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











Publication year range
1.
Gan To Kagaku Ryoho ; 51(4): 368-377, 2024 Apr.
Article in Japanese | MEDLINE | ID: mdl-38644299

ABSTRACT

Companion diagnostics(CDx)are in vitro diagnostic products that are used to predict the efficacy and adverse effects of therapeutic drugs prior to administration, and are co-developed and co-approved with the therapeutic drugs in principle. In Japan, 40 CDx products have been approved by January 2024, and 39 products are used to determine if therapeutic drugs are applicable for cancer treatment. In the CDx products for cancer treatment, PCR, immunohistochemistry, or in situ hybridization is used to clarify the mutations(point mutations, insertions/deletions, fusions, etc.)in cancer-related genes or the expression levels of cancer-related molecules in the cancer tissues. The results of the analysis determine whether a particular therapeutic drug could be used or not for the treatment of the corresponding patient. Recently, several next-generation sequencing(NGS)-based CDx products have been approved and utilized for cancer treatment. The rise of NGS-based diagnostics has made it possible to comprehensively analyze mutations in many cancer-related genes in a single test and to determine whether each of several therapeutic drugs is applicable to the patient at once. On the other hand, with the increase in the number of CDx products, several regulatory issues have arisen, including an issue related to the co-development of CDx and a therapeutic drug and an issue related to the interchangeable use of CDx products that detect the same mutations of the cancer-related genes. The revision of CDx-related guidance is being considered in Japan and overseas in response to this situation.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , Neoplasms/diagnosis , Japan , High-Throughput Nucleotide Sequencing , Mutation
2.
ACS Med Chem Lett ; 13(12): 1885-1891, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36518702

ABSTRACT

Internal tandem duplication (ITD) in the gene encoding FMS-like tyrosine kinase 3 (FLT3) (FLT3-ITD) is the most frequently observed mutation in acute myeloid leukemia (AML). Currently approved FLT3 kinase inhibitors have high efficacy, but drug resistance caused by reactivation of FLT3 kinase activity is often clinically observed. In this study, we developed novel FLT3 degraders by introducing gilteritinib, an FDA-approved FLT3 inhibitor, into targeted protein degradation technology. The most active compound, CRBN(FLT3)-8, potently degraded FLT3-ITD via the ubiquitin-proteasome system and inhibited the proliferation of FLT3-ITD mutant AML cells more effectively than gilteritinib. These findings provide a new lead compound for degradation-based drugs targeting FLT3-ITD-positive cancers.

3.
Cancer Sci ; 113(8): 2828-2838, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35579105

ABSTRACT

BRAF mutations are frequently observed in melanoma and hairy-cell leukemia. Currently approved rapidly accelerated fibrosarcoma (RAF) kinase inhibitors targeting oncogenic BRAF V600 mutations have shown remarkable efficacy in the clinic, but their therapeutic benefits are occasionally hampered by acquired resistance due to RAF dimerization-dependent reactivation of the downstream MAPK pathway, which is known as paradoxical activation. There is also a concern that paradoxical activation of the MAPK pathway may trigger secondary cancer progression. In this study, we developed chimeric compounds, proteolysis targeting chimeras (PROTACs), that target BRAFV600E protein for degradation. CRBN(BRAF)-24, the most effective chimera, potently degraded BRAFV600E in a ubiquitin-proteasome system (UPS)-dependent manner and inhibited the proliferation of BRAFV600E -driven cancer cells. In BRAF wild-type cells, CRBN(BRAF)-24 induced neither BRAFWT degradation nor paradoxical activation of the MAPK pathway. Biochemical analysis revealed that CRBN(BRAF)-24 showed more potent and sustained suppression of MAPK signaling than a BRAFV600E inhibitor, PLX-8394, in BRAFV600E -driven cancer cells. Targeted degradation of BRAFV600E by CRBN(BRAF)-24 could be a promising strategy to evade paradoxical activation of the RAF-MAPK pathway.


Subject(s)
Melanoma , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Cell Line, Tumor , Humans , MAP Kinase Signaling System , Melanoma/drug therapy , Melanoma/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism
4.
Methods Mol Biol ; 2365: 331-347, 2021.
Article in English | MEDLINE | ID: mdl-34432253

ABSTRACT

Inducing degradation of undruggable target proteins by the use of chimeric small molecules, represented by proteolysis-targeting chimeras, is a promising strategy for drug development. We developed a series of chimeric molecules, termed "specific and nongenetic inhibitor of apoptosis protein (IAP)-dependent protein erasers" (SNIPERs) that recruit IAP ubiquitin ligases to induce degradation of target proteins. SNIPERs also induce degradation of some IAPs, including cIAP1 and XIAP, which are antiapoptotic proteins that are overexpressed in many cancers. Such protein degraders have unique properties that could be especially useful in cancer therapy. This chapter describes (1) the design and synthesis of SNIPER compounds, (2) the methods used for the detection of target protein degradation and ubiquitylation, and (3) the protocol to evaluate the antitumor activity of SNIPER.


Subject(s)
Neoplasms , Humans , Ligands , Neoplasms/drug therapy , Proteolysis , Ubiquitination
5.
PLoS One ; 15(3): e0229712, 2020.
Article in English | MEDLINE | ID: mdl-32130260

ABSTRACT

In non-small cell lung cancer (NSCLC), oncogenic driver mutations including those in KRAS and EGFR are typically mutually exclusive. However, recent reports indicate that multiple driver mutations are found in a certain percentage of cancers, and that the therapeutic responses of such cases with co-mutations of driver genes are largely unclear. Here, using CRISPR-Cas9-mediated genome editing, we generated isogenic cell lines harboring one or two copies of an EGFR-activating mutation from the human NSCLC cell line A549, which is known to harbor a homozygous KRAS gene mutation. In comparison with parent cells with KRAS mutation alone, cells with concomitant EGFR mutation exhibited higher sensitivity to EGFR-tyrosine kinase inhibitors (TKIs) but not to conventional anti-cancer drugs. In particular, cells with two copies of EGFR mutation were markedly more sensitive to EGFR-TKIs compared with parent cells. Thus, the presence of concomitant EGFR mutation can affect the TKI response of KRAS-mutated cells, implying that EGFR-TKI may represent an effective treatment option against NSCLC with EGFR/KRAS co-mutation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , A549 Cells , Base Sequence , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Cell Proliferation/drug effects , Clone Cells , ErbB Receptors/genetics , Gene Dosage , Gene Editing , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphorylation/drug effects
6.
Genes Environ ; 42: 8, 2020.
Article in English | MEDLINE | ID: mdl-32071619

ABSTRACT

BACKGROUND: Next Generation Sequencer (NGS) is a powerful tool for a high-throughput sequencing of human genome. It is important to ensure reliability and sensitivity of the sequence data for a clinical use of the NGS. Various cancer-related gene panels such as Oncomine™ or NCC OncoPanel have been developed and used for clinical studies. Because these panels contain multiple genes, it is difficult to ensure the performance of mutation detection for every gene. In addition, various platforms of NGS are developed and their cross-platform validation has become necessity. In order to create mutant standards in a defined background, we have used CRISPR/Cas9 genome-editing system in HEK 293 T/17 cells. RESULTS: Cancer-related genes that are frequently used in NGS-based cancer panels were selected as the target genes. Target mutations were selected based on their frequency reported in database, and clinical significance and on the applicability of CRISPR/Cas9 by considering distance from PAM site, and off-targets. We have successfully generated 88 hetero- and homozygous mutant cell lines at the targeted sites of 36 genes representing a total of 125 mutations. CONCLUSIONS: These knock-in HEK293T/17 cells can be used as the reference mutant standards with a steady and continuous supply for NGS-based cancer panel tests from the JCRB cell bank. In addition, these cell lines can provide a tool for the functional analysis of targeted mutations in cancer-related genes in the isogenic background.

7.
JCI Insight ; 4(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31672935

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) relies on hyperactivated protein synthesis. Consistently, human and mouse PDAC lose expression of the translational repressor and mTOR target 4E-BP1. Using genome-wide polysome profiling, we here explore mRNAs whose translational efficiencies depend on the mTOR/4E-BP1 axis in pancreatic cancer cells. We identified a functional enrichment for mRNAs encoding DNA replication and repair proteins, including RRM2 and CDC6. Consequently, 4E-BP1 depletion favors DNA repair and renders DNA replication insensitive to mTOR inhibitors, in correlation with a sustained protein expression of CDC6 and RRM2, which is inversely correlated with 4E-BP1 expression in PDAC patient samples. DNA damage and pancreatic lesions induced by an experimental pancreatitis model uncover that 4E-BP1/2-deleted mice display an increased acinar cell proliferation and a better recovery than WT animals. Targeting translation, independently of 4E-BP1 status, using eIF4A RNA helicase inhibitors (silvestrol derivatives) selectively modulates translation and limits CDC6 expression and DNA replication, leading to reduced PDAC tumor growth. In summary, 4E-BP1 expression loss during PDAC development induces selective changes in translation of mRNA encoding DNA replication and repair protein. Importantly, targeting protein synthesis by eIF4A inhibitors circumvents PDAC resistance to mTOR inhibition.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Cycle Proteins/genetics , DNA Replication , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Biosynthesis , TOR Serine-Threonine Kinases/antagonists & inhibitors
8.
Front Chem ; 7: 849, 2019.
Article in English | MEDLINE | ID: mdl-31921772

ABSTRACT

Technologies that induce targeted protein degradation by small molecules have been developed recently. Chimeric small molecules such as Proteolysis Targeting Chimeras (PROTACs) and Specific and Non-genetic IAP-dependent Protein Erasers (SNIPERs), and E3 modulators such as thalidomides, hijack the cellular machinery for ubiquitylation, and the ubiquitylated proteins are subjected to proteasomal degradation. This has motivated drug development in industry and academia because "undruggable targets" can now be degraded by targeted protein degradation.

9.
Proc Natl Acad Sci U S A ; 113(44): 12360-12367, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27791185

ABSTRACT

Translational control of gene expression plays a key role during the early phases of embryonic development. Here we describe a transcriptional regulator of mouse embryonic stem cells (mESCs), Yin-yang 2 (YY2), that is controlled by the translation inhibitors, Eukaryotic initiation factor 4E-binding proteins (4E-BPs). YY2 plays a critical role in regulating mESC functions through control of key pluripotency factors, including Octamer-binding protein 4 (Oct4) and Estrogen-related receptor-ß (Esrrb). Importantly, overexpression of YY2 directs the differentiation of mESCs into cardiovascular lineages. We show that the splicing regulator Polypyrimidine tract-binding protein 1 (PTBP1) promotes the retention of an intron in the 5'-UTR of Yy2 mRNA that confers sensitivity to 4E-BP-mediated translational suppression. Thus, we conclude that YY2 is a major regulator of mESC self-renewal and lineage commitment and document a multilayer regulatory mechanism that controls its expression.


Subject(s)
Alternative Splicing/physiology , Cell Differentiation , Cell Self Renewal/physiology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Animals , Blastocyst/metabolism , Carrier Proteins/metabolism , Cell Lineage , Cell Self Renewal/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Introns , Mice , Mice, Knockout , Models, Biological , Octamer Transcription Factor-3/metabolism , Phosphoproteins , Polypyrimidine Tract-Binding Protein/genetics , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/genetics , Transcription, Genetic/physiology , YY1 Transcription Factor/metabolism
11.
Nat Commun ; 7: 11776, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27319316

ABSTRACT

Targeting mTORC1 is a highly promising strategy in cancer therapy. Suppression of mTORC1 activity leads to rapid dephosphorylation of eIF4E-binding proteins (4E-BP1-3) and subsequent inhibition of mRNA translation. However, how the different 4E-BPs affect translation during prolonged use of mTOR inhibitors is not known. Here we show that the expression of 4E-BP3, but not that of 4E-BP1 or 4E-BP2, is transcriptionally induced during prolonged mTORC1 inhibition in vitro and in vivo. Mechanistically, our data reveal that 4E-BP3 expression is controlled by the transcription factor TFE3 through a cis-regulatory element in the EIF4EBP3 gene promoter. CRISPR/Cas9-mediated EIF4EBP3 gene disruption in human cancer cells mitigated the inhibition of translation and proliferation caused by prolonged treatment with mTOR inhibitors. Our findings show that 4E-BP3 is an important effector of mTORC1 and a robust predictive biomarker of therapeutic response to prolonged treatment with mTOR-targeting drugs in cancer.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Breast Neoplasms/genetics , Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic , TOR Serine-Threonine Kinases/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , CRISPR-Cas Systems , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Proliferation , Databases, Genetic , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Female , Gene Editing/methods , HeLa Cells , Hep G2 Cells , Humans , Indoles/pharmacology , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Biosynthesis , Purines/pharmacology , Signal Transduction , Sirolimus/pharmacology , Survival Analysis , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
12.
J Cell Biochem ; 117(2): 500-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26239904

ABSTRACT

PKR-like ER-resident kinase (PERK) phosphorylates eukaryotic translation initiation factor 2 α (eIF2α) under endoplasmic reticulum (ER) stress; this results in repression of general translation and induction of specific gene expression, such as activating transcription factor 4 (ATF4). We previously showed that, upon ER stress, transducin (ß)-like 2 (TBL2) was an ER-localized transmembrane protein and interacted with PERK and that TBL2 was involved in ATF4 expression and cell survival. Here, we show that TBL2 is able to associate with ATF4 mRNA and regulate its translation. The RNA-immunoprecipitation analysis using several TBL2 deletion mutants revealed that the WD40 domain was essential for association with ATF4 mRNA. Importantly, suppression of TBL2 by knockdown or overexpression of the TBL2 mutant with a defective WD40 domain diminished ATF4 induction at the translational level. Thus, our findings indicate that, under ER stress, TBL2 participates in ATF4 translation through its association with the mRNA.


Subject(s)
Activating Transcription Factor 4/genetics , Endoplasmic Reticulum Stress , GTP-Binding Proteins/metabolism , RNA, Messenger/metabolism , Activating Transcription Factor 4/metabolism , HEK293 Cells , Humans , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , Transcriptional Activation
13.
J Biol Chem ; 290(26): 15996-6020, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25940091

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) is a critical regulator of protein synthesis. The best studied targets of mTORC1 in translation are the eukaryotic initiation factor-binding protein 1 (4E-BP1) and ribosomal protein S6 kinase 1 (S6K1). In this study, we identify the La-related protein 1 (LARP1) as a key novel target of mTORC1 with a fundamental role in terminal oligopyrimidine (TOP) mRNA translation. Recent genome-wide studies indicate that TOP and TOP-like mRNAs compose a large portion of the mTORC1 translatome, but the mechanism by which mTORC1 controls TOP mRNA translation is incompletely understood. Here, we report that LARP1 functions as a key repressor of TOP mRNA translation downstream of mTORC1. Our data show the following: (i) LARP1 associates with mTORC1 via RAPTOR; (ii) LARP1 interacts with TOP mRNAs in an mTORC1-dependent manner; (iii) LARP1 binds the 5'TOP motif to repress TOP mRNA translation; and (iv) LARP1 competes with the eukaryotic initiation factor (eIF) 4G for TOP mRNA binding. Importantly, from a drug resistance standpoint, our data also show that reducing LARP1 protein levels by RNA interference attenuates the inhibitory effect of rapamycin, Torin1, and amino acid deprivation on TOP mRNA translation. Collectively, our findings demonstrate that LARP1 functions as an important repressor of TOP mRNA translation downstream of mTORC1.


Subject(s)
Autoantigens/metabolism , Down-Regulation , Membrane Glycoproteins/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Ribonucleoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Autoantigens/genetics , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Membrane Glycoproteins/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding , RNA, Long Noncoding , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Regulatory-Associated Protein of mTOR , Ribonucleoproteins/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , SS-B Antigen
14.
Biochem Biophys Res Commun ; 462(4): 383-8, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25976671

ABSTRACT

Transducin (beta)-like 2 (TBL2) is a poorly characterized protein comprising the N-terminal transmembrane region and the C-terminal WD40 domain. We previously showed that TBL2 is an endoplasmic reticulum (ER)-localized protein that interacts with PKR-like ER-resident kinase (PERK), and under ER stress, it mediates protein expression of activating transcription factor 4 (ATF4). However, further molecular characterization of TBL2 is useful to better understand the function of this molecule. Here, we show that TBL2 associates with the eukaryotic 60S ribosomal subunit but not with the 40S subunit. The association of TBL2 with the 60S subunit was ER stress independent while the TBL2-PERK interaction occurred upon ER stress. Immunoprecipitation analysis using TBL2 deletion mutants revealed that the WD40 domain was essential for the 60S subunit association. These results could provide an important clue to understanding how TBL2 is involved in the expression of specific proteins under ER stress conditions.


Subject(s)
Endoplasmic Reticulum/metabolism , GTP-Binding Proteins/metabolism , Ribosomes/metabolism , Binding Sites , GTP-Binding Proteins/chemistry , HEK293 Cells , Humans
15.
Nat Neurosci ; 18(6): 855-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25915475

ABSTRACT

The circadian (∼24 h) clock is continuously entrained (reset) by ambient light so that endogenous rhythms are synchronized with daily changes in the environment. Light-induced gene expression is thought to be the molecular mechanism underlying clock entrainment. mRNA translation is a key step of gene expression, but the manner in which clock entrainment is controlled at the level of mRNA translation is not well understood. We found that a light- and circadian clock-regulated MAPK/MNK pathway led to phosphorylation of the cap-binding protein eIF4E in the mouse suprachiasmatic nucleus of the hypothalamus, the locus of the master circadian clock in mammals. Phosphorylation of eIF4E specifically promoted translation of Period 1 (Per1) and Period 2 (Per2) mRNAs and increased the abundance of basal and inducible PER proteins, which facilitated circadian clock resetting and precise timekeeping. Together, these results highlight a critical role for light-regulated translational control in the physiology of the circadian clock.


Subject(s)
Behavior, Animal/physiology , Circadian Rhythm/physiology , Eukaryotic Initiation Factor-4E/physiology , Animals , Behavior, Animal/radiation effects , Brain Chemistry/genetics , Brain Chemistry/physiology , Circadian Rhythm/radiation effects , Gene Expression Regulation/radiation effects , Light , MAP Kinase Signaling System/radiation effects , Mice , Mice, Inbred C57BL , Period Circadian Proteins/genetics , Period Circadian Proteins/physiology , Phosphorylation , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/physiology
16.
PLoS One ; 9(11): e112761, 2014.
Article in English | MEDLINE | ID: mdl-25393282

ABSTRACT

Under ER stress, PKR-like ER-resident kinase (PERK) phosphorylates translation initiation factor eIF2α, resulting in repression of global protein synthesis and concomitant upregulation of the translation of specific mRNAs such as activating transcription factor 4 (ATF4). This PERK function is important for cell survival under ER stress and poor nutrient conditions. However, mechanisms of the PERK signaling pathway are not thoroughly understood. Here we identify transducin (beta)-like 2 (TBL2) as a novel PERK-binding protein. We found that TBL2 is an ER-localized type-I transmembrane protein and preferentially binds to the phosphorylated form of PERK, but not another eIF2α kinase GCN2 or ER-resident kinase IRE1, under ER stress. Immunoprecipitation analysis using various deletion mutants revealed that TBL2 interacts with PERK via the N-terminus proximal region and also associates with eIF2α via the WD40 domain. In addition, TBL2 knockdown can lead to impaired ATF4 induction under ER stress or poor nutrient conditions such as glucose and oxygen deprivation. Consistently, TBL2 knockdown rendered cells vulnerable to stresses similarly to PERK knockdown. Thus, TBL2 serves as a potential regulator of the PERK pathway.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Eukaryotic Initiation Factor-2/genetics , GTP-Binding Proteins/genetics , eIF-2 Kinase/genetics , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Apoptosis/drug effects , Binding Sites , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Survival/drug effects , Endoribonucleases/genetics , Endoribonucleases/metabolism , Eukaryotic Initiation Factor-2/metabolism , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Glucose/deficiency , HEK293 Cells , Humans , Oxygen/pharmacology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/metabolism
17.
Mol Cancer Ther ; 11(5): 1082-91, 2012 May.
Article in English | MEDLINE | ID: mdl-22402126

ABSTRACT

Biguanides, including metformin, buformin, and phenformin, are potential antitumorigenic agents and induce cell death during glucose deprivation, a cell condition that occurs in the tumor microenvironment. Here, we show that this selective killing of glucose-deprived cells is coupled with hyperactivation of eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), a negative regulator of translation initiation. We found, in fact, that the 4E-BP1 hyperactivation led to failure of the unfolded protein response (UPR), an endoplasmic reticulum-originated stress signaling pathway for cell survival. We also found that the 4E-BP1-mediated UPR inhibition occurred through a strong inhibition of the mTOR signaling pathway, a proven antitumor target. Importantly, the 4E-BP1 hyperactivation can be also seen in xenografted cancer cells through an in vivo biguanide treatment. Our findings indicate that antitumor action of biguanides can be mediated by 4E-BP1 hyperactivation, which results in UPR inhibition and selective cell killing when glucose is withdrawn.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/toxicity , Biguanides/toxicity , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Death/genetics , Cell Line, Tumor , Glucose/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphoproteins/genetics , Phosphorylation/drug effects , Protein Transport/drug effects , Stress, Physiological , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
18.
Cancer Sci ; 101(5): 1125-32, 2010 May.
Article in English | MEDLINE | ID: mdl-20210797

ABSTRACT

Cancer cells consume large amounts of glucose because of their specific metabolic pathway. However, cancer cells exist in tumor tissue where glucose is insufficient. To survive, cancer cells likely have the mechanism to elude their glucose addiction. Here we show that functional mitochondria are essential if cancer cells are to avoid glucose addiction. Cancer cells with dysfunctional mitochondria, such as mitochondrial DNA-deficient rho(0) cells and electron transport chain blocker-treated cells, were highly sensitive to glucose deprivation. Our data demonstrated that this sensitization was associated with failure of the unfolded protein response (UPR), an adaptive response mediated by the endoplasmic reticulum (ER). This study suggests a link between mitochondria and the ER during the UPR under glucose deprivation conditions and that mitochondria govern cell fate, not only through ATP production and apoptosis regulation, but also through modulating the UPR for cell survival.


Subject(s)
Glucose/metabolism , Mitochondria/physiology , Neoplasms/metabolism , Unfolded Protein Response , Cell Line, Tumor , Cell Survival , Electron Transport/physiology , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Humans , Neoplasms/pathology
19.
J Biol Chem ; 284(40): 27500-10, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19656946

ABSTRACT

NUCB1 (nucleobindin 1) is a Golgi-localized soluble protein with a signal peptide and multiple functional domains. We reported recently that NUCB1 is a negative regulator of the unfolded protein response that activates various endoplasmic reticulum (ER)-originating signaling pathways. In that report, we also showed that Golgi localization of NUCB1 was essential to regulate the unfolded protein response. However, the localization mechanism of NUCB1 is still unknown. Here, we report that the proline residue at the +2-position (Pro(+2)) from the signal peptide cleavage site is the determinant of NUCB1 protein export from the ER and subsequent transport to the Golgi. Fusion of the N-terminal amino acids 1-35 peptide region, including both signal peptide (amino acids 1-26) and Pro(+2), was sufficient for enhanced green fluorescent protein to localize in the Golgi, whereas single amino acid mutation of Pro(+2) resulted in defective export from the ER without affecting the protein maturation process. Furthermore, we demonstrated that Pro(+2) was important for the enhanced green fluorescent protein fusion protein to concentrate at a transport vesicle formation site within the ER, often termed the ER exit site. Interestingly, such a Pro(+2) has also been functionally conserved in other Golgi-localized soluble proteins, Cab45 (Ca(2+)-binding protein of 45 kDa), reticulocalbin 1, and calumenin. Our findings indicate that Pro(+2) can function as a novel ER export signal of some Golgi proteins.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Proline/metabolism , Protein Sorting Signals , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Calcium-Binding Proteins/genetics , Cell Line , Conserved Sequence , DNA-Binding Proteins/genetics , Humans , Mice , Molecular Sequence Data , Mutation , Nerve Tissue Proteins , Nucleobindins , Protein Transport
20.
J Biol Chem ; 284(9): 5478-87, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19112105

ABSTRACT

The cysteine protease caspase-8 plays an essential role in apoptosis induced by death receptors. The protein synthesis inhibitor acetoxycycloheximide (Ac-CHX) has been previously shown to induce rapid apoptosis mediated by the release of cytochrome c in human leukemia Jurkat cells. In this study, the novel molecular mechanism that links caspase-8 to the mitochondrial release of pro-apoptotic proteins has been identified. Jurkat cells deficient in caspase-8 were more resistant to Ac-CHX than wild-type Jurkat cells and manifested decreased apoptosis induction and caspase activation as well as inefficient release of cytochrome c, Smac/DIABLO, and AIF into the cytosol. In contrast to Fas ligand stimulation, the general caspase inhibitor barely prevented the mitochondrial release of these pro-apoptotic proteins in Ac-CHX-treated cells, suggesting that caspase-8 activity is dispensable for triggering the mitochondrial pathway in Ac-CHX-induced apoptosis. Consistent with this notion, caspase-8-deficient Jurkat cells reconstituted with catalytically inactive caspase-8 became sensitive to Ac-CHX and exhibited apoptosis, caspase activation, the liberation of pro-apoptotic proteins into the cytosol, and Bak conformational change as efficiently as wild-type Jurkat cells. Unlike caspase-3, -6, -7, and -9, a small but significant portion of caspase-8 was found to localize in mitochondria before and after exposure to Ac-CHX. These results clearly demonstrate that caspase-8 is able to mediate the mitochondrial release of pro-apoptotic proteins in a manner independent of its proteolytic activity in Ac-CHX-induced apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Caspase 8/metabolism , Cycloheximide/analogs & derivatives , Mitochondria/drug effects , Protein Synthesis Inhibitors/pharmacology , Apoptosis/physiology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Blotting, Western , Caspases/metabolism , Cycloheximide/pharmacology , Cytochromes c/metabolism , Cytosol/drug effects , Cytosol/metabolism , Enzyme Activation/drug effects , Humans , Jurkat Cells , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Protein Conformation , RNA, Small Interfering/pharmacology , Subcellular Fractions , Transfection , bcl-2-Associated X Protein/chemistry , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL