Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 146(19): 13126-13132, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38696488

ABSTRACT

Cisplatin, a cornerstone in cancer chemotherapy, is known for its DNA-binding capacity and forms lesions that lead to cancer cell death. However, the repair of these lesions compromises cisplatin's effectiveness. This study investigates how phosphorylation of HMGB1, a nuclear protein, modifies its binding to cisplatin-modified DNA (CP-DNA) and thus protects it from repair. Despite numerous methods for detecting protein-DNA interactions, quantitative approaches for understanding their molecular mechanism remain limited. Here, we applied click chemistry-based single-molecule force spectroscopy, achieving high-precision quantification of the interaction between phosphorylated HMGB1 and CP-DNA. This method utilizes a synergy of click chemistry and enzymatic ligation for precise DNA-protein immobilization and interaction in the system. Our results revealed that HMGB1 binds to CP-DNA with a significantly high rupture force of ∼130 pN, stronger than most natural DNA-protein interactions and varying across different DNA sequences. Moreover, Ser14 is identified as the key phosphorylation site, enhancing the interaction's kinetic stability by 35-fold. This increase in stability is attributed to additional hydrogen bonding suggested by molecular dynamics (MD) simulations. Our findings not only reveal the important role of phosphorylated HMGB1 in potentially improving cisplatin's therapeutic efficacy but also provide a precise method for quantifying protein-DNA interactions.


Subject(s)
Cisplatin , Click Chemistry , DNA , HMGB1 Protein , Molecular Dynamics Simulation , HMGB1 Protein/metabolism , HMGB1 Protein/chemistry , Cisplatin/chemistry , Cisplatin/pharmacology , Cisplatin/metabolism , Phosphorylation , DNA/chemistry , DNA/metabolism , Humans , Protein Binding , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
2.
J Am Chem Soc ; 146(10): 7052-7062, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38427585

ABSTRACT

Functional DNAs are valuable molecular tools in chemical biology and analytical chemistry but suffer from low activities due to their limited chemical functionalities. Here, we present a chemoenzymatic method for site-specific installation of diverse functional groups on DNA, and showcase the application of this method to enhance the catalytic activity of a DNA catalyst. Through chemoenzymatic introduction of distinct chemical groups, such as hydroxyl, carboxyl, and benzyl, at specific positions, we achieve significant enhancements in the catalytic activity of the RNA-cleaving deoxyribozyme 10-23. A single carboxyl modification results in a 100-fold increase, while dual modifications (carboxyl and benzyl) yield an approximately 700-fold increase in activity when an RNA cleavage reaction is catalyzed on a DNA-RNA chimeric substrate. The resulting dually modified DNA catalyst, CaBn, exhibits a kobs of 3.76 min-1 in the presence of 1 mM Mg2+ and can be employed for fluorescent imaging of intracellular magnesium ions. Molecular dynamics simulations reveal the superior capability of CaBn to recruit magnesium ions to metal-ion-binding site 2 and adopt a catalytically competent conformation. Our work provides a broadly accessible strategy for DNA functionalization with diverse chemical modifications, and CaBn offers a highly active DNA catalyst with immense potential in chemistry and biotechnology.


Subject(s)
DNA, Catalytic , RNA, Catalytic , Base Sequence , Magnesium , DNA, Catalytic/chemistry , DNA , RNA/chemistry , Ions , Nucleic Acid Conformation , Catalysis , RNA, Catalytic/metabolism
3.
Natl Sci Rev ; 11(1): nwae020, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38332843

