Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Biol Pharm Bull ; 46(12): 1787-1796, 2023.
Article in English | MEDLINE | ID: mdl-38044097

ABSTRACT

Obesity is characterized by the excessive accumulation of fat to adipose tissue, which is related to abnormal increasing white adipose tissue (WAT) in the body, and it upregulates the risk of multiple diseases. Here, kuanoniamine C, which is a pyridoacridine alkaloid, suppressed the differentiation of pre-adipose cells into white adipocytes via the modulation of mitochondrial function, and inhibited WAT expansion in the early phase of high-fat-diet-induced obesity model. Pharmacological analysis revealed that inhibition of mitochondrial respiratory complex II, which new target of kuanoniamine C, activated reactive oxygen species (ROS)-extracellular signal-regulated kinase (ERK)-ß-catenin signaling, and this signaling was antagonized by insulin-, IBMX-, and dexamethasone-induced adipogenesis. Therefore, the kuanoniamine C might prevent abnormal WAT expansion even when eating a diet that is not calorie restricted.


Subject(s)
Adipogenesis , Obesity , Animals , Mice , Obesity/drug therapy , Obesity/metabolism , Adipose Tissue , Adipose Tissue, White/metabolism , Diet, High-Fat/adverse effects , Mitochondria , Mice, Inbred C57BL
2.
Anticancer Res ; 43(11): 4887-4895, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37909947

ABSTRACT

BACKGROUND/AIM: P53 is the most frequently mutated tumor suppressor gene among all cancers. In human cancers, specific residues of p53 are mutated at a high frequency, and those mutations are known as hotspot mutations. Mutant p53 promotes tumor progression through the gain-of-function (GOF) mechanism. However, its biological characteristics, especially its metastatic potential, owing to different hotspot mutations in gastric cancer remain unclear. In the present study, we investigated the p53-depended metastatic phenotype. MATERIALS AND METHODS: This study examined the differences in the metastatic potential of wild-type, mutant-p53-R175H, and mutant-p53-R273H NUGC-4 gastric cancer cells in vitro and in vivo. RESULTS: NUGC-4-mutant-p53-R175H cells showed significant cell proliferation, healing and invasive abilities in proliferation, wound healing and invasion assay, respectively, compared to wild-type and mutant-p53-R273H cells. Both NUGC-4-mutant-p53 cell types expressed epithelial-mesenchymal transition (EMT)-related proteins. Furthermore, NUGC-4-mutant-p53-R175H cells showed less attachment to the extracellular matrix and greater expression of EMT-related proteins than NUGC-4-mutant-p53-R273H cells. Regarding the peritoneal dissemination model, NUCG-4-mutant-p53-R175H and NUCG-4-mutant-p53-R273H cells demonstrated less frequent formation of dissemination nodules than NUGC-4-empty cells. In contrast, liver metastases were more frequent and greater in number in NUCG3-mutant-p53-R175H than in the other cell lines. CONCLUSION: Our results suggest that differences in the p53 status, even in the hotspot mutation site, affect not only the characteristics of the cells but also the metastatic ability of gastric cancer.


Subject(s)
Stomach Neoplasms , Humans , Medical Oncology , Phenotype , Stomach Neoplasms/genetics , Tumor Suppressor Protein p53/genetics
3.
Biochem Biophys Res Commun ; 678: 200-206, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37657239

ABSTRACT

Obesity increases the risk of various diseases, and many studies have examined prevention and treatment strategies. Browning of white adipocytes promotes triglyceride (TG) metabolism and is the new focus for treating obesity. This study investigated the role of malonate-a modulator of mitochondrial function-in adipocyte browning, and its potential as a therapeutic agent in obesity. Our findings revealed that malonate increased oxygen consumption without inhibiting ATP synthesis. Malonate induced expression of PRDM16-an important transcription factor for browning-and uncoupling protein 1 (beige adipocyte marker), suggesting that malonate induces browning in white adipocytes. In an obesity mouse model induced by a high-fat diet, malonate significantly reduced body weight and white adipose tissue weight, as well as improved insulin resistance. Importantly, malonate stimulated browning in white adipose tissue and maintained the mass of brown adipose tissue in the high-fat diet-induced obesity mouse model. We propose that manipulation of mitochondrial function by malonate is a promising therapeutic approach for obesity.


Subject(s)
Adipose Tissue, White , Diet, High-Fat , Animals , Mice , Diet, High-Fat/adverse effects , Adipocytes, White , Disease Models, Animal , Malonates/pharmacology , Obesity/etiology , Transcription Factors
4.
iScience ; 26(7): 107113, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37416477

ABSTRACT

Reactive oxygen species (ROS), such as superoxide, are crucial factors involved in the stimulation of cellular aging. Mitochondria, which are important organelles responsible for various metabolic processes in cells, produce ROS. These ROS impair mitochondrial function, thereby accelerating aging-related cellular dysfunction. Herein, we demonstrated that the Spirulina polysaccharide complex (SPC) restores mitochondrial function and collagen production by scavenging superoxide via the upregulation of superoxide dismutase 2 (SOD2) in aging fibroblasts. We observed that SOD2 expression was linked to inflammatory pathways; however, SPC did not upregulate the expression of most inflammatory cytokines produced as a result of induction of LPS in aging fibroblasts, indicating that SPC induces SOD2 without activation of inflammatory pathways. Furthermore, SPC stimulated endoplasmic reticulum (ER) protein folding by upregulating ER chaperones expression. Thus, SPC is proposed to be an antiaging material that rejuvenates aging fibroblasts by increasing their antioxidant potential via the upregulation of SOD2.

5.
Aging (Albany NY) ; 14(19): 7662-7691, 2022 09 26.
Article in English | MEDLINE | ID: mdl-36170016

ABSTRACT

Cell aging attenuates cellular functions, resulting in time-dependent disruption of cellular homeostasis, which maintains the functions of proteins and organelles. Mitochondria are important organelles responsible for cellular energy production and various metabolic processes, and their dysfunction is strongly related to the progression of cellular aging. Here we demonstrate that disruption of proteostasis attenuates mitochondrial function before the induction of DNA damage signaling by proliferative and replicative cellular aging. We found that lotus (Nelumbo nucifera Gaertn.) germ extract clears abnormal proteins and agglutinates via autophagy-mediated restoration of mitochondrial function and cellular aging phenotypes. Pharmacological analyses revealed that DAPK1 expression was suppressed in aging cells, and lotus germ extract upregulated DAPK1 expression by stimulating the acetylation of histones and then induced autophagy by activating the DAPK1-Beclin1 signaling pathway. Furthermore, treatment of aging fibroblasts with lotus germ extract stimulated collagen production and increased contractile ability in three-dimensional cell culture. Thus, time-dependent accumulation of abnormal proteins and agglutinates suppressed mitochondrial function in cells in the early stage of aging, and reactivation of mitochondrial function by restoring proteostasis rejuvenated aging cells. Lotus germ extract rejuvenates aging fibroblasts via the DAPK1-Beclin1 pathway-induced autophagy to clear abnormal proteins and agglutinates.


Subject(s)
Lotus , Proteostasis , Histones , Beclin-1 , Autophagy/physiology , Fibroblasts , Plant Extracts
6.
J Antibiot (Tokyo) ; 74(5): 359-362, 2021 05.
Article in English | MEDLINE | ID: mdl-33469193

ABSTRACT

In our screening program for new biologically active compounds, a new polyene macrolide, lavencidin (1), along with known compound RKGS-A2215A (2), was isolated from the fermentation broth of Streptomyces lavendulae FRI-5 by changing the composition of liquid medium normally used for the strain. Their structures were elucidated by spectral methods (high-resolution fast-atom bombardment mass spectrometry (HRFABMS) and nuclear magnetic resonance (NMR)). Compound 1 includes a conjugated pentaene moiety together with six hydroxy groups and a carboxylic acid as a side chain. Lavencidin (1) showed moderate growth-inhibitory activity against yeast and was cytotoxic against human cancer cell lines with low-micromolar IC50 values.


Subject(s)
Antifungal Agents/pharmacology , Macrolides/chemistry , Macrolides/pharmacology , Streptomyces/metabolism , Yeasts/drug effects , Antifungal Agents/chemistry , Cell Line, Tumor , Humans , Macrolides/metabolism , Magnetic Resonance Spectroscopy
7.
Biochem Biophys Res Commun ; 527(1): 289-296, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446382

ABSTRACT

Osteosarcoma is the most frequent and intractable malignancy of the bone in children and young adults. Surgical operation requires extensive excision of the cancer tissue and neighboring normal tissues. In addition, anticancer drugs and radiation therapy are thought to be almost ineffective. Glucose-regulated protein 78 (GRP78), a cell-protective endoplasmic reticulum (ER) chaperone protein, is one of the most promising anticancer targets for osteosarcoma. Here, by analyzing the molecular mechanisms of kuanoniamine C, we report that kuanoniamine C suppresses GRP78 expression via GRP78 mRNA degradation in an ER stress response-independent manner. Interestingly, kuanoniamine C-induced cell death and downregulation of GRP78 expression was regulated by p53 signaling. Moreover, co-treatment with bortezomib, which is a newly identified anticancer drug for osteosarcoma, and kuanoniamine C suppressed GRP78 protein expression, which is essential for the stimulation of bortezomib-induced cell death. These results suggest that co-treatment with bortezomib and kuanoniamine C is a novel therapeutic strategy for the treatment of osteosarcoma that enhances bortezomib-dependent cell death by the downregulation of GRP78, and this combination selectively targets the major cell population of osteosarcoma, which expresses wild-type p53.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bortezomib/pharmacology , Heat-Shock Proteins/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Osteosarcoma/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Endoplasmic Reticulum Chaperone BiP , HeLa Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Molecular Structure , Osteosarcoma/metabolism , Osteosarcoma/pathology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Structure-Activity Relationship
8.
Cells ; 8(11)2019 10 30.
Article in English | MEDLINE | ID: mdl-31671609

ABSTRACT

Cancer cells modulate their metabolism to proliferate and survive under the metabolic stress condition, which is known as endoplasmic reticulum (ER) stress. Therefore, cancer cells should suppress ER stress-mediated cell death and induce autophagy-which recycles metabolites to provide energy and new macromolecules. In this study, we demonstrate that the ER membrane protein BAP31 acts to suppress adaptation to ER stress conditions, induce cell death, and suppress autophagy by forming a BAP31-STX17 protein complex. The loss of BAP31 stimulates tumor growth in metabolic stress conditions in vivo and enhances invasion activity. Therefore, BAP31 stimulates cell death and inhibits autophagy, and it can be considered a novel tumor suppressor factor that acts by preventing ER stress adaptation.


Subject(s)
Autophagy , Bone Neoplasms/pathology , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Osteosarcoma/pathology , Qa-SNARE Proteins/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Apoptosis , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Osteosarcoma/genetics , Osteosarcoma/metabolism , Qa-SNARE Proteins/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Sci Adv ; 5(6): eaaw1386, 2019 06.
Article in English | MEDLINE | ID: mdl-31206022

ABSTRACT

The endoplasmic reticulum (ER) is composed of large membrane-bound compartments, and its membrane subdomain appears to be in close contact with mitochondria via ER-mitochondria contact sites. Here, I demonstrate that the ER membrane protein, BAP31, acts as a key factor in mitochondrial homeostasis to stimulate the constitution of the mitochondrial complex I by forming an ER-mitochondria bridging protein complex. Within this complex, BAP31 interacts with mitochondria-localized proteins, including Tom40, to stimulate the translocation of NDUFS4, the component of complex I from the cytosol to the mitochondria. Disruption of the BAP31-Tom40 complex inhibits mitochondrial complex I activity and oxygen consumption by the decreased NDUFS4 localization to the mitochondria. Thus, the BAP31-Tom40 ER-mitochondria bridging complex mediates the regulation of mitochondrial function and plays a role as a previously unidentified stress sensor, representing a mechanism for the establishment of ER-mitochondria communication via contact sites between these organelles.


Subject(s)
Cytosol/metabolism , Electron Transport Complex I/genetics , Endoplasmic Reticulum/metabolism , Membrane Proteins/genetics , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Animals , Cell Line , Cell Line, Tumor , Electron Transport Complex I/metabolism , Endoplasmic Reticulum/ultrastructure , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Membrane Proteins/metabolism , Mice , Mitochondria/ultrastructure , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Osteoblasts/cytology , Osteoblasts/metabolism , Protein Binding , Protein Transport , Signal Transduction
10.
J Ind Microbiol Biotechnol ; 46(5): 739-750, 2019 May.
Article in English | MEDLINE | ID: mdl-30788639

ABSTRACT

ß-Carboline alkaloids exhibit a broad spectrum of pharmacological and biological activities and are widely distributed in nature. Genetic information on the biosynthetic mechanism of ß-carboline alkaloids has not been accumulated in bacteria, because there are only a few reports on the microbial ß-carboline compounds. We previously isolated kitasetaline, a mercapturic acid derivative of a ß-carboline compound, from the genetically modified Kitasatospora setae strain and found a plausible biosynthetic gene cluster for kitasetaline. Here, we identified and characterized three kitasetaline (ksl) biosynthetic genes for the formation of the ß-carboline core structure and a gene encoding mycothiol-S-conjugate amidase for the modification of the N-acetylcysteine moiety by using heterologous expression. The proposed model of kitasetaline biosynthesis shows unique enzymatic systems for ß-carboline alkaloids. In addition, feeding fluorotryptophan to the heterologous Streptomyces hosts expressing the ksl genes led to the generation of unnatural ß-carboline alkaloids exerting novel/potentiated bioactivities.


