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











Publication year range
1.
Molecules ; 28(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37298761

ABSTRACT

Brefeldin A has a wide range of anticancer activity against a variety of tumor cells. Its poor pharmacokinetic properties and significant toxicity seriously hinder its further development. In this manuscript, 25 brefeldin A-isothiocyanate derivatives were designed and synthesized. Most derivatives showed good selectivity between HeLa cells and L-02 cells. In particular, 6 exhibited potent antiproliferative activity against HeLa cells (IC50 = 1.84 µM) with no obvious cytotoxic activity to L-02 (IC50 > 80 µM). Further cellular mechanism tests indicated that 6 induced HeLa cell cycle arrest at G1 phase. Cell nucleus fragmentation and decreased mitochondrial membrane potential suggested 6 could induce apoptosis in HeLa cells through the mitochondrial-dependent pathway.


Subject(s)
Antineoplastic Agents , Uterine Cervical Neoplasms , Female , Humans , HeLa Cells , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Brefeldin A/pharmacology , Brefeldin A/therapeutic use , Cell Proliferation , Apoptosis , Isothiocyanates/pharmacology , Isothiocyanates/therapeutic use , Drug Screening Assays, Antitumor , Cell Line, Tumor , Structure-Activity Relationship
2.
Cell Stress Chaperones ; 27(5): 561-572, 2022 09.
Article in English | MEDLINE | ID: mdl-36029373

ABSTRACT

Pulmonary arterial hypertension (PAH) is driven by vascular remodelling due to inflammation and cellular stress, including endoplasmic reticulum stress (ER stress). The main ER-stress chaperone, glucose-regulated protein 78 kDa (GRP78), is known to have protective effects in inflammatory diseases through extracellular signalling. The aim of this study is to investigate its significance in PAH. Human pulmonary arterial smooth muscle cells (PASMC) were stimulated with compounds that induce ER stress, after which the secretion of GRP78 into the cell medium was analysed by western blot. We found that when ER stress was induced in PASMC, there was also a time-dependent secretion of GRP78. Next, naïve PASMC were treated with conditioned medium (CM) from the ER-stressed donor PASMC. Incubation with CM from ER-stressed PASMC reduced the viability, oxidative stress, and expression of inflammatory and ER-stress markers in target cells. These effects were abrogated when the donor cells were co-treated with Brefeldin A to inhibit active secretion of GRP78. Direct treatment of PASMC with recombinant GRP78 modulated the expression of key inflammatory markers. Additionally, we measured GRP78 plasma levels in 19 PAH patients (Nice Group I) and correlated the levels to risk stratification according to ESC guidelines. Here, elevated plasma levels of GRP78 were associated with a favourable risk stratification. In conclusion, GRP78 is secreted by PASMC under ER stress and exhibits protective effects from the hallmarks of PAH in vitro. Circulating GRP78 may serve as biomarker for risk adjudication of patients with PAH. Proposed mechanism of ER-stress-induced GRP78 secretion by PASMC. Extracellular GRP78 can be measured as a circulating biomarker and is correlated with favourable clinical characteristics. Conditioned medium from ER-stressed PASMC reduces extensive viability, ROS formation, inflammation, and ER stress in target cells. These effects can be abolished by blocking protein secretion in donor cells by using Brefeldin A.


Subject(s)
Hypertension, Pulmonary , Pulmonary Artery , Brefeldin A/metabolism , Brefeldin A/pharmacology , Brefeldin A/therapeutic use , Cell Proliferation , Cells, Cultured , Culture Media, Conditioned/pharmacology , Glucose/metabolism , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Inflammation/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Reactive Oxygen Species/metabolism , Vascular Remodeling
3.
Molecules ; 27(9)2022 Apr 28.
Article in English | MEDLINE | ID: mdl-35566172

ABSTRACT

The estrogen receptor α (ERα) is an important biological target mediating 17ß-estradiol driven breast cancer (BC) development. Aiming to develop innovative drugs against BC, either wild-type or mutated ligand-ERα complexes were used as source data to build structure-based 3-D pharmacophore and 3-D QSAR models, afterward used as tools for the virtual screening of National Cancer Institute datasets and hit-to-lead optimization. The procedure identified Brefeldin A (BFA) as hit, then structurally optimized toward twelve new derivatives whose anticancer activity was confirmed both in vitro and in vivo. Compounds as SERMs showed picomolar to low nanomolar potencies against ERα and were then investigated as antiproliferative agents against BC cell lines, as stimulators of p53 expression, as well as BC cell cycle arrest agents. Most active leads were finally profiled upon administration to female Wistar rats with pre-induced BC, after which 3DPQ-12, 3DPQ-3, 3DPQ-9, 3DPQ-4, 3DPQ-2, and 3DPQ-1 represent potential candidates for BC therapy.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Brefeldin A/pharmacology , Brefeldin A/therapeutic use , Estrogen Receptor alpha/metabolism , Female , Humans , Quantitative Structure-Activity Relationship , Rats , Rats, Wistar
4.
Anticancer Agents Med Chem ; 22(6): 1091-1101, 2022.
Article in English | MEDLINE | ID: mdl-34102989

