Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(12): e0295273, 2023.
Article in English | MEDLINE | ID: mdl-38039297

ABSTRACT

We previously reported that macrolide antibiotics, such as clarithromycin (CAM), blocked autophagy flux, and simultaneous proteasome and autophagy inhibition by bortezomib (BTZ) plus CAM resulted in enhanced apoptosis induction in multiple myeloma (MM) cells via increased endoplasmic reticulum (ER) stress loading. However, in actual therapeutic settings, cell adhesion-mediated drug resistance between bone marrow stromal cells (BMSC) and MM cells has been known to be a barrier to treatment. To investigate whether CAM could enhance BTZ-induced cytotoxicity in MM cells under direct cell adhesion with BMSC, we established a co-culture system of EGFP-labeled MM cells with BMSC. The cytotoxic effect of BTZ on MM cells was diminished by its interaction with BMSC; however, the attenuated cytotoxicity was recovered by the co-administration of CAM, which upregulates ER stress loading and NOXA expression. Knockout of NOXA in MM cells canceled the enhanced cell death by CAM, indicating that NOXA is a key molecule for cell death induction by the co-administration of CAM. Since NOXA is degraded by autophagy as well as proteasomes, blocking autophagy with CAM resulted in the sustained upregulation of NOXA in MM cells co-cultured with BMSC in the presence of BTZ. Our data suggest that BMSC-associated BTZ resistance is mediated by the attenuation of ER stress loading. However, the addition of CAM overcomes BMSC-associated resistance via upregulation of NOXA by concomitantly blocking autophagy-mediated NOXA degradation and transcriptional activation of NOXA by ER stress loading.


Subject(s)
Clarithromycin , Multiple Myeloma , Humans , Clarithromycin/pharmacology , Clarithromycin/therapeutic use , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Multiple Myeloma/drug therapy , Cell Line, Tumor , Bortezomib/pharmacology , Bortezomib/therapeutic use , Proteasome Endopeptidase Complex , Autophagy , Stromal Cells , Apoptosis
2.
Oncol Lett ; 22(3): 680, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34345305

ABSTRACT

Following surgery and chemoradiation, ~50% of patients with locally advanced head and neck tumors experience relapse within the first two years, with a poor prognosis. Therefore, a novel therapeutic approach is required. The aim of the present study was to investigate the effect of combination treatment with the proteasome inhibitor bortezomib (BTZ), and ricolinostat (RCS), a specific inhibitor of histone deacetylase 6 (HDAC6), on CAL27 and Detroit562 head and neck cancer cells. BTZ and RCS exhibited cytotoxicity in a dose- and time-dependent manner. Simultaneous treatment with BTZ and RCS resulted in the synergistic enhancement of non-apoptotic cell death and autophagy. The receptor-interacting serine/threonine-protein kinase 1 (RIPK1) inhibitor, necrostatin, but not the autophagy inhibitor, 3-methyladenine, attenuated the cytotoxicity of combined BTZ and RCS treatment. Thus, necroptosis [type-III programmed cell death (PCD)], but not autophagic cell death (type-II PCD), appeared to contribute to the pronounced cytotoxicity. However, no phosphorylation of RIPK1 or mixed lineage kinase domain-like protein was detectable in response to BTZ or RCS. Furthermore, RCS induced α-tubulin acetylation and inhibited BTZ-induced aggresome formation along with endoplasmic reticulum stress loading. Combined treatment with BTZ and RCS enhanced the production of reactive oxygen species (ROS). The ROS scavenger, N-acetyl cysteine, abrogated the increase in cytotoxicity. These results suggest the potential therapeutic value of the dual targeting of the proteasome and HDCA6 for head and neck cancers through the induction of necroptosis-like cell death along with ROS generation.

3.
Cancer Sci ; 112(8): 3324-3337, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34051014

ABSTRACT

Cancer cells use autophagy for growth, survival, and cytoprotection from chemotherapy. Therefore, autophagy inhibitors appear to be good candidates for cancer treatment. Our group previously reported that macrolide antibiotics, especially azithromycin (AZM), have potent autophagy inhibitory effects, and combination treatment with tyrosine kinase inhibitors or proteasome inhibitors enhances their anti-cancer activity. In this study, we evaluated the effect of combination therapy with DNA-damaging drugs and AZM in non-small-cell lung cancer (NSCLC) cells. We found that the cytotoxic activities of DNA-damaging drugs, such as doxorubicin (DOX), etoposide, and carboplatin, were enhanced in the presence of AZM in NSCLC cell lines, whereas AZM alone exhibited almost no cytotoxicity. This enhanced cell death was dependent on wild-type-p53 status and autophagosome-forming ability because TP53 knockout (KO) and ATG5-KO cells attenuated AZM-enhanced cytotoxicity. DOX treatment upregulated lysosomal biogenesis by activating TFEB and led to lysosomal membrane damage as assessed by galectin 3 puncta assay and cytoplasmic leakage of lysosomal enzymes. In contrast, AZM treatment blocked autophagy, which resulted in the accumulation of lysosomes/autolysosomes. Thus, the effects of DOX and AZM were integrated into the marked increase in damaged lysosomes/autolysosomes, leading to prominent lysosomal membrane permeabilization (LMP) for apoptosis induction. Our data suggest that concomitant treatment with DNA-damaging drugs and AZM is a promising strategy for NSCLC treatment via pronounced LMP induction.


Subject(s)
Azithromycin/pharmacology , Carboplatin/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Lysosomes/metabolism , Topoisomerase II Inhibitors/pharmacology , A549 Cells , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage , Drug Synergism , Humans , Lung Neoplasms/drug therapy , Lysosomes/drug effects
4.
Tokai J Exp Clin Med ; 46(1): 7-16, 2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33835469

ABSTRACT

OBJECTIVE: The effects of estrogen on cells are mediated by the estrogen receptor α (ERα) which localizes at the peri-membrane, cytoplasm, and the nucleus of cells. Therefore, we intended to investigate how cytonuclear ERα plays its roles in different cellular activities. METHODS: We used amino acid substituted ERα that localized at the cytoplasm and nucleus but has no direct DNA-binding activities. ERα-negative endometrial carcinoma cells (ERα-) were stably transfected with plasmid of human ERα carrying a substituted phenylalanine at position 445 with alanine (ERα-F445A). Treated with 17ß-estrogen (E2) or bazedoxifene (BDF), cell proliferation, migration, and expression of kinases related to ERα signal transduction pathways were observed. RESULTS: E2 (40 nM) significantly activated proliferation in ERα-F445A cells, but not in ERα- cells. Similarly, E2 significantly activated cell migration in ERα-F445A cells, rather than that in ERα- cells. While no obvious change in the amount of the non-phosphorylated mammalian target of Rapamycin (mTOR), the expression of mTOR phosphorylated at serine 2448 decreased, which was recovered in presence of 17ß-estrogen (E2) in the ERα-F445A cells. On the other hand, the expression of focal adhesion kinase (FAK) phosphorylated at tyrosine at 297 was attenuated in the ERα-F445A cells treated with E2. CONCLUSION: It is suggested that the cytonuclear ERα-F445A induces phosphorylation of kinases in downstream pathways, which regulate cell proliferation and migration.


Subject(s)
Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Endometrial Neoplasms/genetics , Estradiol/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Phosphorylation , TOR Serine-Threonine Kinases/metabolism
5.
Int J Oncol ; 57(6): 1280-1292, 2020 12.
Article in English | MEDLINE | ID: mdl-33173988

ABSTRACT

The proton pump inhibitor lansoprazole (LPZ) inhibits the growth of several cancer cell lines, including A549 and CAL 27. We previously reported that macrolide antibiotics such as azithromycin (AZM) and clarithromycin (CAM) potently inhibit autophagic flux and that combining AZM or CAM with the epidermal growth factor receptor inhibitors enhanced their antitumor effect against various cancer cells. In the present study, we conducted the combination treatment with LPZ and macrolide antibiotics against A549 and CAL 27 cells and evaluated cytotoxicity and morphological changes using cell proliferation and viability assays, flow cytometric analysis, immunoblotting, and morphological assessment. Combination therapy with LPZ and AZM greatly enhanced LPZ­induced cell death, whereas treatment with AZM alone exhibited negligible cytotoxicity. The observed cytotoxic effect was not mediated through apoptosis or necroptosis. Transmission electron microscopy of A549 cells treated with the LPZ + AZM combination revealed morphological changes associated with necrosis and accumulated autolysosomes with undigested contents. Furthermore, the A549 cell line with ATG5 knockout exhibited complete inhibition of autophagosome formation, which did not affect LPZ + AZM treatment­induced cytotoxicity, thus excluding the involvement of autophagy­dependent cell death in LPZ + AZM treatment­induced cell death. A549 cells treated with LPZ + AZM combination therapy retained the endosomal Alexa­dextran for extended duration as compared to untreated control cells, thus indicating impairment of lysosomal digestion. Notably, lysosomal galectin­3 puncta expression induced due to lysosomal membrane permeabilization was increased in cells treated with LPZ + AZM combination as compared to the treatment by either agent alone. Collectively, the present results revealed AZM­induced autolysosome accumulation, potentiated LPZ­mediated necrosis, and lysosomal membrane permeabilization, thus suggesting the potential clinical application of LPZ + AZM combination therapy for cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azithromycin/pharmacology , Lansoprazole/pharmacology , Lysosomes/drug effects , Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Azithromycin/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Gene Knockout Techniques , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/ultrastructure , Lansoprazole/therapeutic use , Lysosomes/pathology , Lysosomes/ultrastructure , Microscopy, Electron, Transmission , Neoplasms/pathology , Permeability/drug effects
6.
Cancer Sci ; 111(6): 2132-2145, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32304130

ABSTRACT

In the cell cycle, the G1 /S transition is controlled by the cyclin-dependent kinase (CDK) 4/6-cyclin D complex. Constitutive activation of CDK4/6 dysregulates G1 /S transition, leading to oncogenic transformation. We found that 3 CDK4/6 inhibitors, abemaciclib, ribociclib, and palbociclib, exerted a cytocidal effect as well as a cytostatic effect at the G1 phase in cancer cell lines, including A549 human non-small cell lung cancer cells. Among these inhibitors, abemaciclib exhibited the most potent cytotoxic effect. The cell-death phenotype induced by abemaciclib, which entailed formation of multiple cytoplasmic vacuoles, was not consistent with apoptosis or necroptosis. Abemaciclib blocked autophagic flux, resulting in accumulation of autophagosomes, however vacuole formation and cell death induced by abemaciclib were independent of autophagy. In addition, methuosis, a cell-death phenotype characterized by vacuole formation induced by excessive macropinocytosis, was excluded because the vacuoles did not incorporate fluorescent dextran. Of note, both formation of vacuoles and induction of cell death in response to abemaciclib were inhibited by vacuolar-type ATPase (V-ATPase) inhibitors such as bafilomycin A1 and concanamycin A. Live-cell imaging revealed that the abemaciclib-induced vacuoles were derived from lysosomes that expanded following acidification. Transmission electron microscopy revealed that these vacuoles contained undigested debris and remnants of organelles. Cycloheximide chase assay revealed that lysosomal turnover was blocked by abemaciclib. Furthermore, mTORC1 inhibition along with partial lysosomal membrane permeabilization occurred after abemaciclib treatment. Together, these results indicate that, in cancer cells, abemaciclib induces a unique form of cell death accompanied by swollen and dysfunctional lysosomes.


Subject(s)
Aminopyridines/pharmacology , Benzimidazoles/pharmacology , Cell Death/drug effects , Cell Proliferation/drug effects , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Lysosomes/drug effects , Vacuoles/drug effects
7.
Biochem Biophys Rep ; 22: 100750, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32195376

ABSTRACT

Tyrosine kinase inhibitors (TKIs) induce autophagy in many types of cancer cells. We previously reported that gefitinib (GEF) and imatinib (IMA) induce autophagy in epidermal growth factor receptor (EGFR) knock-out A549 and non-BCR-ABL-expressing leukemia cell lines, respectively. This evidence suggests that TKI-induced autophagy is independent of the original target molecules. The present study compared the autophagy-inducing abilities of various TKIs, regardless of their targets, by quantitative autophagy flux assay. We established stable clones expressing the GFP-LC3-mCherry-LC3ΔG plasmid in A549, PC-9, and CAL 27 cell lines and assessed autophagy inducibility by monitoring the fluorescent ratios of GFP-LC3 to mCherry-LC3ΔG using an IncuCyte live cell imaging system during exposure to TKIs viz; GEF, osimertinib (OSI), lapatinib (LAP), lenvatinib (LEN), sorafenib (SOR), IMA, dasatinib (DAS), and tivantinib (TIV). Among these TKIs, DAS, GEF, and SOR exhibited prominent autophagy induction in A549 and PC-9 cells. In CAL 27 cells, IMA, SOR, and LEN, but not GEF, TIV, or OSI, exhibited autophagy induction. In the presence of azithromycin (AZM), which showed an inhibitory effect on autophagy flux, TKIs with prominent autophagy inducibility exhibited enhanced cytotoxicity via non-apoptotic cell death relative to effects of TKI alone. Therefore, autophagy inducibility of TKIs differed in the context of cancer cells. However, once induced, they appeared to have cytoprotective functions. Thus, blocking TKI-induced autophagy with AZM may improve the therapeutic effect of TKIs in cancer cells.

