

The main reason for the high mortality rate of non-small cell lung cancer is that patients are usually diagnosed at an advanced stage of the disease. Exosomes, small membrane vesicles secreted by normal cells or tumor cells, play a significant role in the progression of NSCLC. This study successfully optimized the preparation of artificial nanoenzymes self-coupling with horseradish peroxidase (IrO2NPs@HRP-AptCD63), without adding any ligand, demonstrating remarkable catalytic activity suitable for detecting the EGFR protein on the surface of NSCLC exosomes. When fused with the CD63 aptamer for identifying NSCLC exosomes, IrO2NPs@HRP showed enhanced catalytic activity in the 3,3',5,5'-tetramethylbenzidine-H2O2 oxidation-reduction system, thereby enhancing the colorimetric signal. This phenomenon can be distinguished by the naked eye and quantified using a UV-Vis spectrophotometer. Meanwhile, as the redox reaction occurs, the current signal of 3,3',5,5'-tetramethylbenzidine-H2O2, acting as an electrolyte, changes. The developed aptasensor generates dual-mode signal outputs, firstly, to visually assess the samples for their positive or negative status, and subsequently employ more in-depth electrochemical or colorimetric analysis methods for a detailed quantitative analysis of suspected positive samples. The detection limits of electrochemical analysis and colorimetric analysis were 0.9 × 103 particles/mL and 0.14 × 103 particles/mL, respectively. Compared with traditional biomarkers such as CA125, this method exhibits exceptional specificity, capable of simultaneously distinguishing serum exosomes of healthy volunteers, COPD patients, and NSCLC patients, promoting exosome detection in mouse models for tumor monitoring. Additionally, it elucidates the changes in EGFR protein expression on the surface of serum exosomes throughout the developmental trajectory.


Subject(s)
Aptamers, Nucleotide , Carcinoma, Non-Small-Cell Lung , Exosomes , Horseradish Peroxidase , Iridium , Lung Neoplasms , Oxidation-Reduction , Exosomes/chemistry , Exosomes/metabolism , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/metabolism , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Aptamers, Nucleotide/chemistry , Iridium/chemistry , Horseradish Peroxidase/chemistry , Horseradish Peroxidase/metabolism , Animals , Mice , Metal Nanoparticles/chemistry , Biosensing Techniques
14.
Inorg Chem ; 63(28): 13059-13067, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38937959

ABSTRACT

Type I photosensitizers offer an advantage in photodynamic therapy (PDT) due to their diminished reliance on oxygen levels, thus circumventing the challenge of hypoxia commonly encountered in PDT. In this study, we present the synthesis and comprehensive characterization of a novel type I photosensitizer derived from a cyclometalated Ir(III)-rhodamine complex. Remarkably, the complex exhibits a shift in absorption and fluorescence, transitioning from "off" to "on" states in aprotic and protic solvents, respectively, contrary to initial expectations. Upon exposure to light, the complex demonstrates the effective generation of O2- and ·OH radicals via the type I mechanism. Additionally, it exhibits notable photodynamic antibacterial activity against both Gram-positive and Gram-negative bacteria, demonstrated through in vitro and in vivo experiments. This research offers valuable insights for the development of novel type I photosensitizers.


Subject(s)
Anti-Bacterial Agents , Gram-Negative Bacteria , Gram-Positive Bacteria , Iridium , Microbial Sensitivity Tests , Photochemotherapy , Photosensitizing Agents , Rhodamines , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Gram-Negative Bacteria/drug effects , Rhodamines/chemistry , Rhodamines/pharmacology , Iridium/chemistry , Iridium/pharmacology , Gram-Positive Bacteria/drug effects , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Animals , Infrared Rays , Molecular Structure , Mice
15.
Org Biomol Chem ; 22(27): 5603-5607, 2024 07 10.
Article in English | MEDLINE | ID: mdl-38904084

ABSTRACT

The synthesis of the ethyl ester analogue of the ultrapotent antitumour antibiotic seco-duocarmycin SA has been achieved in eleven linear steps from commercially available starting materials. The DSA alkylation subunit can be made in ten linear steps from the same precursor. The route involves C-H activation at the equivalent of the C7 position on indole leading to a borylated intermediate 9 that is stable enough for peptide coupling reactions but can be easily converted to the free hydroxyl analogue.


Subject(s)
Duocarmycins , Indoles , Iridium , Catalysis , Indoles/chemistry , Indoles/chemical synthesis , Iridium/chemistry , Molecular Structure , Pyrroles/chemistry , Pyrroles/chemical synthesis , Boron Compounds/chemistry , Boron Compounds/chemical synthesis
16.
Dalton Trans ; 53(27): 11393-11409, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38899369

ABSTRACT

In this work we disclose a new family of biscyclometallated Ir(III) complexes of the general formula [Ir(C^N)2(N^N)]Cl (IrL1-IrL5), where HC^N is 1-phenyl-ß-carboline and N^N ligands (L1-L5) are different diimine ligands that differ from each other in the number of aromatic rings fused to the bipyridine scaffold. The photophysical properties of IrL1-IrL5 were thoroughly studied, and theoretical calculations were performed for a deeper comprehension of the respective variations along the series. All complexes exhibited high photostability under blue light irradiation. An increase in the number of aromatic rings led to a reduction in the HOMO-LUMO band gap causing a red-shift in the absorbance bands. Although all the complexes generated singlet oxygen (1O2) in aerated aqueous solutions through a photocatalytic process, IrL5 was by far the most efficient photosensitizer. Consequently, IrL5 was highly active in the photocatalytic oxidation of NADH. The formation of aggregates in DMSO at a high concentration (25 mM) was confirmed using different techniques, but was proved to be negligible in the concentration range of biological experiments. Moreover, ICP-MS studies proved that the cellular uptake of IrL2 and IrL3 is much better relative to that of IrL1, IrL4 and IrL5. The antiproliferative activity of IrL1-IrL5 was investigated in the dark and under blue light irradiation against different cancer cell lines. Complexes IrL1-IrL4 were found to be cytotoxic under dark conditions, while IrL5 turned out to be weakly cytotoxic. Despite the low cellular uptake of IrL5, this derivative exhibited a high increase of cytotoxicity upon blue light irradiation resulting in photocytotoxicity indexes (PI) up to 38. IrL1-IrL4 showed lower photocytotoxicity indexes ranging from 1.3 to 17.0. Haemolytic experiments corroborated the compatibility of our complexes with red blood cells. Confocal microscopy studies proved their accumulation in mitochondria, leading to mitochondrial membrane depolarization, and ruled out their localization in lysosomes. Overall, the mitochondria-targeted activity of IrL5, which inhibits considerably the viability of cancer cells upon blue light irradiation, allows us to outline this PS as a new alternative to traditional chemotherapeutic agents.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Iridium , Photochemotherapy , Photosensitizing Agents , Humans , Iridium/chemistry , Iridium/pharmacology , Ligands , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects , Molecular Structure , Cell Survival/drug effects , Cell Line, Tumor , Light , Density Functional Theory
17.
Anal Bioanal Chem ; 416(21): 4705-4715, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38937290

ABSTRACT

3-Methylhistidine (3-MeHis) is increasingly used as an indicator of muscle protein breakdown. The development of a sensitive, simple, and non-invasive method for 3-MeHis assay is important in clinical practice. Herein, a sensitive, simple, and non-invasive electrogenerated chemiluminescence (ECL) method was proposed for the quantitation of 3-MeHis in urine by using an iridium(III) solvent complex ([Ir(dfppy)2(DMSO)Cl], dfppy = 2-(2,4-difluorophenyl)pyridine, Ir-DMSO) as a signal reagent. The photoluminescence (PL) and ECL responses of Ir-DMSO to 3-MeHis were studied. The ECL intensity of Ir-DMSO was enhanced in the presence of 3-MeHis because of the coordination recognition between Ir-DMSO and the imidazole group of 3-MeHis. Based on the enhancement of ECL intensity, 3-MeHis can be sensitively detected in the range of 5 to 25 µM. The detection limit was 0.4 µM. This is the first report of an ECL method for the quantitation of 3-MeHis. Further, to investigate the feasibility of the Ir-DMSO-based ECL method in practical applications, the developed ECL method was applied for 3-MeHis assay in urine samples of 28 healthy volunteers and 2 patients. The urine samples from patients hospitalized with obesity and kidney disease and healthy individuals were distinguished by the ECL responses of Ir-DMSO. The proposed ECL method based on the coordination recognition between iridium(III) solvent complex and the imidazole group of 3-MeHis allows inexpensive, fast, non-invasive, and sensitive detection of 3-MeHis in urine, which is promising for assessing large volumes of patients for routine analysis in clinical practices.


Subject(s)
Iridium , Limit of Detection , Luminescent Measurements , Methylhistidines , Solvents , Humans , Iridium/chemistry , Luminescent Measurements/methods , Methylhistidines/urine , Solvents/chemistry , Coordination Complexes/chemistry , Male , Electrochemical Techniques/methods
18.
Chemosphere ; 362: 142665, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38906192

ABSTRACT

Veterinary antibiotics have become an emerging pollutant in water and wastewater sources due to excess usage, toxicity and resistance to traditional water and wastewater treatment. The present study explored the degradation of a model antibiotic- Florfenicol (FF) using electrochemical oxidation (EO) with Ti-RuO2/IrO2 anode. The anode material was characterized using SEM-EDS studies expressing stable structure and optimal interaction of the neighboring metal oxides with each other. The EDS results showed the presence of Ru, Ir, Ti, O and C elements with 6.44%, 2.57%, 9.61%, 52.74% and 28.64% atomic weight percentages, respectively. Optimization studies revealed pH 5, 30 mA cm-2 current density and 0.05 M Na2SO4 for 5 mg L-1 FF achieved 90% TOC removal within 360 min treatment time. The degradation followed pseudo-first order kinetics. LC-Q-TOF-MS studies revealed six predominant byproducts illustrating hydroxylation, deflourination, and dechlorination to be the major degradation mechanisms during the electrochemical oxidation of FF. Ion chromatography studies revealed an increase in Cl-, F- and NO3- ions as treatment time progressed with Cl- decreasing after the initial phase of the treatment. Toxicity studies using Zebrafish (Danio rerio) embryo showed the treated sample to be toxic inducing developmental disorders such as pericardial edema, yolk sac edema, spinal curvature and tail malformation at 96 h post fertilization (hpf). Compared to control, delayed hatching and coagulation were observed in treated embryos. Overall, this study sets the stage for understanding the effect of mixed metal oxide (MMO) anodes on the degradation of veterinary antibiotic-polluted water and wastewater sources using electrochemical oxidation.


Subject(s)
Anti-Bacterial Agents , Electrodes , Oxidation-Reduction , Oxides , Thiamphenicol , Water Pollutants, Chemical , Water Pollutants, Chemical/chemistry , Water Pollutants, Chemical/toxicity , Thiamphenicol/analogs & derivatives , Thiamphenicol/chemistry , Thiamphenicol/toxicity , Oxides/chemistry , Oxides/toxicity , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/toxicity , Wastewater/chemistry , Electrochemical Techniques , Iridium/chemistry , Iridium/toxicity , Kinetics , Titanium/chemistry , Titanium/toxicity , Metals/chemistry , Metals/toxicity , Ruthenium Compounds
19.
ACS Sens ; 9(6): 3327-3337, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38863381

ABSTRACT

Oxide semiconductor-supported metal nanoparticles often suffer from a high-temperature gas sensing process, resulting in agglomeration and coalescence, which significantly decrease their surface activity and stability. Here, we develop an in situ pyrolysis strategy to redisperse commercial Ir particles (∼15.6 nm) into monodisperse Ir species (∼5.4 nm) on ZnO supports, exhibiting excellent sintering-resistant properties and H2 sensing. We find that large-size Ir nanoparticles can undergo an unexpected splitting decomposition process and spontaneously migrate along the encapsulated carbon layer surface during high-temperature pyrolysis of ZIF-8. This resultant monodisperse status can be integrally reserved, accompanying further oxidation sintering. The final Irred/ZnO-450-based sensor exhibits outstanding stability, H2 response (10-2000 ppm), fast response/recovery capability (7/9.7 s@100 ppm), and good moisture resistance. In situ Raman and ex situ XPS further experimentally verify that highly dispersive Ir species can promote the electron transfer process during the gas sensing process. Our strategy thus provides important insights into the design of agglomeration-resistant gas sensing materials for highly effective H2 detection.


Subject(s)
Carbon , Hydrogen , Iridium , Metal Nanoparticles , Pyrolysis , Zinc Oxide , Hydrogen/chemistry , Hydrogen/analysis , Carbon/chemistry , Metal Nanoparticles/chemistry , Iridium/chemistry , Zinc Oxide/chemistry
20.
J Inorg Biochem ; 259: 112655, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38943844

ABSTRACT

Recent breakthroughs in cancer immunology have propelled immunotherapy to the forefront of cancer research as a promising treatment approach that harnesses the body's immune system to effectively identify and eliminate cancer cells. In this study, three novel cyclometalated Ir(III) complexes, Ir1, Ir2, and Ir3, were designed, synthesized, and assessed in vitro for cytotoxic activity against several tumor-derived cell lines. Among these, Ir1 exhibited the highest cytotoxic activity, with an IC50 value of 0.4 ± 0.1 µM showcasing its significant anticancer potential. Detailed mechanistic analysis revealed that co-incubation of Ir1 with 143B cells led to Ir1 accumulation within mitochondria and the endoplasmic reticulum (ER). Furthermore, Ir1 induced G0/G1 phase cell cycle arrest, while also diminishing mitochondrial membrane potential, disrupting mitochondrial function, and triggering ER stress. Intriguingly, in mice the Ir1-induced ER stress response disrupted calcium homeostasis to thereby trigger immunogenic cell death (ICD), which subsequently activated the host antitumor immune response while concurrently dampening the in vivo tumor-induced inflammatory response.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Immunogenic Cell Death , Iridium , Mitochondria , Iridium/chemistry , Iridium/pharmacology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Immunogenic Cell Death/drug effects , Animals , Mice , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Endoplasmic Reticulum Stress/drug effects , Cell Line, Tumor , Membrane Potential, Mitochondrial/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL