Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
1.
J Dent Res ; 97(7): 779-786, 2018 07.
Article in English | MEDLINE | ID: mdl-29494307

ABSTRACT

This study investigated a case of spindle cell carcinoma (SpCC) in tongue pathological lesions. The patient experienced a local recurrence and distant metastasis after surgical intervention. Although standard chemotherapy was administered, a granulomatous mass continued to develop. This aggressive growth led to survival of the tumor. Secondary debulking surgery was performed to improve the patient's quality of life at the request of the patient. Using a tissue sample derived from the secondary debulking surgery, we performed an analysis of the tumor's cell surface antigens, differentiation potential, metastatic ability, and inhibition potential by anticancer reagents. In vitro analysis revealed that the cell population grown under adherent culture conditions expressed the mesenchymal stem cell (MSC) markers CD73, CD90, and CD105. The cell line established from this SpCC contained colony-forming unit fibroblasts (CFU-Fs) and exhibited multipotent differentiation into several mesenchymal lineages, including bone, cartilage, and fat. The SpCC cells also displayed vigorous mobilization. These characteristics suggested that they had the differentiation potential of mesenchymal cells, especially MSCs, rather than that of epithelial cells. The surgical specimen analyzed in this study resisted the molecular target reagent cetuximab, which is an epidermal growth factor receptor inhibitor. This clinical insight revealed that chemotherapy-resistant SpCC cells have different characteristics compared to most other cancer cells, which are sensitive to cetuximab. Our cell death assay revealed that SpCC cell death was induced by the anticancer drug imatinib, which is known to inhibit protein tyrosine kinase activity of ABL, platelet-derived growth factor receptor α (PDGFRα), and KIT. Here, we report recurrent SpCC with characteristics of MSCs and potential for treatment with imatinib.


Subject(s)
Carcinoma/pathology , Mesenchymal Stem Cells/pathology , Neoplasm Recurrence, Local/pathology , Tongue Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Carcinoma/therapy , Cell Culture Techniques , Cell Death , Cell Differentiation , Cell Movement , Combined Modality Therapy , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Neoplasm Recurrence, Local/therapy , Oral Surgical Procedures , Quality of Life , Stem Cells , Tongue Neoplasms/therapy , Tumor Cells, Cultured
2.
J Cell Biol ; 216(3): 641-656, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28193700

ABSTRACT

Phosphatase and tensin homologue (PTEN) protein levels are critical for tumor suppression. However, the search for a recurrent cancer-associated gene alteration that causes PTEN degradation has remained futile. In this study, we show that Importin-11 (Ipo11) is a transport receptor for PTEN that is required to physically separate PTEN from elements of the PTEN degradation machinery. Mechanistically, we find that the E2 ubiquitin-conjugating enzyme and IPO11 cargo, UBE2E1, is a limiting factor for PTEN degradation. Using in vitro and in vivo gene-targeting methods, we show that Ipo11 loss results in degradation of Pten, lung adenocarcinoma, and neoplasia in mouse prostate with aberrantly high levels of Ube2e1 in the cytoplasm. These findings explain the correlation between loss of IPO11 and PTEN protein in human lung tumors. Furthermore, we find that IPO11 status predicts disease recurrence and progression to metastasis in patients choosing radical prostatectomy. Thus, our data introduce the IPO11 gene as a tumor-suppressor locus, which is of special importance in cancers that still retain at least one intact PTEN allele.


Subject(s)
PTEN Phosphohydrolase/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Suppressor Proteins/metabolism , beta Karyopherins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , HeLa Cells , Humans , Lung Neoplasms/metabolism , Mice , Ubiquitin-Conjugating Enzymes/metabolism
3.
Oncogene ; 36(9): 1276-1286, 2017 03 02.
Article in English | MEDLINE | ID: mdl-27546618

ABSTRACT

The transcription factor nuclear factor-κB (NF-κB) has important roles for tumorigenesis, but how it regulates cancer stem cells (CSCs) remains largely unclear. We identified insulin-like growth factor 2 (IGF2) is a key target of NF-κB activated by HER2/HER3 signaling to form tumor spheres in breast cancer cells. The IGF2 receptor, IGF1 R, was expressed at high levels in CSC-enriched populations in primary breast cancer cells. Moreover, IGF2-PI3K (IGF2-phosphatidyl inositol 3 kinase) signaling induced expression of a stemness transcription factor, inhibitor of DNA-binding 1 (ID1), and IGF2 itself. ID1 knockdown greatly reduced IGF2 expression, and tumor sphere formation. Finally, treatment with anti-IGF1/2 antibodies blocked tumorigenesis derived from the IGF1Rhigh CSC-enriched population in a patient-derived xenograft model. Thus, NF-κB may trigger IGF2-ID1-IGF2-positive feedback circuits that allow cancer stem-like cells to appear. Then, they may become addicted to the circuits. As the circuits are the Achilles' heels of CSCs, it will be critical to break them for eradication of CSCs.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Inhibitor of Differentiation Protein 1/metabolism , Insulin-Like Growth Factor II/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinogenesis , Female , Humans , Inhibitor of Differentiation Protein 1/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/genetics , Mice , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Prognosis , Signal Transduction , Spheroids, Cellular , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 113(11): 3030-5, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929372

ABSTRACT

Loss of the tumor suppressor gene PTEN is implicated in breast cancer progression and resistance to targeted therapies, and is thought to promote tumorigenesis by activating PI3K signaling. In a transgenic model of breast cancer, Pten suppression using a tetracycline-regulatable short hairpin (sh)RNA cooperates with human epidermal growth factor receptor 2 (HER2/neu), leading to aggressive and metastatic disease with elevated signaling through PI3K and, surprisingly, the mitogen-activated protein kinase (MAPK) pathway. Restoring Pten function is sufficient to down-regulate both PI3K and MAPK signaling and triggers dramatic tumor regression. Pharmacologic inhibition of MAPK signaling produces similar effects to Pten restoration, suggesting that the MAPK pathway contributes to the maintenance of advanced breast cancers harboring Pten loss.


Subject(s)
MAP Kinase Signaling System/physiology , Mammary Neoplasms, Experimental/physiopathology , Neoplasm Proteins/physiology , PTEN Phosphohydrolase/deficiency , Receptor, ErbB-2/physiology , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genes, erbB-2 , Humans , MAP Kinase Kinase 1/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/physiology , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Proteins/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/physiology , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/physiology , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/physiology
5.
Oncogene ; 35(34): 4447-58, 2016 08 25.
Article in English | MEDLINE | ID: mdl-26829053

ABSTRACT

Overexpression of MYCN is a hallmark of neuroblastoma (NB). ALK(R1275Q), an activating mutation of ALK (anaplastic lymphoma kinase), has been found in sporadic and familial NB patients. In this report, we demonstrated that ALK(R1275Q) knock-in, MYCN transgenic compound mice developed NB with complete penetrance. Transcriptome analysis revealed that ALK(R1275Q) globally downregulated the expression of extracellular matrix (ECM)- and basement membrane (BM)-associated genes in both primary neuronal cells and NB tumors. Accordingly, ALK(R1275Q)/MYCN tumors exhibited reduced expression of ECM/BM-related proteins as compared with MYCN tumors. In addition, on MYCN transduction, ALK(R1275Q)-expressing neuronal cells exhibited increased migratory and invasive activities. Consistently, enhanced invasion and metastasis were demonstrated in ALK(R1275Q)/MYCN mice. These results collectively indicate that ALK(R1275Q) confers a malignant potential on neuronal cells that overexpress MYCN by impairing normal ECM/BM integrity and enhancing tumor growth and dissemination. Moreover, we found that crizotinib, an ALK inhibitor, almost completely inhibited the growth of ALK(R1275Q)/MYCN tumors in an allograft model. Our findings provided insights into the cooperative mechanism of the mutated ALK and overexpressed MYCN in the pathogenesis of NB and demonstrated the effectiveness of crizotinib on ALK(R1275Q)-positive tumors.


Subject(s)
Extracellular Matrix/metabolism , Mutation , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/etiology , Receptor Protein-Tyrosine Kinases/genetics , Anaplastic Lymphoma Kinase , Animals , Crizotinib , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/physiology
6.
Nat Cell Biol ; 17(6): 816-26, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25985394

