Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Cancers (Basel) ; 15(9)2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37174001

ABSTRACT

The tumor microenvironment (TME) plays a key role in cancer development and progression, as well as contributes to the therapeutic resistance and metastasis of cancer cells. The TME is heterogeneous and consists of multiple cell types, including cancer-associated fibroblasts (CAFs), endothelial cells, and immune cells, as well as various extracellular components. Recent studies have revealed cross talk between cancer cells and CAFs as well as between CAFs and other TME cells, including immune cells. Signaling by transforming growth factor-ß, derived from CAFs, has recently been shown to induce remodeling of tumor tissue, including the promotion of angiogenesis and immune cell recruitment. Immunocompetent mouse cancer models that recapitulate interactions of cancer cells with the TME have provided insight into the TME network and support the development of new anticancer therapeutic strategies. Recent studies based on such models have revealed that the antitumor action of molecularly targeted agents is mediated in part by effects on the tumor immune environment. In this review, we focus on cancer cell-TME interactions in heterogeneous tumor tissue, and we provide an overview of the basis for anticancer therapeutic strategies that target the TME, including immunotherapy.

2.
Biomedicines ; 10(12)2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36551889

ABSTRACT

We previously established mouse models of biliary tract cancer (BTC) based on the injection of cells with biliary epithelial stem cell properties derived from KRAS(G12V)-expressing organoids into syngeneic mice. The resulting mouse tumors appeared to recapitulate the pathological features of human BTC. Here we analyzed images of hematoxylin and eosin (H&E) staining for both the mouse tumor tissue and human cholangiocarcinoma tissue by pixel-level clustering with machine learning. A pixel-clustering model that was established via training with mouse images revealed homologies of tissue structure between the mouse and human tumors, suggesting similarities in tumor characteristics independent of animal species. Analysis of the human cholangiocarcinoma tissue samples with the model also revealed that the entropy distribution of cancer regions was higher than that of noncancer regions, with the entropy of pixels thus allowing discrimination between these two types of regions. Histograms of entropy tended to be broader for noncancer regions of late-stage human cholangiocarcinoma. These analyses indicate that our mouse BTC models are appropriate for investigation of BTC carcinogenesis and may support the development of new therapeutic strategies. In addition, our pixel-level clustering model is highly versatile and may contribute to the development of a new BTC diagnostic tool.

3.
Cancers (Basel) ; 14(10)2022 May 12.
Article in English | MEDLINE | ID: mdl-35625983

ABSTRACT

Germline mutations of NF1 cause neurofibromatosis type 1 (NF1) through the activation of the RAS signaling pathway, and some NF1 patients develop malignant peripheral nerve sheath tumors (MPNSTs). Here, we established subclones of the human NF1-MPNST cell line sNF96.2 that manifest increased tumorigenic activity and increased phosphorylation of the protein kinases MEK and Akt relative to the parental cells. Genomic DNA sequencing identified 14 additional heterozygous mutations within the coding regions of 13 cancer- and other disease-related genes in these subclones. One of these genes, PTPN11, encodes SHP-2, and the forced expression of the identified G503V mutant of SHP-2 increased both tumorigenic activity and MEK phosphorylation in parental sNF96.2 cells, suggesting that the combination of PTPN11 and NF1 mutations induces the pathological activation of the RAS pathway. These effects of SHP-2 (G503V) were inhibited by the coexpression of the G370A mutant of BRAP, which was also detected in the highly malignant subclones, and this inhibition was accompanied by the calpain-dependent cleavage of SHP-2 (G503V). The cleavage of SHP-2 (G503V) and suppression of MEK phosphorylation mediated by BRAP (G370A) were not detected in NF1-intact (HeLa) cells. Tumor promotion by SHP-2 (G503V) and its suppression by BRAP (G370A) may serve as a basis for the development of new treatment strategies for NF1.

4.
Cancer Res ; 81(18): 4751-4765, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34289987

ABSTRACT

Invasive lung adenocarcinoma (LADC) can be classified histologically as lepidic, acinar, papillary, micropapillary, or solid. Most LADC tumors manifest several of these histological subtypes, with heterogeneity being related to therapeutic resistance. We report here that in immunodeficient mice, human LADC cells form tumors with distinct histological features, MUC5AC-expressing solid-type or cytokeratin 7 (CK7)-expressing acinar-type tumors, depending on the site of development, and that a solid-to-acinar transition (SAT) could be induced by the tumor microenvironment. The TGFß-Smad signaling pathway was activated in both tumor and stromal cells of acinar-type tumors. Immortalized cancer-associated fibroblasts (CAF) derived from acinar-type tumors induced SAT in 3D cocultures with LADC cells. Exogenous TGFß1 or overexpression of an active form of TGFß1 increased CK7 expression and reduced MUC5AC expression in LADC cells, and knockdown of Tgfb1 mRNA in CAFs attenuated SAT induction. RNA-sequencing analysis suggested that angiogenesis and neutrophil recruitment are associated with SAT in vivo. Our data indicate that CAF-mediated paracrine TGFß signaling induces remodeling of tumor tissue and determines the histological pattern of LADC, thereby contributing to tumor heterogeneity. SIGNIFICANCE: CAFs secrete TGFß to induce a solid-to-acinar transition in lung cancer cells, demonstrating how the tumor microenvironment influences histological patterns and tumor heterogeneity in lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Cancer-Associated Fibroblasts/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Adenocarcinoma of Lung/etiology , Animals , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Disease Models, Animal , Female , Fluorescent Antibody Technique , Heterografts , Humans , Immunohistochemistry , Interleukin-8/genetics , Interleukin-8/metabolism , Mice , Models, Biological , Neoplasm Grading
5.
Cancer Sci ; 112(5): 1822-1838, 2021 May.
Article in English | MEDLINE | ID: mdl-33068050

ABSTRACT

Biliary tract cancer (BTC) arises from biliary epithelial cells (BECs) and includes intrahepatic cholangiocarcinoma (IHCC), gallbladder cancer (GC), and extrahepatic cholangiocarcinoma (EHCC). Although frequent KRAS mutations and epigenetic changes at the INK4A/ARF locus have been identified, the molecular pathogenesis of BTC is unclear and the development of corresponding anticancer agents remains inadequate. We isolated epithelial cell adhesion molecule (EpCAM)-positive BECs from the mouse intrahepatic bile duct, gallbladder, and extrahepatic bile duct, and established organoids derived from these cells. Introduction of activated KRAS and homozygous deletion of Ink4a/Arf in the cells of each organoid type conferred the ability to form lethal metastatic adenocarcinoma with differentiated components and a pronounced desmoplastic reaction on cell transplantation into syngeneic mice, indicating that the manipulated cells correspond to BTC-initiating cells. The syngeneic mouse models recapitulate the pathological features of human IHCC, GC, and EHCC, and they should therefore prove useful for the investigation of BTC carcinogenesis and the development of new therapeutic strategies. Tumor cells isolated from primary tumors formed organoids in three-dimensional culture, and serial syngeneic transplantation of these cells revealed that their cancer stem cell properties were supported by organoid culture, but not by adherent culture. Adherent culture thus attenuated tumorigenic activity as well as the expression of both epithelial and stem cell markers, whereas the expression of epithelial-mesenchymal transition (EMT)-related transcription factor genes and mesenchymal cell markers was induced. Our data show that organoid culture is important for maintenance of epithelial cell characteristics, stemness, and tumorigenic activity of BTC-initiating cells.


Subject(s)
Biliary Tract Neoplasms/genetics , Cholangiocarcinoma/genetics , Epithelial Cells/physiology , Genes, ras , Organoids , Stem Cells/physiology , ADP-Ribosylation Factor 1/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Extrahepatic/anatomy & histology , Bile Ducts, Extrahepatic/cytology , Bile Ducts, Intrahepatic/cytology , Biliary Tract Neoplasms/pathology , Cholangiocarcinoma/pathology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Models, Animal , Epithelial Cell Adhesion Molecule , Epithelial Cells/chemistry , Epithelial-Mesenchymal Transition , Female , Gallbladder/anatomy & histology , Gallbladder/cytology , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/pathology , Gene Deletion , Genes, Tumor Suppressor , Liver/anatomy & histology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation/methods , Organoids/metabolism , Organoids/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Tissue Array Analysis/methods , Tumor Microenvironment/physiology
6.
Cancers (Basel) ; 12(12)2020 Dec 17.
Article in English | MEDLINE | ID: mdl-33348616

ABSTRACT

Somatic mutations in EGFR and KRAS as well as chromosome rearrangements affecting ALK, ROS1, and RET have been identified in human lung adenocarcinoma (LUAD). We here developed organoid-based orthotopic and syngeneic mouse models for studies of the pathogenesis and treatment of LUAD. We isolated EpCAM-positive epithelial cells from mouse lungs and cultured them as organoids to maintain epithelial stem cell properties. These cells were transformed by KRAS(G12V) or EML4-ALK and then transplanted via the trachea into the lungs of the syngeneic mice, where they formed tumors that expressed the lung lineage marker TTF-1 and which closely recapitulated the pathology of human LUAD. Treatment with crizotinib suppressed the growth of tumors formed by the EML4-ALK-expressing lung epithelial cells in a subcutaneous transplantation model. Organoid culture of normal lung epithelial cells resulted in enrichment of EpCAM+SCA-1(Ly6a)+ cells as well as in that of cells expressing another member of the Ly6 protein family, Ly6d, which was found to be required for the growth of the LUAD-initiating cells expressing KRAS(G12V) or EML4-ALK. We also found that a high expression level of LY6D was associated with poor prognosis in human LUAD. Our results thus suggest that LY6D is a potential lung cancer stem cell marker.

7.
Cancer Metastasis Rev ; 39(3): 711-720, 2020 09.
Article in English | MEDLINE | ID: mdl-32399646

ABSTRACT

Metastasis of cancer cells to the brain occurs frequently in patients with certain subtypes of breast cancer. In particular, patients with HER2-positive or triple-negative breast cancer are at high risk for the development of brain metastases. Despite recent advances in the treatment of primary breast tumors, the prognosis of breast cancer patients with brain metastases remains poor. A better understanding of the molecular and cellular mechanisms underlying brain metastasis might be expected to lead to improvements in the overall survival rate for these patients. Recent studies have revealed complex interactions between metastatic cancer cells and their microenvironment in the brain. Such interactions result in the activation of various signaling pathways related to metastasis in both cancer cells and cells of the microenvironment including astrocytes and microglia. In this review, we focus on such interactions and on their role both in the metastatic process and as potential targets for therapeutic intervention.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Animals , Brain Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Female , Humans , Molecular Targeted Therapy , Receptor, ErbB-2/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
8.
Cancer Sci ; 111(8): 2689-2695, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32462706

ABSTRACT

Chemoresistance is a hallmark of cancer stem cells (CSCs). To develop novel therapeutic strategies that target CSCs, we established osteosarcoma-initiating (OSi) cells by introducing the c-Myc gene into bone marrow stromal cells derived from Ink4a/Arf KO mice. These OSi cells include bipotent committed cells (similar to osteochondral progenitor cells) with a high tumorigenic activity as well as tripotent cells (similar to mesenchymal stem cells) of low tumorigenicity. We recently showed that the tripotent OSi cells are highly resistant to chemotherapeutic agents, and that depolymerization of the actin cytoskeleton in these cells induces their terminal adipocyte differentiation and suppresses their tumorigenicity. We here provide an overview of modulation of actin cytoskeleton dynamics associated with terminal adipocyte differentiation in osteosarcoma as well as discuss the prospects for new therapeutic strategies that target chemoresistant CSCs by inducing their differentiation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/drug effects , Cell Differentiation/drug effects , Neoplastic Stem Cells/drug effects , Osteosarcoma/drug therapy , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/pathology , Adipocytes/drug effects , Adipocytes/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Genetic Heterogeneity , Humans , Mice , Neoplastic Stem Cells/pathology , Osteosarcoma/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
9.
Int J Mol Sci ; 19(8)2018 Jul 24.
Article in English | MEDLINE | ID: mdl-30042341

ABSTRACT

Intratumoral human epidermal growth factor receptor 2 (HER2) heterogeneity has been reported in 16⁻36% of HER2-positive breast cancer and its clinical impact is under discussion. We examined the biological effects of HER2-heterogeneity on mouse models and analyzed metastatic brains by RNA sequence analysis. A metastatic mouse model was developed using 231-Luc (triple negative cells) and 2 HER2-positive cell lines, namely, HER2-60 and HER2-90 which showed heterogeneous and monotonous HER2 expressions, respectively. Metastatic lesions developed in 3 weeks in all the mice injected with HER2-60 cells, and in 69% of the mice injected with HER2-90 and 87.5% of the mice injected with 231-Luc. The median survival days of mice injected with 231-Luc, HER2-60, and HER2-90 cells were 29 (n = 24), 24 (n = 22) and 30 (n = 13) days, respectively. RNA sequence analysis showed that CASP-1 and its related genes were significantly downregulated in metastatic brain tumors with HER2-60 cells. The low expression of caspase-1 could be a new prognostic biomarker for early relapse in HER2-positive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/mortality , Genetic Heterogeneity , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Caspase 1/genetics , Caspase 1/metabolism , Female , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Prognosis , Recurrence , Sequence Analysis, RNA
10.
Sci Rep ; 8(1): 6069, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29666462

ABSTRACT

Neurofibromatosis type 1 (NF1) is caused by germline mutations in the NF1 gene and is characterized by café au lait spots and benign tumours known as neurofibromas. NF1 encodes the tumour suppressor protein neurofibromin, which negatively regulates the small GTPase Ras, with the constitutive activation of Ras signalling resulting from NF1 mutations being thought to underlie neurofibroma development. We previously showed that knockdown of neurofibromin triggers epithelial-mesenchymal transition (EMT) signalling and that such signalling is activated in NF1-associated neurofibromas. With the use of a cell-based drug screening assay, we have now identified the antiallergy drug tranilast (N-(3,4-dimethoxycinnamoyl) anthranilic acid) as an inhibitor of EMT and found that it attenuated the expression of mesenchymal markers and angiogenesis-related genes in NF1-mutated sNF96.2 cells and in neurofibroma cells from NF1 patients. Tranilast also suppressed the proliferation of neurofibromin-deficient cells in vitro more effectively than it did that of intact cells. In addition, tranilast inhibited sNF96.2 cell migration and proliferation in vivo. Knockdown of type III collagen (COL3A1) also suppressed the proliferation of neurofibroma cells, whereas expression of COL3A1 and SOX2 was increased in tranilast-resistant cells, suggesting that COL3A1 and the transcription factor SOX2 might contribute to the development of tranilast resistance.


Subject(s)
Anti-Allergic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Neovascularization, Physiologic/drug effects , Neurofibromin 1/genetics , ortho-Aminobenzoates/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Gene Deletion , Genes, Neurofibromatosis 1/drug effects , Germ-Line Mutation , HeLa Cells , Humans , Mice, SCID , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/prevention & control , Neoplasms/drug therapy , Neoplasms/genetics , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics
11.
Cancer Sci ; 109(5): 1447-1454, 2018 May.
Article in English | MEDLINE | ID: mdl-29498146

ABSTRACT

Metastasis is the leading cause of cancer death. A tumor-supportive microenvironment, or premetastatic niche, at potential secondary tumor sites plays an important role in metastasis, especially in tumor cell colonization. Although a fibrotic milieu is known to promote tumorigenesis and metastasis, the underlying molecular contributors to this effect have remained unclear. Here we show that periostin, a component of the extracellular matrix that functions in tissue remodeling, has a key role in formation of a fibrotic environment that promotes tumor metastatic colonization. We found that periostin was widely expressed in fibrotic lesions of mice with bleomycin-induced lung fibrosis, and that up-regulation of periostin expression coincided with activation of myofibroblasts positive for α-smooth muscle actin. We established a lung metastasis model for B16 murine melanoma cells and showed that metastatic colonization of the lung by these cells was markedly promoted by bleomycin-induced lung fibrosis. Inhibition of periostin expression by giving an intratracheal antisense oligonucleotide targeting periostin mRNA was found to suppress bleomycin-induced lung fibrosis and thereby to attenuate metastatic colonization of the lung by melanoma cells. Our results indicate that periostin is a key player in the development of bleomycin-induced fibrosis and consequent enhancement of tumor cell colonization in the lung. Our results therefore implicate periostin as a potential target for prevention or treatment of lung metastasis.


Subject(s)
Bleomycin/adverse effects , Cell Adhesion Molecules/antagonists & inhibitors , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Oligonucleotides, Antisense/administration & dosage , Pulmonary Fibrosis/therapy , Actins/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Extracellular Matrix/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Oligonucleotides, Antisense/pharmacology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Tumor Microenvironment , Up-Regulation , Xenograft Model Antitumor Assays
12.
Dev Dyn ; 247(3): 462-472, 2018 03.
Article in English | MEDLINE | ID: mdl-28960588

ABSTRACT

Lung cancer is the most common cancer worldwide. Treatment options for lung cancer include surgery, radiation therapy, chemotherapy, molecularly targeted therapy including epidermal growth factor receptor or anaplastic lymphoma kinase inhibitors, and immunotherapy. These treatments can be administered alone or in combination. Despite therapeutic advances, however, lung cancer remains the leading cause of cancer death. Recent studies have indicated that epithelial-mesenchymal transition (EMT) is associated with malignancy in various types of cancer, and activation of EMT signaling in cancer cells is widely considered to contribute to metastasis, recurrence, or therapeutic resistance. In this review, we provide an overview of the role of EMT in the progression of lung cancer. We also discuss the prospects for new therapeutic strategies that target EMT signaling in lung cancer. Developmental Dynamics 247:462-472, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Epithelial-Mesenchymal Transition , Lung Neoplasms/drug therapy , Signal Transduction/drug effects , Animals , Disease Progression , Humans
13.
Cancer Med ; 6(11): 2635-2645, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28980419

ABSTRACT

Therapeutic options for malignant brain tumors are limited, with new drugs being continuously evaluated. Organotypic brain slice culture has been adopted for neuroscience studies as a system that preserves brain architecture, cellular function, and the vascular network. However, the suitability of brain explants for anticancer drug evaluation has been unclear. We here adopted a mouse model of malignant glioma based on expression of H-RasV12 in Ink4a/Arf-/- neural stem/progenitor cells to establish tumor-bearing brain explants from adult mice. We treated the slices with cisplatin, temozolomide, paclitaxel, or tranilast and investigated the minimal assays required to assess drug effects. Serial fluorescence-based tumor imaging was sufficient for evaluation of cisplatin, a drug with a pronounced cytotoxic action, whereas immunostaining of cleaved caspase 3 (a marker of apoptosis) and of Ki67 (a marker of cell proliferation) was necessary for the assessment of temozolomide action and immunostaining for phosphorylated histone H3 (a marker of mitosis) allowed visualization of paclitaxel-specific effects. Staining for cleaved caspase 3 was also informative in the assessment of drug toxicity for normal brain tissue. Incubation of explants with fluorescently labeled antibodies to CD31 allowed real-time imaging of the microvascular network and complemented time-lapse imaging of tumor cell invasion into surrounding tissue. Our results suggest that a combination of fluorescence imaging and immunohistological staining allows a unified assessment of the effects of various classes of drug on the survival, proliferation, and invasion of glioma cells, and that organotypic brain slice culture is therefore a useful tool for evaluation of antiglioma drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Drug Screening Assays, Antitumor/methods , Glioma/drug therapy , Tissue Culture Techniques , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain Neoplasms/pathology , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Disease Models, Animal , Fluorescent Antibody Technique , Genes, ras , Glioma/pathology , Immunohistochemistry , Ki-67 Antigen/metabolism , Mice , Microscopy, Confocal , Neoplasm Invasiveness , Neural Stem Cells , Paclitaxel/pharmacology , Spheroids, Cellular/drug effects , Temozolomide , ortho-Aminobenzoates/pharmacology
15.
Biomed Res Int ; 2017: 8032910, 2017.
Article in English | MEDLINE | ID: mdl-28210624

ABSTRACT

Metastasis is the main cause of treatment failure and death in cancer patients. Metastasis of tumor cells to the brain occurs frequently in individuals with breast cancer, non-small cell lung cancer, or melanoma. Despite recent advances in our understanding of the causes and in the treatment of primary tumors, the biological and molecular mechanisms underlying the metastasis of cancer cells to the brain have remained unclear. Metastasizing cancer cells interact with their microenvironment in the brain to establish metastases. We have now developed mouse models of brain metastasis based on intracardiac injection of human breast cancer or melanoma cell lines, and we have performed RNA sequencing analysis to identify genes in mouse brain tissue and the human cancer cells whose expression is associated specifically with metastasis. We found that the expressions of the mouse genes Tph2, Sspo, Ptprq, and Pole as well as those of the human genes CXCR4, PLLP, TNFSF4, VCAM1, SLC8A2, and SLC7A11 were upregulated in brain tissue harboring metastases. Further characterization of such genes that contribute to the establishment of brain metastases may provide a basis for the development of new therapeutic strategies and consequent improvement in the prognosis of cancer patients.


Subject(s)
Brain Neoplasms/genetics , Breast Neoplasms/genetics , Melanoma/genetics , Neoplasm Proteins/biosynthesis , Animals , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Melanoma/pathology , Mice , Neoplasm Metastasis , Neoplasm Proteins/genetics , Prognosis , Sequence Analysis, RNA , Tumor Microenvironment/genetics
16.
Genes Cells ; 21(11): 1244-1252, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27658801

ABSTRACT

Phyllodes tumors (PTs) are rare fibroepithelial tumors of the breast with epithelial and stromal components, and surgical resection is the standard and only available treatment for malignant PTs. To provide a better understanding of these tumors, we developed mouse models that recapitulate the pathological and clinical properties of human malignant PTs. Mouse undifferentiated mammary gland cells were infected with a retrovirus encoding the human oncoprotein H-RasG12V , and the infected cells were transplanted orthotopically into the mammary fat pads of syngeneic mice. The transplanted cells showed a high tumorigenic activity, with the resulting tumors manifesting pathological characteristics including stromal overgrowth similar to those of human malignant PTs. The tumors also showed high rates of both local recurrence and lung metastasis. Our models may prove useful for studies of the pathophysiology of malignant PTs as well as facilitate the development of new treatments.


Subject(s)
Disease Models, Animal , Genes, ras , Mammary Glands, Animal/cytology , Phyllodes Tumor , Animals , Cell Differentiation , Cell Transplantation , Female , Gene Expression , Gene Transfer Techniques , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/virology , Mice , Mice, Inbred C57BL , Phyllodes Tumor/genetics , Phyllodes Tumor/pathology , Retroviridae/genetics
17.
Stem Cells ; 34(8): 1997-2007, 2016 08.
Article in English | MEDLINE | ID: mdl-27251010

ABSTRACT

Cancer stem cells (CSCs) constitute a small subpopulation of cancer cells with stem-like properties that are able to self-renew, generate differentiated daughter cells, and give rise to heterogeneous tumor tissue. Tumor heterogeneity is a hallmark of cancer and underlies resistance to anticancer therapies and disease progression. The epithelial-mesenchymal transition (EMT) is a reversible phenomenon that is mediated by EMT-inducing transcription factors (EMT-TFs) and plays an important role in normal organ development, wound healing, and the invasiveness of cancer cells. Recent evidence showing that overexpression of several EMT-TFs is associated with stemness in cancer cells has suggested the existence of a link between EMT and CSCs. In this review, we focus on the roles of CSCs and EMT signaling in driving tumor heterogeneity. A better understanding of the dynamics of both CSCs and EMT-TFs in the generation of tumor heterogeneity may provide a basis for the development of new treatment options for cancer patients. Stem Cells 2016;34:1997-2007.


Subject(s)
Epithelial-Mesenchymal Transition , Molecular Targeted Therapy , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Humans , Models, Biological , Signal Transduction
18.
Sci Rep ; 6: 24413, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27074716

ABSTRACT

Marfan syndrome (MFS) is an autosomal dominant genetic disease caused by abnormal formation of the extracellular matrix with an incidence of 1 in 3, 000 to 5, 000. Patients with Marfan syndrome experience poor quality of life caused by skeletal disorders such as scoliosis, and they are at high risk of sudden death from cardiovascular impairment. Suitable animal models of MFS are essential for conquering this intractable disease. In particular, studies employing pig models will likely provide valuable information that can be extrapolated to humans because of the physiological and anatomical similarities between the two species. Here we describe the generation of heterozygous fibrillin-1 (FBN1) mutant cloned pigs (+/Glu433AsnfsX98) using genome editing and somatic cell nuclear transfer technologies. The FBN1 mutant pigs exhibited phenotypes resembling those of humans with MFS, such as scoliosis, pectus excavatum, delayed mineralization of the epiphysis and disrupted structure of elastic fibres of the aortic medial tissue. These findings indicate the value of FBN1 mutant pigs as a model for understanding the pathogenesis of MFS and for developing treatments.


Subject(s)
Fibrillin-1/genetics , Heterozygote , Organisms, Genetically Modified , Swine/genetics , Animals , Clone Cells , Disease Models, Animal , Fibroblasts/physiology , Gene Editing , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Nuclear Transfer Techniques
19.
Genet Test Mol Biomarkers ; 18(11): 722-35, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25325900

ABSTRACT

AIMS: We assessed the validity of a next-generation sequencing protocol using in-solution hybridization-based enrichment to identify NF1 mutations for the diagnosis of 86 patients with a prototypic genetic syndrome, neurofibromatosis type 1. In addition, other causative genes for classic genetic syndromes were set as the target genes for coverage analysis. RESULTS: The protocol identified 30 nonsense, 19 frameshift, and 8 splice-site mutations, together with 10 nucleotide substitutions that were previously reported to be pathogenic. In the remaining 19 samples, 10 had single-exon or multiple-exon deletions detected by a multiplex ligation-dependent probe amplification method and 3 had missense mutations that were not observed in the normal Japanese SNP database and were predicted to be pathogenic. Coverage analysis of the genes other than the NF1 gene included on the same diagnostic panel indicated that the mean coverage was 115-fold, a sufficient depth for mutation detection. CONCLUSIONS: The overall mutation detection rate using the currently reported method in 86 patients who met the clinical diagnostic criteria was 92.1% (70/76) when 10 patients with large deletions were excluded. The results validate the clinical utility of this next-generation sequencing-based method for the diagnosis of neurofibromatosis type 1. Comparable detection rates can be expected for other genetic syndromes, based on the results of the coverage analysis.


Subject(s)
Exons , Genes, Neurofibromatosis 1 , High-Throughput Nucleotide Sequencing , Molecular Diagnostic Techniques/methods , Mutation , Neurofibromatosis 1/genetics , DNA Mutational Analysis/methods , Female , Humans , Male
20.
Cancer Sci ; 105(7): 779-87, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24754246

ABSTRACT

Human epidermal growth factor receptor 2 (HER2)-positive breast cancer is treated with HER2-targeted agents, such as trastuzumab and lapatinib, that suppress signaling by phosphatidylinositol 3-kinase (PI3K)-Akt and MAPK pathways. However, resistance to HER2-targeted therapy remains a major clinical problem. Overexpression of CD24 has been detected in many cancers and is associated with a poor prognosis in women with breast cancer. HER2-positive breast tumors are predominantly positive for CD24, suggesting that the expression of the two molecules is related. To investigate the relation between HER2 and CD24, we overexpressed HER2 in breast cancer cells that were triple-negative for the estrogen receptor, progesterone receptor and HER2. We found that expression of CD24 was increased by stable overexpression of HER2. Flow cytometry thus revealed that the percentage of CD24-positive cells was markedly higher in the HER2-positive fraction than in the HER2-negative fraction. Knockdown of CD24 in breast cancer cells positive for endogenous HER2 downregulated HER2 expression, whereas knockdown of HER2 did not affect the expression of CD24. Knockdown of CD24 also suppressed the phosphorylation of Akt, which functions downstream of HER2 and PI3K to promote cell survival. Moreover, knockdown of CD24 increased the sensitivity of HER2-positive breast cancer cells to lapatinib treatment. Our results thus indicate that CD24 supports both the expression of HER2 and the consequent activation of PI3K-Akt signaling. Furthermore, CD24 may contribute to resistance to HER2-targeted therapy and, therefore, is itself a potential therapeutic target in HER2-positive breast cancer.


Subject(s)
Breast Neoplasms/metabolism , CD24 Antigen/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor/drug effects , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lapatinib , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL