Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Small ; 18(49): e2203659, 2022 12.
Article in English | MEDLINE | ID: mdl-36310137

ABSTRACT

Photoimmunotherapy is attractive for cancer treatment due to its spatial controllability and sustained responses. This work presents a ferrocene-containing Ir(III) photosensitizer (IrFc1) that can bind with transferrin and be transported into triple-negative breast cancer (TNBC) cells via a transferrin receptor-mediated pathway. When the ferrocene in IrFc1 is oxidized by reactive oxygen species, its capability to photosensitize both type I (electron transfer) and type II (energy transfer) pathways is activated through a self-amplifying process. Upon irradiation, IrFc1 induces the generation of lipid oxidation to cause ferroptosis in TNBC cells, which promotes immunogenic cell death (ICD) under both normoxia and hypoxia. In vivo, IrFc1 treatment elicits a CD8+ T-cell response, which activates ICD in TNBC resulting in enhanced anticancer immunity. In summary, this work reports a small molecule-based photosensitizer with enhanced cancer immunotherapeutic properties by eliciting ferroptosis through a self-amplifying process.


Subject(s)
Neoplasms , Photosensitizing Agents , Photosensitizing Agents/pharmacology , Iridium , Receptors, Transferrin
2.
Angew Chem Int Ed Engl ; 61(16): e202115247, 2022 04 11.
Article in English | MEDLINE | ID: mdl-34965011

ABSTRACT

Ferroptosis is a programmed cell death pathway discovered in recent years, and ferroptosis-inducing agents have great potential as new antitumor candidates. Here, we report a IrIII complex (Ir1) containing a ferrocene-modified diphosphine ligand that localizes in lysosomes. Under the acidic environments of lysosomes, Ir1 can effectively catalyze Fenton-like reaction, produce hydroxyl radicals, induce lipid peroxidation, down-regulate glutathione peroxidase 4, and result in ferroptosis. RNA sequencing analysis shows that Ir1 can significantly affect pathways related to ferroptosis and cancer immunity. Accordingly, Ir1 can induce immunogenic cells death and suppress tumor growth in vitro, regulate T cell activity and immune microenvironments in vivo. In conclusion, we show the potential of small molecules with ferroptosis-inducing capabilities for effective cancer immunotherapy.


Subject(s)
Ferroptosis , Neoplasms , Humans , Immunotherapy , Iridium/pharmacology , Lipid Peroxidation , Metallocenes , Neoplasms/pathology , Tumor Microenvironment
3.
Angew Chem Int Ed Engl ; 61(15): e202114600, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35132748

ABSTRACT

The nucleus is considered the ideal target for anti-tumor therapy because DNA and some enzymes in the nucleus are the main causes of cell canceration and malignant proliferation. However, nuclear target drugs with good biosafety and high efficiency in cancer treatment are rare. Herein, a nuclear-targeted material MeTPAE with aggregation-induced emission (AIE) characteristics was developed based on a triphenylamine structure skeleton. MeTPAE can not only interact with histone deacetylases (HDACs) to inhibit cell proliferation but also damage telomere and nucleic acids precisely through photodynamic treatment (PDT). The cocktail strategy of MeTPAE caused obvious cell cycle arrest and showed excellent PDT anti-tumor activity, which offered new opportunities for the effective treatment of malignant tumors.


Subject(s)
Neoplasms , Photochemotherapy , Cell Cycle Checkpoints , Drug Delivery Systems , Humans , Neoplasms/drug therapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use
4.
Angew Chem Int Ed Engl ; 61(43): e202210988, 2022 Oct 24.
Article in English | MEDLINE | ID: mdl-35979672

ABSTRACT

Activation of the cyclic GMP-AMP synthase-stimulator of the interferon gene (cGAS-STING) pathway is a potent anticancer immunotherapeutic strategy, and the induction of pyroptosis is a feasible way to stimulate the anticancer immune responses. Herein, two PtII complexes (Pt1 and Pt2) were designed as photoactivators of the cGAS-STING pathway. In response to light irradiation, Pt1 and Pt2 could damage mitochondrial/nuclear DNA and the nuclear envelope to activate the cGAS-STING pathway, and concurrently induce pyroptosis in cancer cells, which evoked an intense anticancer immune response in vitro and in vivo. Overall, we present the first photoactivator of the cGAS-STING pathway, which may provide an innovative design strategy for anticancer immunotherapy.


Subject(s)
Neoplasms , Nucleotidyltransferases , Nucleotidyltransferases/metabolism , Interferons/pharmacology , Platinum/pharmacology , Pyroptosis , Membrane Proteins/metabolism , Signal Transduction , Immunotherapy , DNA/metabolism , Antiviral Agents/pharmacology , Neoplasms/therapy
5.
J Am Chem Soc ; 143(30): 11370-11381, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34291952

ABSTRACT

Phase separation of DNA is involved in chromatin packing for the regulation of gene transcription. Visualization and manipulation of DNA phase separation in living cells present great challenges. Herein, we present a Ru(II) complex (Ru1) with high DNA binding affinity and DNA "light-switch" behavior that can induce and monitor DNA phase separation both in vitro and in living cells. Molecular dynamics simulations indicate that the two phen-PPh3 ligands with positively charged lipophilic triphenylphosphine substituents and flexible long alkyl chains in Ru1 play essential roles in the formation of multivalent binding forces between DNA molecules to induce DNA phase separation. Importantly, the unique environmental sensitive emission property of Ru1 enables direct visualization of the dynamic process of DNA phase separation in living cells by two-photon phosphorescent lifetime imaging. Moreover, Ru1 can change the gene expression pattern by modulating chromatin accessibility as demonstrated by integrating RNA-sequencing and transposase-accessible chromatin with high-throughput sequencing. In all, we present here the first small-molecule-based tracer and modulator of DNA phase separation in living cells and elucidate its impact on the chromatin state and transcriptome.


Subject(s)
Coordination Complexes/chemistry , DNA/isolation & purification , Light , Ruthenium/chemistry , A549 Cells , Chromatin/chemistry , DNA/chemistry , Humans , Ligands , Molecular Dynamics Simulation , Molecular Structure
6.
Small ; 17(1): e2005086, 2021 01.
Article in English | MEDLINE | ID: mdl-33284508

ABSTRACT

The epigenetic dysregulation and hypoxia are two important factors that drive tumor malignancy, and N6 -methyladenosine (m6 A) in mRNA is involved in the regulation of gene expression. Herein, a nanocatalyst OsSx -PEG (PEG = poly(ethylene glycol)) nanoparticles (NPs) as O2 modulator is developed to improve tumor hypoxia. OsSx -PEG NPs can significantly downregulate genes involved in hypoxia pathway. Interestingly, OsSx -PEG NPs elevate RNA m6 A methylation levels to cause the m6 A-dependent mRNA degradation of the hypoxia-related genes. Moreover, OsSx -PEG NPs can regulate the expression of RNA m6 A methyltransferases and demethylases. Finally, DOX@OsSx -PEG (DOX = doxorubicin; utilized as a model drug) NPs modulate tumor hypoxia and regulate mRNA m6 A methylation of hypoxia-related genes in vivo. As the first report about relationship between catalytic nanomaterials and RNA modifications, the research opens a new avenue for unveiling the underlying action mechanisms of hypoxia-modulating nanomaterials and shows potential of regulating RNA modification to overcome chemoresistance.


Subject(s)
Nanoparticles , Neoplasms , Cell Line, Tumor , Doxorubicin , Humans , Hypoxia , Methylation , Polyethylene Glycols
7.
Angew Chem Int Ed Engl ; 60(27): 15095-15100, 2021 06 25.
Article in English | MEDLINE | ID: mdl-33835669

ABSTRACT

Ferroptosis regulates cell death through reactive oxygen species (ROS)-associated lipid peroxide accumulation, which is expected to affect the structure and polarity of lipid droplets (LDs), but with no clear evidence. Herein, we report the first example of an LD/nucleus dual-targeted ratiometric fluorescent probe, CQPP, for monitoring polarity changes in the cellular microenvironment. Due to the donor-acceptor structure of CQPP, it offers ratiometric fluorescence emission and fluorescence lifetime signals that reflect polarity variations. Using nucleus imaging as a reference, CQPP was applied to report the increase in LD polarity and the homogenization of polarity between LDs and cytoplasm in the ferroptosis model. This LD/nucleus dual-targeted fluorescent probe shows the great potential of using fluorescence imaging to study ferroptosis and ferroptosis-related diseases.


