Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Oncol Res ; 32(5): 911-923, 2024.
Article in English | MEDLINE | ID: mdl-38686054

ABSTRACT

Photodynamic therapy (PDT) is a promising cancer treatment. This study investigated the antitumor effects and mechanisms of a novel photosensitizer meso-5-[ρ-diethylene triamine pentaacetic acid-aminophenyl]-10,15,20-triphenyl-porphyrin (DTP) mediated PDT (DTP-PDT). Cell viability, reactive oxygen species (ROS), and apoptosis were measured with a Cell Counting Kit-8 assay, DCFH-DA fluorescent probe, and Hoechst staining, respectively. Cell apoptosis- and autophagy-related proteins were examined using western blotting. RNA sequencing was used to screen differentially expressed mRNAs (DERs), and bioinformatic analysis was performed to identify the major biological events after DTP-PDT. Our results show that DTP-PDT inhibited cell growth and induced ROS generation in MCF-7 and SGC7901 cells. The ROS scavenger N-acetyl-L-cysteine (NAC) and the P38 MAPK inhibitor SB203580 alleviated DTP-PDT-induced cytotoxicity. DTP-PDT induced cell apoptosis together with upregulated Bax and downregulated Bcl-2, which could also be inhibited by NAC or SB203580. The level of LC3B-II, a marker of autophagy, was increased by DTP-PDT. A total of 3496 DERs were obtained after DTP-PDT. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses indicated that DERs included those involved in cytosolic ribosomes, the nuclear lumen, protein binding, cell cycle, protein targeting to the endoplasmic reticulum, and ribosomal DNA replication. Disease Ontology and Reactome enrichment analyses indicated that DERs were associated with a variety of cancers and cell cycle checkpoints. Protein-protein interaction results demonstrated that cdk1 and rps27a ranked in the top 10 interacting genes. Therefore, DTP-PDT could inhibit cell growth and induce cell apoptosis and autophagy, partly through ROS and the P38 MAPK signaling pathway. Genes associated with the cell cycle, ribosomes, DNA replication, and protein binding may be the key changes in DTP-PDT-mediated cytotoxicity.


Subject(s)
Apoptosis , Photochemotherapy , Photosensitizing Agents , Reactive Oxygen Species , Transcriptome , Humans , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Porphyrins/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Autophagy/drug effects , MCF-7 Cells , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/genetics , Gene Expression Profiling
2.
Chin J Nat Med ; 20(11): 854-862, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36427919

ABSTRACT

For the purpose of seeking new antibiotics, researchers usually modify the already-existing ones. However, this strategy has been extensively used and is close to its limits, especially in the case of aminoglycosides, and it is difficult to find a proper aminoglycoside antibiotic for novel modification. In this paper, we reported the design, synthesis, and evaluation of a series of 5-epi-neamine derivatives based on the structural information of bacterial 16S RNA A-site binding with aminoglycosides. Bioassay results showed that our design strategy was feasible. Our study offers a new way to search for structurally novel aminoglycosides. Meanwhile, our study provides valuable structure-activity relationship information, which will lead to better understanding and exploitation of the drug target, and improved development of new aminoglycoside antibiotics.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Aminoglycosides/pharmacology , Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , RNA, Ribosomal, 16S/metabolism , Structure-Activity Relationship , Biological Assay
3.
Molecules ; 27(21)2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36364310

ABSTRACT

Chemical modification of old drugs is an important way to obtain new ones, and it has been widely used in developing new aminoglycoside antibiotics. However, many of the previous modifying strategies seem arbitrary for their lack of support from structural biological detail. In this paper, based on the structural information of aminoglycoside and its drug target, we firstly analyzed the reason that some 2'-N-acetylated products of aminoglycosides caused by aminoglycoside-modifying enzyme AAC(2') can partially retain activity, and then we designed, synthesized, and evaluated a series of 2'-modified kanamycin A derivatives. Bioassay results showed our modifying strategy was feasible. Our study provided valuable structure-activity relationship information, which would help researchers to develop new aminoglycoside antibiotics more effectively.


Subject(s)
Aminoglycosides , Kanamycin , Kanamycin/pharmacology , Kanamycin/chemistry , Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Structure-Activity Relationship , Biological Assay , Acetyltransferases
6.
Exp Hematol ; 43(9): 770-4.e2, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25937048

ABSTRACT

Mantle-cell lymphoma (MCL) remains incurable despite numerous therapeutic advances. OSU-2S, a novel nonimmunosuppressive FTY720 (Fingolimod) derivative, exhibits potent cytotoxicity in MCL cell lines and primary cells. OSU-2S increased the surface expression of CD74, a therapeutic antibody target in MCL cells. OSU-2S, in combination with anti-CD74 antibody milatuzumab, enhanced cytotoxicity in MCL. Moreover, MCL tumor antigen receptor tyrosine kinase-like orphan receptor 1 (ROR1) targeted immunonanoparticle-carrying OSU-2S (2A2-OSU-2S-ILP)-mediated selective cytotoxicity of MCL in vitro, as well as activity in a xenografted mouse model of MCL in vivo. The newly developed OSU-2S delivery using ROR1-directed immunonanoparticles provide selective targeting of OSU-2S to MCL and other ROR1(+) malignancies, sparing normal B cells.


Subject(s)
Cytotoxins/pharmacology , Drug Delivery Systems , Lymphoma, Mantle-Cell/metabolism , Propylene Glycols/pharmacology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Sphingosine/analogs & derivatives , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Neoplasm/metabolism , Fingolimod Hydrochloride , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Lymphoma, Mantle-Cell/genetics , Mice , Mice, Inbred NOD , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Sphingosine/pharmacology , Xenograft Model Antitumor Assays
7.
J Med Chem ; 58(5): 2290-8, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25689347

ABSTRACT

Previously, we reported that Akt inactivation by γ-tocopherol (2) in PTEN-negative prostate cancer cells resulted from its unique ability to facilitate membrane co-localization of Akt and PHLPP1 (PH domain leucine-rich repeat protein phosphatase isoform 1), a Ser473-specific Akt phosphatase, through pleckstrin homology (PH) domain binding. This finding provided a basis for exploiting 2 to develop a novel class of PHLPP1-targeted Akt inhibitors. Here, we used 3 (γ-VE5), a side chain-truncated 2 derivative, as a scaffold for lead optimization. The proof-of-concept of this structural optimization was obtained by 20, which exhibited higher antitumor efficacy than 3 in PTEN-negative cancer cells through PHLPP1-facilitated Akt inactivation. Like 3, 20 preferentially recognized the PH domains of Akt and PHLPP1, as its binding affinities for other PH domains, including those of ILK and PDK1, were an order-of-magnitude lower. Moreover, 20 was orally active in suppressing xenograft tumor growth in nude mice, which underlines the translational potential of this new class of Akt inhibitor in PTEN-deficient cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Membrane/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , gamma-Tocopherol/chemistry , gamma-Tocopherol/pharmacology , Animals , Antineoplastic Agents/chemistry , Antioxidants/chemistry , Antioxidants/pharmacology , Cell Survival/drug effects , Drug Design , Humans , Immunoblotting , Immunoprecipitation , Male , Mice , Mice, Nude , Phosphorylation/drug effects , Protein Binding , Surface Plasmon Resonance , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
J Pharm Biomed Anal ; 98: 160-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24927402

ABSTRACT

OSU-2S is a novel anti-cancer and immune modulatory agent designed specifically to avert the immunosuppressive effects and related toxicities observed in clinical studies with its predecessor analog, FTY720. To characterize its preclinical pharmacokinetics, a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed and validated for the quantification of OSU-2S in mouse plasma. Ethyl acetate extraction of samples containing OSU-2S and the internal standard, Sph-17, was followed by separation with a 6min gradient (water/0.1% formic acid and methanol/0.1% formic acid) on a reverse-phase C18 column at room temperature. Selected reaction monitoring was used for detection on a triple quadrupole mass spectrometer with positive ionization. The assay was linear over the concentration range 3-3000ng/mL with accuracy ranging from 103 to 111%, and both within- and between-run precision (CV%) ≤11%. All stability samples were within ±15% of nominal values, and replicates were within 15% CV. The assay was successfully applied to a mouse pharmacokinetic study of OSU-2S with intravenous and intraperitoneal administration. OSU-2S non-compartmental pharmacokinetic parameters, area under the concentration-time curve, clearance, and elimination half-life were estimated at 1522hµg/L, 3.06L/h/kg and 15.6h, respectively, for intravenous injection. Systemic availability after intraperitoneal injection was approximately 46%. These data demonstrate the OSU-2S compound displays acceptable pharmacokinetic properties for further in vivo pharmacologic evaluation, which can be facilitated by the validated LC-MS/MS assay.


Subject(s)
Plasma/chemistry , Propylene Glycols/chemistry , Sphingosine/analogs & derivatives , Animals , Chromatography, Liquid/methods , Female , Fingolimod Hydrochloride , Half-Life , Mice , Mice, Inbred ICR , Sphingosine/chemistry , Tandem Mass Spectrometry
9.
Carcinogenesis ; 35(10): 2203-13, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24879635

ABSTRACT

Gemcitabine resistance remains a significant clinical challenge. Here, we used a novel glucose transporter (Glut) inhibitor, CG-5, as a proof-of-concept compound to investigate the therapeutic utility of targeting the Warburg effect to overcome gemcitabine resistance in pancreatic cancer. The effects of gemcitabine and/or CG-5 on viability, survival, glucose uptake and DNA damage were evaluated in gemcitabine-sensitive and gemcitabine-resistant pancreatic cancer cell lines. Mechanistic studies were conducted to determine the molecular basis of gemcitabine resistance and the mechanism of CG-5-induced sensitization to gemcitabine. The effects of CG-5 on gemcitabine sensitivity were investigated in a xenograft tumor model of gemcitabine-resistant pancreatic cancer. In contrast to gemcitabine-sensitive pancreatic cancer cells, the resistant Panc-1 and Panc-1(GemR) cells responded to gemcitabine by increasing the expression of ribonucleotide reductase M2 catalytic subunit (RRM2) through E2F1-mediated transcriptional activation. Acting as a pan-Glut inhibitor, CG-5 abrogated this gemcitabine-induced upregulation of RRM2 through decreased E2F1 expression, thereby enhancing gemcitabine-induced DNA damage and inhibition of cell survival. This CG-5-induced inhibition of E2F1 expression was mediated by the induction of a previously unreported E2F1-targeted microRNA, miR-520f. The addition of oral CG-5 to gemcitabine therapy caused greater suppression of Panc-1(GemR) xenograft tumor growth in vivo than either drug alone. Glut inhibition may be an effective strategy to enhance gemcitabine activity for the treatment of pancreatic cancer.


Subject(s)
Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Thiazolidinediones/pharmacology , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Deoxycytidine/pharmacology , E2F1 Transcription Factor , Female , Glucose/metabolism , Humans , Mice , Mice, Nude , MicroRNAs/genetics , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Xenograft Model Antitumor Assays , Gemcitabine , Pancreatic Neoplasms
10.
J Med Chem ; 55(8): 3827-36, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22468970

ABSTRACT

On the basis of our finding that the antitumor effect of 5-{4-[(1-methylcyclohexyl)methoxy]benzyl}thiazolidine-2,4-dione, a thiazolidinedione peroxisome proliferator-activated receptor (PPAR)γ agonist, was, in part, attributable to its ability to block glucose uptake independently of PPARγ, we used its PPARγ-inactive analogue to develop a novel class of glucose transporter (GLUT) inhibitors. This lead optimization led to compound 30 {5-(4-hydroxy-3-trifluoromethylbenzylidene)-3-[4,4,4-trifluoro-2-methyl-2-(2,2,2-trifluoroethyl)butyl]thiazolidine-2,4-dione} as the optimal agent, which exhibited high antitumor potency through the suppression of glucose uptake (IC(50), 2.5 µM), while not cytotoxic to prostate and mammary epithelial cells. This glucose uptake inhibition was associated with the inhibition of GLUT1 (IC(50), 2 µM). Moreover, the mechanism of antitumor action of compound 30 was validated by its effect on a series of energy restriction-associated cellular responses. Homology modeling analysis suggests that the inhibitory effect of compound 30 on glucose entry was attributable to its ability to bind to the GLUT1 channel at a site distinct from that of glucose.


Subject(s)
Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Glucose Transporter Type 1/antagonists & inhibitors , Thiazolidinediones/chemical synthesis , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Glucose Transport Proteins, Facilitative/metabolism , Humans , Male , PPAR gamma/agonists , Thiazolidinediones/pharmacology
11.
Bioorg Med Chem ; 19(1): 30-40, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21177112

ABSTRACT

Based on the structural information of biomacromolecule-aminoglycoside complexes, a series of kanamycin B analogues were rationally designed and synthesized. A convenient approach to the construction of kanamycin derivatives, in which the C4'-position on ring I of neamine moiety was modified, was developed. Most synthetic analogues exhibited good to excellent antibiotic activity against some typical drug-resistant bacteria. The disclosed results suggested that the C4'-position of aminoglycosides such as kanamycin may be an ideal site for modification to gain new modifying enzyme-resistant aminoglycoside antibiotics.


Subject(s)
Aminoglycosides/chemical synthesis , Aminoglycosides/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Drug Design , Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Bacteria/drug effects , Carbohydrate Sequence , Drug Resistance, Microbial , Models, Molecular , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...