8.
Breast Cancer ; 27(2): 225-235, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31625014

ABSTRACT

BACKGROUND: Vitamin K2 (VK2) has been reported to induce apoptosis in many types of cancer cells including leukemia. However, there are no precise reports regarding the breast cancer cells. From the stand point of clinical implications of VK2 including chemoprevention, we investigated the effects of VK2 on breast cancer cell lines. METHODS: Breast cancer cell lines were cultured with VK2, and the cytotoxicity and cell death phenotype were examined. The HL-60 leukemia cells were used as a control for VK2-induced apoptosis. RESULTS: VK2 exhibited the cytotoxic effect, especially in triple negative breast cancer cell lines, namely, MDA-MB-231 and MDA-MB-468. However, in contrast to HL-60 cells, typical features of the cells undergoing apoptosis, such as chromatin condensation, nuclear fragments, and cleavage of caspase-3 were not detected. Transmission electron microscopy exhibited an increased number of autophagosomes/autolysosomes with plasma membrane integrity. An autophagy inhibitor, 3-methyladenine, apparently attenuated VK2-induced cytotoxicity, which indicated the involvement of autophagy-dependent cell death. Interestingly, both VK2-induced non-apoptotic cell death in MDA-MB-231 cells and VK2-induced apoptosis in HL-60 cells were suppressed in the presence of reactive oxygen species (ROS) scavengers. Therefore, ROS production by VK2 seems to be located up-stream in the molecular machinery for both the types of cell death execution. CONCLUSION: The VK2 induced non-apoptotic cell death along with autophagy, in triple negative breast cancer cell lines. Cell death phenotype induced by VK2 appears to differ among the type of cancers. This suggests the possibility of using VK2 for the breast cancer therapy.


Subject(s)
Autophagosomes/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Vitamin K 2/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Cell Death/drug effects , Cell Line, Tumor , Female , HL-60 Cells , Humans , MCF-7 Cells , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/metabolism
9.
Oncol Rep ; 42(1): 231-242, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31059070

ABSTRACT

Epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase and mutations in this gene are major drivers of lung cancer development. EGFR tyrosine kinase inhibitors (TKIs) are standard first­line therapies for patients with advanced non­small cell lung cancer (NSCLC) with activating EGFR mutations, but are not effective in patients with wild­type EGFR. In the present study, the cytotoxic effects of various TKIs against EGFR were investigated in wild­type NSCLC cells as single treatments or in combination with Fingolimod (FTY720), which has been approved for treating multiple sclerosis and has cytotoxic effects against several tumor cell lines. It was found that the combined treatment with TKIs lapatinib (Lap) or sorafenib (Sor) and FTY720 synergistically suppressed the viability of the NSCLC cell lines A549 and H596. Additionally, FTY720 inhibited lysosomal acidification and suppressed autophagy flux. Immunoblotting and reverse transcription­quantitative polymerase chain reaction showed that FTY720 combined with Lap or Sor, enhanced endoplasmic reticulum (ER) stress loading and cell cycle arrest in A549 cells. The enhancement of ER stress loading and cell cycle arrest induced by combined treatment with Lap or Sor and FTY720, which was associated with the cytotoxicity induced by the combination of these drugs. These findings suggested that FTY720 improved TKI therapy in NSCLC patients with wild­type EGFR, by sensitizing NSCLC cells to TKIs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle/drug effects , Drug Resistance, Neoplasm/drug effects , Fingolimod Hydrochloride/pharmacology , Lung Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , A549 Cells , Autophagy-Related Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Repositioning , Drug Synergism , ErbB Receptors/genetics , Humans , Lapatinib/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Sorafenib/pharmacology
10.
Int J Oncol ; 52(4): 1165-1177, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29484439

ABSTRACT

The maintenance of the intracellular level of amino acids is crucial for cellular homeostasis. This is carried out via the regulation of both the influx from the extracellular environment and the recycling of intracellular resources. Since epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors, including gefitinib (GEF) have been reported to induce the apoptosis of several cancer cell lines, in the present study, we examined whether the cytotoxic effects of GEF are further enhanced under amino acid starvation (AAS) culture conditions. Under AAS culture conditions, the cell killing effect of GEF was synergistically pronounced in the EGFR-expressing cell lines, namely, CAL 27, Detroit 562, A549 and PANC-1 cells compared with those treated with either GEF or AAS alone. The addition of essential amino acids, but not non-essential amino acids to the cell culture medium resulted in the cancellation of this pronounced cytotoxicity. The knockdown of L-type amino acid transporter 1 (LAT-1) by siRNA also enhanced GEF-induced cytotoxicity. Therefore, the shortage of the intracellular amino acid pool appears to determine the sensitivity to GEF. Notably, this enhanced cytotoxicity is not mediated by the induction of apoptosis, but is accompanied by the pronounced induction of autophagy. The presence of necrostatin-1, an inhibitor of receptor-interacting serine/threonine-protein kinase 1 (RIPK­1), but not that of Z-VAD-fmk, attenuated the cytotoxic effects of GEF under AAS culture conditions. Electron microscopy demonstrated that the CAL 27 cells treated with GEF under AAS culture conditions exhibited swelling of the cytosol and organelles with an increased number of autophagosomes and autolysosomes, but without chromatin condensation and nuclear fragmentation. Autophagic cell death was excluded as the inhibition of autophagy did not attenuate the cytotoxicity. These results strongly suggest the induction of necroptosis in response to GEF under AAS culture conditions. However, we could not detect any phosphorylation of RIPK-1 and mixed lineage kinase domain like pseudokinase (MLKL), as well as any necrosome formation. Therefore, the enhanced cytotoxic effect of GEF under AAS culture conditions is thought to be mediated by atypical necroptosis.


Subject(s)
Amino Acids/metabolism , Antineoplastic Agents/pharmacology , Cell Culture Techniques/methods , Cell Death/physiology , Quinazolines/pharmacology , Cell Death/drug effects , Cell Line, Tumor , ErbB Receptors/metabolism , Gefitinib , Humans
11.
Leuk Lymphoma ; 59(6): 1439-1450, 2018 06.
Article in English | MEDLINE | ID: mdl-28918692

ABSTRACT

The D-type cyclin (CCND)-cyclin-dependent kinase 4/6 (CDK4/6) complex has been implicated in multiple myeloma development. We investigated the biological activity of CDK4/6 inhibitor abemaciclib on cell growth and survival in three myeloma cell lines, KMS-12-PE, RPMI 8226, and IM-9. Abemaciclib inhibited myeloma cell growth in a dose-dependent manner in all cell lines, with significant differences seen at a concentration of 320 nM. Treatment with 1 µM abemaciclib increased the fraction of cells in the G0/G1 phase and decreased the fraction in the S-G2/M phases. Further, treatment with abemaciclib at a concentration of 3.2 µM or more showed apparent cytocidal activity accompanied with cytoplasmic vacuolization against myeloma cells. Importantly, abemaciclib induced autophagy in a dose-dependent manner in all three cell lines. These results indicate that the CCND-CDK4/6 complex is closely tied to myeloma cell growth and survival.


Subject(s)
Aminopyridines/pharmacology , Autophagy/drug effects , Benzimidazoles/pharmacology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Multiple Myeloma/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Protein Kinase Inhibitors/pharmacology
12.
Biochem Biophys Rep ; 11: 174-181, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28955782

ABSTRACT

In this study, we showed that the dual addition of glucosyl ceramide synthase and ceramidase inhibitors to A549 cell culture led to the possibility of ceramide channel formation via endogenous palmitoyl-ceramide accumulation with an increase in cholesterol contents in the lysosome membrane as an initial step prior to initiation of necrotic cell death. In addition, the dual addition led to black circular structures of 10-20 nm, interpreted as stain-filled cylindrical channels on transmission electron microscopy. The formation of palmitoyl-ceramide channels in the lysosome membrane causes the liberation of cathepsin B from lysosomes for necrotic cell death. On the other hand, necrotic cell death in the dual addition was not caused by oxidative stress or cathepsin B activity, and the cell death was free from the contribution of the translation of Bax protein to the lysosome membrane.

13.
PLoS One ; 11(12): e0164529, 2016.
Article in English | MEDLINE | ID: mdl-27977675

ABSTRACT

Autophagy, a self-digestive system for cytoplasmic components, is required to maintain the amino acid pool for cellular homeostasis. We previously reported that the macrolide antibiotics azithromycin (AZM) and clarithromycin (CAM) have an inhibitory effect on autophagy flux, and they potently enhance the cytocidal effect of various anticancer reagents in vitro. This suggests that macrolide antibiotics can be used as an adjuvant for cancer chemotherapy. Since cancer cells require a larger metabolic demand than normal cells because of their exuberant growth, upregulated autophagy in tumor cells has now become the target for cancer therapy. In the present study, we examined whether macrolides exhibit cytotoxic effect under an amino acid-starving condition in head and neck squamous cancer cell lines such as CAL 27 and Detroit 562 as models of solid tumors with an upregulated autophagy in the central region owing to hypovascularity. AZM and CAM induced cell death under the amino acid-depleted (AAD) culture condition in these cell lines along with CHOP upregulation, although they showed no cytotoxicity under the complete culture medium. CHOP knockdown by siRNA in the CAL 27 cells significantly suppressed macrolide-induced cell death under the AAD culture condition. CHOP-/- murine embryonic fibroblast (MEF) cell lines also attenuated AZM-induced cell death compared with CHOP+/+ MEF cell lines. Using a tet-off atg5 MEF cell line, knockout of atg5, an essential gene for autophagy, also induced cell death and CHOP in the AAD culture medium but not in the complete culture medium. This suggest that macrolide-induced cell death via CHOP induction is dependent on autophagy inhibition. The cytotoxicity of macrolide with CHOP induction was completely cancelled by the addition of amino acids in the culture medium, indicating that the cytotoxicity is due to the insufficient amino acid pool. These data suggest the possibility of using macrolides for "tumor-starving therapy".


Subject(s)
Anti-Bacterial Agents/pharmacology , Autophagy/drug effects , Azithromycin/pharmacology , Carcinoma, Squamous Cell/pathology , Cell Death/drug effects , Clarithromycin/pharmacology , Head and Neck Neoplasms/pathology , Macrolides/pharmacology , Animals , Cell Line, Tumor , Humans , Mice
14.
Sci Rep ; 6: 35599, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27752123

ABSTRACT

There are various autoimmunogenic antigens (AIs) in testicular germ cells (TGCs) recognized as foreign by the body's immune system. However, there is little information of TGC-specific AIs being available. The aim of this study is to identify TGC-specific AIs. We have previously established that immunization using viable syngeneic TGC can also induce murine experimental autoimmune orchitis (EAO) without using any adjuvant. This study is to identify TGC-specific AIs by TGC liquid chromatography-tandem mass spectrometry analysis, followed by two-dimensional gel electrophoresis that reacted with serum IgG from EAO mice. In this study, we identified 11 TGC-specific AIs that reacted with serum from EAO mice. Real-time RT-PCR analysis showed that the mRNA expressions of seven TGC-specific AIs were significantly higher in only mature testis compared to other organs. Moreover, the recombinant proteins of identified 10 (except unnamed protein) TGC-specific AIs were created by using human embryonic kidney 293 (HEK293) cells and these antigencities were reconfirmed by Western blot using EAO serum reaction. These results indicated Atp6v1a, Hsc70t, Fbp1 and Dazap1 were candidates for TGC-specific AIs. Identification of these AIs will facilitate new approaches for understanding infertility and cancer pathogenesis and may provide a basis for the development of novel therapies.


Subject(s)
Autoantigens/blood , Germ Cells/cytology , Immunization , Testis/cytology , Animals , Autoantibodies/blood , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Epididymis/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Immunoglobulin G/metabolism , Male , Mice , Orchitis/blood , Orchitis/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Spermatozoa/metabolism
15.
Int J Oncol ; 49(5): 1848-1858, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27601063

ABSTRACT

The ubiquitin-proteasome and autophagy-lysosome pathways are two major self-digestive systems for cellular proteins. Ubiquitinated misfolded proteins are degraded mostly by proteasome. However, when ubiquitinated proteins accumulate beyond the capacity of proteasome clearance, they are transported to the microtubule-organizing center (MTOC) along the microtubules to form aggresomes, and subsequently some of them are degraded by the autophagy-lysosome system. We previously reported that macrolide antibiotics such as azithromycin and clarithromycin block autophagy flux, and that concomitant treatment with the proteasome inhibitor bortezomib (BZ) and macrolide enhances endoplasmic reticulum (ER) stress-mediated apoptosis in breast cancer cells. As ubiquitinated proteins are concentrated at the aggresome upon proteasome failure, we focused on the microtubule as the scaffold of this transport pathway for aggresome formation. Treatment of metastatic breast cancer cell lines (e.g., MDA-MB­231 cells) with BZ resulted in induction of aggresomes, which immunocytochemistry detected as a distinctive eyeball-shaped vimentin-positive inclusion body that formed in a perinuclear lesion, and that electron microscopy detected as a sphere of fibrous structure with some dense amorphous deposit. Vinorelbine (VNR), which inhibits microtubule polymerization, more effectively suppressed BZ-induced aggresome formation than paclitaxel (PTX), which stabilizes microtubules. Combined treatment using BZ and VNR, but not PTX, enhanced the cytotoxic effect and apoptosis induction along with pronounced ER stress loading such as upregulation of GRP78 and CHOP/GADD153. The addition of azithromycin to block autophagy flux in the BZ plus VNR-containing cell culture further enhanced the cytotoxicity. These data suggest that suppression of BZ-induced aggresome formation using an inhibitory drug such as VNR for microtubule polymerization is a novel strategy for metastatic breast cancer therapy.


Subject(s)
Apoptosis/drug effects , Bortezomib/pharmacology , Breast Neoplasms/pathology , Endoplasmic Reticulum Stress/drug effects , Proteasome Endopeptidase Complex/drug effects , Vinblastine/analogs & derivatives , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Tumor Cells, Cultured , Vinblastine/pharmacology , Vinorelbine
16.
Int J Oncol ; 48(1): 45-54, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26718641

ABSTRACT

Pancreatic cancer is one of the most difficult types of cancer to treat because of its high mortality rate due to chemotherapy resistance. We previously reported that combined treatment with gefitinib (GEF) and clarithromycin (CAM) results in enhanced cytotoxicity of GEF along with endoplasmic reticulum (ER) stress loading in non-small cell lung cancer cell lines. An epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) such as GEF induces autophagy in a pro-survival role, whereas CAM inhibits autophagy flux in various cell lines. Pronounced GEF-induced cytotoxicity therefore appears to depend on the efficacy of autophagy inhibition. In the present study, we compared the effect on autophagy inhibition among such macrolides as CAM, azithromycin (AZM), and EM900, a novel 12-membered non-antibiotic macrolide. We then assessed the enhanced GEF-induced cytotoxic effect on pancreatic cancer cell lines BxPC-3 and PANC-1. Autophagy flux analysis indicated that AZM is the most effective autophagy inhibitor of the three macrolides. CAM exhibits an inhibitory effect but less than AZM and EM900. Notably, the enhancing effect of GEF-induced cytotoxicity by combining macrolides correlated well with their efficient autophagy inhibition. However, this pronounced cytotoxicity was not due to upregulation of apoptosis induction, but was at least partially mediated through necroptosis. Our data suggest the possibility of using macrolides as 'chemosensitizers' for EGFR-TKI therapy in pancreatic cancer patients to enhance non-apoptotic tumor cell death induction.


Subject(s)
Autophagy/drug effects , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Apoptosis/drug effects , Autophagy/genetics , Azithromycin/administration & dosage , Cell Line, Tumor , Clarithromycin/administration & dosage , ErbB Receptors/genetics , Erythromycin/administration & dosage , Erythromycin/analogs & derivatives , Gefitinib , Gene Expression Regulation, Neoplastic/drug effects , Humans , Macrolides/administration & dosage , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Quinazolines/administration & dosage
17.
J Reprod Dev ; 62(1): 51-8, 2016.
Article in English | MEDLINE | ID: mdl-26498204

ABSTRACT

We previously established trophoblast stem cells from mouse androgenetic embryos (AGTS cells). In this study, to further characterize AGTS cells, we compared cell proliferation activity between trophoblast stem (TS) cells and AGTS cells under fibroblast growth factor 4 (FGF4) signaling. TS cells continued to proliferate and maintained mitotic cell division in the presence of FGF4. After FGF4 deprivation, the cell proliferation stopped, the rate of M-phase cells decreased, and trophoblast giant cells formed. In contrast, some of AGTS cells continued to proliferate, and the rate of M-phase cells did not decrease after FGF4 deprivation, although the other cells differentiated into giant cells. RO3306, an ATP competitor that selectively inhibits CDK1, inhibited the cell proliferation of both TS and AGTS cells. Under RO3306 treatment, cell death was induced in AGTS cells but not in TS cells. These results indicate that RO3306 caused TS cells to shift mitotic cell division to endoreduplication but that some of AGTS cells did not shift to endoreduplication and induced cell death. In conclusion, the paternal genome facilitated the proliferation of trophoblast cells without FGF4 signaling.


Subject(s)
Androgens/metabolism , Fibroblast Growth Factor 4/genetics , Fibroblast Growth Factor 4/metabolism , Gene Expression Regulation, Developmental , Stem Cells/cytology , Trophoblasts/cytology , Animals , Cell Death , Cell Proliferation , Cell Survival , Female , Genome , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mitosis , Quinolines/chemistry , Signal Transduction , Thiazoles/chemistry
18.
Biochem Biophys Res Commun ; 461(1): 28-34, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25858318

ABSTRACT

Gefitinib (GEF), an inhibitor for EGFR tyrosine kinase, potently induces autophagy in non-small cell lung cancer (NSCLC) cell lines such as PC-9 cells expressing constitutively activated EGFR kinase by EGFR gene mutation as well as A549 and H226 cells with wild-type EGFR. Unexpectedly, GEF-induced autophagy was also observed in non-NSCLC cells such as murine embryonic fibroblasts (MEF) and leukemia cell lines K562 and HL-60 without EGFR expression. Knockout of EGFR gene in A549 cells by CRISPR/Cas9 system still exhibited autophagy induction after treatment with GEF, indicating that the autophagy induction by GEF is not mediated through inhibiting EGFR kinase activity. Combined treatment with GEF and clarithromycin (CAM), a macrolide antibiotic having the effect of inhibiting autophagy flux, enhances the cytotoxic effect in NSCLC cell lines, although treatment with CAM alone exhibits no cytotoxicity. GEF treatment induced up-regulation of endoplasmic reticulum (ER)-stress related genes such as CHOP/GADD153 and GRP78. Knockdown of CHOP in PC-9 cells and Chop-knockout MEF both exhibited less sensitivity to GEF than controls. Addition of CAM in culture medium resulted in further pronounced GEF-induced ER stress loading, while CAM alone exhibited no effect. These data suggest that GEF-induced autophagy functions as cytoprotective and indicates the potential therapeutic possibility of using CAM for GEF therapy. Furthermore, it is suggested that the intracellular signaling for autophagy initiation in response to GEF can be completely dissociated from EGFR, but unknown target molecule(s) of GEF for autophagy induction might exist.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Clarithromycin/administration & dosage , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Gefitinib , Humans , Lung Neoplasms/pathology , Quinazolines/administration & dosage , Treatment Outcome
19.
Int J Oncol ; 46(2): 474-86, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25422130

ABSTRACT

The inhibitory effects of macrolide antibiotics including clarithromycin (CAM) on autophagy flux have been reported. Although a macrolide antibiotic exhibits no cytotoxicity, its combination with bortezomib (BZ), a proteasome inhibitor, for the simultaneous blocking of the ubiquitin (Ub)­proteasome and autophagy­lysosome pathways leads to enhanced multiple myeloma (MM) cell apoptosis induction via stress overloading of the endoplasmic reticulum (ER). As misfolded protein cargo is recruited by histone deacetylase 6 (HDAC6) to dynein motors for aggresome transport, serving to sequester misfolded proteins, we further investigated the cellular effects of targeting proteolytic pathways and aggresome formation concomitantly in MM cells. Pronounced apoptosis was induced by the combination of vorinostat [suberoylanilide hydroxamic acid (SAHA); potently inhibits HDAC6] with CAM and BZ compared with each reagent or a 2­reagent combination. CAM/BZ treatment induced vimentin positive­aggresome formation along with the accumulation of autolysosomes in the perinuclear region, whereas they were inhibited in the presence of SAHA. The SAHA/CAM/BZ combination treatment maximally upregulated genes related to ER stress including C/EBP homologous protein (CHOP). Similarly to MM cell lines, enhanced cytotoxicity with CHOP upregulation following SAHA/CAM/BZ treatment was shown by a wild­type murine embryonic fibroblast (MEF) cell line; however, a CHOP­deficient MEF cell line almost completely canceled this pronounced cytotoxicity. Knockdown of HDAC6 with siRNA exhibited further enhanced CAM/BZ­induced cytotoxicity and CHOP induction along with the cancellation of aggresome formation. Targeting the integrated networks of aggresome, proteasome, and autophagy is suggested to induce efficient ER stress­mediated apoptosis in MM cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Multiple Myeloma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols , Boronic Acids/administration & dosage , Bortezomib , Cell Line, Tumor , Clarithromycin/administration & dosage , Drug Synergism , Endoplasmic Reticulum/drug effects , Histone Deacetylase 6 , Histone Deacetylases/drug effects , Humans , Hydroxamic Acids/administration & dosage , Mice , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Proteasome Endopeptidase Complex/drug effects , Pyrazines/administration & dosage , Vorinostat
20.
Int J Oncol ; 43(5): 1456-66, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24008433

ABSTRACT

2-Aminophenoxazine-3-one (Phx-3) induces apoptosis in several types of cancer cell lines. However, the mechanism of apoptosis induction by Phx-3 has not been fully elucidated. In this study, we investigated the anticancer effects of Phx-3 in the glioblastoma cell line LN229 and analyzed its molecular mechanism. The results indicated that 6- and 20-h treatment with Phx-3 significantly induced apoptosis in LN229 cells, with downregulation of survivin and XIAP. Both ERK and JNK, which are the members of the MAPK family, were activated after treatment with Phx-3. Inhibition of ERK using the specific inhibitor U0126 blocked the Phx-3-induced apoptosis only in part. However, inhibition of JNK using the specific inhibitor SP600125 completely prevented Phx-3-induced apoptosis and restored the phosphorylation states of ERK to the control levels. Enhanced generation of reactive oxygen species (ROS) was detected after 3-h treatment with Phx-3. In addition, the ROS scavenger melatonin almost completely blocked Phx-3-induced JNK activation and apoptosis. This suggests that JNK activation was mediated by Phx-3-induced ROS generation. Although SP600125 and melatonin completely blocked the reduction of mitochondrial membrane potential after a 3-h treatment with Phx-3, extension of Phx-3 exposure time to 20 h resulted in no cancelation of mitochondrial depolarization by these reagents. These reagents also had little effect on the decreased expression of survivin and XIAP during a 3-20-h exposure to Phx-3. These results indicate that the production of ROS following JNK activation is the main axis of Phx-3-induced apoptosis in LN229 cells for short-term exposure to Phx-3, whereas alternative mechanism(s) appear to be involved in apoptosis induction during long-term exposure to Phx-3.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/pathology , Glioblastoma/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Potential, Mitochondrial/drug effects , Oxazines/pharmacology , Reactive Oxygen Species/metabolism , Aromatase Inhibitors/pharmacology , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Mitochondria/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...