144 results on '"Ibaragi S"'
Search Results
102. Anti-EGFR antibody cetuximab is secreted by oral squamous cell carcinoma and alters EGF-driven mesenchymal transition.
- Author
-
Fujiwara T, Eguchi T, Sogawa C, Ono K, Murakami J, Ibaragi S, Asaumi JI, Okamoto K, Calderwood SK, and Kozaki KI
- Subjects
- Antibodies, Monoclonal, Humanized metabolism, Carcinoma, Squamous Cell pathology, Cell Line, Tumor, Cetuximab metabolism, Drug Resistance, Neoplasm drug effects, Epidermal Growth Factor metabolism, Epithelial-Mesenchymal Transition drug effects, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Extracellular Vesicles drug effects, Extracellular Vesicles metabolism, Humans, Mouth Neoplasms pathology, Antibodies, Monoclonal, Humanized pharmacology, Carcinoma, Squamous Cell metabolism, Cetuximab pharmacology, Epidermal Growth Factor antagonists & inhibitors, Mouth Neoplasms metabolism
- Abstract
Genetic amplification, overexpression, and increased signaling from the epidermal growth factor receptor (EGFR) are often found in oral squamous cell carcinoma (OSCC) and thus EGFR is frequently targeted molecularly by the therapeutic antibody cetuximab. We assessed effects of cetuximab in control of EGF-driven malignant traits of OSCC cells. EGF stimulation promoted progression level of mesenchymal traits in OSCC cells, which were attenuated by cetuximab but incompletely. We pursued a potential mechanism underlying such incomplete attenuation of OSCC malignant traits. Cetuximab promoted secretion of EGFR-EVs by OSCC cells and failed to inhibit EGF-driven secretion of EGFR-EVs. Cetuximab was also found to be robustly secreted with the EGFR-EVs by the OSCC cells. Thus, EGF promotes the level of mesenchymal traits of OSCC cells and secretion of EGFR-EVs, which involve cetuximab resistance., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
103. Low-intensity pulsed ultrasound stimulation promotes osteoblast differentiation through hedgehog signaling.
- Author
-
Matsumoto K, Shimo T, Kurio N, Okui T, Ibaragi S, Kunisada Y, Obata K, Masui M, Pai P, Horikiri Y, Yamanaka N, Takigawa M, and Sasaki A
- Subjects
- Animals, Male, Mice, Osteoblasts cytology, Zinc Finger Protein GLI1 metabolism, Zinc Finger Protein Gli2 metabolism, Cell Differentiation, Hedgehog Proteins metabolism, Osteoblasts metabolism, Signal Transduction, Ultrasonic Waves
- Abstract
Low-intensity pulsed ultrasound (LIPUS) has been used as an adjunct to fracture healing therapies, but the mechanisms underlying its action are not known. We reported that sonic hedgehog (SHH) signaling was activated in osteoblasts at the dynamic remodeling site of a bone fracture. Mechanical stimulation is a crucial factor in bone remodeling, and it is related to the primary cilia as a sensor of hedgehog signaling. Here we observed that LIPUS promoted callus formation in accord with Gli2-positive cells after 14 days at the mouse femur fractured site compared with a control group. An immunofluorescence analysis showed that the numbers of primary cilia and cilia/osterix double-positive osteoblasts were increased at the fracture site by LIPUS. LIPUS stimulated not only the number and the length of primary cilia, but also the levels of ciliated protein, Ift88 mRNA, and SHH, Gli1, and Gli2 in MC3T3-E1 cells. Further experiments revealed that LIPUS stimulated osteogenic differentiation in the presence of smoothened agonist (SAG) treatment. These results indicate that LIPUS stimulates osteogenic differentiation and the maturation of osteoblasts by a primary cilium-mediated activation of hedgehog signaling., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
104. Ammonium tetrathiomolybdate enhances the antitumor effects of cetuximab via the suppression of osteoclastogenesis in head and neck squamous carcinoma.
- Author
-
Morisawa A, Okui T, Shimo T, Ibaragi S, Okusha Y, Ono M, Nguyen TTH, Hassan NMM, and Sasaki A
- Subjects
- Animals, Antineoplastic Agents, Immunological therapeutic use, Bone Neoplasms pathology, Bone Neoplasms secondary, Bone Resorption pathology, Carcinoma, Squamous Cell pathology, Cetuximab pharmacology, Cetuximab therapeutic use, Chelating Agents therapeutic use, Drug Synergism, Female, Head and Neck Neoplasms pathology, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Nude, Molybdenum pharmacology, Molybdenum therapeutic use, Neoplasm Invasiveness pathology, Osteoclasts physiology, Protein-Lysine 6-Oxidase metabolism, RANK Ligand metabolism, Squamous Cell Carcinoma of Head and Neck, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Agents, Immunological pharmacology, Bone Neoplasms drug therapy, Bone Resorption drug therapy, Carcinoma, Squamous Cell drug therapy, Chelating Agents pharmacology, Copper metabolism, Head and Neck Neoplasms drug therapy, Osteoclasts drug effects
- Abstract
Head and neck squamous cell carcinoma (HNSCC) poses a significant challenge clinically where one of the mechanisms responsible for the invasion into facial bones occurs via the activation of osteoclasts. Copper has been demonstrated to play a key role in skeletal remodeling. However, the role of copper in cancer-associated bone destruction is thus far unknown. Lysyl oxidase (LOX) is a copper-dependent enzyme that promotes osteoclastogenesis. In the present study, we investigated the effects of copper on HNSCC with bone invasion by the copper chelator, ammonium tetrathiomolybdate (TM) in vitro and in vivo. We demonstrate that TM blocks the proliferation of HNSCC cells, inhibits LOX activation and decreases the expression of the receptor activator of nuclear factor-κB ligand (RANKL) in osteoblasts and osteocytes, subsequently suppressing bone destruction. These findings suggest that copper is a potential target for the treatment of HNSCCs associated with bone destruction.