ABSTRACT

BACKGROUND: Chronic Myeloid Leukemia (CML) is a myeloproliferative disease caused by BCR-ABL oncoprotein. Tyrosine kinase inhibitors have been developed to inhibit the activity of BCR-ABL; however, drug resistance and side effect occur in clinic application. Therefore, it is urgent to find novel drugs for CML treatment. Under the guidance of cytotoxic activity, crude extracts of 55 fungal strains from the medicinal mangrove Acanthus ilicifolius were evaluated, and one potent cytotoxic natural compound, brefeldin A (BFA), was discovered from Penicillium sp. (HS-N-29). OBJECTIVE: This study was aimed to determine the cytotoxic activity of BFA and the effect on the activation and expression of BCR-ABL in K562 cells. METHODS: We evaluated cytotoxic activity by MTT assay and soft agar clone assay; apoptosis and cell cycle distribution by Muse cell analyzer. The protein level of BCR-ABL and signaling molecules was detected by western blotting, and the mRNA level of BCR-ABL was determined by RT-PCR. RESULTS: BFA inhibited cell proliferation, induced G2/M cell cycle arrest, and stimulated cell apoptosis in K562 cells. Importantly, for the first time, we revealed that BFA inhibited the activation of BCR-ABL and consequently inhibited the activation of its downstream signaling molecules in K562 cells. Moreover, we found BFA degraded BCR-ABL without affecting its transcription in K562 cells, and BFA-induced BCR-ABL degradation was related to caspase activation, while not to autophagy or ubiquitinated proteasome degradation pathway. CONCLUSION: Our present results indicate that BFA acts as a dual functional inhibitor and degrader of BCR-ABL, and BFA is a potential compound for chemotherapeutics to overcome CML.


Subject(s)
Antineoplastic Agents , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Brefeldin A/pharmacology , Brefeldin A/therapeutic use , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/metabolism , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
5.
Article in English | MEDLINE | ID: mdl-30397071

ABSTRACT

Antifungal agents directed against novel therapeutic targets are required for treating invasive, chronic, and allergic Aspergillus infections. Competitive fitness profiling technologies have been used in a number of bacterial and yeast systems to identify druggable targets; however, the development of similar systems in filamentous fungi is complicated by the fact that they undergo cell fusion and heterokaryosis. Here, we demonstrate that cell fusion in Aspergillus fumigatus under standard culture conditions is not predominately constitutive, as with most ascomycetes, but can be induced by a range of extracellular stressors. Using this knowledge, we have developed a barcode-free genetic profiling system that permits high-throughput parallel determination of strain fitness in a collection of diploid A. fumigatus mutants. We show that heterozygous cyp51A and arf2 null mutants have reduced fitness in the presence of itraconazole and brefeldin A, respectively, and a heterozygous atp17 null mutant is resistant to brefeldin A.


Subject(s)
Antifungal Agents/therapeutic use , Aspergillus fumigatus/drug effects , Brefeldin A/therapeutic use , Cell Fusion/methods , Drug Resistance, Multiple, Fungal/genetics , Itraconazole/therapeutic use , ADP-Ribosylation Factors/genetics , Aspergillosis/drug therapy , Aspergillus fumigatus/genetics , Aspergillus fumigatus/physiology , Cytochrome P-450 Enzyme System/genetics , Fungal Proteins/genetics , Gene Knockout Techniques , Humans , Microbial Sensitivity Tests , Mitochondrial Proton-Translocating ATPases/genetics
6.
Nat Rev Cancer ; 10(12): 842-57, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21102635

ABSTRACT

There is now considerable and increasing evidence for a causal role for aberrant activity of the Ras superfamily of small GTPases in human cancers. These GTPases function as GDP-GTP-regulated binary switches that control many fundamental cellular processes. A common mechanism of GTPase deregulation in cancer is the deregulated expression and/or activity of their regulatory proteins, guanine nucleotide exchange factors (GEFs) that promote formation of the active GTP-bound state and GTPase-activating proteins (GAPs) that return the GTPase to its GDP-bound inactive state. In this Review, we assess the association of GEFs and GAPs with cancer and their druggability for cancer therapeutics.


Subject(s)
GTPase-Activating Proteins/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Brefeldin A/therapeutic use , Drug Discovery , GTPase-Activating Proteins/physiology , Guanine Nucleotide Exchange Factors/physiology , Humans , Neoplasms/etiology , Neoplasms/metabolism , Proto-Oncogene Proteins/physiology , T-Lymphoma Invasion and Metastasis-inducing Protein 1
7.
World J Gastroenterol ; 15(7): 836-44, 2009 Feb 21.
Article in English | MEDLINE | ID: mdl-19230044

ABSTRACT

AIM: To investigate the effect of detachment of esophageal cancer cells from extracellular matrix on the localization of death receptor 5 (DR5) and apoptosis. METHODS: Anchorage-dependent EC9706 cells of esophageal squamous cell carcinoma were pretreated or not treated with brefeldin A. Detached cells were harvested by ethylenediaminetetraacetic acid digestion. Expression and localization of DR5 in these cells were determined by immunocytochemical and immunofluorescence assays, as well as flow cytometry analysis. Apoptosis of EC9706 cells was detected by flow cytometry after stained with fluorescein isothiocyanate-labeled annexin V/propidium iodide. Activation of caspase 8 was detected by Western blot analysis. RESULTS: Immunocytochemical assay indicated that DR5 was predominantly perinuclear in adherent cells but was mainly localized in cell membrane in detached cells. In addition, immunofluorescence assay also confirmed the above-mentioned results, and further demonstrated that DR5 was present in the form of coarse granules in detached cells, but in the form of fine granules in adherent cells. Cytometry analysis revealed higher levels of DR5 expression on the surfaces of brefeldin-A-untreated cells than on the surfaces of brefeldin-A-treated cells, but brefeldin A treatment did not affect the total DR5 expression levels. Moreover, nocodazole did not influence the extracelluar DR5 expression levels in EC9706 cells. Apoptosis assay revealed that detached cells were more sensitive to DR5 antibody-induced apoptosis than adherent cells. Western blotting showed that caspase 8 was activated in temporarily detached cells 4 h earlier than in adherent cells. CONCLUSION: Progress from adhesion to detachment of EC9706 cells causes DR5 relocalization, and promotes cytoplasmic translocation of DR5 to cell surfaces via a Golgi-dependent pathway. Moreover, it might also result in DR5 aggregation to render apoptosis of detached cells.


Subject(s)
Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Extracellular Matrix/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis , Brefeldin A/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Enzyme Activation , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/metabolism , Extracellular Matrix/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
8.
Cancer Detect Prev ; 26(2): 114-20, 2002.
Article in English | MEDLINE | ID: mdl-12102145

ABSTRACT

We have found that an increase in the ganglioside GM3 is a prerequisite for the induction of terminal differentiation, cuhninating in death by apoptosis, of human colonic carcinoma cells in vitro. To evaluate the therapeutic effect of increasing GM3 in human colonic carcinoma cells, we examined whether treated cells lose their tumorigenic activity and whether this approach is effective against cancer cells growing in vivo. Cells of the human colonic carcinoma cell line HCT 116 not only differentiated but also lost their tumorigenic activity by an artificial increase in GM3. When HCT 116 tumors growing in nude mice were treated with a drug that increases GM3, an appreciable increase in GM3 and induction of apoptosis were clearly observed. The growth of treated tumors was greatly suppressed. These results suggest that the modulation of ganglioside expression to introduce gangliosides with biological activity into cancer cells could be a novel effective approach for cancer therapy.


Subject(s)
Adenocarcinoma/drug therapy , Brefeldin A/therapeutic use , Colonic Neoplasms/drug therapy , G(M3) Ganglioside/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Cell Differentiation , Cell Division , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Tumor Cells, Cultured/drug effects
9.
Gastroenterology ; 121(5): 1101-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11677202

ABSTRACT

BACKGROUND & AIMS: Abetalipoproteinemia and Anderson's disease are hereditary lipid malabsorption syndromes. In abetalipoproteinemia, lipoprotein assembly is defective because of mutations in the microsomal triglyceride transfer protein. Here, we evaluated the intracellular transport of apolipoprotein B48 to localize the defect in Anderson's disease. METHODS: Asparagine-linked oligosaccharide processing of apolipoprotein B48 in normal and affected individuals was determined by the endoglycosidase H and F sensitivities of the protein after metabolic labeling of intestinal explants in organ culture. Cell ultrastructure was evaluated with electron microscopy. RESULTS: In Anderson's disease as in normal individuals, there was a time-dependent transformation of high mannose endoglycosidase H-sensitive oligosaccharides, of endoplasmic reticulum origin, to complex endoglycosidase H-resistant oligosaccharides, added in the Golgi network. In contrast, despite the translocation of apolipoprotein B48 into the endoplasmic reticulum in patients with abetalipoproteinemia and in biopsies treated with Brefeldin A, which blocks anterograde transport between the endoplasmic reticulum and the Golgi network, there was no transformation of endoglycosidase H-sensitive oligosaccharides. CONCLUSIONS: In abetalipoproteinemia and Anderson's disease, apolipoprotein B48 is completely translocated into the endoplasmic reticulum, but only in Anderson's disease is the protein transported to the Golgi apparatus. This suggests that Anderson's disease is caused by a post-Golgi cargo-specific secretion defect.


Subject(s)
Abetalipoproteinemia/metabolism , Apolipoproteins B/metabolism , Lipid Metabolism , Abetalipoproteinemia/pathology , Apolipoprotein B-48 , Apolipoproteins B/deficiency , Biological Transport , Brefeldin A/therapeutic use , Carrier Proteins/physiology , Endoplasmic Reticulum/metabolism , Glycosylation , Golgi Apparatus/metabolism , Humans , Intestinal Mucosa/pathology
10.
J Urol ; 164(3 Pt 1): 836-41, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10953164

ABSTRACT

PURPOSE: The objective of this study was to investigate growth-inhibitory and apoptotic activity of the experimental antitumor drug, brefeldin A (BFA), on primary cultures of human epithelial cells derived from prostatic adenocarcinomas. MATERIALS AND METHODS: Clonal assays were performed to evaluate the effects of BFA on growth of prostatic cancer cell strains. Loss of cell viability in response to BFA was assessed by trypan blue exclusion. Induction of apoptosis by BFA was evaluated by morphologic criteria, electrophoretic assay of DNA fragmentation, and a cell death ELISA. Immunoblots were used to monitor p53 and pRB expression in response to BFA. RESULTS: BFA was growth-inhibitory at a half-maximal concentration of 5 ng./ml. (18 nM). Morphological manifestations of apoptosis were evident by 24 hours of treatment. Cell viability declined and the cell death ELISA indicated an 18-fold increase in apoptosis in BFA-treated versus untreated cells at 48 hours. DNA fragmentation was also seen at 48 hours. Levels of p53 were not altered by BFA, but pRB was maintained in a hypophosphorylated state by BFA treatment. CONCLUSIONS: BFA is a potent inducer of apoptosis in prostatic cancer cells via a p53-independent mechanism. Cells derived from low-grade as well as high-grade cancers responded similarly to BFA. Since p53-mediated pathways of apoptosis may frequently be abrogated in prostatic cancer cells, agents such as BFA that induce p53-independent cell death may be promising candidates for chemotherapeutic agents.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Brefeldin A/therapeutic use , Genes, p53/genetics , Prostatic Neoplasms/drug therapy , Protein Synthesis Inhibitors/therapeutic use , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Apoptosis/genetics , Cell Death/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Clone Cells/drug effects , Coloring Agents , DNA Fragmentation , Electrophoresis, Agar Gel , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/drug effects , Humans , Immunoblotting , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Retinoblastoma Protein/drug effects , Trypan Blue , Tumor Cells, Cultured , Tumor Suppressor Protein p53/drug effects
11.
Cancer J Sci Am ; 2(1): 52-8, 1996.
Article in English | MEDLINE | ID: mdl-9166499

ABSTRACT

PURPOSE: An empiric in vitro screen of human tumor cell lines found brefeldin A inhibited the growth of immortalized human cell lines, with particular sensitivity to brefeldin in a series of immortalized melanoma cell lines and nonimmortalized prostate carcinoma explants. Brefeldin A alters the morphology and function of the Golgi apparatus, endosomal, and trans-Golgi compartments in different cell types. The studies presented here sought to obtain evidence of in vivo antitumor activity by brefeldin A. METHODS: Antiproliferative activity was studied in prostate carcinoma cells in vitro using cell counts, protein, and viable stains. Activity was also studied in vivo against subcutaneous and subrenal capsule melanoma models. RESULTS: Protracted exposures in vitro (between 24 and 72 hours) are necessary to cause persistent growth inhibition of immortalized PC3 prostate carcinoma cells. In human melanoma athymic mouse xenografts, brefeldin A showed antitumor activity in vivo when given 16 to 64 mg/kg/injection intraperitoneally q 7 h x 2, daily for 5 days. Activity was also observed in the intraperitoneal LOX IMVI (65%-100% increase in life span, with 17%-50% day 60 survivors); early-stage subcutaneous LOX IMVI and SK-MEL-5 (86%-100% growth inhibition), and subrenal capsule SK-MEL-5 and M19-MEL models. CONCLUSIONS: Brefeldin A possesses noteworthy antitumor activity in vivo and antiproliferative effects in vitro in certain cell types. Strategies to allow protracted exposure of tumor cells to brefeldin A while preserving a therapeutic index are needed to assess the clinical potential of brefeldin A.


Subject(s)
Antineoplastic Agents/therapeutic use , Brefeldin A/therapeutic use , Cell Proliferation/drug effects , Neoplasms, Experimental/drug therapy , Protein Synthesis Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , In Vitro Techniques , Male , Melanoma/drug therapy , Mice , Mice, Nude , Prostatic Neoplasms/drug therapy , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL