Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Type of study
Publication year range
1.
J Gen Physiol ; 150(12): 1747-1757, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30352794

ABSTRACT

Modulators of insulin secretion could be used to treat diabetes and as tools to investigate ß cell regulatory pathways in order to increase our understanding of pancreatic islet function. Toward this goal, we previously used an insulin-linked luciferase that is cosecreted with insulin in MIN6 ß cells to perform a high-throughput screen of natural products for chronic effects on glucose-stimulated insulin secretion. In this study, using multiple phenotypic analyses, we found that one of the top natural product hits, chromomycin A2 (CMA2), potently inhibited insulin secretion by at least three potential mechanisms: disruption of Wnt signaling, interference of ß cell gene expression, and partial suppression of Ca2+ influx. Chronic treatment with CMA2 largely ablated glucose-stimulated insulin secretion even after washout, but it did not inhibit glucose-stimulated generation of ATP or Ca2+ influx. However, by using the KATP channel opener diazoxide, we uncovered defects in depolarization-induced Ca2+ influx that may contribute to the suppressed secretory response. Glucose-responsive ERK1/2 and S6 phosphorylation were also disrupted by chronic CMA2 treatment. By querying the FUSION bioinformatic database, we revealed that the phenotypic effects of CMA2 cluster with a number of Wnt-GSK3 pathway-related genes. Furthermore, CMA2 consistently decreased GSK3ß phosphorylation and suppressed activation of a ß-catenin activity reporter. CMA2 and a related compound, mithramycin, are known to have DNA interaction properties, possibly abrogating transcription factor binding to critical ß cell gene promoters. We observed that CMA2 but not mithramycin suppressed expression of PDX1 and UCN3. However, neither expression of INSI/II nor insulin content was affected by chronic CMA2. The mechanisms of CMA2-induced insulin secretion defects may involve components both proximal and distal to Ca2+ influx. Therefore, CMA2 is an example of a chemical that can simultaneously disrupt ß cell function through both noncytotoxic and cytotoxic mechanisms. Future therapeutic applications of CMA2 and similar aureolic acid analogues should consider their potential effects on pancreatic islet function.


Subject(s)
Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Plicamycin/analogs & derivatives , Animals , Cell Line , Gene Expression/drug effects , Humans , Mice , Plicamycin/isolation & purification , Plicamycin/pharmacology , Primary Cell Culture , Signal Transduction/drug effects , Streptomyces/chemistry
2.
Nat Prod Commun ; 12(4): 571-577, 2017 Apr.
Article in English | MEDLINE | ID: mdl-30520599

ABSTRACT

A new antibiotic complex of six aureolic acids was isolated from the marine sediment-associated strain Streptomyces sp. KMM 9048. Four of the compounds (3-6) were found to be similar but not identical to the known chromomycins A2, A3, demethyl chromomycin A3 and A4. The two remaining.compounds; A2₋1 (1) and A3₋1 (2), were established as novel chromomycin analogs, which did not contain sugar B. Spectroscopic methods including ID and 2D NMR, and HRMS and MS/MS were applied for structure elucidation. Compounds 1-5 showed strong antimicrobial activity against Gram-positive indicatory bacteria Enterococcusfaecium, Staphylococcus aureus, S. epidernzidis, and Bacillus subtilis. Antitumor assay indicated that all tested compounds, in different manners, inhibited colony formation of RPMI-7951 and SK-Mel-28 cancer cells. This is the first study reporting the inhibitory effects of chromomycin analogs 1-5 on the colony formation of the investigated cancer cell lines. Compound 3, in a concentration of 5 nM, inhibited colony formation of RPMI-7951 and SK-Mel-28 cells by 82 % and 72 %, respectively. Our finding indicated that, of the compounds tested, 3 and 4 are promising anticancer and antimicrobial agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Geologic Sediments/microbiology , Plicamycin/pharmacology , Streptomyces/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Cell Line, Tumor , Chromomycins/chemistry , Chromomycins/isolation & purification , Chromomycins/pharmacology , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , Humans , Microbial Sensitivity Tests , Plicamycin/chemistry , Plicamycin/isolation & purification , Streptomyces/genetics , Streptomyces/isolation & purification , Streptomyces/metabolism , Tandem Mass Spectrometry
3.
Mar Drugs ; 14(8)2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27537898

ABSTRACT

Targeting autophagic pathways might play a critical role in designing novel chemotherapeutic approaches in the treatment of human cancers, and the prevention of tumor-derived chemoresistance. Marine compounds were found to decrease tumor cell growth in vitro and in vivo. Some of them were shown to induce autophagic flux in tumor cells. In this study, we observed that the selected marine life-derived compounds (Chromomycin A2, Psammaplin A, and Ilimaquinone) induce expression of several autophagic signaling intermediates in human squamous cell carcinoma, glioblastoma, and colorectal carcinoma cells in vitro through a transcriptional regulation by tumor protein (TP)-p53 family members. These conclusions were supported by specific qPCR expression analysis, luciferase reporter promoter assay, and chromatin immunoprecipitation of promoter sequences bound to the TP53 family proteins, and silencing of the TP53 members in tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , Aquatic Organisms/chemistry , Autophagy/drug effects , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chromatin Immunoprecipitation , Disulfides/chemistry , Disulfides/isolation & purification , Disulfides/pharmacology , Humans , Plicamycin/analogs & derivatives , Plicamycin/chemistry , Plicamycin/isolation & purification , Plicamycin/pharmacology , Quinones/chemistry , Quinones/isolation & purification , Quinones/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Sesquiterpenes/chemistry , Sesquiterpenes/isolation & purification , Sesquiterpenes/pharmacology , Tumor Suppressor Protein p53/genetics , Tyrosine/analogs & derivatives , Tyrosine/chemistry , Tyrosine/isolation & purification , Tyrosine/pharmacology
4.
Mar Drugs ; 12(6): 3466-76, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24905484

ABSTRACT

A biological screening study of an actinomycetes strain assembly was conducted using a cell-based cytotoxicity assay. The CKK1019 strain was isolated from a sea sand sample. Cytotoxicity-guided fractionation of the CKK1019 strain culture broth, which exhibited cytotoxicity, led to the isolation of chromomycins A2 (1) and A3 (2). 1 and 2 showed potent cytotoxicity against the human gastric adenocarcinoma (AGS) cell line (IC50 1; 1.7 and 2; 22.1 nM), as well as strong inhibitory effects against TCF/ß-catenin transcription (IC50 1; 1.8 and 2; 15.9 nM). 2 showed the ability to overcome tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) resistance. To the best of our knowledge, the effects of chromomycins A2 (1) and A3 (2) on TRAIL resistance-overcoming activity, and on the Wnt signaling pathway, have not been reported previously. Thus, 1 and 2 warrant potential drug lead studies in relation to TRAIL-resistant and Wnt signal-related diseases and offer potentially useful chemical probes for investigating TRAIL resistance and the Wnt signaling pathway.


Subject(s)
Actinobacteria/metabolism , Adenocarcinoma/drug therapy , Chromomycin A3/pharmacology , Plicamycin/analogs & derivatives , Stomach Neoplasms/drug therapy , Adenocarcinoma/pathology , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chromomycin A3/isolation & purification , Geologic Sediments/microbiology , Humans , Inhibitory Concentration 50 , Plicamycin/isolation & purification , Plicamycin/pharmacology , Stomach Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Wnt Signaling Pathway/drug effects
6.
J Antibiot (Tokyo) ; 51(3): 261-6, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9589060

ABSTRACT

We developed a microbial prescreen using Bacillus stearothermophilus NUB3620 and bacteriophage TP-68 to detect potential antitumor compounds acting on DNA or topoisomerases. During the course of screening microbial cultures for their antibacteriophage activities, we found that Streptomyces sp. isolated from a soil sample collected in Iwakuni city, Yamaguchi prefecture, Japan, produced a new antitumor antibiotic, UCH9. UCH9 was isolated from culture broth by a combination of EtOAc extraction and column chromatography. UCH9 has a new structure related to the antitumor antibiotic chromomycins. It exhibited antimicrobial activity against Gram-positive organisms. UCH9 also showed cytotoxic activity against HeLa S3 cells with an IC50 value of 13 nM and exhibited antitumor activity in vivo against mouse leukemia P388.


Subject(s)
Antibiotics, Antineoplastic/isolation & purification , Fermentation , Plicamycin/analogs & derivatives , Streptomyces/classification , Animals , Antibiotics, Antineoplastic/pharmacology , HeLa Cells , Humans , Leukemia P388/drug therapy , Male , Mice , Plicamycin/isolation & purification , Plicamycin/pharmacology , Streptomyces/metabolism
8.
Antibiotiki ; 27(2): 83-7, 1982 Feb.
Article in Russian | MEDLINE | ID: mdl-6461290

ABSTRACT

Variamycin B, a new antitumor antibiotic was isolated from metabolic products of Streptomyces olivovariabilis sp. nov. The empirical formula of variamycin B is C52H76O24, the melting point is 163-165 degrees C (dec.), (alpha)20D--30 +/- 2 degrees (C 0.5, alcohol), the UV-spectrum: lambda max 230, 279, 317, 330, 415 (lg E 4.27, 4.6, 3.83, 3.75, 3.95), IR-spectrum: 1080, 1170, 1380, 1570, 1640, 1720, 3400 cm-1. Variamycin B is classified as belonging to the group of antibiotic analogs of aureolic acid. Chromomycinone, tetrazide and residues of two 2,6-didesoxysugars, i.e. olivose and variose were obtained as a result of complete and partial acid degradation of variamycin B. It was shown with the data of quantitative analysis of the hydrolysates obtained on complete hydrolysis of variamycin B that the ratio between the residues of variose and olivose in the antibiotic molecule was 1 : 4. Oxidation of variamycin B with Fremi salt resulted in formation of chinone having chromomycinone-chinone, 2 residues of olivose and 1 residue of variose in its composition. Investigation of the products of partial hydrolysis provided isolation of a substance having aglicone-chromomycenone and 4 residues of olivose in its composition. A partial structure of variamycin B is proposed.


Subject(s)
Antibiotics, Antineoplastic/isolation & purification , Plicamycin/analogs & derivatives , Streptomyces/metabolism , Antibiotics, Antineoplastic/analysis , Antibiotics, Antineoplastic/biosynthesis , Chemical Phenomena , Chemistry, Physical , Plicamycin/analysis , Plicamycin/biosynthesis , Plicamycin/isolation & purification , Spectrophotometry, Ultraviolet
SELECTION OF CITATIONS
SEARCH DETAIL
...