- Published
- 2018
- Full Text
- View/download PDF
105. The Prognostic Implications of Bone Invasion in Gingival Squamous Cell Carcinoma.
- Author
-
Yoshida S, Shimo T, Murase Y, Takabatake K, Kishimoto K, Ibaragi S, Yoshioka N, Okui T, Nagatsuka H, and Sasaki A
- Subjects
- Adult, Aged, Aged, 80 and over, Bone Neoplasms surgery, Carcinoma, Squamous Cell surgery, Female, Follow-Up Studies, Gingival Neoplasms surgery, Humans, Male, Middle Aged, Neoplasm Invasiveness, Neoplasm Recurrence, Local surgery, Prognosis, Retrospective Studies, Survival Rate, Bone Neoplasms secondary, Carcinoma, Squamous Cell pathology, Gingival Neoplasms pathology, Neoplasm Recurrence, Local pathology
- Abstract
Background/aim: This study evaluated the associations between bone invasion of gingival squamous cell carcinoma (SCC) and clinicopathological manifestations, and aimed to determine whether bone invasion is an independent prognostic factor in gingival SCC., Patients and Methods: The study was a retrospective review of 78 patients with gingival SCC who underwent surgery with curative intent. The level of bone invasion was pathologically categorized as medullary, cortical or no bone invasion., Results: Cortical and medullary bone invasion was present in 29 and 22 patients, respectively. There was a significant association between medullary bone invasion and tumor size (p=0.017), pathological N classification (p<0.001), differentiation (p=0.017) and lymphovascular invasion (p=0.007). Medullary bone invasion and lymphovascular invasion were independent predictors of reduced overall survival (p=0.015, 0.048); medullary bone invasion was also an independent predictor of reduced disease-specific survival (p=0.018)., Conclusion: Pathologically-proven medullary bone invasion and lymphovascular invasion were found to be key prognostic factors in gingival SCC. The results suggest that it is necessary to consider adjuvant therapy in patients with medullary bone invasion., (Copyright© 2018, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
106. Palonosetron Prevents Highly Emetogenic Chemotherapy-induced Nausea and Vomiting in Oral Cancer Patients.
- Author
-
Sento S, Kitamura N, Yamamoto T, Nakashiro K, Hamakawa H, Ibaragi S, Sasaki A, Takamaru N, Miyamoto Y, Kodani I, Ryoke K, Mishima K, and Ueyama Y
- Subjects
- Administration, Intravenous, Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Cisplatin administration & dosage, Cisplatin adverse effects, Drug Therapy, Combination, Female, Humans, Isoquinolines administration & dosage, Male, Middle Aged, Nausea chemically induced, Palonosetron, Quinuclidines administration & dosage, Serotonin Antagonists administration & dosage, Serotonin Antagonists therapeutic use, Treatment Outcome, Vomiting chemically induced, Antineoplastic Agents therapeutic use, Isoquinolines therapeutic use, Mouth Neoplasms drug therapy, Nausea prevention & control, Quinuclidines therapeutic use, Vomiting prevention & control
- Abstract
Background/aim: To evaluate the efficacy of palonosetron in preventing acute and delayed nausea and vomiting in patients receiving highly emetogenic chemotherapy (HEC) in oral cancer patients., Patients and Methods: Oral cancer patients receiving HEC were enrolled; among the 40 patients, 87 courses of chemotherapy were administered. On day 1, 0.75 mg palonosetron was intravenously administrated just before chemotherapy., Results: The primary endpoint was the proportion of patients with a complete response (CR) and the secondary endpoint was the proportion of patients with complete control (CC) during the acute and delayed phase. During the acute phase, 86 of 87 courses (98.9%) had CR and 84 of 87 courses (96.6%) had CC. During the delayed phase, 84 of 87 courses (96.6%) had CR and 70 of 87 courses (80.5%) had CC., Conclusion: Palonosetron is effective at preventing HEC-induced chemotherapy-induced nausea and vomiting (CINV) in oral cancer chemotherapeutic regimens in the acute and delayed phases., (Copyright© 2017, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
