Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 96: 129498, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37804994

ABSTRACT

Neurodegenerative disease is a debilitating and incurable condition that affects millions of people around the world. The loss of functions or malfunctions of neural cells are the causes of mortality. A proteosome inhibitor, MG132, is well known to cause neurodegeneration in vitro when model neuronal-derived cell lines are exposed to it. Niclosamide, an anthelmintic drug, which has been used to treat tapeworm infections for more than 50 years, has recently attracted renewed attention in drug repurposing because it has been found to be a good candidate in many drug development screenings. We recently found that all markers of MG132-induced neuronal cell toxicity, including the accumulation of ubiquitinated proteins, were prevented by the presence of niclosamide. In addition, niclosamide was shown to enhance autophagy induced by MG132. There results suggested that niclosamide could act as a neuroprotective agent. In the present study, niclosamide derivatives were synthesized, and the structure-activity relationship (SAR) were determined with respect to protein ubiquitination induced by MG132 and effect on cell survival signaling pathways for neuroprotective function. Our results indicate that phenol OH plays a significant role in neuroprotective activity while the niclosamide derivatives without Cl (5- or 2'-Cl) showed almost the same neuroprotective effect. 4'-NO2 can be replaced by N3 or CF3 whereas NH2 significantly decreased activity. These findings provide guidance for the development of new niclosamide analogues against neurodegenerative diseases including Parkinson's disease.


Subject(s)
Neuroblastoma , Neurodegenerative Diseases , Neuroprotective Agents , Humans , Niclosamide/pharmacology , Neurodegenerative Diseases/drug therapy , Cell Line, Tumor , Neuroblastoma/drug therapy , Neuroprotective Agents/pharmacology , Structure-Activity Relationship , Apoptosis
2.
Oncotarget ; 13: 642-658, 2022.
Article in English | MEDLINE | ID: mdl-35548329

ABSTRACT

Glioblastoma multiforme (GBM) is one of the deadliest cancers of the brain. Its ability to infiltrate healthy brain tissues renders it difficult to remove surgically. Furthermore, it exhibits high rates of radio- and chemoresistance, making the survival rates of patients with GBM poor. Therefore, novel effective therapies for GBM remain urgently in demand. Niclosamide is an anti-helminthic drug and recently it has been receiving attention due to its reported anticancer effects in cancer models, including GBM. Furthermore, camptothecin (CPT) is a naturally-occurring alkaloid and has been previously reported to be a potential chemotherapeutic agent by targeting the nuclear topoisomerase I. In the present study, the possible combined chemotherapeutic effects of niclosamide and CPT on the human glioblastoma cell line U87 MG was investigated by MTT assay and western blot analysis. Niclosamide exhibited synergistic activities with CPT to suppress the proliferation of U87 MG cells. Additionally, niclosamide suppressed cell proliferation and induced cell death mainly by triggering ER stress and autophagy, whilst CPT induced cell apoptosis mainly through p53-mediated mitochondrial dysfunction and activation of the MAPK (ERK/JNK) pathways. Overall, these findings suggest that co-administration of niclosamide and CPT may provide a novel therapeutic treatment strategy for GBM.


Subject(s)
Antineoplastic Agents , Brain Neoplasms , Glioblastoma , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Niclosamide/pharmacology , Niclosamide/therapeutic use , Tumor Suppressor Protein p53/metabolism
3.
PLoS One ; 12(9): e0184324, 2017.
Article in English | MEDLINE | ID: mdl-28877265

ABSTRACT

Glioblastoma is the most common and lethal malignant primary brain tumor for which the development of efficacious chemotherapeutic agents remains an urgent need. The anti-helminthic drug niclosamide, which has long been in use to treat tapeworm infections, has recently attracted renewed interest due to its apparent anticancer effects in a variety of in vitro and in vivo cancer models. However, the mechanism(s) of action remains to be elucidated. In the present study, we found that niclosamide induced cell toxicity in human glioblastoma cells corresponding with increased protein ubiquitination, ER stress and autophagy. In addition, niclosamide treatment led to down-regulation of Wnt/ß-catenin, PI3K/AKT, MAPK/ERK, and STAT3 pro-survival signal transduction pathways to further reduce U-87 MG cell viability. Taken together, these results provide new insights into the glioblastoma suppressive capabilities of niclosamide, showing that niclosamide can target multiple major cell signaling pathways simultaneously to effectively promote cell death in U-87 MG cells. Niclosamide constitutes a new prospect for a therapeutic treatment against human glioblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Niclosamide/pharmacology , Ubiquitination , Anthelmintics/chemistry , Apoptosis , Brain Neoplasms/drug therapy , Cadaverine/analogs & derivatives , Cadaverine/chemistry , Cell Line, Tumor , Cell Proliferation , Cell Survival , Drug Screening Assays, Antitumor , Glioblastoma/drug therapy , Humans , STAT3 Transcription Factor/metabolism , Signal Transduction
4.
Parkinsons Dis ; 2016: 6564212, 2016.
Article in English | MEDLINE | ID: mdl-27774335

ABSTRACT

Ubiquitin proteasome system (UPS) dysfunction has been implicated in the development of many neuronal disorders, including Parkinson's disease (PD). Previous studies focused on individual neuroprotective agents and their respective abilities to prevent neurotoxicity following a variety of toxic insults. However, the effects of the antioxidant N-acetylcysteine (NAC) on proteasome impairment-induced apoptosis have not been well characterized in human neuronal cells. The aim of this study was to determine whether cotreatment of NAC and insulin-like growth factor-1 (IGF-1) efficiently protected against proteasome inhibitor-induced cytotoxicity in SH-SY5Y cells. Our results demonstrate that the proteasome inhibitor, MG132, initiates poly(ADP-ribose) polymerase (PARP) cleavage, caspase 3 activation, and nuclear condensation and fragmentation. In addition, MG132 treatment leads to endoplasmic reticulum (ER) stress and autophagy-mediated cell death. All of these events can be attenuated without obvious reduction of MG132 induced protein ubiquitination by first treating the cells with NAC and IGF-1 separately or simultaneously prior to exposure to MG132. Moreover, our data demonstrated that the combination of the two proved to be significantly more effective for neuronal protection. Therefore, we conclude that the simultaneous use of growth/neurotrophic factors and a free radical scavenger may increase overall protection against UPS dysfunction-mediated cytotoxicity and neurodegeneration.

5.
Virus Genes ; 52(3): 432-5, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26941234

ABSTRACT

The cold-active bacteriophage VMY22, belonging to the Podoviridae family, was isolated from Mingyong Glacier in China. Sequence analysis revealed that the genome is 18,609 bp long, with an overall G + C content of 36.4 mol%, and 25 open reading frames (ORFs). The sequence contains 46 potential promoters, 6 transcription terminators, and no tRNAs. Most of the ORFs show a high degree of similarity to B103 (NC_004165). Two noteworthy findings were made. First, one of the predicted proteins, ORF 19, shows high sequence similarity to the bacteriocin biosynthesis protein from Bacillus cereus. From this information, we propose that the VMY22 phage is at an intermediate phase in its coevolution with its bacterial host. Second, seven of the hypothetical proteins appear to be unique to this cold-active B. cereus phage (i.e., not found in temperate-active B. cereus phages). These observations add to our current knowledge about the coevolution of bacteriophages and their hosts. The identification of a novel group of gene and protein structures and functions will lead to a better understanding of cold-adaptation mechanisms in bacteria and their bacteriophages.


Subject(s)
Bacillus Phages/chemistry , Bacillus Phages/genetics , Bacillus cereus/virology , DNA, Viral/chemistry , DNA, Viral/genetics , Genome, Viral , Anti-Bacterial Agents/chemistry , Bacillus Phages/classification , Bacillus Phages/isolation & purification , Bacterial Proteins/chemistry , Base Composition , China , Chromosome Mapping , Evolution, Molecular , Open Reading Frames , Phylogeny , Promoter Regions, Genetic , Sequence Analysis, DNA , Transcription Termination, Genetic , Viral Proteins/chemistry
7.
Clin Cancer Res ; 19(7): 1681-92, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23554355

ABSTRACT

PURPOSE: G protein-coupled estrogen receptor 1 (GPER1), previously named GPR30, is a membrane receptor reported to mediate nongenomic estrogen responses. We investigated if GPER1 expression correlates with any clinicopathologic variables and distant disease-free survival (DDFS) in patients with breast cancer, if any prognostic impact of the receptor is dependent on estrogen receptor-α (ER-α) status, and if the receptor impacts apoptotic signaling in ER-positive breast cancer cells. EXPERIMENTAL DESIGN: GPER1 expression was analyzed by immunohistochemistry in breast tumors from 273 pre- and postmenopausal stage II patients, all treated with adjuvant tamoxifen for 2 years (cohort I) and from 208 premenopausal lymph node-negative patients, of which 87% were not subjected to any adjuvant systemic treatment (cohort II). GPER1-dependent proapoptotic signaling was analyzed in MCF7 cells with and without GPER1 knockdown, T47D cells, HEK293 cells (HEK), and HEK stably expressing GPER1 (HEK-R). RESULTS: GPER1 positively correlates with ER and progesterone receptor expression. Multivariate analysis showed that GPER1 is an independent prognostic marker of increased 10-year DDFS in the ER-positive subgroup. HEK-R has higher basal proapoptotic signaling compared with HEK including increased cytochrome C release, caspase-3 cleavage, PARP cleavage, and decreased cell viability. Treating HEK-R with the proteasome inhibitor epoxomicin, to decrease GPER1 degradation, further increases receptor-dependent proapoptotic signaling. Also, GPER1 knockdown decreases basal and agonist-stimulated proapoptotic receptor signaling in MCF7 cells. CONCLUSIONS: GPER1 is a prognostic indicator for increased DDFS in ER-positive breast cancer, which may be associated with constitutive GPER1-dependent proapoptotic signaling in ER-positive breast cancer cells.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , HEK293 Cells , Humans , Middle Aged , Neoplasm Staging , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics
8.
Neurochem Int ; 62(1): 31-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23142153

ABSTRACT

Inhibition of proteasome activity and the resulting protein accumulation are now known to be important events in the development of many neurological disorders, including Alzheimer's and Parkinson's diseases. Abnormal or over expressed proteins cause endoplasmic reticulum and oxidative stress leading to cell death, thus, normal proteasome function is critical for their removal. We have shown previously, with cultured SH-SY5Y neuroblastoma cells, that proteasome inhibition by the drug epoxomicin results in accumulation of ubiquitinated proteins. This causes obligatory loading of the mitochondria with calcium (Ca(2+)), resulting in mitochondrial damage and cytochrome c release, followed by programmed cell death (PCD). In the present study, we demonstrate that all-trans-retinoic acid (RA) pretreatment of SH-SY5Y cells protects them from PCD death after subsequent epoxomicin treatment which causes proteasome inhibition. Even though ubiquitinated protein aggregates are present, there is no evidence to suggest that autophagy is involved. We conclude that protection by RA is likely by mechanisms that interfere with cell stress-PCD pathway that otherwise would result from protein accumulation after proteasome inhibition. In addition, although RA activates both the AKT and ERK phosphorylation signaling pathways, only pretreatment with LY294002, an inhibitor of PI3-kinase in the AKT pathway, removed the protective effect of RA from the cells. This finding implies that RA activation of the AKT signaling cascade takes precedence over its activation of ERK1/2 phosphorylation, and that this selective effect of RA is key to its protection of epoxomicin-treated cells. Taken together, these findings suggest that RA treatment of cultured neuroblastoma cells sets up conditions under which proteasome inhibition, and the resultant accumulation of ubiquitinated proteins, loses its ability to kill the cells and may likely play a therapeutic role in neurodegenerative diseases.


Subject(s)
Cell Death/drug effects , Oncogene Protein v-akt/physiology , Proteasome Endopeptidase Complex/drug effects , Signal Transduction/drug effects , Tretinoin/pharmacology , Anti-Bacterial Agents/pharmacology , Blotting, Western , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Cell Death/genetics , Cell Line , Chromones/pharmacology , Data Interpretation, Statistical , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Humans , Membrane Potential, Mitochondrial/drug effects , Microscopy, Confocal , Morpholines/pharmacology , Oligopeptides/pharmacology , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proteasome Endopeptidase Complex/genetics
9.
Mol Cell Neurosci ; 47(3): 181-90, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21545837

ABSTRACT

The proteasome is an enzyme complex responsible for targeted intracellular proteolysis. Alterations in proteasome-mediated protein clearance have been implicated in the pathogenesis of aging, Alzheimer's disease (AD) and Parkinson's disease (PD). In such diseases, proteasome inhibition may contribute to formation of abnormal protein aggregates, which in turn activate intracellular unfolded protein responses that cause oxidative stress and apoptosis. In this study, we investigated the protective effect of Insulin-like Growth Factor-I (IGF-1) for neural SH-SY5Y cells treated with the proteasomal inhibitor, Epoxomicin. In SH-SY5Y cells, Epoxomicin treatment results in accumulation of intracellular ubiquitinated proteins and cytochrome c release from damaged mitochondria, leading to cell death, in Epoxomicin time- and dose-dependent manner. In cells treated with small amounts of IGF-1, the same dosages of Epoxomicin reduced both mitochondrial damage (cytochrome c release) and reduced caspase-3 activation and PARP cleavage, both of which are markers of apoptosis. Notably, however, IGF-1-treated SH-SY5Y cells still contained ubiquitinated protein aggregates. This result indicates that IGF-1 blocks the downstream apoptotic consequences of Epoxomicin treatment leading to decreased proteasome function. Clues as to the mechanism for this protective effect come from (a) increased AKT phosphorylation observed in IGF-1-protected cells, vs. cells exposed to Epoxomicin without IGF-1, and (b) reduction of IGF-1 protection by pretreatment of the cells with LY294002 (an inhibitor of PI3-kinase). Together these findings suggest that activation of PI3/AKT pathways by IGF-1 is involved in IGF-1 neuroprotection against apoptosis following proteasome inhibition.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Mitochondria/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Proteasome Inhibitors , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytoprotection/physiology , Dose-Response Relationship, Drug , Humans , Insulin-Like Growth Factor I/metabolism , Mitochondria/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
10.
J Biol Chem ; 278(44): 43146-56, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-12939279

ABSTRACT

Osteocytes embedded in the matrix of bone are thought to be mechanosensory cells that translate mechanical strain into biochemical signals that regulate bone modeling and remodeling. We have shown previously that fluid flow shear stress dramatically induces prostaglandin release and COX-2 mRNA expression in osteocyte-like MLO-Y4 cells, and that prostaglandin E2 (PGE2) released by these cells functions in an autocrine manner to regulate gap junction function and connexin 43 (Cx43) expression. Here we show that fluid flow regulates gap junctions through the PGE2 receptor EP2 activation of cAMP-dependent protein kinase A (PKA) signaling. The expression of the EP2 receptor, but not the subtypes EP1,EP3, and EP4, increased in response to fluid flow. Application of PGE2 or conditioned medium from fluid flow-treated cells to non-stressed MLO-Y4 cells increased expression of the EP2 receptor. The EP2 receptor antagonist, AH6809, suppressed the stimulatory effects of PGE2 and fluid flow-conditioned medium on the expression of the EP2 receptor, on Cx43 protein expression, and on gap junction-mediated intercellular coupling. In contrast, the EP2 receptor agonist butaprost, not the E1/E3 receptor agonist sulprostone, stimulated the expression of Cx43 and gap junction function. Fluid flow conditioned medium and PGE2 stimulated cAMP production and PKA activity suggesting that PGE2 released by mechanically stimulated cells is responsible for the activation of cAMP and PKA. The adenylate cyclase activators, forskolin and 8-bromo-cAMP, enhanced intercellular connectivity, the number of functional gap junctions, and Cx43 protein expression, whereas the PKA inhibitor, H89, inhibited the stimulatory effect of PGE2 on gap junctions. These studies suggest that the EP2 receptor mediates the effects of autocrine PGE2 on the osteocyte gap junction in response to fluid flow-induced shear stress. These data support the hypothesis that the EP2 receptor, cAMP, and PKA are critical components of the signaling cascade between mechanical strain and gap junction-mediated communication between osteocytes.


Subject(s)
Gap Junctions , Osteocytes/metabolism , Receptors, Prostaglandin E/physiology , Sulfonamides , Adenylyl Cyclases/metabolism , Animals , Blotting, Western , Cell Line , Cells, Cultured , Connexin 43/metabolism , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclooxygenase 2 , Dinoprostone/metabolism , Isoenzymes/metabolism , Isoquinolines/pharmacology , Models, Biological , Prostaglandin Antagonists/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP2 Subtype , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stress, Mechanical , Time Factors , Xanthones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...