Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Oncogene ; 41(43): 4779-4794, 2022 10.
Article in English | MEDLINE | ID: mdl-36127398

ABSTRACT

Genetic alteration of Rho GTPase-activating proteins (ARHGAP) and GTPase RhoA is a hallmark of diffuse-type gastric cancer and elucidating its biological significance is critical to comprehensively understanding this malignancy. Here, we report that gene fusions of ARHGAP6/ARHGAP26 are frequent genetic events in peritoneally-metastasized gastric and pancreatic cancer. From the malignant ascites of patients, we established gastric cancer cell lines that spontaneously gain hotspot RHOA mutations or four different ARHGAP6/ARHGAP26 fusions. These alterations critically downregulate RhoA-ROCK-MLC2 signaling, which elicits cell death. Omics and functional analyses revealed that the downstream signaling initiates actin stress fibers and reinforces intercellular junctions via several types of catenin. E-cadherin-centered homotypic adhesion followed by lysosomal membrane permeabilization is a pivotal mechanism in cell death. These findings support the tumor-suppressive nature of ARHGAP-RhoA signaling and might indicate a new avenue of drug discovery against this refractory cancer.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Actins/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Cadherins/metabolism , Catenins/metabolism , Cell Death , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
2.
Pathobiology ; 89(4): 222-232, 2022.
Article in English | MEDLINE | ID: mdl-35272288

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDA) is associated with very poor prognoses. Therefore, new therapies and preclinical models are urgently needed. In the present study, we sought to develop more realistic experimental models for use in PDA research. METHODS: We developed patient-derived xenografts (PDXs), established PDX-derived cell lines (PDCLs), and generated cell line-derived xenografts (CDXs), which we integrated to create 13 matched "trios" - i.e., patient-derived tumor models of PDA. We then compared and contrasted histological and molecular alterations between these three model systems. RESULTS: Orthotopic implantation (OI) of the PDCLs resulted in tumorigenesis and metastases to the liver and peritoneum. Morphological comparisons of OI-CDXs and OI-PDXs with passaged tumors revealed that the histopathological features of the original tumor were maintained in both models. Molecular alterations in PDX tumors (including those to KRAS, TP53, SMAD4, and CDKN2A) were similar to those in the respective PDCLs and CDX tumors. When gene expression levels in the PDCLs, ectopic tumors, and OI tumors were compared, the distant metastasis-promoting gene CXCR4 was specifically upregulated in OI tumors, whose immunohistochemical profiles suggested epithelial-mesenchymal transition and adeno-squamous trans-differentiation. CONCLUSION: These patient-derived tumor models provide useful tools for monitoring responses to antineoplastic agents and for studying PDA biology.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Humans , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
3.
Oncogene ; 41(16): 2326-2339, 2022 04.
Article in English | MEDLINE | ID: mdl-35250028

ABSTRACT

Oncogenic signalling confers tumour-progression advantages; thus, its pharmacological blockade is the best strategy for cancer chemotherapy. However, drug resistance and heterogeneous dependency of tumour hamper their therapeutic potential, suggesting the necessity for a new ubiquitous modality based on evading drug resistance. Here, we proposed a de novo addiction to oncogenic signalling (Dead-On) concept, wherein specific blockade of target molecules forces cancer cells to develop dependency on an oncogenic signalling. In cervical squamous cell carcinoma cells, Aurora A/B dual blockade elicited rapid addiction to EGFR-Erk signalling, and its pharmacological/genetic inhibition synergistically enhanced anti-cancer activities in vitro, in vivo, and in a patient-derived organoid model. The signal activation was independent of EGFR genetic status, it was triggered by receptor accumulation on the plasma membrane via Rab11-mediated endocytic recycling machinery. These findings support our novel Dead-On concept which may lead to drug discovery as well as expand the adaptation of approved targeted drugs.


Subject(s)
Carcinoma, Squamous Cell , Uterine Cervical Neoplasms , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , ErbB Receptors/metabolism , Female , Humans , Oncogenes , Signal Transduction , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics
4.
Pharmaceuticals (Basel) ; 15(2)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35215239

ABSTRACT

In the field of drug repurposing, the use of statins for treating dyslipidemia is considered promising in ovarian cancer treatment based on epidemiological studies and basic research findings. Biomarkers should be established to identify patients who will respond to statin treatment to achieve clinical application. In the present study, we demonstrated that statins have a multifaceted mode of action in ovarian cancer and involve pathways other than protein prenylation. To identify biomarkers that predict the response to statins, we subjected ovarian cancer cells to microarray analysis and calculated Pearson's correlation coefficients between gene expression and cell survival after statin treatment. The results showed that VDAC1 and LDLRAP1 were positively and negatively correlated with the response to statins, respectively. Histoculture drug response assays revealed that statins were effective in clinical samples. We also confirmed the synergistic effects of statins with paclitaxel and panobinostat and determined that statins are hematologically safe to administer to statin-treated mice. Future clinical trials based on the expression of the biomarkers identified in this study for repurposing statins for ovarian cancer treatment are warranted.

5.
Nat Cancer ; 2(9): 962-977, 2021 09.
Article in English | MEDLINE | ID: mdl-35121863

ABSTRACT

Peritoneal metastasis, a hallmark of incurable advanced gastric cancer (GC), presently has no curative therapy and its molecular features have not been examined extensively. Here we present a comprehensive multi-omic analysis of malignant ascitic fluid samples and their corresponding tumor cell lines from 98 patients, including whole-genome sequencing, RNA sequencing, DNA methylation and enhancer landscape. We identify a higher frequency of receptor tyrosine kinase and mitogen-activated protein kinase pathway alterations compared to primary GC; moreover, approximately half of the gene alterations are potentially treatable with targeted therapy. Our analyses also stratify ascites-disseminated GC into two distinct molecular subtypes: one displaying active super enhancers (SEs) at the ELF3, KLF5 and EHF loci, and a second subtype bearing transforming growth factor-ß (TGF-ß) pathway activation through SMAD3 SE activation and high expression of transcriptional enhancer factor TEF-1 (TEAD1). In the TGF-ß subtype, inhibition of the TEAD pathway circumvents therapy resistance, suggesting a potential molecular-guided therapeutic strategy for this subtype of intractable GC.


Subject(s)
Peritoneal Neoplasms , Stomach Neoplasms , Ascites/genetics , Cell Line, Tumor , Humans , Peritoneal Neoplasms/genetics , Stomach Neoplasms/genetics , Transforming Growth Factor beta/genetics
6.
Int J Oncol ; 56(3): 835-847, 2020 03.
Article in English | MEDLINE | ID: mdl-32124945

ABSTRACT

Definitive chemoradiotherapy (CRT) is a less invasive therapy compared with surgery for some types of cancer; however, the 5­year survival rate of patients with stages II­III esophageal squamous cell carcinoma (ESCC) is only 37%. Therefore, prediction of CRT responders is necessary. Unfortunately, no definitive biomarker exists that is useful to predict survival outcome following CRT. From our previous microarray study, CD24 and keratin 4 (KRT4), which encodes cytokeratin 4 (CK4), were overexpressed in the favorable prognostic epithelial subtype with SIM bHLH transcription factor 2 (SIM2) expression. This study investigated the association between their mRNA and protein expression levels, and clinicopathological characteristics, and also investigated the functions of CD24 in SIM2­mediated tumor differentiation and CRT sensitivity. High CD24 and KRT4 mRNA expression was associated with a favorable prognosis following CRT. Multivariate analyses revealed that high CD24 and CK4 protein expression, as determined by immunohistochemistry, and differentiated type were independent factors for predicting a favorable prognosis in response to CRT. Notably, in cases with low CD24 or CK4, surgery was suggested to be a good therapeutic modality compared with CRT. CD24 and KRT4 were expressed preferentially in differentiated layers of the normal esophageal mucosa, and their mRNA expression in 3D cultured ESCC cells was induced by SIM2 transfection, thus suggesting that CD24 and KRT4 were downstream differentiation markers of SIM2. Furthermore, CD24 small interfering RNA increased the mRNA expression levels of superoxide dismutase 2 and enhanced H2O2 resistance, thus indicating the involvement of CD24 in the radiosensitivity of patients with ESCC; however, it had no effect on cisplatin sensitivity. In conclusion, the two markers CD24 and CK4 may be considered predictive biomarkers for definitive CRT.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers, Tumor/genetics , CD24 Antigen/genetics , Carcinoma, Small Cell/therapy , Esophageal Neoplasms/therapy , Keratin-4/genetics , Up-Regulation , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers, Tumor/metabolism , CD24 Antigen/metabolism , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Chemoradiotherapy , Digestive System Surgical Procedures , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Keratin-4/metabolism , Male , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , Survival Analysis , Survival Rate , Treatment Outcome
7.
Biochem Biophys Res Commun ; 522(2): 342-347, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31761322

ABSTRACT

ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, increases the intracellular levels of glutathione (GSH) by upregulating solute carrier family 7 member 11 (SLC7A11). Diffuse-type gastric cancer is an aggressive tumor that is frequently associated with ARID1A deficiency. Here, we investigated the efficacy of GSH inhibition for the treatment of diffuse-type gastric cancer with ARID1A deficiency using ARID1A-proficient or -deficient patient-derived cells (PDCs). ARID1A-deficient PDCs were selectively sensitive to the GSH inhibitor APR-246, the GCLC inhibitor buthionine sulfoximine, and the SLC7A11 inhibitor erastin. Expression of SLC7A11, which is required for incorporation of cystine, and the basal level of GSH were lower in ARID1A-deficient than in ARID1A-proficient PDCs. Treatment with APR-246 decreased intracellular GSH levels, leading to the excessive production of reactive oxygen species (ROS), and these phenotypes are suppressed by supply of cystine and GSH compensators. Taken together, vulnerability of ARID1A-deficient gastric cancer cells to GSH inhibition is caused by decreased GSH synthesis due to diminished SLC7A11 expression. The present results suggest that GSH inhibition is a promising strategy for the treatment of diffuse-type gastric cancers with ARID1A deficiency.


Subject(s)
DNA-Binding Proteins/deficiency , Glutathione/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Transcription Factors/deficiency , Amino Acid Transport System y+/metabolism , Animals , Ascites/metabolism , Ascites/pathology , DNA-Binding Proteins/metabolism , Female , Glutathione/metabolism , Humans , Mice, Nude , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
8.
Cell Death Dis ; 10(12): 876, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31748500

ABSTRACT

Cervical cancer is the most common gynecological malignancy in the world; however, the survival rates of advanced-stage and recurrent cervical cancer patients remain poor. The multifaced protein insulin-like growth factor 2 receptor (IGF2R) has various ligands, represented as IGF-2 and mannose-6-phosphate (M6P)-tagged proteins. Regarding its antagonistic activity as an IGF1R signal, IGF2R is currently considered a tumor suppressor gene, whereas its significance as an M6P receptor is still unclear. Here, on the basis of transcriptome analysis of TCGA and GEO open datasets, we show that IGF2R is upregulated and correlated with poor prognosis in cervical cancer. Several experiments using cervical cancer cell lines revealed that IGF2R depletion induced apoptosis, decreased cell viability, and increased vulnerability to certain anticancer drug cisplatin. In contrast to its negligible impact in IGF1R signaling, loss of IGF2R disrupted the Golgi-to-lysosome transport of M6P-tagged cathepsins, resulting in decreased lysosomal activity, with their abnormal accumulation and dysfunction of both autophagy and mitophagy, which cause the accumulation of misfolded proteins and production of reactive oxygen species. Taken together, IGF2R has an oncogenic role through transportation of M6P-tagged cargo in cervical cancer and can be used as a predictive biomarker for prognostic classification.


Subject(s)
Cathepsins/metabolism , Receptor, IGF Type 2/metabolism , Uterine Cervical Neoplasms/metabolism , Apoptosis/physiology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Gene Knockdown Techniques , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , Humans , Lysosomes/genetics , Lysosomes/metabolism , Lysosomes/pathology , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Receptor, IGF Type 1/metabolism , Up-Regulation , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
9.
Cancer Sci ; 109(4): 1121-1134, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29427302

ABSTRACT

Degree of histological differentiation is an important characteristic of cancers and may be associated with malignant potential. However, in squamous cell carcinomas, a key transcriptional factor regulating tumor differentiation is largely unknown. Chemoradiotherapy (CRT) is a standard treatment for locally advanced esophageal squamous cell carcinoma; however, the survival rate is still below 40%. From microarray data, single-minded 2 (SIM2) was overexpressed in the epithelial subtype. Here, we investigated the correlation between SIM2 expression and its clinical implication, and in vitro and in vivo functions of SIM2 in tumor differentiation and in CRT sensitivity. Although SIM2 was suppressed in cancerous tissues, SIM2-high ESCC showed a favorable prognosis in CRT. Transient SIM2 expression followed by 3D culture induced expression of differentiation markers and suppressed epithelial-mesenchymal transition- and basal-cell markers. Levels of PDPN-high tumor basal cells and of expression of genes for DNA repair and antioxidant enzymes were reduced in stable transfectants, and they showed high CDDP and H2 O2 sensitivities, and their xenografts showed a well-differentiated histology. Reduction of tumor basal cells was restored by knockdown of aryl hydrocarbon receptor nuclear translocator (ARNT) that interacted with SIM2. Together, SIM2 increases CRT sensitivity through tumor differentiation by cooperation with ARNT.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Animals , Antioxidants/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Biomarkers, Tumor/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , DNA Repair/physiology , Epithelial-Mesenchymal Transition/physiology , Esophageal Squamous Cell Carcinoma , Female , Gene Expression Regulation/physiology , Humans , Mice , Survival Rate , Transfection/methods
10.
Sci Rep ; 7(1): 14574, 2017 11 06.
Article in English | MEDLINE | ID: mdl-29109451

ABSTRACT

Despite chemoradiotherapy being one of the most important modalities in advanced cervical cancer, there is a lack of both usable biomarkers to predict treatment outcome and of knowledge about the mechanism of refractoriness to the therapy. Here we identified a transcriptional factor Single-minded homolog 2 (SIM2) as an independent predictive biomarker for uterine cervical squamous cell carcinoma (CvSCC). The retrospective study showed that high expression level of SIM2 was correlated to good survival in CvSCC patients. SIM2 knockdown in CvSCC cell lines showed resistance to hypoxia with increased expression of HIF1A and its target genes. Loss of SIM2 also caused growth promotion, resistance to ROS, and radiation in 3D culture. Furthermore, SIM2 knockdown suppressed tumor growth with increased HIF-1α expression and angiogenesis in vivo. On the other hand, SIM2 long isoform (SIM2l)-overexpressed cells had contrary results, indicating the long isoform plays a key role for maintenance of these phenotypes. These data indicated that SIM2l has a potential to be precision medicine for CvSCC patients and that anti-angiogenesis therapy might be usable for SIM2lLow poor survivors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Squamous Cell/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Uterine Cervical Neoplasms/metabolism , Biomarkers/metabolism , Carcinoma, Squamous Cell/diagnosis , Female , Gene Knockdown Techniques , Humans , Hypoxia/metabolism , Middle Aged , Prognosis , Retrospective Studies , Uterine Cervical Neoplasms/diagnosis
11.
Cancer Sci ; 108(2): 216-225, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27987372

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors. Although improvement in both surgical techniques and neoadjuvant chemotherapy has been achieved, the 5-year survival rate of locally advanced tumors was, at best, still 55%. Therefore, elucidation of mechanisms of the malignancy is eagerly awaited. Epithelial-mesenchymal transition (EMT) by transforming growth factor-ß (TGF-ß) has been reported to have critical biological roles for cancer cell stemness, whereas little is known about it in ESCC. In the current study, a transcriptional factor SIX1 was found to be aberrantly expressed in ESCCs. SIX1 cDNA transfection induced overexpression of transforming growth factors (TGFB1 and TGFB2) and its receptor (TGFBR2). Cell invasion was reduced by SIX1 knockdown and was increased in stable SIX1-transfectants. Furthermore, the SIX1-transfectants highly expressed tumor basal cell markers such as NGFR, SOX2, ALDH1A1, and PDPN. Although mock-transfectants had only a 20% PDPN-high population, SIX1-transfectants had 60-70%. In two sets of 42 and 85 ESCC patients receiving surgery alone or neoadjuvant chemoradiotherapy followed by surgery, the cases with high SIX1 mRNA and protein expression level significantly showed a poor prognosis compared with those with low levels. These SIX1 high cases also expressed the above basal cell markers, but suppressed the differentiation markers. Finally, TGF-ß signaling blockade suppressed ESCC cell growth in association with the reduction of PDPN-positive tumor basal cell population. The present results suggest that SIX1 accelerates self-renewal of tumor basal cells, resulting in a poor prognosis for ESCC patients.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/metabolism , Homeodomain Proteins/metabolism , Neoplasm Proteins/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Homeodomain Proteins/genetics , Humans , Membrane Glycoproteins/metabolism , Neoplasm Proteins/genetics , Prognosis , Receptors, Transforming Growth Factor beta/genetics , Transfection , Transforming Growth Factor beta/genetics
12.
Oncotarget ; 8(7): 11020-11029, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28038457

ABSTRACT

Epidermal growth factor receptor (EGFR) is a well-validated oncological target molecule for monoclonal antibody therapies and Sym004 is a novel anti-EGFR antibody mixture comprising two recombinant chimeric IgG1 antibodies against non-overlapping epitopes of EGFR. Because EGFR is highly expressed in the majority of esophageal squamous cell carcinomas (ESCCs), we investigated the efficacy of Sym004 in human ESCC cell lines. Forty eight ESCC cell lines were treated with three kinds of anti-EGFR antibodies (Sym004, cetuximab, and panitumumab). Genetic background was investigated by next generation sequencing. The internalization of anti-EGFR antibodies into ESCC cells and inhibition of the EGFR signaling cascade by anti-EGFR antibodies were investigated in vitro. Furthermore, growth inhibition by anti-EGFR antibody treatment was investigated in vitro and in vivo. Sym004 treatments were more effective at inducing EGFR internalization and degradation than the two other anti-EGFR antibodies. Sym004 was more sensitive significantly to cell lines with EGFR gene amplification than those without amplification (P = 0.002). Growth inhibition of Sym004 was greater than in that of cetuximab or panitumumab in vitro and in vivo. These studies showed that Sym004 exhibited antitumor activity in some ESCC cell lines in preclinical settings and warrant a clinical evaluation in patients with ESCC. EGFR amplification is a potential biomarker of response to Sym004.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/metabolism , Esophageal Neoplasms/drug therapy , Mice, Inbred BALB C , Xenograft Model Antitumor Assays/methods , Animals , Antibodies, Monoclonal/immunology , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cetuximab/pharmacology , ErbB Receptors/genetics , ErbB Receptors/immunology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Gene Amplification , Humans , Mice, Nude , Panitumumab , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/genetics
13.
PLoS One ; 10(12): e0143804, 2015.
Article in English | MEDLINE | ID: mdl-26625258

ABSTRACT

Definitive chemoradiotherapy (CRT) is a less invasive therapy for esophageal squamous cell carcinoma (ESCC). Five-year survival rate of locally advanced ESCC patients by definitive CRT were 37%. We previously reported that tumor-specific cytotoxic T-lymphocyte (CTL) activation signatures were preferentially found in long-term survivors. However, it is unknown whether the CTL activation is actually driven by CRT. We compared gene expression profiles among pre- and post-treatment biopsy specimens of 30 ESCC patients and 121 pre-treatment ESCC biopsy specimens. In the complete response (CR) cases, 999 overexpressed genes including at least 234 tumor-specific CTL-activation associated genes such as IFNG, PRF1, and GZMB, were found in post-treatment biopsy specimens. Clustering analysis using expression profiles of these 234 genes allowed us to distinguish the immune-activated cases, designating them as I-type, from other cases. However, despite the better CR rate in the I-type, overall survival was not significantly better in both these 30 cases and another 121 cases. Further comparative study identified a series of epithelial to mesenchymal transition-related genes overexpressed in the early relapse cases. Importantly, the clinical outcome of CDH2-negative cases in the I-type was significantly better than that of the CDH2-positive cases in the I-type. Furthermore, NK cells, which were activated by neutrophils-producing S100A8/S100A9, and CTLs were suggested to cooperatively enhance the effect of CRT in the CDH2-negative I-type. These results suggested that CTL gene activation may provide a prognostic advantage in ESCCs with epithelial characteristics.


Subject(s)
Carcinoma, Squamous Cell/therapy , Chemoradiotherapy , Esophageal Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/prevention & control , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Esophageal Neoplasms/prevention & control , Esophageal Squamous Cell Carcinoma , Humans , Middle Aged , Secondary Prevention , Transcriptome
14.
PLoS One ; 10(6): e0130808, 2015.
Article in English | MEDLINE | ID: mdl-26110809

ABSTRACT

Diffuse-type solid tumors are often composed of a high proportion of rarely proliferating (i.e., dormant) cancer cells, strongly indicating the involvement of cancer stem cells (CSCs) Although diffuse-type gastric cancer (GC) patients have a poor prognosis due to high-frequent development of peritoneal dissemination (PD), it is limited knowledge that the PD-associated CSCs and efficacy of CSC-targeting therapy in diffuse-type GC. In this study, we established highly metastatic GC cell lines by in vivo selection designed for the enrichment of PD-associated GC cells. By microarray analysis, we found C-X-C chemokine receptor type 4 (CXCR4) can be a novel marker for highly metastatic CSCs, since CXCR4-positive cells can grow anchorage-independently, initiate tumors in mice, be resistant to cytotoxic drug, and produce differentiated daughter cells. In clinical samples, these CXCR4-positive cells were found from not only late metastasis stage (accumulated ascites) but also earlier stage (peritoneal washings). Moreover, treatment with transforming growth factor-ß enhanced the anti-cancer effect of docetaxel via induction of cell differentiation/asymmetric cell division of the CXCR4-positive gastric CSCs even in a dormant state. Therefore, differentiation inducers hold promise for obtaining the maximum therapeutic outcome from currently available anti-cancer drugs through re-cycling of CSCs.


Subject(s)
Neoplastic Stem Cells/metabolism , Receptors, CXCR4/metabolism , Stomach Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Humans , Mice , Neoplastic Stem Cells/pathology , Stomach Neoplasms/pathology , Taxoids/pharmacology , Transforming Growth Factor beta/pharmacology
15.
PLoS One ; 10(4): e0123407, 2015.
Article in English | MEDLINE | ID: mdl-25853862

ABSTRACT

The dormancy of tumor cells is a major problem in chemotherapy, since it limits the therapeutic efficacy of anti-tumor drugs that only target dividing cells. One potential way to overcome chemo-resistance is to "wake up" these dormant cells. Here we show that the opioid antagonist methylnaltrexone (MNTX) enhances the effect of docetaxel (Doc) by blocking a cell growth-suppressive pathway. We found that PENK, which encodes opioid growth factor (OGF) and suppresses cell growth, is predominantly expressed in diffuse-type gastric cancers (GCs). The blockade of OGF signaling by MNTX releases cells from their arrest and boosts the effect of Doc. In comparison with the use of Doc alone, the combined use of Doc and MNTX significantly prolongs survival, alleviates abdominal pain, and diminishes Doc-resistant spheroids on the peritoneal membrane in model mice. These results suggest that blockade of the pathways that suppress cell growth may enhance the effects of anti-tumor drugs.


Subject(s)
Naltrexone/analogs & derivatives , Receptors, Opioid/biosynthesis , Stomach Neoplasms/drug therapy , Taxoids/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Docetaxel , Drug Synergism , Growth Inhibitors/biosynthesis , Growth Inhibitors/genetics , Humans , Mice , Naltrexone/administration & dosage , Narcotic Antagonists/administration & dosage , Quaternary Ammonium Compounds/administration & dosage , Receptors, Opioid/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
16.
Chem Biol Drug Des ; 85(3): 356-68, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25082146

ABSTRACT

Several decades of cumulated research evidence have proven that aggregation of beta-amyloid 42 (Aß42) is the main cause of neuronal death in the brains of patients with Alzheimer's disease. Therefore, inhibition of Aß42 aggregation holds great promise for the prevention and treatment of Alzheimer's disease. To this end, we used a systematic in vitro evolution including a paired peptide library method. We identified two peptides with high binding affinity (with Kd in the nm range) for Aß42. Functionally, these peptides strongly inhibited the aggregation of Aß42 as shown by the thioflavin T assay and atomic force microscopy. Moreover, these peptides rescued PC12 cells from the cytotoxic effect of aggregated Aß42 in vitro. Our results suggest that these novel peptides may be potential therapeutic seeds for the treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Peptides/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/pharmacology , Animals , Cell Survival/drug effects , Fluorescein/chemistry , Kinetics , Microscopy, Atomic Force , Molecular Sequence Data , PC12 Cells , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptide Library , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Rats , Surface Plasmon Resonance
18.
J Pept Sci ; 18(12): 711-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23109368

ABSTRACT

Improving a particular function of molecules is often more difficult than identifying such molecules ab initio. Here, a method to acquire higher affinity and/or more functional peptides was developed as a progressive library selection method. The primary library selection products were utilized to build a secondary library composed of blocks of 4 amino acids, of which selection led to peptides with increased activity. These peptides were further converted to randomly generate paired peptides. Cathepsin E-inhibitors thus obtained exhibited the highest activities and affinities (pM order). This was also the case with cathepsin E-activating peptides, proving the methodological effectiveness. The primary, secondary, and tertiary library selections can be regarded as module-finding, module-shuffling, and module-pairing, respectively, which resembles the progression of the natural evolution of proteins. The mode of peptide binding to their target proteins is discussed in analogy to antibodies and epitopes of an antigen.


Subject(s)
Aptamers, Peptide/chemistry , Cathepsin E/antagonists & inhibitors , Protease Inhibitors/chemistry , Amino Acid Sequence , Directed Molecular Evolution , Hydrogen-Ion Concentration , Molecular Sequence Data , Peptide Library , Protein Binding
19.
Int J Pept ; 2012: 316432, 2012.
Article in English | MEDLINE | ID: mdl-23365585

ABSTRACT

Enzymes are regulated by their activation and inhibition. Enzyme activators can often be effective tools for scientific and medical purposes, although they are more difficult to obtain than inhibitors. Here, using the paired peptide method, we report on protease-cathepsin-E-activating peptides that are obtained at neutral pH. These selected peptides also underwent molecular evolution, after which their cathepsin E activation capability improved. Thus, the activators we obtained could enhance cathepsin-E-induced cancer cell apoptosis, which indicated their potential as cancer drug precursors.

20.
Protein Pept Lett ; 18(6): 642-50, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21342094

ABSTRACT

Recent studies have been supporting that the generation of Aß42 oligomers is responsible for Alzheimer's disease. Therefore, those peptides which bind to Aß42 are scientifically interesting and can be possible candidates for the diagnosis and therapy of Alzheimer's disease. A systemic in vitro evolution, developed recently and the designated progressive library method (PLM), was applied to obtain Ab42-binding aptamers peptides. As a result, high affinity peptide aptamers made of 8 or 9 amino acids could be identified by this approach, endorsing the methodological effectiveness. Namely, the selection products from the secondary library of diversified peptides, which was constructed based on the information obtained from the primary library selection, were confirmed to be superior to those selected from the primary library as had been reported previously. The affinities of those peptides measured by SPR (surface plasmon resonance) were comparable to or higher than that of those peptides so far reported (K(d) of 10⁻7). The other peptides selected were confirmed of their binding by a novel mode of gel shift assay (fluorescence enhancement caused by the binding). Thus, novel Aß42-binding peptides with high affinity were provided for the future Alzheimer's disease study. The demonstration of the effectiveness of the systemic in vitro evolution of PLM is very encouraging for the study of identifying novel functional peptides.


Subject(s)
Amyloid beta-Peptides/metabolism , Aptamers, Peptide/genetics , Aptamers, Peptide/metabolism , Directed Molecular Evolution/methods , Peptide Fragments/metabolism , Peptide Library , Amino Acid Sequence , Aptamers, Peptide/chemistry , Base Sequence , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...