107. Oral Squamous Cell Carcinoma-derived Sonic Hedgehog Promotes Angiogenesis.
- Author
-
Kuroda H, Kurio N, Shimo T, Matsumoto K, Masui M, Takabatake K, Okui T, Ibaragi S, Kunisada Y, Obata K, Yoshioka N, Kishimoto K, Nagatsuka H, and Sasaki A
- Subjects
- Animals, Carcinoma, Squamous Cell blood supply, Carcinoma, Squamous Cell drug therapy, Cell Line, Tumor, Cell Proliferation drug effects, Cells, Cultured, Female, Humans, Immunohistochemistry, Mice, Inbred BALB C, Mice, Nude, Mouth Neoplasms blood supply, Mouth Neoplasms drug therapy, Neovascularization, Pathologic prevention & control, Patched-1 Receptor metabolism, Signal Transduction drug effects, Veratrum Alkaloids pharmacology, Xenograft Model Antitumor Assays, Zinc Finger Protein GLI1 metabolism, Zinc Finger Protein Gli2 metabolism, Carcinoma, Squamous Cell metabolism, Hedgehog Proteins metabolism, Mouth Neoplasms metabolism, Neovascularization, Pathologic metabolism
- Abstract
Background: Sonic hedgehog (SHH) signaling is related to the pathogenesis of oral squamous cell carcinoma (OSCC), but its role in OSCC is not yet well understood. In this study, we analyzed the role of SHH signaling in OSCC., Materials and Methods: We examined the expression pattern of SHH and its signal proteins in clinically resected OSCC samples by immunohistochemistry. We also evaluated the function of SHH signaling using the hedgehog signaling inhibitor cyclopamine in vivo and in vitro by proliferation, migration and angiogenesis analyses., Results: We found that SHH was highly expressed in human tongue OSCC, whereas patched (PTCH1), glioma-associated oncogene 1 (GLI1) and GLI2 proteins were expressed in the microvascular cells in the tumor invasive front. Administration of cyclopamine to mice suppressed the growth and angiogenesis of OSCC xenografts in vivo. Moreover, cyclopamine inhibited endothelial cell proliferation and migration, and reduced aorta vascular length in the rat., Conclusion: These findings suggest that OSCC-derived SHH stimulates angiogenesis at the tumor invasive front., (Copyright© 2017, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
108. Role of Neurokinin 3 Receptor Signaling in Oral Squamous Cell Carcinoma.
- Author
-
Obata K, Shimo T, Okui T, Matsumoto K, Takada H, Takabatake K, Kunisada Y, Ibaragi S, Yoshioka N, Kishimoto K, Nagatsuka H, and Sasaki A
- Subjects
- Animals, Apoptosis drug effects, Bone Resorption drug therapy, Bone Resorption metabolism, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell metabolism, Cell Movement drug effects, Cell Proliferation drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Nude, Mouth Neoplasms drug therapy, Mouth Neoplasms metabolism, Prognosis, Quinolines pharmacology, Receptors, Neurokinin-3 antagonists & inhibitors, Retrospective Studies, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Bone Resorption pathology, Carcinoma, Squamous Cell pathology, Mouth Neoplasms pathology, Receptors, Neurokinin-3 metabolism
- Abstract
Background/aim: The neurokinin 3 receptor (NK-3R) is differentially expressed in the central nervous system including cases of human oral squamous cell carcinoma. However, the role of NK-3R signaling in oral squamous cell carcinoma is not well known., Materials and Methods: NK-3R expression in surgically resected oral squamous cell carcinoma was examined immunohistochemically and the strength of the expression was quantified. We evaluated the function of NK-3R signaling using NK-3R antagonist in human oral squamous cell carcinoma bone invasion mouse model., Results: NK-3R was significantly expressed in tumor cells that had invaded the bone matrix compared to the oral side tumor cells. SB222200, a selective antagonist of NK-3R, significantly suppressed the radiographic osteolytic lesion and tumorigenesis., Conclusion: NK-3R signaling is a potential target for the treatment of oral squamous cell carcinoma in cases of bone destruction., (Copyright© 2017, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
109. Semaphorin 4D promotes bone invasion in head and neck squamous cell carcinoma.
- Author
-
Takada H, Ibaragi S, Eguchi T, Okui T, Obata K, Masui M, Morisawa A, Takabatake K, Kawai H, Yoshioka N, Hassan NMM, Shimo T, Hu GF, Nagatsuka H, and Sasaki A
- Subjects
- Animals, Bone Neoplasms pathology, Bone Neoplasms secondary, Carcinoma, Squamous Cell pathology, Cell Line, Tumor, Cell Movement genetics, Gene Expression Regulation, Neoplastic genetics, Head and Neck Neoplasms pathology, Humans, Mice, Neoplasm Invasiveness genetics, Neovascularization, Pathologic pathology, Oligodendroglia metabolism, Osteoclasts pathology, RANK Ligand genetics, Semaphorins metabolism, Squamous Cell Carcinoma of Head and Neck, Xenograft Model Antitumor Assays, Antigens, CD genetics, Bone Neoplasms genetics, Carcinoma, Squamous Cell genetics, Head and Neck Neoplasms genetics, Insulin-Like Growth Factor I genetics, Neovascularization, Pathologic genetics, Semaphorins genetics
- Abstract
Head and neck squamous cell carcinomas (HNSCCs) frequently invade the bones of the facial skeleton. Semaphorin 4D (Sema4D) is an axon guidance molecule produced by oligodendrocytes. Sema4D was also identified in the bone microenvironment and many cancer tissues including HNSCC. To date, however, the role of Sema4D in cancer-associated bone disease is still unknown. This is the first study to demonstrate the role of Sema4D in bone invasion of cancer. In the clinical tissue samples of bone lesion of HNSCC, Sema4D was detected at high levels, and its expression was correlated with insulin-like growth factor-I (IGF-I) expression. In vitro experiments showed that IGF-I regulates Sema4D expression and Sema4D increased proliferation, migration and invasion in HNSCC cells. Sema4D also regulated the expression of receptor activator of nuclear factor κβ ligand (RANKL) in osteoblasts, and this stimulated osteoclastgenesis. Furthermore, knockdown of Sema4D in HNSCC cells inhibited tumor growth and decreased the number of osteoclasts in a mouse xenograft model. Taken together, IGF-I-driven production of Sema4D in HNSCCs promotes osteoclastogenesis and bone invasion.
- Published
- 2017
- Full Text
- View/download PDF
110. Perimandibular abscess associated with bisphosphonate-related osteonecrosis of the jaw.
- Author
-
Ocho K, Iwamuro M, Hagiya H, Ibaragi S, and Otsuka F
- Published
- 2017
- Full Text
- View/download PDF
111. Orthognathic surgery during breast cancer treatment-A case report.
- Author
-
Shimo T, Yoshioka N, Nakamura M, Ibaragi S, Okui T, Kunisada Y, Masui M, Yao M, Kishimoto K, Yoshida S, Nishiyama A, Kamioka H, and Sasaki A
- Abstract
Introduction: In recent years, patients with orthognathic surgery in middle-aged and elderly people have come to be a more frequent occurrence. Breast cancer is the most frequently diagnosed cancer in woman worldwide, and its prevalence rate is steadily increasing., Presentation of Case: We report a case of a 47-year-old Japanese woman in whom left-side breast cancer (Stage 1) was unexpectedly found just before orthognathic surgery in April 2012. Breast-conserving surgery was performed (estrogen receptor+, progesterone receptor+, HER2 -, surgical margin+, sentinel lymph node +) that May. From June to August docetaxel (75mg/m
2 ) and cyclophosphamide (600mg/m2 ) were administrated four times every 21days and thereafter radiotherapy (total 60Gy) was completed. The cancer surgeon declared the prognosis good and the patient had a strong desire to undergo orthognathic surgery, so in November we performed a bimaxillary osteotomy, and administration of tamoxifen began 6 weeks after the osteotomy., Discussion: There are breast cancer cases in which the prognosis is sufficiently good for a planned orthognathic surgery to proceed. Good communication among surgeons and the patient is important., Conclusion: We experienced a case in which breast cancer was found just before the orthognathic surgery; we performed a bimaxillary osteotomy, including follow-up tamoxifen administration, during breast cancer treatment., (Copyright © 2017 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
112. Tachykinin Receptor 3 Distribution in Human Oral Squamous Cell Carcinoma.
- Author
-
Obata K, Shimo T, Okui T, Matsumoto K, Takada H, Takabatake K, Kunisada Y, Ibaragi S, Nagatsuka H, and Sasaki A
- Subjects
- Humans, Carcinoma, Squamous Cell metabolism, Mouth Neoplasms metabolism, Receptors, Neurokinin-3 metabolism, Receptors, Tachykinin metabolism
- Abstract
Background: Tachykinin 3 (TAC3) and its preferred tachykinin receptor 3 (TACR3) that are prominently detected in the central nervous system, play significant roles in physiological development and specifically in the human reproductive system. The roles of TAC3/TACR3 in oral squamous cell carcinoma are unknown., Materials and Methods: We examined the expression pattern of TAC3/TACR3 in clinically-resected oral squamous cell carcinoma samples using immunohistochemistry and immunofluorescence analysis., Results: We found that even though the expression level of TACR3 was negative in the normal epithelium, it was highly elevated in tumor cells. A more intense signal was observed in the invasive front of tumor cells that had migrated into the mandible bone matrix. TAC3 was not detected in tumor cells, but was expressed in PGP-9.5-positive sensory nerves in the mandible., Conclusion: Our results suggest that peripheral sensory nerve-derived TAC3 may affect gingival oral squamous cell carcinoma cells through TACR3 in the bone matrix., (Copyright© 2016 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
113. Autologous Blood Injection for the Treatment of Recurrent Temporomandibular Joint Dislocation.
- Author
-
Yoshioka N, Shimo T, Ibaragi S, and Sasaki A
- Subjects
- Adult, Clinical Protocols, Humans, Joint Dislocations therapy, Joint Instability therapy, Research Design, Blood Transfusion, Autologous methods, Temporomandibular Joint Disorders therapy
- Abstract
Temporomandibular joint (TMJ) dislocation can occur during daily activities and negatively affect a patient's quality of life. Although both nonsurgical and surgical techniques have been used to treat recurrent TMJ dislocation, the former is not always successful and the latter, although having a high success rate, is invasive and requires hospitalization. Recently, autologous blood injection has been used to treat recurrent TMJ dislocation. However, this technique is not yet widely used in clinical practice. We designed this study to obtain further information as to efficacy, safety and stability of autologous blood injection for recurrent TMJ dislocation.
- Published
- 2016
- Full Text
- View/download PDF
114. Novel Midkine Inhibitor iMDK Inhibits Tumor Growth and Angiogenesis in Oral Squamous Cell Carcinoma.
- Author
-
Masui M, Okui T, Shimo T, Takabatake K, Fukazawa T, Matsumoto K, Kurio N, Ibaragi S, Naomoto Y, Nagatsuka H, and Sasaki A
- Subjects
- Animals, Carcinoma, Squamous Cell blood supply, Cell Differentiation drug effects, Cell Line, Tumor, Cytokines physiology, Female, Humans, Mice, Mice, Inbred BALB C, Midkine, Mouth Neoplasms blood supply, Antineoplastic Agents therapeutic use, Carcinoma, Squamous Cell drug therapy, Coumarins therapeutic use, Cytokines antagonists & inhibitors, Mouth Neoplasms drug therapy, Neovascularization, Pathologic drug therapy, Thiazoles therapeutic use
- Abstract
Midkine is a heparin-binding growth factor highly expressed in various human malignant tumors. However, its role in the growth of oral squamous cell carcinoma is not well understood. In this study, we analyzed the antitumor effect of a novel midkine inhibitor (iMDK) against oral squamous cell carcinoma. Administration of iMDK induced a robust antitumor response and suppressed cluster of differentiation 31 (CD31) expression in oral squamous cell carcinoma HSC-2 cells and SAS cells xenograft models. iMDK inhibited the proliferation of these cells dose-dependently, as well as the expression of midkine and phospho-extracellular signal-regulated kinase in HSC-2 and SAS cells. Moreover, iMDK significantly inhibited vascular endothelial growth factor and induced tube growth of human umbilical vein endothelial cells in a dose-dependent fashion. These findings suggest that midkine is critically involved in oral squamous cell carcinoma and iMDK can be effectively used for the treatment of oral squamous cell carcinoma., (Copyright© 2016 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved.)
- Published
- 2016
115. Synthetic Terrein Inhibits Progression of Head and Neck Cancer by Suppressing Angiogenin Production.
- Author
-
Shibata A, Ibaragi S, Mandai H, Tsumura T, Kishimoto K, Okui T, Hassan NM, Shimo T, Omori K, Hu GF, Takashiba S, Suga S, and Sasaki A
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Disease Progression, Head and Neck Neoplasms metabolism, Human Umbilical Vein Endothelial Cells, Humans, Neovascularization, Pathologic prevention & control, Ribonuclease, Pancreatic biosynthesis, Antineoplastic Agents pharmacology, Cyclopentanes pharmacology, Head and Neck Neoplasms pathology, Ribonuclease, Pancreatic antagonists & inhibitors
- Abstract
Background/aim: Head and neck cancers are the fifth most common cancer type worldwide, affecting more than half a million patients annually. Development of effective therapeutic drugs is, therefore, required for this type of disease. This study assessed the effects of synthetic terrein on head and neck cancer., Materials and Methods: Synthetic terrein was prepared by using the modified Altenhach's procedure. The effect of synthetic terrein on cell proliferation of head and neck cancer cells and HUVECs was assessed. Angiogenin secretion and ribosome biogenesis were examined by ELISA and silver staining of the nucleolar organizer region. A mouse xenograft model was prepared by inoculating mice with suspensions of cells of the human head and neck cancer cell line OSC-19 subcutaneously into the dorsal region of each mouse. Ki-67, CD31 and angiogenin expression in xenografted tumors was examined by immunohistochemistry., Results: Synthetic terrein inhibited the growth of various head and neck cancer cells. In addition, an in vivo experiment revealed that synthetic terrein inhibited a xenograft tumor growth in athymic mice. Immunohistochemical analysis revealed that expression of Ki-67, CD31 and ANG was down-regulated in synthetic terrein-treated tumors, compared to controls. Synthetic terrein suppressed the ANG secretion and ribosome biogenesis in cancer cells, and cell proliferation in vascular endothelial cells., Conclusion: The mechanism underlying the anti-tumor effects of synthetic terrein against head and neck cancer consists of the inhibition of both tumor cell proliferation and angiogenesis via the suppression of ANG production., (Copyright© 2016 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved.)
- Published
- 2016
116. The Role of Sonic Hedgehog Signaling in Osteoclastogenesis and Jaw Bone Destruction.
- Author
-
Shimo T, Matsumoto K, Takabatake K, Aoyama E, Takebe Y, Ibaragi S, Okui T, Kurio N, Takada H, Obata K, Pang P, Iwamoto M, Nagatsuka H, and Sasaki A
- Subjects
- Animals, Carcinoma, Squamous Cell pathology, Cell Differentiation physiology, Cell Line, Cell Proliferation, Humans, Mandibular Neoplasms pathology, Mice, Carcinoma, Squamous Cell physiopathology, Hedgehog Proteins physiology, Mandibular Neoplasms physiopathology, Osteoclasts cytology, Signal Transduction physiology
- Abstract
Sonic hedgehog (SHH) and its signaling have been identified in several human cancers, and increased levels of its expression appear to correlate with disease progression and metastasis. However, the role of SHH in bone destruction associated with oral squamous cell carcinomas is still unclear. In this study we analyzed SHH expression and the role played by SHH signaling in gingival carcinoma-induced jawbone destruction. From an analysis of surgically resected lower gingival squamous cell carcinoma mandible samples, we found that SHH was highly expressed in tumor cells that had invaded the bone matrix. On the other hand, the hedgehog receptor Patched and the signaling molecule Gli-2 were highly expressed in the osteoclasts and the progenitor cells. SHH stimulated osteoclast formation and pit formation in the presence of the receptor activator for nuclear factor-κB ligand (RANKL) in CD11b+ mouse bone marrow cells. SHH upregulated phosphorylation of ERK1/2 and p38 MAPK, NFATc1, tartrate-resistant acid phosphatase (TRAP), and Cathepsin K expression in RAW264.7 cells. Our results suggest that tumor-derived SHH stimulated the osteoclast formation and bone resorption in the tumor jawbone microenvironment.
- Published
- 2016
- Full Text
- View/download PDF
117. Chemically Modified Interleukin-6 Aptamer Inhibits Development of Collagen-Induced Arthritis in Cynomolgus Monkeys.
- Author
-
Hirota M, Murakami I, Ishikawa Y, Suzuki T, Sumida S, Ibaragi S, Kasai H, Horai N, Drolet DW, Gupta S, Janjic N, and Schneider DJ
- Subjects
- Amino Acid Sequence, Animals, Arthritis, Experimental chemically induced, Cells, Cultured, Female, Humans, Interleukin-6 chemistry, Macaca fascicularis, Molecular Sequence Data, Phosphorylation, STAT3 Transcription Factor metabolism, Sequence Homology, Amino Acid, T-Lymphocytes metabolism, Aptamers, Peptide chemistry, Arthritis, Experimental prevention & control, Collagen adverse effects, Interleukin-6 blood
- Abstract
Interleukin-6 (IL-6) is a potent mediator of inflammatory and immune responses, and a validated target for therapeutic intervention of inflammatory diseases. Previous studies have shown that SL1026, a slow off-rate modified aptamer (SOMAmer) antagonist of IL-6, neutralizes IL-6 signaling in vitro. In the present study, we show that SL1026 delays the onset and reduces the severity of rheumatoid symptoms in a collagen-induced arthritis model in cynomolgus monkeys. SL1026 (1 and 10 mg/kg), administered q.i.d., delayed the progression of arthritis and the concomitant increase in serum IL-6 levels compared to the untreated control group. Furthermore, SL1026 inhibited IL-6-induced STAT3 phosphorylation ex vivo in T lymphocytes from human blood and IL-6-induced C-reactive protein and serum amyloid A production in human primary hepatocytes. Importantly, SOMAmer treatment did not elicit an immune response, as evidenced by the absence of anti-SOMAmer antibodies in plasma of treated monkeys. These results demonstrate that SOMAmer antagonists of IL-6 may be attractive agents for the treatment of IL-6-mediated diseases, including rheumatoid arthritis.
- Published
- 2016
- Full Text
- View/download PDF
118. Expression pattern of sonic hedgehog signaling and calcitonin gene-related peptide in the socket healing process after tooth extraction.
- Author
-
Pang P, Shimo T, Takada H, Matsumoto K, Yoshioka N, Ibaragi S, and Sasaki A
- Subjects
- Animals, Immunohistochemistry, Kruppel-Like Transcription Factors metabolism, Mice, Mice, Inbred C57BL, Patched Receptors, Patched-1 Receptor, Receptors, Cell Surface metabolism, Sensory Receptor Cells metabolism, Signal Transduction, Time Factors, Tooth Socket innervation, Zinc Finger Protein Gli2, Calcitonin Gene-Related Peptide metabolism, Hedgehog Proteins metabolism, Tooth Extraction, Tooth Socket metabolism, Wound Healing physiology
- Abstract
Sonic Hedgehog (SHH), a neural development inducer, plays a significant role in the bone healing process. Calcitonin gene-related peptide (CGRP), a neuropeptide marker of sensory nerves, has been demonstrated to affect bone formation. The roles of SHH signaling and CGRP-positive sensory nerves in the alveolar bone formation process have been unknown. Here we examined the expression patterns of SHH signaling and CGRP in mouse socket by immunohistochemistry and immunofluorescence analysis. We found that the expression level of SHH peaked at day 3 and was then decreased at 5 days after tooth extraction. CGRP, PTCH1 and GLI2 were each expressed in a similar pattern with their highest expression levels at day 5 and day 7 after tooth extraction. CGRP and GLI2 were co-expressed in some inflammatory cells and bone forming cells. In some areas, CGRP-positive neurons expressed GLI2. In conclusion, SHH may affect alveolar bone healing by interacting with CGRP-positive sensory neurons and thus regulate the socket's healing process after tooth extraction., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
119. Expression and roles of CCN2 in dental epithelial cells.
- Author
-
Shimo T, Koyama E, Kurio N, Matsumoto K, Okui T, Ibaragi S, Yoshioka N, and Sasaki A
- Subjects
- Animals, Biomarkers, Cattle, Cells, Cultured, Immunohistochemistry, Tooth Germ embryology, Tooth Germ metabolism, Connective Tissue Growth Factor genetics, Connective Tissue Growth Factor metabolism, Epithelial Cells metabolism, Gene Expression, Odontogenesis genetics, Tooth cytology
- Abstract
Connective tissue growth factor (CCN2) regulates diverse cellular functions, including tooth development. In order to delineate the precise role of CCN2 in the epithelium during odontogenesis, we investigated how it is expressed and what roles it may have in primary cultures of epithelial cells derived from developing tooth germ of the bovine fetus. Ccn2 mRNA and protein were strongly expressed in the inner dental epithelium, which is consistent with the expression of transforming growth factor-β2 mRNA and proliferating cell nuclear antigen. Bone morphogenetic protein 4 (BMP4) and fibroblast growth factor 2 (FGF2) were also expressed in the inner dental epithelium, indicating that CCN2 functionally interacts with these factors in the epithelium. The stimulatory effects of FGF2 on cell proliferation and BMP4 on cell differentiation were additively up-regulated by CCN2 in a newly-established dental epithelium cell culture. Taken together, our data provide clear evidence that CCN2 is synthesized by inner dental epithelial cells, and appears to act as an autocrine factor, which regulates dental epithelial cell proliferation and differentiation in concert with growth factors., (Copyright © 2015 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved.)
- Published
- 2015
120. A case of adenoid cystic carcinoma associated with IgG4-related disease.
- Author
-
Shimo T, Yao M, Takebe Y, Ono Y, Obata K, Kurio N, Ibaragi S, Yoshioka N, Kishimoto K, Yanagi Y, Nagatsuka H, and Sasaki A
- Abstract
Introduction: Immunoglobulin G4-related disease (IgG4-RD) is an inflammatory condition associated with elevated serum IgG4 levels and tissue infiltration by IgG4-expressing plasma cells. We present a case of adenoid cystic carcinoma (ACC) of the submandibular gland with possible involvement of IgG4-RD., Presentation of Case: The patient was a 59-year-old man presenting with a swollen right submandibular gland. Laboratory tests revealed IgG4 levels of 176mg/dl (reference range: 4.8-105). An initial open biopsy for histological diagnosis showed chronic sialadenitis. The region was monitored on an outpatient basis, and finally the right submandibular was totally resected because malignant tumor could not be excluded. Histological examination of the submandibular gland showed an ACC with lymphocytic infiltration containing many IgG4-positive plasma cells in the tumor stroma., Discussion: We have described a case that indicated a possible involvement of ACC with IgG4-RD. This allows us to speculate that longstanding IgG4-RD may progress to malignancy or infiltration of IgG4-positive plasma cells through the signals of tumor stimuli. Further investigations are required to determine the potential pathogenic mechanism underlying this unique tumor., Conclusion: This case underscores that caution is needed in the diagnosis of masses with high serum IgG4 levels, as the differential diagnosis includes malignancy., (Copyright © 2015 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
121. Synthetic (+)-terrein suppresses interleukin-6/soluble interleukin-6 receptor induced-secretion of vascular endothelial growth factor in human gingival fibroblasts.
- Author
-
Mandai H, Omori K, Yamamoto D, Tsumura T, Murota K, Yamamoto S, Mitsudo K, Ibaragi S, Sasaki A, Maeda H, Takashiba S, and Suga S
- Subjects
- Cyclopentanes chemistry, Dose-Response Relationship, Drug, Fibroblasts metabolism, Humans, Interleukin-6 metabolism, Molecular Structure, Receptors, Interleukin-6 metabolism, Solubility, Stereoisomerism, Structure-Activity Relationship, Cyclopentanes chemical synthesis, Cyclopentanes pharmacology, Fibroblasts drug effects, Gingiva cytology, Interleukin-6 antagonists & inhibitors, Receptors, Interleukin-6 antagonists & inhibitors, Vascular Endothelial Growth Factors metabolism
- Abstract
Interleukin (IL)-6 is a proinflammatory cytokine that performs a wide variety of biological functions, including important roles in the progression of chronic inflammatory diseases such as periodontal disease. (+)-Terrein, a secondary bioactive fungal metabolite isolated from Aspergillus terreus, has various biological activities; however, its anti-inflammatory effects are still unknown. The purpose of this study was to examine the effect of synthetic (+)-terrein on IL-6 signaling and related protein production in human gingival fibroblasts. To our knowledge, this study is the first to report that synthetic (+)-terrein is not cytotoxic at concentrations less than 20 μM and suppresses IL-6/soluble IL-6 receptor (sIL-6R)-induced phosphorylation of signal transducer and activator of transcription-3, extracellular signal-regulated kinase 1/2, and c-jun N-terminal kinase 1/2-signaling proteins that are downstream of IL-6 signaling. In addition, synthetic (+)-terrein suppresses IL-6/sIL-6R-induced vascular endothelial growth factor (VEGF) secretion in a concentration-dependent manner (p<0.01). These data suggest that synthetic (+)-terrein has potential anti-IL-6 signaling activity and suppresses VEGF-associated inflammatory disease progression., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
122. Neamine inhibits oral cancer progression by suppressing angiogenin-mediated angiogenesis and cancer cell proliferation.
