Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Chemistry ; 30(32): e202400153, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38566460

ABSTRACT

This paper presents a green and efficient aqueous-phase method for the synthesis of thiosulfonates, which has the benefits of no need for catalysts or redox reagents and a short reaction time, providing a method with great economic value for synthesizing thiosulfonates. Furthermore, 3-Sulfenylindoles can be easily synthesized using this method, which expands the potential applications of this reaction.

2.
Mol Pharm ; 21(1): 346-357, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38015620

ABSTRACT

To overcome the limitations of traditional platinum (Pt)-based drugs and further improve the targeting ability and therapeutic efficacy in vivo, we proposed to design a human serum albumin (HSA)-Pt agent complex nanoparticle (NP) for cancer treatment by multimodal action against the tumor microenvironment. We not only synthesized a series of Pt(II) di-2-pyridone thiosemicarbazone compounds and obtained a Pt(II) agent [Pt(Dp44mT)Cl] with significant anticancer activity but also successfully constructed a novel HSA-Pt(Dp44mT) complex nanoparticle delivery system. The structure of the HSA-Pt(Dp44mT) complex revealed that Pt(Dp44mT)Cl binds to the IIA subdomain of HSA and coordinates with His-242. The HSA-His242-Pt-Dp44mT NPs had an obvious effect on the inhibition of tumor growth, which was superior to that of Dp44mT and Pt(Dp44mT)Cl, and they had almost no toxicity. In addition, the HSA-His242-Pt-Dp44mT NPs were found to kill cancer cells by inducing apoptosis, autophagy, and inhibiting angiogenesis.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Humans , Serum Albumin, Human/chemistry , Platinum , Tumor Microenvironment , Neoplasms/drug therapy , Neoplasms/pathology , Antineoplastic Agents/chemistry , Cell Line, Tumor
3.
Angew Chem Int Ed Engl ; 63(10): e202317257, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38128012

ABSTRACT

Lignin represents the largest aromatic carbon resource in plants, holding significant promise as a renewable feedstock for bioaromatics and other cyclic hydrocarbons in the context of the circular bioeconomy. However, the methoxy groups of aryl methyl ethers, abundantly found in technical lignins and lignin-derived chemicals, limit their pertinent chemical reactivity and broader applicability. Unlocking the phenolic hydroxyl functionality through O-demethylation (ODM) has emerged as a valuable approach to mitigate this need and enables further applications. In this review, we provide a comprehensive summary of the progress in the valorization of technical lignin and lignin-derived chemicals via ODM, both catalytic and non-catalytic reactions. Furthermore, a detailed analysis of the properties and potential applications of the O-demethylated products is presented, accompanied by a systematic overview of available ODM reactions. This review primarily focuses on enhancing the phenolic hydroxyl content in lignin-derived species through ODM, showcasing its potential in the catalytic funneling of lignin and value-added applications. A comprehensive synopsis and future outlook are included in the concluding section of this review.

4.
Carcinogenesis ; 44(7): 549-561, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37466677

ABSTRACT

Immunotherapy is the only approved systemic therapy for advanced cutaneous squamous cell carcinoma (cSCC), however, roughly 50% of patients do not respond to the therapy and resistance often occurs over time to those who initially respond. Immunosuppression could have a critical role in developing treatment resistance, thus, understanding the mechanisms of how immunosuppression is developed and regulated may be the key to improving clinical diagnosis and treatment strategies for cSCC. Here, through using a series of immunocompetent genetically engineered mouse models, we demonstrate that miR-22 promotes cSCC development by establishing regulatory T cells (Tregs)-mediated immunosuppressive tumor microenvironment (TME) in a tumor cell autonomous manner. Mechanism investigation revealed that miR-22 elicits the constitutive activation of JAK/STAT3 signaling by directly targeting its suppressor SOCS3, which augments cancer cell-derived chemokine secretion and Tregs recruitment. Epithelial-specific and global knockouts of miR-22 repress papilloma and cSCC development and progression, manifested with reduced Tregs infiltration and elevated CD8+ T cell activation. Transcriptomic analysis and functional rescue study confirmed CCL17, CCL20 and CCL22 as the main affected chemokines that mediate the chemotaxis between miR-22 highly expressing keratinocyte tumor cells and Tregs. Conversely, overexpression of SOCS3 reversed miR-22-induced Tregs recruitment toward tumor cells. Clinically, gradually increasing Tregs infiltration during cSCC progression was negatively correlated with SOCS3 abundance, supported by previously documented elevated miR-22 levels. Thus, our study uncovers a novel miR-22-SOCS3-JAK/STAT3-chemokines regulatory mechanism in defining the immunosuppressive TME and highlights the promising clinical application value of miR-22 as a common targeting molecule against JAK/STAT3 signaling and immune escape in cSCC.

5.
Sensors (Basel) ; 21(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34770368

ABSTRACT

As a newly developed pile foundation, the snowflake shaped steel sheet pile is composed of three Y-shaped sections with an included angle of 120° and has a large specific surface area, which can give full play to the side friction of pile and improve the bearing capacity of single pile. At the same time, the snowflake shaped steel sheet pile has a high strength, relatively few materials, and it has good prospects with engineering applications. In order to accurately grasp the mechanical characteristics of the snowflake shaped steel sheet pile, this paper carried out the model test of snowflake shaped steel sheet pile based on OFDR (optical frequency domain reflector) distributed optical fiber sensor technology. The results show that: (1) OFDR distributed optical fiber sensing technology can effectively monitor the strain of snowflake steel sheet pile; (2) under the vertical load, the strain of snowflake steel sheet pile decreases along the length of the pile; (3) the strain of the same section of snowflake steel sheet pile is different at different positions, the strain at the junction between web and web is basically the same as the junction between web and flange, and the strain of the pile shaft on the flange edge is larger.

6.
J Biol Chem ; 294(5): 1437-1450, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30498087

ABSTRACT

The inner centromere region of a mitotic chromosome critically regulates sister chromatid cohesion and kinetochore-microtubule attachments. However, the molecular mechanism underlying inner centromere assembly remains elusive. Here, using CRISPR/Cas9-based gene editing in HeLa cells, we disrupted the interaction of Shugoshin 1 (Sgo1) with histone H2A phosphorylated on Thr-120 (H2ApT120) to selectively release Sgo1 from mitotic centromeres. Interestingly, cells expressing the H2ApT120-binding defective mutant of Sgo1 have an elevated rate of chromosome missegregation accompanied by weakened centromeric cohesion and decreased centromere accumulation of the chromosomal passenger complex (CPC), an integral part of the inner centromere and a key player in the correction of erroneous kinetochore-microtubule attachments. When artificially tethered to centromeres, a Sgo1 mutant defective in binding protein phosphatase 2A (PP2A) is not able to support proper centromeric cohesion and CPC accumulation, indicating that the Sgo1-PP2A interaction is essential for the integrity of mitotic centromeres. We further provide evidence indicating that Sgo1 protects centromeric cohesin to create a binding site for the histone H3-associated protein kinase Haspin, which not only inhibits the cohesin release factor Wapl and thereby strengthens centromeric cohesion but also phosphorylates histone H3 at Thr-3 to position CPC at inner centromeres. Taken together, our findings reveal a positive feedback-based mechanism that ensures proper assembly of the functional inner centromere during mitosis. They further suggest a causal link between centromeric cohesion defects and chromosomal instability in cancer cells.


Subject(s)
Cell Cycle Proteins/metabolism , Centromere/metabolism , Feedback, Physiological , Histones/metabolism , Mitosis , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kinetochores/metabolism , Microtubules/metabolism , Phosphorylation , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Cohesins
7.
Mol Pharm ; 17(4): 1405-1414, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32096645

ABSTRACT

To cause tumor regression by acting against cancer cells and inhibiting neovascularization in the tumor microenvironment, we constructed human serum albumin (HSA)-based delivery systems of 2-acetylpyridine-4,4-dimethyl-3-thiosemicarbazone-copper(II) [Cu(Ap44mT)]Cl and paclitaxel to improve both the therapeutic efficacy and the targeting ability in vivo. X-ray crystallography and matrix-assisted laser desorption/ionization time-of-flight mass spectra confirmed that [Cu(Ap44mT)]Cl complexed with HSA, whereas paclitaxel was tethered to the HSA complex by a linker sensitive to the active matrix metalloproteinase 2 (MMP2) protein. Up to 78% of paclitaxel was released from HSA within 2 h owing to MMP2 protein cleavage. In addition, a large amount of Cu(Ap44mT) was released from HSA in a pH 4.7 buffer. In vivo results revealed the following: (1) the tumor inhibitory rates of the HSA conjugate and the two-agent combination were 72.1 and 50.7%, respectively; (2) the inhibition rate of tumor angiogenesis of the HSA conjugate (73.3%) was higher than that of the two-agent combination (52.4%); (3) the increased amount of Cu in the tumor treated with the HSA conjugate was about 2-fold that in the tumor treated with the two-agent combination. Obviously, the HSA conjugate not only possessed a stronger capacity to inhibit neovascularization and the growth of liver tumors but also improved the targeting ability compared to the combination of the two agents alone.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Neovascularization, Pathologic/drug therapy , Serum Albumin, Human/chemistry , Tumor Microenvironment/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy/methods , Copper/chemistry , Drug Delivery Systems/methods , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/metabolism , Paclitaxel/chemistry , Paclitaxel/pharmacology
8.
EMBO Rep ; 19(4)2018 04.
Article in English | MEDLINE | ID: mdl-29491004

ABSTRACT

Heterochromatin protein-1 (HP1) is a key component of heterochromatin. Reminiscent of the cohesin complex which mediates sister-chromatid cohesion, most HP1 proteins in mammalian cells are displaced from chromosome arms during mitotic entry, whereas a pool remains at the heterochromatic centromere region. The function of HP1 at mitotic centromeres remains largely elusive. Here, we show that double knockout (DKO) of HP1α and HP1γ causes defective mitosis progression and weakened centromeric cohesion. While mutating the chromoshadow domain (CSD) prevents HP1α from protecting sister-chromatid cohesion, centromeric targeting of HP1α CSD alone is sufficient to rescue the cohesion defects in HP1 DKO cells. Interestingly, HP1-dependent cohesion protection requires Haspin, an antagonist of the cohesin-releasing factor Wapl. Moreover, HP1α CSD directly binds the N-terminal region of Haspin and facilitates its centromeric localization. The need for HP1 in cohesion protection can be bypassed by centromeric targeting of Haspin or inhibiting Wapl activity. Taken together, these results reveal a redundant role for HP1α and HP1γ in the protection of centromeric cohesion through promoting Haspin localization at mitotic centromeres in mammalian cells.


Subject(s)
Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Heterochromatin/metabolism , Animals , Centromere/genetics , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Gene Knockout Techniques , HeLa Cells , Heterochromatin/genetics , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Mammals , Mitosis/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Transport
9.
EMBO Rep ; 19(1): 43-56, 2018 01.
Article in English | MEDLINE | ID: mdl-29138236

ABSTRACT

Sister-chromatid cohesion mediated by the cohesin complex is fundamental for precise chromosome segregation in mitosis. Through binding the cohesin subunit Pds5, Wapl releases the bulk of cohesin from chromosome arms in prophase, whereas centromeric cohesin is protected from Wapl until anaphase onset. Strong centromere cohesion requires centromeric localization of the mitotic histone kinase Haspin, which is dependent on the interaction of its non-catalytic N-terminus with Pds5B. It remains unclear how Haspin fully blocks the Wapl-Pds5B interaction at centromeres. Here, we show that the C-terminal kinase domain of Haspin (Haspin-KD) binds and phosphorylates the YSR motif of Wapl (Wapl-YSR), thereby directly inhibiting the YSR motif-dependent interaction of Wapl with Pds5B. Cells expressing a Wapl-binding-deficient mutant of Haspin or treated with Haspin inhibitors show centromeric cohesion defects. Phospho-mimetic mutation in Wapl-YSR prevents Wapl from binding Pds5B and releasing cohesin. Forced targeting Haspin-KD to centromeres partly bypasses the need for Haspin-Pds5B interaction in cohesion protection. Taken together, these results indicate a kinase-dependent role for Haspin in antagonizing Wapl and protecting centromeric cohesion in mitosis.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , DNA-Binding Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Amino Acid Motifs , Anaphase , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Centromere/metabolism , Centromere/ultrastructure , Chromatids/metabolism , Chromatids/ultrastructure , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Mutation , Nuclear Proteins/metabolism , Phosphorylation , Prophase , Protein Binding , Protein Serine-Threonine Kinases/deficiency , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Cohesins
10.
J Org Chem ; 85(20): 13029-13036, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32954735

ABSTRACT

An intramolecular decarboxylative coupling reaction for the construction of 2-(1,3,4-oxadiazol-2-yl)aniline derivatives was developed from readily available isatins and hydrazides by virtue of electrochemistry. In this reaction, isatins were employed as amino-attached C1 sources, providing a variety of 2-(1,3,4-oxadiazol-2-yl)aniline derivatives with moderate to good yields.

11.
Mol Pharm ; 15(6): 2180-2193, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29722993

ABSTRACT

Designing a multitarget anticancer drug with improved delivery and therapeutic efficiency in vivo presents a great challenge. Thus, we proposed to design an anticancer multitarget metal pro-drug derived from thiosemicarbazone based on the His146 residue in the IB subdomain of palmitic acid (PA)-modified human serum albumin (HSA-PA). The structure-activity relationship of six Cu(II) compounds with 6-methyl-2-formylpyridine-4N-substituted thiosemicarbazones were investigated, and then the multitarget capability of 4b was confirmed in cancer cell DNA and proteins. The structure of the HSA-PA-4b complex (HSA-PA-4b) revealed that 4b is bound to the IB subdomain of modified HSA, and that His146 replaces the nitrate ligand in 4b, coordinating with Cu2+, whereas PA is complexed with the IIA subdomain by its carboxyl forming hydrogen bonds with Lys199 and His242. In vivo data showed that 4b and the HSA-PA-4b complex inhibit lung tumor growth, and the targeting ability and therapeutic efficacy of the PA-modified HSA complex was stronger than 4b alone.


Subject(s)
Antineoplastic Agents/pharmacology , Copper/chemistry , Neoplasms/drug therapy , Prodrugs/pharmacology , Serum Albumin, Human/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , DNA, Neoplasm/metabolism , Drug Design , Histidine/chemistry , Humans , Mice , Mice, Nude , Molecular Docking Simulation , Molecular Targeted Therapy/methods , Neoplasms/pathology , Prodrugs/chemistry , Prodrugs/therapeutic use , Protein Domains , Structure-Activity Relationship , Xenograft Model Antitumor Assays
12.
Mol Pharm ; 14(6): 1861-1873, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28471669

ABSTRACT

We not only modified the types and numbers of coordinated ligands in a metal agent to enhance its anticancer activity, but we also designed a metal prodrug based on the N-donor residues of the human serum albumin (HSA) IIA subdomain to improve its delivery efficiency and selectivity in vivo. However, there may be a conflict in simultaneously achieving the two goals because Lys199 and His242 in the IIA subdomain of HSA can replace its two coordinated ligands, which will decrease its anticancer activity relative to the original metal agent. Thus, to improve the delivery efficiency of the metal agent and simultaneously avoid decreasing its anticancer activity in vivo, we decided to develop an anticancer metal prodrug by regulating its pharmacophore ligand so that it would not be displaced by the Lys199 residue of the folic acid (FA)-functionalized HSA nanoparticle (NP) carrier. To this end, we first synthesized two (E)-N'-(5-chloro-2-hydroxybenzylidene)benzohydrazide Schiff base (HL) Cu(II) compounds by designing a second ligand with a different coordinating atom with Cu2+/Cu(L)(QL)(Br) [C1, QL = quinolone] and Cu(L)(DMF)(Br) [C2, DMF = N,N-dimethylformamide]. As revealed by the structures of the two HSA complexes, the Cu compounds bind to the hydrophobic cavity in the HSA IIA subdomain. The QL ligand of C1 is replaced by Lys199, which coordinates with Cu2+, whereas the DMF ligand of C2 is kept intact and His242 is replaced with Br- of C2 and coordinates with Cu2+. The cytotoxicity of the Cu compounds was enhanced by the FA-HSA NPs in the Bel-7402 cells approximately 2-4-fold; however, they raise the cytotoxicity levels in the normal cells in vitro, and the FA-HSA NPs did not. Importantly, the in vivo data showed that FA-HSA-C2 NPs increased selectivity and the capacity to inhibit tumor growth and were less toxic than HSA-C2 NPs and C2. Moreover, C2/HSA-C2 NPs/FA-HSA-C2 NPs induced Bel-7402 cell death by potentially multiple mechanisms.


Subject(s)
Antineoplastic Agents/pharmacology , Copper/pharmacology , Drug Delivery Systems/methods , Nanoparticles/chemistry , Prodrugs/pharmacology , Serum Albumin, Human/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Copper/chemistry , Female , Folic Acid/chemistry , Hemolysis/drug effects , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Male , Mice , Mice, Nude , Prodrugs/chemistry , X-Ray Diffraction
13.
Mol Pharm ; 13(5): 1501-7, 2016 05 02.
Article in English | MEDLINE | ID: mdl-27017838

ABSTRACT

To increase delivery efficiency, anticancer activity, and selectivity of anticancer metal agents in vivo, we proposed to develop the anticancer metal pro-drug based on His242 residue of the human serum albumin (HSA) carrier IIA subdomain. To confirm our hypothesis, we prepared two Cu(II) compounds [Cu(P4 mT)Cl and Cu(Bp44 mT)Cl] by modifying Cu(II) compound ligand structure. Studies with two HSA complex structures revealed that Cu(P4 mT)Cl bound to the HSA subdomain IIA via hydrophobic interactions, but Cu(Bp44 mT)Cl bound to the HSA subdomain IIA via His242 replacement of a Cl atom of Cu(Bp44 mT)Cl, and a coordination to Cu(2+). Furthermore, Cu(II) compounds released from HSA could be regulated at different pHs. In vivo data revealed that the HSA-Cu(Bp44 mT) complex increased copper's selectivity and capacity of inhibiting tumor growth compared to Cu(Bp44 mT)Cl alone.


Subject(s)
Antineoplastic Agents/chemistry , Copper/chemistry , Prodrugs/chemistry , Serum Albumin/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites , Copper/pharmacology , Drug Delivery Systems/methods , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Ligands , Neoplasms/drug therapy , Prodrugs/pharmacology , Serum Albumin/pharmacology
14.
J Inorg Biochem ; 250: 112403, 2024 01.
Article in English | MEDLINE | ID: mdl-37866112

ABSTRACT

To develop the next-generation metal agents for efficiently inhibiting tumor growth, a series of novel mononuclear, binuclear and trinuclear copper (Cu) thiophene-2-formaldehyde thiosemicarbazone complexes and a tetranuclear Cu 1,2,4-triazole-derived complex have been synthesized and their structure-activity relationships have been studied. The trinucleated Cu complex showed the strongest inhibitory activity against T24 cells among all the Cu complexes. Its antitumor effect in vivo was superior to that of cisplatin, with reduced side effects. Further studies on the antitumor mechanism have showed that Cu complexes not only induced apoptosis of cancer cells but also inhibited tumor angiogenesis by inhibiting the migration and invasion of vascular endothelial cells, blocking the cell cycle in the G1 phase, and inducing autophagy.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Humans , Antineoplastic Agents/pharmacology , Copper/pharmacology , Models, Molecular , Endothelial Cells , Neoplasms/drug therapy , Apoptosis , Coordination Complexes/pharmacology , Cell Line, Tumor , Cell Proliferation
15.
J Med Chem ; 67(5): 3843-3859, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38442035

ABSTRACT

To develop a potential theranostic metal agent to reverse the resistance of cancer cells to cisplatin and effectively inhibit tumor growth and metastasis, we proposed to design a cyclometalated iridium (Ir) complex based on the properties of the tumor environment (TME). To the end, we designed and synthesized a series of Ir(III) 2-hydroxy-1-naphthaldehyde thiosemicarbazone complexes by modifying the hydrogen atom(s) of the N-3 position of 2-hydroxy-1-naphthaldehyde thiosemicarbazone compounds and the structure of cyclometalated Ir(III) dimers and then investigated their structure-activity and structure-fluorescence relationships to obtain an Ir(III) complex (Ir5) with remarkable fluorescence and cytotoxicity to cancer cells. Ir5 not only possesses mitochondria-targeted properties but also overcomes cisplatin resistance and effectively inhibits tumor growth and metastasis in vivo. Besides, we confirmed the anticancer mechanisms of Ir5 acting on different components in the TME: directly killing liver cancer cells by inducing necroptosis and activating the necroptosis-related immune response.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Naphthalenes , Neoplasms , Thiosemicarbazones , Humans , Cisplatin/pharmacology , Cisplatin/therapeutic use , Antineoplastic Agents/chemistry , Iridium/pharmacology , Iridium/chemistry , Precision Medicine , Necroptosis , Neoplasms/drug therapy , Mitochondria , Coordination Complexes/chemistry , Cell Line, Tumor
16.
J Med Chem ; 67(7): 5744-5757, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38553427

ABSTRACT

To develop a next-generation metal agent and dual-agent multitargeted combination therapy, we developed a copper (Cu) compound based on the properties of the human serum albumin (HSA)-indomethacin (IND) complex to remodel the tumor microenvironment (TME). We optimized a series of Cu(II) isopropyl 2-pyridyl ketone thiosemicarbazone compounds to obtain a Cu(II) compound (C4) with significant cytotoxicity and then constructed an HSA-IND-C4 complex (HSA-IND-C4) delivery system. IND and C4 bind to the hydrophobic cavities of the IB and IIA domains of HSA, respectively. In vivo, the HSA-IND-C4 not only showed enhanced antitumor efficacy relative to C4 and C4 + IND but also improved their targeting ability and decreased their side effects. The antitumor mechanism of C4 + IND involved acting on the different components of the TME. IND inhibited tumor-related inflammation, while C4 not only induced apoptosis and autophagy of cancer cells but also inhibited tumor angiogenesis.


Subject(s)
Antineoplastic Agents , Neoplasms , Prodrugs , Thiosemicarbazones , Humans , Serum Albumin, Human/chemistry , Copper/chemistry , Serum Albumin/chemistry , Thiosemicarbazones/pharmacology , Thiosemicarbazones/therapeutic use , Indomethacin/therapeutic use , Tumor Microenvironment , Prodrugs/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Neoplasms/drug therapy
17.
J Med Chem ; 67(11): 9091-9103, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38778566

ABSTRACT

Induction of cuproptosis and targeting of multiple signaling pathways show promising applications in tumor therapy. In this study, we synthesized two thiosemicarbazone-copper complexes ([CuII(L)Cl] 1 and [CuII2CuI(L)2Cl3] 2, where HL is the (E)-N-methyl-2-(phenyl(pyridin-2-yl)methylene ligand), to assess their antilung cancer activities. Both copper complexes showed better anticancer activity than cisplatin and exhibited hemolysis comparable to that of cisplatin. In vivo experiments showed that complex 2 retarded the A549 cell growth in a mouse xenograft model with low systemic toxicity. Primarily, complex 2 kills lung cancer cells in vitro and in vivo by triggering multiple pathways, including cuproptosis. Complex 2 is the first mixed-valent Cu(I/II) complex to induce cellular events consistent with cuproptosis in cancer cells, which may stimulate the development of mixed-valent copper complexes and provide effective cancer therapy.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Copper , Lung Neoplasms , Thiosemicarbazones , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/therapeutic use , Humans , Copper/chemistry , Animals , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Coordination Complexes/therapeutic use , Mice , Mice, Nude , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Cell Line, Tumor , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , A549 Cells , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Signal Transduction/drug effects , Structure-Activity Relationship , Hemolysis/drug effects , Drug Screening Assays, Antitumor , Mice, Inbred BALB C
18.
Nat Commun ; 15(1): 4108, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750011

ABSTRACT

MAPK pathway-driven tumorigenesis, often induced by BRAFV600E, relies on epithelial dedifferentiation. However, how lineage differentiation events are reprogrammed remains unexplored. Here, we demonstrate that proteostatic reactivation of developmental factor, TBX3, accounts for BRAF/MAPK-mediated dedifferentiation and tumorigenesis. During embryonic development, BRAF/MAPK upregulates USP15 to stabilize TBX3, which orchestrates organogenesis by restraining differentiation. The USP15-TBX3 axis is reactivated during tumorigenesis, and Usp15 knockout prohibits BRAFV600E-driven tumor development in a Tbx3-dependent manner. Deleting Tbx3 or Usp15 leads to tumor redifferentiation, which parallels their overdifferentiation tendency during development, exemplified by disrupted thyroid folliculogenesis and elevated differentiation factors such as Tpo, Nis, Tg. The clinical relevance is highlighted in that both USP15 and TBX3 highly correlates with BRAFV600E signature and poor tumor prognosis. Thus, USP15 stabilized TBX3 represents a critical proteostatic mechanism downstream of BRAF/MAPK-directed developmental homeostasis and pathological transformation, supporting that tumorigenesis largely relies on epithelial dedifferentiation achieved via embryonic regulatory program reinitiation.


Subject(s)
Carcinogenesis , Proto-Oncogene Proteins B-raf , T-Box Domain Proteins , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Animals , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Mice , Cell Differentiation , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , MAP Kinase Signaling System/genetics , Gene Expression Regulation, Neoplastic , Mice, Knockout , Female , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism
19.
Energy Fuels ; 38(10): 8740-8748, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38774064

ABSTRACT

Pyrolysis of lignocellulosic biomass and waste plastics has been intensely studied in the last few decades to obtain renewable fuels and chemicals. Various pyrolysis devices have been developed for use in a laboratory setting, operated either in batch or continuously at scales ranging from milligrams per hour to tenths of g per hour. We report here the design and operation of a novel staged free-fall (catalytic) pyrolysis unit and demonstrate that the concept works very well for the (catalytic) pyrolysis of pinewood sawdust, paper sludge, and polypropylene as representative feeds. The unit consists of a vertical tube with a pretreatment section, a pyrolysis section, a solid residue collection section, a gas-liquid separation/collection section, and a catalytic reaction section to optionally perform ex situ catalytic upgrading of the pyrolysis vapor. The sample is placed in a tube, which is transported by gravity through various sections of the unit. It allows for rapid testing with semicontinuous feeding (e.g., 50 g h-1) and the opportunity to perform reactions under an (inert) gas (e.g., N2) at atmospheric as well as elevated pressure (e.g., 50 bar). Liquid yields for noncatalytic sawdust pyrolysis at optimized conditions (475 °C and atmospheric pressure) were 63 wt % on biomass intake. A lower yield of 51 wt % (on a biomass basis) was obtained for the noncatalytic pyrolysis of paper sludge, likely due to the presence of minerals (e.g., CaCO3) in the feed. The possibility of using the unit for ex situ catalytic pyrolysis (pyrolysis at 475 °C and catalytic upgrading at 550 °C) was also successfully demonstrated using paper sludge as the feed and H-ZSM-5 as the catalyst (21 wt % catalyst on biomass). This resulted in a biphasic liquid product with 25.6 wt % of an aqueous phase and 11 wt % of an oil phase. The yield of benzene, toluene, and xylenes was 1.9 wt % (on a biomass basis). Finally, the concept was also proven for a representative polyolefin (polypropylene), both noncatalytic as well as in situ catalytic pyrolysis using H-ZSM-5 as the catalyst at 500 °C. The liquid yield of thermal, noncatalytic plastic pyrolysis was as high as 77 wt % on plastic intake, while in situ catalytic pyrolysis gave a combined 7.8 wt % yield of benzene, toluene, and xylenes on plastic intake.

20.
Chemistry ; 19(18): 5542-5, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23494927

ABSTRACT

Electrify your chemistry! Direct electrosynthesis of ketones from benzylic methylenes in an undivided cell was realized in moderate to good yields. In this electrosynthesis, electrons instead of conventional oxidants and catalysts are employed to make the reaction environmentally benign. Moreover, the reaction intermediate radical was detected by ESR spectroscopy and the reaction mechanism was clarified.

SELECTION OF CITATIONS
SEARCH DETAIL