ABSTRACT

Pancreatic ductal adenocarcinomas (PDACs) are highly metastatic with poor prognosis, mainly due to delayed detection. We hypothesized that intercellular communication is critical for metastatic progression. Here, we show that PDAC-derived exosomes induce liver pre-metastatic niche formation in naive mice and consequently increase liver metastatic burden. Uptake of PDAC-derived exosomes by Kupffer cells caused transforming growth factor ß secretion and upregulation of fibronectin production by hepatic stellate cells. This fibrotic microenvironment enhanced recruitment of bone marrow-derived macrophages. We found that macrophage migration inhibitory factor (MIF) was highly expressed in PDAC-derived exosomes, and its blockade prevented liver pre-metastatic niche formation and metastasis. Compared with patients whose pancreatic tumours did not progress, MIF was markedly higher in exosomes from stage I PDAC patients who later developed liver metastasis. These findings suggest that exosomal MIF primes the liver for metastasis and may be a prognostic marker for the development of PDAC liver metastasis.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Exosomes/metabolism , Liver Neoplasms/pathology , Macrophage Migration-Inhibitory Factors/biosynthesis , Pancreatic Neoplasms/pathology , Animals , Base Sequence , Bone Marrow Cells/immunology , Cell Line, Tumor , Cell Movement , Female , Fibronectins/biosynthesis , Gene Expression Regulation, Neoplastic , Hepatic Stellate Cells/pathology , Humans , Liver/cytology , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Precancerous Conditions/pathology , RNA Interference , RNA, Small Interfering , Sequence Analysis, RNA , Signal Transduction , Transforming Growth Factor beta/metabolism
7.
Nature ; 518(7538): 240-4, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25409150

ABSTRACT

Broad and deep tumour genome sequencing has shed new light on tumour heterogeneity and provided important insights into the evolution of metastases arising from different clones. There is an additional layer of complexity, in that tumour evolution may be influenced by selective pressure provided by therapy, in a similar fashion to that occurring in infectious diseases. Here we studied tumour genomic evolution in a patient (index patient) with metastatic breast cancer bearing an activating PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha, PI(3)Kα) mutation. The patient was treated with the PI(3)Kα inhibitor BYL719, which achieved a lasting clinical response, but the patient eventually became resistant to this drug (emergence of lung metastases) and died shortly thereafter. A rapid autopsy was performed and material from a total of 14 metastatic sites was collected and sequenced. All metastatic lesions, when compared to the pre-treatment tumour, had a copy loss of PTEN (phosphatase and tensin homolog) and those lesions that became refractory to BYL719 had additional and different PTEN genetic alterations, resulting in the loss of PTEN expression. To put these results in context, we examined six other patients also treated with BYL719. Acquired bi-allelic loss of PTEN was found in one of these patients, whereas in two others PIK3CA mutations present in the primary tumour were no longer detected at the time of progression. To characterize our findings functionally, we examined the effects of PTEN knockdown in several preclinical models (both in cell lines intrinsically sensitive to BYL719 and in PTEN-null xenografts derived from our index patient), which we found resulted in resistance to BYL719, whereas simultaneous PI(3)K p110ß blockade reverted this resistance phenotype. We conclude that parallel genetic evolution of separate metastatic sites with different PTEN genomic alterations leads to a convergent PTEN-null phenotype resistant to PI(3)Kα inhibition.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Thiazoles/pharmacology , Alleles , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Class I Phosphatidylinositol 3-Kinases , Drug Resistance, Neoplasm/drug effects , Female , Humans , Loss of Heterozygosity/drug effects , Loss of Heterozygosity/genetics , Mice , Mice, Nude , PTEN Phosphohydrolase/metabolism , Thiazoles/therapeutic use , Xenograft Model Antitumor Assays
8.
PLoS One ; 9(4): e95236, 2014.
Article in English | MEDLINE | ID: mdl-24743474

ABSTRACT

Tetracycline or doxycycline (dox)-regulated control of genetic elements allows inducible, reversible and tissue specific regulation of gene expression in mice. This approach provides a means to investigate protein function in specific cell lineages and at defined periods of development and disease. Efficient and stable regulation of cDNAs or non-coding elements (e.g. shRNAs) downstream of the tetracycline-regulated element (TRE) requires the robust expression of a tet-transactivator protein, commonly the reverse tet-transactivator, rtTA. Most rtTA strains rely on tissue specific promoters that often do not provide sufficient rtTA levels for optimal inducible expression. Here we describe the generation of two mouse strains that enable Cre-dependent, robust expression of rtTA3, providing tissue-restricted and consistent induction of TRE-controlled transgenes. We show that these transgenic strains can be effectively combined with established mouse models of disease, including both Cre/LoxP-based approaches and non Cre-dependent disease models. The integration of these new tools with established mouse models promises the development of more flexible genetic systems to uncover the mechanisms of development and disease pathogenesis.


Subject(s)
Gene Expression Regulation/genetics , Models, Genetic , Repressor Proteins , Response Elements , Transgenes , Animals , Mice , Mice, Transgenic , Repressor Proteins/genetics , Repressor Proteins/metabolism
9.
Oncogene ; 33(4): 440-8, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-23376849

ABSTRACT

Intratumoral heterogeneity within individual breast tumors is a well-known phenomenon that may contribute to drug resistance. This heterogeneity is dependent on several factors, such as types of oncogenic drivers and tumor precursor cells. The purpose of our study was to engineer a mouse mammary tumor model with intratumoral heterogeneity by using defined genetic perturbations. To achieve this, we used mice with knockout (-/-) of Ink4a/Arf, a tumor suppressor locus; these mice are known to be susceptible to non-mammary tumors such as fibrosarcoma. To induce mammary tumors, we retrovirally introduced an oncogene, HRAS(G12V), into Ink4a/Arf(-/-) mammary cells in vitro, and those cells were inoculated into syngeneic mice mammary fat pads. We observed 100% tumorigenesis. The tumors formed were negative for estrogen receptor, progesterone receptor and HER2. Further, they had pathological features similar to those of human triple-negative breast cancer (TNBC) (for example, pushing borders, central necrosis). The tumors were found to be heterogeneous and included two subpopulations: CD49f(-) quiescent cells and CD49f(+)cells. Contrary to our expectation, CD49f(-) quiescent cells had high tumor-initiating potential and CD49f(+)cells had relatively low tumor-initiating potential. Gene expression analysis revealed that CD49f(-) quiescent cells overexpressed epithelial-to-mesenchymal transition-driving genes, reminiscent of tumor-initiating cells and claudin-low breast cancer. Our animal model with intratumoral heterogeneity, derived from defined genetic perturbations, allows us to test novel molecular targeted drugs in a setting that mimics the intratumoral heterogeneity of human TNBC.


Subject(s)
Cell Transformation, Neoplastic/genetics , Integrin alpha6/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Female , Flow Cytometry , Immunohistochemistry , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins p21(ras)/genetics , Real-Time Polymerase Chain Reaction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
10.
Oncogene ; 33(29): 3820-9, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-23975421

ABSTRACT

Helicobacter pylori infection induces chronic inflammation that contributes to gastric tumorigenesis. Tumor necrosis factor (TNF-α) is a proinflammatory cytokine, and polymorphism in the TNF-α gene increases the risk of gastric cancer. We herein investigated the role of TNF-α in gastric tumorigenesis using Gan mouse model, which recapitulates human gastric cancer development. We crossed Gan mice with TNF-α (Tnf) or TNF-α receptor TNFR1 (Tnfrsf1a) knockout mice to generate Tnf-/- Gan and Tnfrsf1a-/- Gan mice, respectively, and examined their tumor phenotypes. Notably, both Tnf-/- Gan mice and Tnfrsf1a-/- Gan mice showed similar, significant suppression of gastric tumor growth compared with control Tnf+/+ or Tnfrsf1a+/+ Gan mice. These results indicate that TNF-α signaling through TNFR1 is important for gastric tumor development. Bone marrow (BM) transplantation experiments showed that TNF-α expressed by BM-derived cells (BMDCs) stimulates the TNFR1 on BMDCs by an autocrine or paracrine manner, which is important for gastric tumor promotion. Moreover, the microarray analysis and colony formation assay indicated that NADPH oxidase organizer 1 (Noxo1) and Gna14 are induced in tumor epithelial cells in a TNF-α-dependent manner, and have an important role in tumorigenicity and tumor-initiating cell property of gastric cancer cells. Accordingly, it is possible that the activation of TNF-α/TNFR1 signaling in the tumor microenvironment promotes gastric tumor development through induction of Noxo1 and Gna14, which contribute to maintaining the tumor cells in an undifferentiated state. The present results indicate that targeting the TNF-α/TNFR1 pathway may be an effective preventive or therapeutic strategy for gastric cancer.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Cell Transformation, Neoplastic/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Hyaluronan Receptors/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Tumor Necrosis Factor-alpha/genetics
11.
Oncogene ; 33(33): 4236-41, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-24336323

ABSTRACT

Chronic inflammation is frequently associated with tumorigenesis in elderly people. By contrast, young people without chronic inflammation often develop tumors considered independent of chronic inflammation but driven instead by mutations. Thus, whether inflammation has a significant role in tumor progression in tumors driven by mutations remains largely unknown. Here we show that TNFα is required for the tumorigenesis of osteosarcoma, the most common tumor in children and adolescents. We show that transplantation of AX osteosarcoma cells, which harbor mutations driving c-Myc overexpression and Ink4a-deficiency, in wild-type mice promotes lethal tumorigenesis accompanied by ectopic bone formation and multiple metastases, phenotypes seen in osteosarcoma patients. Such tumorigenesis was completely abrogated in TNFα-deficient mice. AX cells have the capacity to undergo osteoblastic differentiation; however, that activity was significantly inhibited by TNFα treatment, suggesting that TNFα maintains AX cells in an undifferentiated state. TNFα inhibition of AX cell osteoblastic differentiation occurred through ERK activation, and a pharmacological TNFα inhibitor effectively inhibited both AX cell tumorigenesis and increased osteoblastic gene expression and increased survival of tumor-bearing mice. Lethal tumorigenesis of AX cells was also abrogated in IL-1α/IL-1ß doubly deficient mice. We found that both TNFα and IL-1 maintained AX cells in an undifferentiated state via ERK activation. Thus, inflammatory cytokines are required to promote tumorigenesis even in mutation-induced tumors, and TNFα/IL-1 and ERK may represent therapeutic targets for osteosarcoma.


Subject(s)
Bone Neoplasms/metabolism , Osteosarcoma/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Bone Neoplasms/pathology , Cell Differentiation , Disease Progression , Fibroblasts/physiology , Humans , Interleukin-6/metabolism , MAP Kinase Signaling System , Mice , Mice, Knockout , Neoplasm Transplantation , Osteoblasts/metabolism , Osteosarcoma/pathology , Receptors, Tumor Necrosis Factor/metabolism , Up-Regulation
12.
Br J Cancer ; 110(4): 958-66, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24300972

ABSTRACT

BACKGROUND: Circulating tumour cells (CTCs) have an important role in metastatic processes, but details of their basic characteristics remain elusive. We hypothesised that CD44-expressing CTCs show a mesenchymal phenotype and high potential for survival in hepatocellular carcinoma (HCC). METHODS: Circulating CD44(+)CD90(+) cells, previously shown to be tumour-initiating cells, were sorted from human blood and their genetic characteristics were compared with those of tumour cells from primary tissues. The mechanism underlying the high survival potential of CD44-expressing cells in the circulatory system was investigated in vitro. RESULTS: CD44(+)CD90(+) cells in the blood acquired epithelial-mesenchymal transition, and CD44 expression remarkably increased from the tissue to the blood. In Li7 and HLE cells, the CD44(high) population showed higher anoikis resistance and sphere-forming ability than did the CD44(low) population. This difference was found to be attributed to the upregulation of Twist1 and Akt signal in the CD44(high) population. Twist1 knockdown showed remarkable reduction in anoikis resistance, sphere formation, and Akt signal in HLE cells. In addition, mesenchymal markers and CD44s expression were downregulated in the Twist1 knockdown. CONCLUSIONS: CD44s symbolises the acquisition of a mesenchymal phenotype regulating anchorage-independent capacity. CD44s-expressing tumour cells in peripheral blood are clinically important therapeutic targets in HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Hyaluronan Receptors/metabolism , Liver Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Nuclear Proteins/genetics , Twist-Related Protein 1/genetics , Anoikis/genetics , Apoptosis , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Cell Survival , Down-Regulation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/genetics , Liver Neoplasms/metabolism , Mesoderm/cytology , Nuclear Proteins/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , RNA Interference , RNA, Small Interfering , Thy-1 Antigens/metabolism , Twist-Related Protein 1/biosynthesis
13.
Br J Cancer ; 109(2): 379-86, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23778530

ABSTRACT

BACKGROUND: Multiple early gastric cancers (EGCs) may develop in 6-14% of patients even after achieving curative endoscopic submucosal dissection (ESD); however, a useful biomarker for predicting recurrence is not available. The present study investigated whether the expression of CD44 variant 9 (CD44v9), a functional cancer stem cell marker, in the primary gastric cancer tissue represents an indicator of recurrence. METHODS: Eighty-eight patients who underwent ESD for EGC from 2008 to 2010 were enrolled and monitored for recurrence for 3 years. The expression levels of CD44v9 in the tissue of initial EGCs were evaluated by immunohistochemistry, and the recurrence rate was compared between CD44v9-positive and CD44v9-negative groups. The mucin phenotype and expression of microRNA-21 (miR-21) and programmed cell death protein 4 (PDCD4) were also analysed. RESULTS: The recurrence rate of EGC was significantly higher in the CD44v9-positive group than in the CD44v9-negative group (hazard ratio (HR), 21.8; 95% confidence interval (CI), 5.71-83.1). However, mucin phenotypes and the expression of miR-21 and PDCD4 did not predict recurrence after ESD. Meanwhile, grade of gastric atrophy was also identified as a significant marker of multiple recurrence (HR, 4.95; 95% CI, 1.30-18.8). CONCLUSION: CD44 variant 9 expression represents a potential predictive marker for recurrence in EGC.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor , Hyaluronan Receptors/physiology , Neoplasm Recurrence, Local/diagnosis , Stomach Neoplasms/diagnosis , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Disease Progression , Endoscopy , Female , Gastrectomy/methods , Humans , Hyaluronan Receptors/metabolism , Male , Middle Aged , Prognosis , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Treatment Outcome
14.
Oncogene ; 32(44): 5191-8, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-23334333

ABSTRACT

Increasing evidence indicates that several types of solid tumor are hierarchically organized and sustained by a distinct population of cancer stem cells (CSCs). CSCs possess enhanced mechanisms of protection from stress induced by reactive oxygen species (ROS) that render them resistant to chemo- and radiotherapy. Expression of CD44, especially variant isoforms (CD44v) of this major CSC marker, contributes to ROS defense through upregulation of the synthesis of reduced glutathione (GSH), the primary intracellular antioxidant. CD44v interacts with and stabilizes xCT, a subunit of the cystine-glutamate transporter xc(-), and thereby promotes cystine uptake for GSH synthesis. Given that cancer cells are often exposed to high levels of ROS during tumor progression, the ability to avoid the consequences of such exposure is required for cancer cell survival and propagation in vivo. CSCs, in which defense against ROS is enhanced by CD44v are thus thought to drive tumor growth, chemoresistance and metastasis. Therapy targeted to the CD44v-xCT system may therefore impair the ROS defense ability of CSCs and thereby sensitize them to currently available treatments.


Subject(s)
Hyaluronan Receptors/physiology , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Biological Transport , Cell Hypoxia , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Oxidation-Reduction , Protein Isoforms/physiology , Reactive Oxygen Species/metabolism
15.
Placenta ; 34(2): 110-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23199791

ABSTRACT

Gestational choriocarcinoma is a malignant trophoblastic tumor that usually occurs in the uterus after pregnancy. The tumor is curable with advanced chemotherapy, but the molecular mechanism of choriocarcinoma tumorigenesis remains unclear. This is partly because the low incidence makes it difficult to obtain clinical samples for investigation and because an appropriate choriocarcinoma cell model to study the tumorigenesis has not been developed. We have established a new choriocarcinoma cell line, induced choriocarcinoma cell-1 (iC(3)-1), that possesses unique characteristics compared to other choriocarcinoma cell lines, including production of tumors that consist of the two types of cells commonly found in choriocarcinoma and mimicking of the clinical pathology. Existing trophoblast cell lines utilized in previous choriocarcinoma studies have had significantly dissimilar gene expression profiles. Therefore, it is important to choose an appropriate cell line for a particular study based on the characteristics of the cell line. In this study, to clarify the genetic characteristics of iC(3)-1 and to explore the tumorigenesis mechanism, we examined the gene profile of iC(3)-1 compared to those of existing cell lines and normal placental tissue. Bioinformatics analysis showed that several characteristic genes, IGF1R, CHFR, MUC3A, TAF7, PARK7, CDC123 and PSMD8, were significantly upregulated in iC(3)-1 compared to BeWo and JEG3 cells. Interestingly, HAS2, CD44 and S100P were significantly upregulated in iC(3)-1 compared to parental HTR8/SVneo cells and normal third trimester placenta. Choriocarcinoma samples also showed immunoreactivity to HAS2, CD44 and S100. In summary, the gene expression profile of iC(3)-1 suggests that studies using this cell line can make an important contribution to improved understanding of choriocarcinoma tumorigenesis.


Subject(s)
Choriocarcinoma/genetics , Placenta/metabolism , Uterine Neoplasms/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Choriocarcinoma/metabolism , Choriocarcinoma/pathology , Female , Gene Expression Regulation, Neoplastic , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Hyaluronan Synthases , Intracellular Signaling Peptides and Proteins/genetics , Mucin-3/genetics , Neoplasm Proteins/genetics , Oncogene Proteins/genetics , Poly-ADP-Ribose Binding Proteins , Pregnancy , Proteasome Endopeptidase Complex/genetics , Protein Deglycase DJ-1 , Receptor, IGF Type 1/genetics , S100 Proteins/genetics , S100 Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Transcriptome , Ubiquitin-Protein Ligases , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
16.
Oncogene ; 31(23): 2849-61, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-21986948

ABSTRACT

The cell of origin of tumors and the factors determining the cell of origin remain unclear. In this study, a mouse model of precursor B acute lymphoblastic leukemia/lymphoma (pre-B ALL/LBL) was established by retroviral transduction of Myc genes (N-Myc or c-Myc) into mouse bone marrow cells. Hematopoietic stem cells (HSCs) exhibited the highest susceptibility to N-Myc-induced pre-B ALL/LBL versus lymphoid progenitors, myeloid progenitors and committed progenitor B cells. N-Myc was able to induce pre-B ALL/LBL directly from progenitor B cells in the absence of Ink4a and Arf. Arf was expressed higher in progenitor B cells than Ink4a. In addition, N-Myc induced pre-B ALL/LBL from Arf(-/-) progenitor B cells suggesting that Arf has a predominant role in determining the cell of origin of pre-B ALL/LBL. Tumor cells derived from Ink4a/Arf(-/-) progenitor B cells exhibited a higher rate of proliferation and were more chemoresistant than those derived from wild-type HSCs. Furthermore, the Mdm2 inhibitor Nutlin-3 restored p53 and induced massive apoptosis in mouse pre-B ALL/LBL cells derived from Ink4a/Arf(-/-) cells and human B-ALL cell lines lacking Ink4a and Arf expression, suggesting that Mdm2 inhibition may be a novel therapeutic approach to the treatment of Ink4a/Arf(-/-) B-ALL/LBL, such as is frequently found in Ph(+) ALL and relapsed ALL. Collectively, these findings indicate that Ink4a and Arf are critical determining factors of the cell of origin and the therapeutic sensitivity of Myc-induced lymphoid tumors.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/physiology , Cytarabine/pharmacology , Genes, myc , Imidazoles/pharmacology , Piperazines/pharmacology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Suppressor Protein p14ARF/physiology , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cells, Cultured , Disease Models, Animal , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Knockout , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/drug effects , Precursor Cells, B-Lymphoid/metabolism
17.
Oncogene ; 29(42): 5687-99, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20676132

ABSTRACT

The development of cancer is due to the growth and proliferation of transformed normal cells. Recent evidence suggests that the nature of oncogenic stress and the state of the cell of origin critically affect both tumorigenic activity and tumor histological type. However, this mechanistic relationship in mesenchymal tumors is currently largely unexplored. To clarify these issues, we established a mouse osteosarcoma (OS) model through overexpression of c-MYC in bone marrow stromal cells (BMSCs) derived from Ink4a/Arf (-/-) mice. Single-cell cloning revealed that c-MYC-expressing BMSCs are composed of two distinctly different clones: highly tumorigenic cells, similar to bipotent-committed osteochondral progenitor cells, and low-tumorigenic tripotent cells, similar to mesenchymal stem cells (MSCs). It is noteworthy that both bipotent and tripotent cells were capable of generating histologically similar, lethal OS, suggesting that both committed progenitor cells and MSCs can become OS cells of origin. Shifting mesenchymal differentiation by depleting PPARγ in tripotent MSC-like cells and overexpressing PPARγ in bipotent cells affected cell proliferation and tumorigenic activity. Our findings indicate that differentiation potential has a key role in OS tumorigenic activity, and that the suppression of adipogenic ability is a critical factor for the development of OS.


Subject(s)
Adipogenesis/physiology , Bone Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Osteosarcoma/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Adipocytes/metabolism , Adipocytes/pathology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Differentiation/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression , Gene Expression Profiling , Humans , Immunoblotting , Immunohistochemistry , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteosarcoma/genetics , PPAR gamma/biosynthesis , PPAR gamma/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Stem Cells/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
18.
Oncogene ; 29(13): 1941-51, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20101228

ABSTRACT

Malignant pleural mesothelioma (MPM) is resistant to chemotherapy and thus shows a dismal prognosis. Osteopontin (OPN), a secreted noncollagenous and phosphoprotein, is suggested to be involved in the pathogenesis of MPM. However, the precise role of OPN, especially in the multidrug resistance of MPM, remains to be elucidated. We therefore established stable transfectants (ACC-MESO-1/OPN), which constitutively express OPN, to determine its role in the chemoresistance observed in MPM. The introduction of the OPN gene provides MPM cells with upregulated multidrug resistance through the mechanism of enhanced hyaluronate (HA) binding. The expression of CD44 variant isoforms, which inhibit HA binding, significantly decreased in ACC-MESO-1/OPN cells in comparison to control transfectants. Interestingly, the inhibition of the HA-CD44 interaction abrogated multidrug resistance in the ACC-MESO-1/OPN, thus suggesting the involvement of the surviving signal emanating from the HA-CD44 interaction. An enhanced level of the p-Akt in ACC-MESO-1/OPN cells was observed, and was diminished by CD44 siRNA. Inhibition of the Akt phosphorylation increased in number of the cells underwent apoptosis induced by NVB, VP-16 and GEM. Collectively, these results indicate that OPN is strongly involved in multidrug resistance by enhancing the CD44 binding to HA.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Hyaluronic Acid/pharmacology , Mesothelioma/pathology , Osteopontin/metabolism , Pleural Neoplasms/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronic Acid/metabolism , Mesothelioma/metabolism , Pleural Neoplasms/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
19.
Oncogene ; 28(31): 2806-20, 2009 Aug 06.
Article in English | MEDLINE | ID: mdl-19503101

ABSTRACT

BubR1 is a critical component of the mitotic checkpoint that delays the onset of anaphase until all chromosomes have established bipolar attachment to the microtubules. We previously reported that mutations of the BUB1B gene (encoding BubR1) caused premature chromatid separation (PCS) syndrome, a condition characterized by constitutional aneuploidy and a high risk of childhood cancer. We here report that the cells from PCS syndrome patients have loss of regulation of the centrosome duplication machinery, resulting in centrosome amplification and multipolar mitosis. PCS syndrome cells show increased activity of Polo-like kinase 1 (Plk1), whose knockdown suppresses centrosome amplification. BubR1 localizes to centrosomes, physically interacts with Plk1 and inhibits Plk1 phosphorylation and its kinase activity during interphase. These results unravel a crucial role of BubR1 in preventing centrosome reduplication through negative regulation of Plk1 in interphase cells.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Cell Cycle Proteins/genetics , Cell Line , Chromatids/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HCT116 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Interphase , Microscopy, Fluorescence , Mutation , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spindle Apparatus/metabolism , Syndrome , Transfection , Polo-Like Kinase 1
20.
Oncogene ; 27(31): 4305-14, 2008 Jul 17.
Article in English | MEDLINE | ID: mdl-18372918

ABSTRACT

Aurora A mitotic kinase is frequently overexpressed in various human cancers and is widely considered to be an oncoprotein. However, the cellular contexts in which Aurora A induces malignancy in vivo are still unclear. We previously reported a mouse model in which overexpression of human Aurora A in the mammary gland leads to small hyperplastic changes but not malignancy because of the induction of p53-dependent apoptosis. To study the additional factors required for Aurora A-associated tumorigenesis, we generated a new Aurora A overexpression mouse model that lacks p53. We present evidence here that Aurora A overexpression in primary mouse embryonic fibroblasts (MEFs) that lack p53 overrides postmitotic checkpoint and leads to the formation of multinucleated polyploid cells. Induction of Aurora A overexpression in the mammary glands of p53-deficient mice resulted in development of precancerous lesions that were histologically similar to atypical ductal hyperplasia in human mammary tissue and showed increased cellular senescence and p16 expression. We further observed DNA damage in p53-deficient primary MEFs after Aurora A overexpression. Our results suggest that Aurora A overexpression in mammary glands is insufficient for the development of malignant tumors in p53-deficient mice because of the induction of cellular senescence. Both p53 and p16 are critical in preventing mammary gland tumorigenesis in the Aurora A overexpression mouse model.


Subject(s)
Breast Neoplasms/genetics , Cellular Senescence , Gene Expression Regulation, Neoplastic , Genes, p53 , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Protein Serine-Threonine Kinases/metabolism , Animals , Aurora Kinase A , Aurora Kinases , Breast Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage , Fibroblasts/metabolism , Humans , Mice , Mice, Transgenic , Milk Proteins/metabolism , Neoplasm Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...