Subject(s)
Alkaloids/chemistry , Carbolines/chemistry , Fluorine/chemistry , Streptomyces/metabolism , Acetylcysteine , Amidohydrolases/metabolism , Cell Line, Tumor , Humans , Multigene Family , Mutation , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Tryptophan/chemistry
11.
J Antibiot (Tokyo) ; 71(10): 854-861, 2018 10.
Article in English | MEDLINE | ID: mdl-29973681

ABSTRACT

ß-Carboline alkaloids and related compounds show a broad spectrum of biological activities. We previously identified new members of the ß-carboline alkaloid family by using an engineered Kitasatospora setae strain and a heterologous Streptomyces host expressing the plausible biosynthetic genes, including the hypothetical gene kse_70640 (kslB). Here, we elucidated the chemical structure of a new tetrahydro-ß-carboline compound (named kitasetalic acid) that appeared in a heterologous Streptomyces host expressing the kslB gene alone. Kitasetalic acid suppressed the expression of glucose-regulated protein 78 (GRP78) without inducing cell death. This is the first report to show that a tetrahydro-ß-carboline compound regulates the expression of the GRP78 protein in cancer cell lines.


Subject(s)
Carbolines/metabolism , Genetic Engineering , Glucose/pharmacology , Streptomycetaceae/metabolism , Carbolines/chemistry , Carbolines/pharmacology , Cell Line , Endoplasmic Reticulum Chaperone BiP , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Molecular Structure , Streptomycetaceae/genetics
12.
Int J Oncol ; 53(2): 761-770, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29845212

ABSTRACT

The tumor suppressor gene p53 encodes a transcription factor that regulates various cellular functions, including DNA repair, apoptosis and cell cycle progression. Approximately half of all human cancers carry mutations in p53 that lead to loss of tumor suppressor function or gain of functions that promote the cancer phenotype. Thus, targeting mutant p53 as an anticancer therapy has attracted considerable attention. In the current study, a small-molecule screen identified andrographlide (ANDRO) as a mutant p53 suppressor. The effects of ANDRO, a small molecule isolated from the Chinese herb Andrographis paniculata, on tumor cells carrying wild-type or mutant p53 were examined. ANDRO suppressed expression of mutant p53, induced expression of the cyclin-dependent kinase inhibitor p21 and pro-apoptotic proteins genes, and inhibited the growth of cancer cells harboring mutant p53. ANDRO also induced expression of the heat-shock protein (Hsp70) and increased binding between Hsp70 and mutant p53 protein, thus promoting proteasomal degradation of p53. These results provide novel insights into the mechanisms regulating the function of mutant p53 and suggest that activation of Hsp70 may be a new strategy for the treatment of cancers harboring mutant p53.


Subject(s)
Diterpenes/administration & dosage , HSP70 Heat-Shock Proteins/genetics , Mutation , Neoplasms, Experimental/drug therapy , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Andrographis/chemistry , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Diterpenes/pharmacology , HCT116 Cells , HSP70 Heat-Shock Proteins/metabolism , Humans , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Plant Extracts/chemistry , Proteolysis , Xenograft Model Antitumor Assays
13.
Biochem Biophys Res Commun ; 492(1): 33-40, 2017 10 07.
Article in English | MEDLINE | ID: mdl-28811106

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most difficult cancers to treat owing to the lack of effective chemotherapeutic methods. Sorafenib, the first-line and only available treatment for HCC, extends patient overall survival by several months, with a response rate below 10%. Thus, the identification of an agent that enhances the anticancer effect of sorafenib is critical for the development of therapeutic options for HCC. Endoplasmic reticulum (ER) stress response is one of the methods of sorafenib-induced cell death. Here we report that questiomycin A suppresses expression of GRP78, a cell-protective ER chaperone protein. Analysis of the molecular mechanisms of questiomycin A revealed that this compound stimulated GRP78 protein degradation in an ER stress response-independent manner. Cotreatment with sorafenib and questiomycin A suppressed GRP78 protein expression, which is essential for the stimulation of sorafenib-induced cell death. Moreover, our in vivo study demonstrated that the coadministration of sorafenib and questiomycin A suppressed tumor formation in HCC-induced xenograft models. These results suggest that cotreatment with sorafenib and questiomycin A is a novel therapeutic strategy for HCC by enhancing sorafenib-dependent ER stress-induced cell death, and downregulation of GRP78 is a new target for the stimulation of the therapeutic effects of sorafenib in HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death/drug effects , Heat-Shock Proteins/antagonists & inhibitors , Niacinamide/analogs & derivatives , Oxazines/pharmacology , Phenylurea Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Niacinamide/chemistry , Niacinamide/pharmacology , Oxazines/chemistry , Phenylurea Compounds/chemistry , Sorafenib , Structure-Activity Relationship
16.
Chem Biol ; 22(9): 1206-16, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26320861

ABSTRACT

TP53 is the most frequently mutated gene in human cancer, and small-molecule reactivation of mutant p53 function represents an important anticancer strategy. A cell-based, high-throughput small-molecule screen identified chetomin (CTM) as a mutant p53 R175H reactivator. CTM enabled p53 to transactivate target genes, restored MDM2 negative regulation, and selectively inhibited the growth of cancer cells harboring mutant p53 R175H in vitro and in vivo. We found that CTM binds to Hsp40 and increases the binding capacity of Hsp40 to the p53 R175H mutant protein, causing a potential conformational change to a wild-type-like p53. Thus, CTM acts as a specific reactivator of the p53 R175H mutant form through Hsp40. These results provide new insights into the mechanism of reactivation of this specific p53 mutant.


Subject(s)
Antineoplastic Agents/pharmacology , Disulfides/pharmacology , HSP40 Heat-Shock Proteins/metabolism , Indole Alkaloids/pharmacology , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Disulfides/chemistry , Drug Screening Assays, Antitumor , HCT116 Cells , High-Throughput Screening Assays , Humans , Indole Alkaloids/chemistry , Mice , Mice, Nude , Mutation , Small Molecule Libraries/chemistry , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
17.
Oncotarget ; 6(24): 19990-20001, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26254280

ABSTRACT

Altered regulation of ER stress response has been implicated in a variety of human diseases, such as cancer and metabolic diseases. Excessive ER function contributes to malignant phenotypes, such as chemoresistance and metastasis. Here we report that the tumor suppressor p53 regulates ER function in response to stress. We found that loss of p53 function activates the IRE1α/XBP1 pathway to enhance protein folding and secretion through upregulation of IRE1α and subsequent activation of its target XBP1. We also show that wild-type p53 interacts with synoviolin (SYVN1)/HRD1/DER3, a transmembrane E3 ubiquitin ligase localized to ER during ER stress and removes unfolded proteins by reversing transport to the cytosol from the ER, and its interaction stimulates IRE1α degradation. Moreover, IRE1α inhibitor suppressed protein secretion, induced cell death in p53-deficient cells, and strongly suppressed the formation of tumors by p53-deficient human tumor cells in vivo compared with those that expressed wild-type p53. Therefore, our data imply that the IRE1α/XBP1 pathway serves as a target for therapy of chemoresistant tumors that express mutant p53.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/physiology , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/deficiency , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , HCT116 Cells , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Regulatory Factor X Transcription Factors , Signal Transduction , Sulfonamides/pharmacology , Thiophenes/pharmacology , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , X-Box Binding Protein 1
18.
Mar Drugs ; 13(4): 2376-89, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25894488

ABSTRACT

Avarol is a sesquiterpenoid hydroquinone with potent cytotoxicity. Although resolving endoplasmic reticulum (ER) stress is essential for intracellular homeostasis, erratic or excessive ER stress can lead to apoptosis. Here, we reported that avarol selectively induces cell death in pancreatic ductal adenocarcinomas (PDAC), which are difficult to treat owing to the availability of few chemotherapeutic agents. Analyses of the molecular mechanisms of avarol-induced apoptosis indicated upregulation of ER stress marker BiP and ER stress-dependent apoptosis inducer CHOP in PDAC cells but not in normal cells, suggesting that avarol selectively induces ER stress responses. We also showed that avarol activated the PERK-eIF2α pathway but did not affect the IRE1 and ATF6 pathways. Moreover, CHOP downregulation was significantly suppressed by avarol-induced apoptosis. Thus, the PERK-eIF2α-CHOP signaling pathway may be a novel molecular mechanism of avarol-induced apoptosis. The present data indicate that avarol has potential as a chemotherapeutic agent for PDAC and induces apoptosis by activating the PERK-eIF2α pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , Transcription Factor CHOP/agonists , eIF-2 Kinase/metabolism , Animals , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Biomarkers/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cell Line , Cell Line, Tumor , Dysidea/chemistry , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/agonists , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/agonists , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Inhibitory Concentration 50 , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , RNA Interference , Sesquiterpenes/adverse effects , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Up-Regulation/drug effects , eIF-2 Kinase/chemistry
19.
Reprod Sci ; 22(10): 1272-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25736325

ABSTRACT

We screened a library of 528 approved drugs to identify candidate compounds with therapeutic potential as preeclampsia treatments via their proangiogenic properties. Using human umbilical vein endothelial cells (HUVECs), we assessed whether the screened drugs induced placental growth factor (PIGF) and restored damaged endothelial cell function. Enzyme-linked immunosorbent assays (ELISAs) were carried out to measure levels of PlGF in conditioned media treated with each drug (100 µmol/L) in the drug library. Tube formation assays were performed using HUVECs to evaluate the angiogenic effects of drugs that induced PlGF. We also performed ELISA, quantitative reverse transcription polymerase chain reaction, and tube formation assays after treatment with a range of concentrations of the candidate drug. Of the drugs that induced PlGF, vardenafil was the only compound that significantly facilitated tube formation in comparison with the control cells (P < .01). Treatment with vardenafil at concentrations of 50, 100, and 250 µmol/L increased expression of PlGF in a dose-dependent manner. Vardenafil (250 µmol/L) significantly improved tube formation which was inhibited in the presence of soluble fms-like tyrosine kinase 1 (100 ng/mL) and/or soluble endoglin (100 ng/mL). Production of PlGF from HUVECs in the presence of sera derived from patients with preeclampsia was significantly elevated by administration of vardenafil (250 µmol/L). By assessing drug repositioning through screening a library of approved drugs, we identified vardenafil as a potential protective agent against preeclampsia. The therapeutic mechanism of vardenafil may involve inhibition of the systemic maternal antiangiogenic state that leads to preeclampsia, in addition to its vasodilating effect. As concentrations used are high and unlikely to be useful clinically, further work is needed before testing it in humans.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Drug Repositioning , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Pre-Eclampsia/drug therapy , Pregnancy Proteins/metabolism , Vardenafil Dihydrochloride/pharmacology , Case-Control Studies , Cells, Cultured , Dose-Response Relationship, Drug , Female , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Placenta Growth Factor , Pre-Eclampsia/blood , Pre-Eclampsia/enzymology , Pre-Eclampsia/physiopathology , Pregnancy , Pregnancy Proteins/genetics , Up-Regulation , Vascular Endothelial Growth Factor Receptor-1/metabolism
20.
Cell Rep ; 5(2): 331-9, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24139803

ABSTRACT

Resolved endoplasmic reticulum (ER) stress response is essential for intracellular homeostatic balance, but unsettled ER stress can lead to apoptosis. Here, we show that a proapoptotic p53 target, CDIP1, acts as a key signal transducer of ER-stress-mediated apoptosis. We identify B-cell-receptor-associated protein 31 (BAP31) as an interacting partner of CDIP1. Upon ER stress, CDIP1 is induced and enhances an association with BAP31 at the ER membrane. We also show that CDIP1 binding to BAP31 is required for BAP31 cleavage upon ER stress and for BAP31-Bcl-2 association. The recruitment of Bcl-2 to the BAP31-CDIP1 complex, as well as CDIP1-dependent truncated Bid (tBid) and caspase-8 activation, contributes to BAX oligomerization. Genetic knockout of CDIP1 in mice leads to impaired response to ER-stress-mediated apoptosis. Altogether, our data demonstrate that the CDIP1/BAP31-mediated regulation of mitochondrial apoptosis pathway represents a mechanism for establishing an ER-mitochondrial crosstalk for ER-stress-mediated apoptosis signaling.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 8/metabolism , Cell Line , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/chemistry , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...