ABSTRACT

Mounting evidence suggests that strategies combining DNA-damaging agents and stimulator of interferon genes (STING) agonists are promising cancer therapeutic regimens because they can amplify STING activation and remodel the immunosuppressive tumor microenvironment. However, a single molecular entity comprising both agents has not yet been developed. Herein, we designed two PtIV-MSA-2 conjugates (I and II) containing the DNA-damaging chemotherapeutic drug cisplatin and the innate immune-activating STING agonist MSA-2; these conjugates showed great potential as multispecific small-molecule drugs against pancreatic cancer. Mechanistic studies revealed that conjugate I upregulated the expression of transcripts associated with innate immunity and metabolism in cancer cells, significantly differing from cisplatin and MSA-2. An analysis of the tumor microenvironment demonstrated that conjugate I could enhance the infiltration of natural killer (NK) cells into tumors and promote the activation of T cells, NK cells and dendritic cells in tumor tissues. These findings indicated that conjugate I, which was created by incorporating a Pt chemotherapeutic drug and STING agonist into one molecule, is a promising and potent anticancer drug candidate, opening new avenues for small-molecule-based cancer metalloimmunotherapy.

4.
Angew Chem Int Ed Engl ; 63(9): e202317613, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38195970

ABSTRACT

Most of the anticancer compounds synthesized by chemists are primarily evaluated for their direct cytotoxic effects at the cellular level, often overlooking the critical role of the immune system. In this study, we developed a patient-derived, T-cell-retaining tumor organoid model that allows us to evaluate the anticancer efficacy of chemical drugs under the synergistic paradigm of antigen-specific T-cell-dependent killing, which may reveal the missed drug hits in the simple cytotoxic assay. We evaluated clinically approved platinum (Pt) drugs and a custom library of twenty-eight PtIV compounds. We observed low direct cytotoxicity of Pt drugs, but variable synergistic effects in combination with immune checkpoint inhibitors (ICIs). In contrast, the majority of PtIV compounds exhibited potent tumor-killing capabilities. Interestingly, several PtIV compounds went beyond direct tumor killing and showed significant immunosynergistic effects with ICIs, outstanding at sub-micromolar concentrations. Among these, Pt-19, PtIV compounds with cinnamate axial ligands, emerged as the most therapeutically potent, demonstrating pronounced immunosynergistic effects by promoting the release of cytotoxic cytokines, activating immune-related pathways and enhancing T cell receptor (TCR) clonal expansion. Overall, this initiative marks the first use of patient-derived immunocompetent tumor organoids to explore and study chemotherapy, advancing their path toward more effective small molecule drug discovery.


Subject(s)
Antineoplastic Agents , Humans , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Platinum/chemistry , T-Lymphocytes , Organoids
5.
Nat Commun ; 14(1): 800, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36781887

ABSTRACT

Temporal control of delivery and release of drugs in tumors are important in improving therapeutic outcomes to patients. Here, we report a sequential stimuli-triggered in situ self-assembly and disassembly strategy to direct delivery and release of theranostic drugs in vivo. Using cisplatin as a model anticancer drug, we design a stimuli-responsive small-molecule cisplatin prodrug (P-CyPt), which undergoes extracellular alkaline phosphatase-triggered in situ self-assembly and succeeding intracellular glutathione-triggered disassembly process, allowing to enhance accumulation and elicit burst release of cisplatin in tumor cells. Compared with cisplatin, P-CyPt greatly improves antitumor efficacy while mitigates off-target toxicity in mice with subcutaneous HeLa tumors and orthotopic HepG2 liver tumors after systemic administration. Moreover, P-CyPt also produces activated near-infrared fluorescence (at 710 nm) and dual photoacoustic imaging signals (at 700 and 750 nm), permitting high sensitivity and spatial-resolution delineation of tumor foci and real-time monitoring of drug delivery and release in vivo. This strategy leverages the advantages offered by in situ self-assembly with those of intracellular disassembly, which may act as a general platform for the design of prodrugs capable of improving drug delivery for cancer theranostics.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Prodrugs , Animals , Mice , Cisplatin/pharmacology , Precision Medicine , Cell Line, Tumor , Neoplasms/drug therapy
6.
Angew Chem Int Ed Engl ; 62(2): e202213337, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36259513

ABSTRACT

Triggering receptor expressed on myeloid cells-2 (TREM2) is a key pro-tumorigenic marker of tumor-infiltrating macrophages, showing potent immunosuppressive activity in tumor microenvironment. A platinum(IV) complex OPA derived from oxaliplatin (OP) and artesunate (ART) exhibited direct cytotoxicity against human colon cancer cells and immunomodulatory activity to inhibit TREM2 on macrophages in vitro and vivo. Furthermore, OPA deterred the tumor growth in mouse models bearing MC38 colorectal tumor by reducing the number of CD206+ and CX3 CR1+ immunosuppressive macrophages; it also promoted the expansion and infiltration of immunostimulatory dendritic, cytotoxic T, and natural killer cells. OPA is the first small-molecular TREM2 inhibitor capable of relieving immunosuppressive tumor microenvironment and enhancing chemical anticancer efficiency of a platinum drug, thus showing typical characteristics of a chemoimmunotherapeutic agent.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Animals , Mice , Humans , Platinum/pharmacology , Platinum/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Myeloid Cells/pathology , Macrophages , Immunosuppressive Agents/pharmacology , Colonic Neoplasms/drug therapy , Tumor Microenvironment , Membrane Glycoproteins , Receptors, Immunologic
7.
ACS Synth Biol ; 11(11): 3874-3885, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36278399

ABSTRACT

Threose nucleic acid (TNA) is considered a potential RNA progenitor due to its chemical simplicity, base pairing property, and capability of folding into a functional tertiary structure. However, it is unknown whether the functional property can be maintained during transition from TNA to RNA. Here, we use a toggle in vitro selection to identify nucleic acid catalyst sequences that are active in both TNA and RNA backbones. One such nucleic acid enzyme with exchangeable backbone (CAMELEON) catalyzes an RNA cleavage reaction when prepared as TNA (T) and RNA (R). Further biochemical characterization reveals that CAMELEON R and T exhibit different catalytic behaviors such as rate enhancement and magnesium dependence. Structural probing and mutagenesis experiments suggest that they likely fold into distinct tertiary structures. This work demonstrates that the catalytic activity can be preserved during backbone transition from TNA to RNA and provides further experimental support for TNA as an RNA precursor in evolution.


Subject(s)
Nucleic Acids , RNA, Catalytic , Nucleic Acids/chemistry , RNA/genetics , RNA/chemistry , Tetroses/chemistry , Base Pairing , Nucleic Acid Conformation , RNA, Catalytic/genetics
8.
Eur J Med Chem ; 242: 114691, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36029563

ABSTRACT

Platinum(IV) prodrugs c,c,t-[PtCl2(NH3)2(OH)(amlexanox)] (MAP) and c,c,t-[PtCl2(NH3)2(amlexanox)2] (DAP) were synthesized by reacting amlexanox with oxoplatin and characterized by NMR, HR-MS, HPLC, and elemental analysis. The complexes could be reduced to platinum(II) species and amlexanox to exert antitumor activity. Generally, MAP was more potent than DAP and cisplatin towards various human cancer cell lines; particularly, it was active in cisplatin-resistant Caov-3 ovarian cancer and A549/DDP lung cancer cells. MAP induced serious damage to DNA, remarkable change in mitochondrial morphology, decrease in mitochondrial membrane potential, release of cytochrome c from mitochondria, and up-regulation of pro-apoptotic protein Bax in Caov-3 cells, thereby leading to evident apoptosis. Meanwhile, MAP markedly promoted the autophagic flux, including affecting the expression of microtubule-associated protein light chain 3 (LC3) and autophagy adaptor protein p62 in Caov-3 cells, with an increase in the ratio of LC3-II/LC3-I and a decrease in p62, thus trigging the occurrence of autophagy. The MAP-induced bimodal cell death mode is uncommon for platinum complexes, which presents a new possibility to invent anticancer drugs with unique mechanism of action.


Subject(s)
Antineoplastic Agents , Neoplasms , Prodrugs , Aminopyridines , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Apoptosis Regulatory Proteins , Autophagy , Cell Line, Tumor , Cisplatin/pharmacology , Cytochromes c/metabolism , DNA/pharmacology , Humans , Microtubule-Associated Proteins , Platinum/chemistry , Prodrugs/chemistry , bcl-2-Associated X Protein
9.
Nat Chem ; 14(3): 350-359, 2022 03.
Article in English | MEDLINE | ID: mdl-34916596

ABSTRACT

Threose nucleic acid has been considered a potential evolutionary progenitor of RNA because of its chemical simplicity, base pairing properties and capacity for higher-order functions such as folding and specific ligand binding. Here we report the in vitro selection of RNA-cleaving threose nucleic acid enzymes. One such enzyme, Tz1, catalyses a site-specific RNA-cleavage reaction with an observed pseudo first-order rate constant (kobs) of 0.016 min-1. The catalytic activity of Tz1 is maximal at 8 mM Mg2+ and remains relatively constant from pH 5.3 to 9.0. Tz1 preferentially cleaves a mutant epidermal growth factor receptor RNA substrate with a single point substitution, while leaving the wild-type intact. We demonstrate that Tz1 mediates selective gene silencing of the mutant epidermal growth factor receptor in eukaryotic cells. The identification of catalytic threose nucleic acids provides further experimental support for threose nucleic acid as an ancestral genetic and functional material. The demonstration of Tz1 mediating selective knockdown of intracellular RNA suggests that functional threose nucleic acids could be developed for future biomedical applications.


Subject(s)
Nucleic Acids , ErbB Receptors/metabolism , Nucleic Acid Conformation , Nucleic Acids/chemistry , Point Mutation , RNA/chemistry , Tetroses
10.
Eur J Med Chem ; 229: 114047, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34915428

ABSTRACT

Phagocytosis of cancer cells by antigen presenting cells (APCs) is critical to activate the host's immune responses. However, the targeting ability of APCs to cancer cells is limited by the upregulation of transmembrane protein CD47 on the cancer cell surface. Blocking CD47 can affect the macrophage-mediated phagocytosis. Two platinum-based immunomodulators MUP and DMUP were synthesized to enhance the phagocytic activity of macrophages by blocking the CD47-SIRPα axis. These PtIV complexes not only showed high antiproliferative activity against a panel of human cancer cell lines, but also cooperated with human peripheral blood mononuclear cells (PBMCs) to suppress cancer cells. They acted as immune checkpoint inhibitors to modulate the immune responses of both cancer and immune cells. In particular, DMUP decreased the expression of CD47 in tumor tissues and promoted the polarization of macrophages from M2 to M1 phenotype in a mouse model of non-small cell lung cancer, thereby enhancing the anticancer effect. By interfering with DNA synthesis and stimulating immune system, DMUP takes the advantage of chemotherapy and immunotherapy to inhibit cancer cells. The dual efficacy of DMUP makes it a potential chemoimmunotherapeutic agent in cancer therapy.


Subject(s)
Antineoplastic Agents/chemistry , CD47 Antigen/antagonists & inhibitors , Coordination Complexes/chemistry , Platinum/chemistry , Receptors, Immunologic/antagonists & inhibitors , Animals , Antigens, Differentiation/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , CD47 Antigen/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Drug Screening Assays, Antitumor , Humans , Immune Checkpoint Inhibitors/chemistry , Immune Checkpoint Inhibitors/pharmacology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred ICR , Neoplasms/drug therapy , Neoplasms/therapy , Phagocytosis/drug effects , Reactive Oxygen Species/metabolism , Receptors, Immunologic/metabolism
11.
J Am Chem Soc ; 143(21): 8154-8163, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34028252

ABSTRACT

Threose nucleic acid (TNA) has been considered a potential RNA progenitor in evolution due to its chemical simplicity and base pairing property. Catalytic TNA sequences with RNA ligase activities might have facilitated the transition to the RNA world. Here we report the isolation of RNA ligase TNA enzymes by in vitro selection. The identified TNA enzyme T8-6 catalyzes the formation of a 2'-5' phosphoester bond between a 2',3'-diol and a 5'-triphosphate group, with a kobs of 1.1 × 10-2 min-1 (40 mM Mg2+, pH 9.0). For efficient reaction, T8-6 requires UA|GA at the ligation junction and tolerates variations at other substrate positions. Functional RNAs such as hammerhead ribozyme can be prepared by T8-6-catalyzed ligation, with site-specific introduction of a 2'-5' linkage. Together, this work provides experimental support for TNA as a plausible pre-RNA genetic polymer and also offers an alternative molecular tool for biotechnology.


Subject(s)
Nucleic Acids/metabolism , RNA Ligase (ATP)/metabolism , Tetroses/metabolism , Nucleic Acid Conformation , Nucleic Acids/chemistry , RNA Ligase (ATP)/chemistry , Tetroses/chemistry
12.
Dalton Trans ; 50(10): 3516-3522, 2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33433537

ABSTRACT

Platinum-based anticancer drugs constitute the cornerstone of chemotherapy for various cancers. Although cytotoxic agents are considered to have immunosuppressive effects, increasing evidence suggests that some cytotoxic compounds can effectively stimulate the antitumor immune response by inducing a special type of apoptosis called immunogenic cell death (ICD). A platinum(iv) complex (DCP) modified with the derivative of synthetic capsaicin (nonivamide) was designed to elicit ICD. The complex exhibited high cytotoxicity against a panel of human cancer cell lines including pancreas (PANC-1), breast (MCF-7), and liver (HepG2) cancer cells, and osteosarcoma (MG-63) cells. In addition to causing DNA damage, DCP also triggered the translocation of calreticulin (CRT) as well as the release of ATP and HMGB1 protein in PANC-1 cells, thus manifesting an efficient ICD-inducing effect on cancer cells. Furthermore, the DCP-treated PANC-1 cell-conditioned culture medium promoted the release of IFN-γ and TNF-α to induce the immune response of human peripheral blood mononuclear cells, thereby increasing their cytotoxicity to cancer cells. Concurrently, the phagocytosis of PANC-1 cells by macrophages was also augmented by DCP. The results demonstrate that DCP is an effective inducer of ICD and a potential agent for chemoimmunotherapy of cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Capsaicin/pharmacology , Coordination Complexes/pharmacology , Leukocytes, Mononuclear/drug effects , Photosensitizing Agents/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Capsaicin/chemical synthesis , Capsaicin/chemistry , Cattle , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , DNA/chemistry , DNA/drug effects , DNA Damage , Drug Screening Assays, Antitumor , Humans , Leukocytes, Mononuclear/immunology , Molecular Structure , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry
13.
Metallomics ; 12(11): 1834-1840, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33151228

ABSTRACT

Cisplatin and oxaliplatin are widely used anti-tumour chemotherapeutic agents with different spectra of activity. The therapeutic efficacy of such platinum-based drug is believed to, at least in part, result from formation of Pt-DNA adducts, followed by DNA damage response and ultimately apoptosis. However, it remains unclear whether these DNA lesions caused by cisplatin and oxaliplatin elicit distinct reactions in cellular signaling pathways. Here, a label-free comparative proteomic study was performed to profile the protein phosphorylation patterns using Pt-DNA probes with different ligand identities and geometries. Phosphorylated proteins recognizing different cisplatin- and oxaliplatin-DNA lesions were enriched and analyzed on LC-MS/MS. Proteomic analysis revealed that cisplatin mainly affected proteins involved in mRNA processing, while chromatin organization and rRNA processing are two major biological processes influenced by oxaliplatin. Changes to site-specific phosphorylation levels of two proteins YBX1 and UBF1 were also validated by Western blotting. In particular, platinum drug treatment in colon and liver cancer cell lines down-regulated S484 phosphorylation of UBF1, which is an essential transcription factor responsible for ribosomal DNA transcription activation, implying that inhibition of ribosome biogenesis might be involved in the cytotoxic mechanism of platinum drugs. Collectively, these results directly reflected distinct protein phosphorylation patterns triggered by cisplatin and oxaliplatin, and could also provide valuable resources for future mechanistic studies of platinum-based anti-tumour agents.


Subject(s)
Cisplatin/pharmacology , DNA Adducts/metabolism , Oxaliplatin/pharmacology , Platinum/metabolism , Proteins/metabolism , Proteomics , Base Sequence , Cell Line, Tumor , Gene Ontology , Humans , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Interaction Maps/drug effects
14.
ACS Appl Mater Interfaces ; 12(48): 53592-53597, 2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33206496

ABSTRACT

DNA nanotechnology is powerful in constructing programmable nanostructures with distinct dimensions, sizes, and shapes. However, natural DNA molecules are prone to nuclease degradation, thus limiting the in vivo applications of such DNA nanostructures. 2'-Fluoroarabinonucleic acid (FANA) is a chemically modified oligonucleotide with similar base pairing properties to DNA and exhibits superior physical and chemical stabilities. In this work, FANA molecules were used to construct double crossover nanostructures, and it was demonstrated that incorporation of FANA conferred nucleic acid nanostructures with increased thermal stability and stronger nuclease resistance. More importantly, FANA nanostructures were able to maintain the structural integrity in the strongly acidic environment (pH 1.2). Last, such FANA nanostructures functioned well in acting as stable carriers of small-molecule cargoes for cellular delivery in simulated gastric fluid, while the DNA counterparts were mostly degraded. Collectively, these results demonstrated that FANA self-assembly was not only a substantial complement to the structural DNA nanotechnology but also an appealing molecular tool for in vivo biomedical applications.

15.
Angew Chem Int Ed Engl ; 59(51): 23313-23321, 2020 12 14.
Article in English | MEDLINE | ID: mdl-32897000

ABSTRACT

Breast cancer (BC) is one of the most common malignancies in women and often accompanied by inflammatory processes. Cyclooxygenase-2 (COX-2) plays a vital role in the progression of BC, correlating with the expression of programmed death-ligand 1 (PD-L1). Overexpression of PD-L1 contributes to the immune escape of cancer cells, and its blockade would stimulate anticancer immunity. Two multispecific platinum(IV) complexes DNP and NP were prepared using non-steroidal antiinflammatory drug naproxen (NPX) as axial ligand(s) to inhibit the BC cells. DNP exhibited high cytotoxicity and antiinflammatory properties superior over NP, cisplatin and NPX; moreover, it displayed potent antitumor activity and almost no general toxicity in mice bearing triple-negative breast cancer (TNBC). Mechanistic studies revealed that DNP could downregulate the expression of COX-2 and PD-L1 in vitro and vivo, inhibit the secretion of prostaglandin, reduce the expression of BC-associated protein BRD4 and phosphorylation of extracellular signal-regulated kinases 1/2 (Erk1/2), and block the oncogene c-Myc in BC cells. These findings demonstrate that DNP is capable of intervening in inflammatory, immune, and metastatic processes of BC, thus presenting a new mechanism of action for anticancer platinum(IV) complexes. The multispecificity offers a special superiority for DNP to treat TNBC by combining chemotherapy and immunotherapy in one molecule.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cyclooxygenase 2 Inhibitors/pharmacology , Immunosuppression Therapy , Inflammation/drug therapy , Organoplatinum Compounds/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Breast Neoplasms/diagnosis , Breast Neoplasms/immunology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase 2/immunology , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/chemistry , Drug Screening Assays, Antitumor , Female , Humans , Inflammation/immunology , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry
16.
Inorg Chem ; 59(15): 10920-10927, 2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32654480

ABSTRACT

A reversible fluorescent probe DRhFe is devised by linking spirolactam rhodamine and dansylamide through an Fe3+ ionophore N2-hydroxyethyldiethylenetriamine, where Fe3+ chelation generating rhodamine B can switch intramolecular fluorescence resonance energy transfer for ratiometric Fe3+ sensing. Probe DRhFe exhibits an Fe3+-specific emission shift from 483 to 576 nm, and this ratiometric response has been successfully applied to monitor labile Fe3+ fluctuations in cells undergoing ferroptosis.


Subject(s)
Ferric Compounds/analysis , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Dansyl Compounds/chemistry , Ferroptosis , Fluorescent Dyes/chemical synthesis , HeLa Cells , Humans , Molecular Structure , Optical Imaging , Rhodamines/chemistry
17.
Inorg Chem ; 58(19): 13150-13160, 2019 Oct 07.
Article in English | MEDLINE | ID: mdl-31539237

ABSTRACT

Mitochondrial DNA (mtDNA) is an attractive cellular target for anticancer agents in addition to nuclear DNA (nDNA). The cationic platinum(II) complex cis-[Pt(NP)(NH3)2Cl]NO3 (PtNP, NP = N-(2-ethylpyridine)-1,8-naphthalimide) bearing the DNA-intercalating moiety NP was designed. The structure of PtNP was fully characterized by single-crystal X-ray crystallography, NMR, and HRMS. PtNP is superior to cisplatin in both in vitro and in vivo anticancer activities with low systemic toxicity. The interaction of PtNP with CT-DNA demonstrated that PtNP could effectively bind to DNA through both covalent and noncovalent double binding modes. In addition to causing significant damage to nDNA and remarkable inhibition to DNA damage repair, PtNP also distributed in mitochondria, inducing mtDNA damage and affecting the downstream transcriptional level of mitochondrion-encoded genes. In addition, PtNP disturbed the physiological processes of mitochondria by reducing the mitochondrial membrane potential and promoting the generation of reactive oxygen species. Mechanistic studies demonstrate that PtNP induced apoptosis via mitochondrial pathways by upregulating Bax and Puma and downregulating Bcl-2 proteins, leading to the release of cytochrome c and activation of caspase-3 and caspase-9. As a dual-DNA-damage agent, PtNP is able to improve the anticancer activity by damaging both nuclear and mitochondrial DNA, thus providing a new anticancer mechanism of action for the naphthalimide monofunctional platinum(II) complexes.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage/drug effects , Intercalating Agents/pharmacology , Organoplatinum Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Crystallography, X-Ray , Humans , Intercalating Agents/chemistry , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred ICR , Models, Molecular , Naphthalimides/chemistry , Naphthalimides/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics , Organoplatinum Compounds/chemistry , Pyridines/chemistry , Pyridines/pharmacology
18.
Inorg Chem ; 58(9): 6507-6516, 2019 May 06.
Article in English | MEDLINE | ID: mdl-31013065

ABSTRACT

Cancer is characterized by abnormal cellular energy metabolism, which preferentially switches to aerobic glycolysis rather than oxidative phosphorylation as a means of glucose metabolism. Many key enzymes involved in the abnormal glycolysis are potential targets of anticancer drugs. Platinum(IV) complexes are potential anticancer prodrugs and kinetically more inert than the platinum(II) counterparts, which offer an opportunity to be modified by functional ligands for activation or targeted delivery. A novel platinum(IV) complex, c, c, t-[Pt(NH3)2Cl2(C10H15N2O3S)(C2HO2Cl2)] (DPB), was designed to explore the effects of axial ligands on the reactivity and bioactivity of the complex as well as on tumor energy metabolism. The complex was characterized by electrospray ionization mass spectrometry and multinuclear (1H, 13C, and 195Pt) NMR spectroscopy. The introduction of dichloroacetate (DCA) markedly increases the lipophilicity, reactivity, and cytotoxicity of the complex and blocks the growth of cancer cells having active glycolysis, and the introduction of biotin (C10H16N2O3S) enhances the tumor-targeting potential of the complex. The cytotoxicity of DPB is increased dramatically in a variety of cancer cell lines as compared with the platinum(IV) complex PB without the DCA group. DPB alters the mitochondrial membrane potential and disrupts the mitochondrial morphology. The levels of mitochondrial and cellular reactive oxygen species are also decreased. Furthermore, the mitochondrial function of tumor cells was impaired by DPB, leading to the inhibition of both glycolysis and glucose oxidation and finally to the death of cancer cells via a mitochondria-mediated apoptotic pathway. These findings demonstrate that DPB suppresses cancer cells mainly through altering metabolic pathways and highlight the importance of dual-targeting for the efficacy of anticancer drugs.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Energy Metabolism/drug effects , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , HCT116 Cells , HeLa Cells , Hep G2 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...