Subject(s)
Cell Nucleus/metabolism , Fluorescent Dyes/chemistry , Lipid Droplets/metabolism , Ferroptosis , Fluorescent Dyes/chemical synthesis , Humans , Lipid Droplets/chemistry , Molecular Structure , Reactive Oxygen Species/metabolism
8.
Inorg Chem ; 59(17): 12632-12642, 2020 Sep 08.
Article in English | MEDLINE | ID: mdl-32838518

ABSTRACT

Multifunctional platinumIV anticancer prodrugs have the potential to enrich the anticancer properties and overcome the clinical problems of drug resistance and side effects of platinumII anticancer agents. Herein, we develop dual and triple action platinumIV complexes with targeted and biological active functionalities. One complex (PFL) that consists of cisplatin, tegafur, and lonidamine exhibits strong cytotoxicity against triple negative breast cancer (TNBC) cells. Cellular uptake and distribution studies reveal that PFL mainly accumulates in mitochondria. As a result, PFL disrupts the mitochondrial ultrastructure and induces significant alterations in the mitochondrial membrane potential, which further leads to an increase in production of reactive oxygen species (ROS) and a decrease in ATP synthesis in MDA-MB-231 TNBCs. Western blot analysis reveals the formation of ternary complex of thymidylate synthase, which shows the intracellular conversion of tegafur into 5-FU after its release from PFL. Furthermore, treatment with PFL impairs the mitochondrial function, leading to the inhibition of glycolysis and mitochondrial respiration and induction of apoptosis through the mitochondrial pathway. The RNA-sequencing experiment shows that PFL can perturb the pathways involved in DNA synthesis, DNA damage, metabolism, and transcriptional activity. These findings demonstrate that PFL intervenes in several cellular processes including DNA damage, thymidylate synthase inhibition, and perturbation of the mitochondrial bioenergetics to kill the cancer cells. The results highlight the significance of a triple-action prodrug for efficient anticancer therapy for TNBCs.


Subject(s)
Coordination Complexes/chemistry , Coordination Complexes/metabolism , Enzyme Inhibitors/chemistry , Platinum/chemistry , Prodrugs/metabolism , Thymidylate Synthase/antagonists & inhibitors , Triple Negative Breast Neoplasms/pathology , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage , Drug Liberation , Fluorouracil/metabolism , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Prodrugs/chemistry , Reactive Oxygen Species/metabolism , Transcription, Genetic/drug effects
9.
Angew Chem Int Ed Engl ; 59(42): 18755-18762, 2020 10 12.
Article in English | MEDLINE | ID: mdl-32634290

ABSTRACT

The development and malignancy of cancer cells are closely related to the changes of the epigenome. In this work, a mitochondria-targeted rhenium(I) complex (DFX-Re3), integrating the clinical iron chelating agent deferasirox (DFX), has been designed. By relocating iron to the mitochondria and changing the key metabolic species related to epigenetic modifications, DFX-Re3 can elevate the methylation levels of histone, DNA, and RNA. As a consequence, DFX-Re3 affects the events related to apoptosis, RNA polymerases, and T-cell receptor signaling pathways. Finally, it is shown that DFX-Re3 induces immunogenic apoptotic cell death and exhibits potent antitumor activity in vivo. This study provides a new approach for the design of novel epigenetic drugs that can recode the cancer epigenome by intervening in mitochondrial metabolism and iron homeostasis.


Subject(s)
Coordination Complexes/chemistry , Iron/metabolism , Mitochondria/metabolism , Rhenium/chemistry , Animals , Apoptosis/drug effects , Cell Line, Tumor , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Deferasirox/chemistry , Drug Evaluation, Preclinical , Epigenomics , Histones/metabolism , Humans , Iron Chelating Agents/chemistry , Methylation/drug effects , Mice , Mitochondria/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , RNA Polymerase II/metabolism , Reactive Oxygen Species/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/drug effects
10.
Chemistry ; 25(28): 7012-7022, 2019 May 17.
Article in English | MEDLINE | ID: mdl-30913329

ABSTRACT

The chemo-anti-inflammatory strategy is attracting ever more attention for the treatment of cancer. Here, two cyclometalated IrIII complexes Ir2 and Ir3 formed by conjugation of Ir1 with two antiphlogistics (aspirin and salicylic acid) have been designed. Ir2 and Ir3 exhibit higher antitumor and anti-inflammatory potencies than a mixture of Ir1 and aspirin/salicylic acid. We show that they can be hydrolyzed, accumulate in mitochondria, and induce mitochondrial dysfunction. Due to their intense long-lived phosphorescence, Ir2 and Ir3 can track mitochondrial morphological changes. Phosphorescence lifetime imaging shows that Ir2 and Ir3 can aggregate during mitochondrial dysfunction. As expected, Ir2 and Ir3 exhibit immunomodulatory properties by regulating the activity of immune factors. Both Ir2 and Ir3 can induce caspase-dependent apoptosis and caspase-independent paraptosis and inhibit several events related to metastasis. Moreover, Ir2 and Ir3 show potent tumor growth inhibition in vivo. Our study demonstrates that the combination of mitochondrial-targeting and immunomodulatory activities is feasible to develop multifunctional metal-based anticancer agents.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Aspirin/therapeutic use , Coordination Complexes/therapeutic use , Immunomodulation/drug effects , Iridium/therapeutic use , Neoplasms/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aspirin/chemistry , Aspirin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Female , Humans , Iridium/chemistry , Iridium/pharmacology , Luminescent Measurements/methods , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/pathology , Neoplasms/diagnostic imaging , Neoplasms/pathology , Optical Imaging/methods
11.
Chemistry ; 24(71): 18971-18980, 2018 Dec 17.
Article in English | MEDLINE | ID: mdl-30264929

ABSTRACT

Emerging studies have shown that mitochondrial DNA (mtDNA) is an attractive target for anticancer therapeutics. Herein, a heterobimetallic complex [Ru(dip)2 (µ-bpm)PtCl2 ]Cl2 (RuPt; dip=4,7-diphenyl-1,10-phenanthroline; bpm=2,2'-bipyrimidine) and the corresponding mononuclear complex [Ru(dip)2 (bpm)]Cl2 (Ru) have been designed and synthesized. RuPt can bind to mtDNA and damage it both in the dark and upon visible light irradiation. By using a variety of methods, it was demonstrated that RuPt can interfere with the function of mtDNA by decreasing the amplification and copy number of mtDNA, and affecting the transcriptional level of mitochondria-encoded genes. Furthermore, RuPt can disturb the physiological processes of mitochondria and induce caspase-dependent apoptosis in the presence of light. In addition, RuPt shows low systemic toxicity and potent in vivo anticancer potency upon light irradiation. This study provides strong evidence that mtDNA is an important molecular target of RuPt, and photodamaging mtDNA is an effective strategy to overcome cisplatin resistance.


Subject(s)
DNA Damage/drug effects , DNA, Mitochondrial/genetics , Neoplasms/drug therapy , Organoplatinum Compounds/pharmacology , Photosensitizing Agents/pharmacology , Ruthenium/pharmacology , A549 Cells , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cisplatin/chemistry , Cisplatin/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , HeLa Cells , Humans , Neoplasms/genetics , Organoplatinum Compounds/chemistry , Phenanthrolines/chemistry , Phenanthrolines/pharmacology , Photochemotherapy , Photosensitizing Agents/chemistry , Pyrimidines/chemistry , Pyrimidines/pharmacology , Ruthenium/chemistry
12.
Chemistry ; 23(60): 15166-15176, 2017 Oct 26.
Article in English | MEDLINE | ID: mdl-28833658

ABSTRACT

Valproic acid (VPA) is a short-chain, fatty acid type histone deacetylase inhibitor (HDACi), which can cause growth arrest and induce differentiation of transformed cells. Phosphorescent cyclometalated IrIII complexes have emerged as potential anticancer agents. By conjugation of VPA to IrIII complexes through an ester bond, VPA-functionalized cyclometalated iridium(III) complexes 1 a-3 a were designed and synthesized. These complexes display excellent two-photon properties, which are favorable for live-cell imaging. The ester bonds in 1 a-3 a can be hydrolyzed quickly by esterase and display similar inhibition of HDAC activity to VPA. Notably, 1 a-3 a can overcome cisplatin resistance effectively and are about 54.5-89.7 times more cytotoxic than cisplatin against cisplatin-resistant human lung carcinoma (A549R) cells. Mechanistic studies indicate that 1 a-3 a can penetrate into human cervical carcinoma (HeLa) cells quickly and efficiently, accumulate in mitochondria, and induce a series of cell-death-related events mediated by mitochondria. This study gives insights into the design and anticancer mechanisms of multifunctional anticancer agents.


Subject(s)
Antineoplastic Agents/toxicity , Coordination Complexes/chemistry , Iridium/chemistry , Mitochondria/drug effects , Valproic Acid/chemistry , A549 Cells , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cisplatin/toxicity , Coordination Complexes/chemical synthesis , Coordination Complexes/toxicity , Crystallography, X-Ray , Drug Resistance, Neoplasm/drug effects , HeLa Cells , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/toxicity , Humans , Microscopy, Fluorescence, Multiphoton , Molecular Conformation , Reactive Oxygen Species/metabolism
13.
Chemistry ; 22(23): 7800-9, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27106876

ABSTRACT

Elucidation of relationship among chemical structure, cellular uptake, localization, and biological activity of anticancer metal complexes is important for the understanding of their mechanisms of action. Organometallic rhenium(I) tricarbonyl compounds have emerged as potential multifunctional anticancer drug candidates that can integrate therapeutic and imaging capabilities in a single molecule. Herein, two mononuclear phosphorescent rhenium(I) complexes (Re1 and Re2), along with their corresponding dinuclear complexes (Re3 and Re4), were designed and synthesized as potent anticancer agents. The subcellular accumulation of Re1-Re4 was conveniently analyzed by confocal microscopy in situ in live cells by utilizing their intrinsic phosphorescence. We found that increased lipophilicity of the bidentate ligands could enhance their cellular uptake, leading to improved anticancer efficacy. The dinuclear complexes were more potent than the mononuclear counterparts. The molecular anticancer mechanisms of action evoked by Re3 and Re4 were explored in detail. Re3 with a lower lipophilicity localizes to lysosomes and induces caspase-independent apoptosis, whereas Re4 with higher lipophilicity specially accumulates in mitochondria and induces caspase-independent paraptosis in cancer cells. Our study demonstrates that subcellular localization is crucial for the anticancer mechanisms of these phosphorescent rhenium(I) complexes.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Rhenium/chemistry , Rhenium/pharmacology , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Humans , Membrane Potential, Mitochondrial/drug effects , Models, Molecular , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Organometallic Compounds/pharmacokinetics , Reactive Oxygen Species/metabolism , Rhenium/pharmacokinetics , Transcriptome/drug effects
14.
Angew Chem Int Ed Engl ; 53(45): 12137-41, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25244053

ABSTRACT

During autophagy, the intracellular components are captured in autophagosomes and delivered to lysosomes for degradation and recycling. Changes in lysosomal trafficking and contents are key events in the regulation of autophagy, which has been implicated in many physiological and pathological processes. In this work, two iridium(III) complexes (LysoIr1 and LysoIr2) are developed as theranostic agents to monitor autophagic lysosomes. These complexes display lysosome-activated phosphorescence and can specifically label lysosomes with high photostability. Simultaneously, they can induce autophagy potently without initiating an apoptosis response. We demonstrate that LysoIr2 can effectively implement two functions, namely autophagy induction and lysosomal tracking, in the visualization of autophagosomal-lysosomal fusion. More importantly, they display strong two-photon excited fluorescence (TPEF), which is favorable for live cell imaging and in vivo applications.


Subject(s)
Autophagy , Iridium/chemistry , Lysosomes/metabolism , Cell Line, Tumor , Green Fluorescent Proteins/metabolism , Humans , Photons , Proton Magnetic Resonance Spectroscopy
15.
Eur J Med Chem ; 275: 116638, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38950489

ABSTRACT

The cGAS (cyclic GMP-AMP synthase)-STING (stimulator of interferon genes) pathway promotes antitumor immune responses by sensing cytosolic DNA fragments leaked from nucleus and mitochondria. Herein, we designed a highly charged ruthenium photosensitizer (Ru1) with a ß-carboline alkaloid derivative as the ligand for photo-activating of the cGAS-STING pathway. Due to the formation of multiple non-covalent intermolecular interactions, Ru1 can self-assemble into carrier-free nanoparticles (NPs). By incorporating the triphenylphosphine substituents, Ru1 can target and photo-damage mitochondrial DNA (mtDNA) to cause the cytoplasmic DNA leakage to activate the cGAS-STING pathway. Finally, Ru1 NPs show potent antitumor effects and elicit intense immune responses in vivo. In conclusion, we report the first self-assembling mtDNA-targeted photosensitizer, which can effectively activate the cGAS-STING pathway, thus providing innovations for the design of new photo-immunotherapeutic agents.


Subject(s)
Antineoplastic Agents , Immunotherapy , Membrane Proteins , Nucleotidyltransferases , Photosensitizing Agents , Ruthenium , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Humans , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Animals , Ruthenium/chemistry , Ruthenium/pharmacology , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Molecular Structure , Dose-Response Relationship, Drug , Nanoparticles/chemistry , Structure-Activity Relationship , Drug Screening Assays, Antitumor , DNA, Mitochondrial/metabolism , Cell Proliferation/drug effects , Cell Line, Tumor , Neoplasms/drug therapy , Neoplasms/pathology
16.
Natl Sci Rev ; 11(8): nwae234, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39114378

ABSTRACT

Ruthenium polypyridyl complexes are promising anticancer candidates, while their cellular targets have rarely been identified, which limits their clinical application. Herein, we design a series of Ru(II) polypyridyl complexes containing bioactive ß-carboline derivatives as ligands for anticancer evaluation, among which Ru5 shows suitable lipophilicity, high aqueous solubility, relatively high anticancer activity and cancer cell selectivity. The subsequent utilization of a photo-clickable probe, Ru5a, serves to validate the significance of ATP synthase as a crucial target for Ru5 through photoaffinity-based protein profiling. Ru5 accumulates in mitochondria, impairs mitochondrial functions and induces mitophagy and ferroptosis. Combined analysis of mitochondrial proteomics and RNA-sequencing shows that Ru5 significantly downregulates the expression of the chloride channel protein, and influences genes related to ferroptosis and epithelial-to-mesenchymal transition. Finally, we prove that Ru5 exhibits higher anticancer efficacy than cisplatin in vivo. We firstly identify the molecular targets of ruthenium polypyridyl complexes using a photo-click proteomic method coupled with a multiomics approach, which provides an innovative strategy to elucidate the anticancer mechanisms of metallo-anticancer candidates.

17.
Chemistry ; 19(36): 12152-60, 2013 Sep 02.
Article in English | MEDLINE | ID: mdl-23878093

ABSTRACT

A series of Ru(II)-arene complexes (1-6) of the general formula [(η(6)-arene)Ru(L)Cl]PF6 (arene=benzene or p-cymene; L=bidentate ß-carboline derivative, an indole alkaloid with potential cyclin-dependent kinases (CDKs) inhibitory activities) is reported. All the complexes were fully characterized by classical analytical methods, and three were characterized by X-ray crystallography. Hydrolytic studies show that ß-carboline ligands play a vital role in their aqueous behaviour. These complexes are highly active in vitro, with the most active complex 6 displaying a 3- to 12-fold higher anticancer activity than cisplatin against several cancer cell lines. Interestingly, the complexes are able to overcome cross-resistance to cisplatin, and show much lower cytotoxicity against normal cells. Complexes 1-6 may directly target CDK1, because they can block cells in the G2M phase, down-regulate the expression of CDK1 and cyclin B1, and inhibit CDK1/cyclin B in vitro. Further mechanism studies show that the complexes can effectively induce apoptosis through mitochondrial-related pathways and intracellular reactive oxygen species (ROS) elevation.


Subject(s)
Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , CDC2 Protein Kinase/antagonists & inhibitors , Carbolines/chemistry , Carbolines/chemical synthesis , Cisplatin/chemistry , Enzyme Inhibitors/chemical synthesis , Organometallic Compounds/chemistry , Organometallic Compounds/chemical synthesis , Antineoplastic Agents/chemistry , Carbolines/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Organometallic Compounds/pharmacology , Ruthenium/chemistry
18.
Chemistry ; 19(31): 10160-9, 2013 Jul 29.
Article in English | MEDLINE | ID: mdl-23828334

ABSTRACT

Histone deacetylases inhibitors (HDACis) have gained much attention as a new class of anticancer agents in recent years. Herein, we report a series of fluorescent ruthenium(II) complexes containing N(1)-hydroxy-N(8)-(1,10-phenanthrolin-5-yl)octanediamide (L), a suberoylanilide hydroxamic acid (SAHA) derivative, as a ligand. As expected, these complexes show interesting chemiphysical properties, including relatively high quantum yields, large Stokes shifts, and long emission lifetimes. The in vitro inhibitory effect of the most effective drug, [Ru(DIP)2L](PF6)2 (3; DIP: 4,7-diphenyl-1,10-phenanthroline), on histone deacetylases (HDACs) is approximately equivalent in activity to that of SAHA, and treatment with complex 3 results in increased levels of the acetylated histone H3. Complex 3 is highly active against a panel of human cancer cell lines, whereas it shows relatively much lower toxicity to normal cells. Further mechanism studies show that complex 3 can elicit cell cycle arrest and induce apoptosis through mitochondria-related pathways and the production of reactive oxygen species. These data suggest that these fluorescent ruthenium(II)-HDACi conjugates may represent a promising class of anticancer agents for potential dual imaging and therapeutic applications targeting HDACs.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacology , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Ruthenium/chemistry , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , HeLa Cells , Hep G2 Cells , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Ligands , Molecular Structure , Organometallic Compounds/chemistry , Reactive Oxygen Species/analysis , Vorinostat
19.
Curr Opin Chem Biol ; 74: 102308, 2023 06.
Article in English | MEDLINE | ID: mdl-37088005

ABSTRACT

The realization that liquid-liquid phase separation (LLPS) underlies the formation of membraneless compartments in cells has motivated efforts to modulate the condensation process of biomolecules. Increasing evidence shows that metals and inorganic molecules abundantly distributed in cells play important roles in the regulation of biomolecular condensation. Herein, we briefly reviewed the background of biomacromolecular phase separation and summarized the recent research progress on the roles of metals and inorganic molecules in regulating protein and nucleic acid phase separation in vitro and in cells.


Subject(s)
Nucleic Acids , Proteins/metabolism
20.
Chem Sci ; 14(25): 6890-6903, 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37389261

ABSTRACT

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon (STING) pathway is a key mediator of innate immunity involved in cancer development and treatment. The roles of mitochondrial DNA (mtDNA) in cancer immunotherapy have gradually emerged. Herein, we report a highly emissive rhodium(iii) complex (Rh-Mito) as the mtDNA intercalator. Rh-Mito can specifically bind to mtDNA to cause the cytoplasmic release of mtDNA fragments to activate the cGAS-STING pathway. Moreover, Rh-Mito activates the mitochondrial retrograde signaling by disturbing the key metabolites involved in epigenetic modifications, which alters the nuclear genome methylation landscape to influence the expression of genes related to immune signaling pathways. Finally, we demonstrate that ferritin-encapsulated Rh-Mito elicits potent anticancer activities and evokes intense immune responses in vivo by intravenous injection. Overall, we report for the first time that small molecules targeting mtDNA can activate the cGAS-STING pathway, which gives insights into the development of biomacromolecule-targeted immunotherapeutic agents.

SELECTION OF CITATIONS
SEARCH DETAIL