- Author
-
Kishimoto K, Yoshida S, Ibaragi S, Yoshioka N, Hu GF, and Sasaki A
- Subjects
- Animals, Cell Line, Tumor, Disease Progression, Humans, Immunohistochemistry, In Situ Nick-End Labeling, Mice, Mice, Nude, Mouth Neoplasms blood supply, Mouth Neoplasms pathology, Neovascularization, Pathologic pathology, Protein Transport drug effects, Ribonuclease, Pancreatic metabolism, Xenograft Model Antitumor Assays, Angiogenesis Inducing Agents pharmacology, Cell Proliferation drug effects, Framycetin pharmacology, Mouth Neoplasms metabolism, Neovascularization, Pathologic metabolism
- Abstract
Background: Angiogenin undergoes nuclear translocation and stimulates ribosomal RNA transcription in both endothelial and cancer cells. Consequently, angiogenin has a dual effect on cancer progression by inducing both angiogenesis and cancer cell proliferation. The aim of this study was to assess whether neamine, a blocker of nuclear translocation of angiogenin, possesses antitumor activity toward oral cancer., Materials and Methods: The antitumor effect of neamine on oral cancer cells was examined both in vitro and in vivo., Results: Neamine inhibited the proliferation of HSC-2, but not that of SAS oral cancer cells in vitro. Treatment with neamine effectively inhibited growth of HSC-2 and SAS cell xenografts in athymic mice. Neamine treatment resulted in a significant decrease in tumor angiogenesis, accompanied by a decrease in angiogenin- and proliferating cell nuclear antigen-positive cancer cells, especially of HSC-2 tumors., Conclusion: Neamine effectively inhibits oral cancer progression through inhibition of tumor angiogenesis. Neamine also directly inhibits proliferation of certain types of oral cancer cells. Therefore, neamine has potential as a lead compound for oral cancer therapy.
- Published
- 2014
123. Angiogenin mediates androgen-stimulated prostate cancer growth and enables castration resistance.
- Author
-
Li S, Hu MG, Sun Y, Yoshioka N, Ibaragi S, Sheng J, Sun G, Kishimoto K, and Hu GF
- Subjects
- Androgens metabolism, Animals, Cell Line, Tumor, Cell Proliferation drug effects, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Heterografts, Humans, Male, Mice, Mice, SCID, Neoplasm Transplantation, Promoter Regions, Genetic, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology, Protein Binding, Receptors, Androgen genetics, Receptors, Androgen metabolism, Ribonuclease, Pancreatic antagonists & inhibitors, Transcription, Genetic, Angiogenesis Inducing Agents metabolism, Prostatic Neoplasms metabolism, Prostatic Neoplasms, Castration-Resistant metabolism, RNA, Ribosomal genetics, Ribonuclease, Pancreatic metabolism
- Abstract
Unlabelled: The androgen receptor (AR) is a critical effector of prostate cancer development and progression. Androgen-dependent prostate cancer is reliant on the function of AR for growth and progression. Most castration-resistant prostate cancer (CRPC) remains dependent on AR signaling for survival and growth. Ribosomal RNA (rRNA) is essential for both androgen-dependent and castration-resistant growth of prostate cancer cells. During androgen-dependent growth of prostate cells, androgen-AR signaling leads to the accumulation of rRNA. However, the mechanism by which AR regulates rRNA transcription is unknown. Here, investigation revealed that angiogenin (ANG), a member of the secreted ribonuclease superfamily, is upregulated in prostate cancer and mediates androgen-stimulated rRNA transcription in prostate cancer cells. Upon androgen stimulation, ANG undergoes nuclear translocation in androgen-dependent prostate cancer cells, where it binds to the rDNA promoter and stimulates rRNA transcription. ANG antagonists inhibit androgen-induced rRNA transcription and cell proliferation in androgen-dependent prostate cancer cells. Interestingly, ANG also mediates androgen-independent rRNA transcription through a mechanism that involves its constitutive nuclear translocation in androgen-insensitive prostate cancer cells, resulting in a constant rRNA overproduction and thereby stimulating cell proliferation. Critically, ANG overexpression in androgen-dependent prostate cancer cells enables castration-resistant growth of otherwise androgen-dependent cells. Thus, ANG-stimulated rRNA transcription is not only an essential component for androgen-dependent growth of prostate cancer but also contributes to the transition of prostate cancer from androgen-dependent to castration-resistant growth status., Implications: The ability of angiogenin to regulate rRNA transcription and prostate cancer growth makes it a viable target for therapy.
- Published
- 2013
- Full Text
- View/download PDF
124. Novel HSP90 inhibitor NVP-AUY922 enhances the anti-tumor effect of temsirolimus against oral squamous cell carcinoma.
- Author
-
Okui T, Shimo T, Fukazawa T, Mohammad Monsur Hassan N, Honami T, Ibaragi S, Takaoka M, Naomoto Y, and Sasaki A
- Subjects
- Angiogenesis Inhibitors administration & dosage, Angiogenesis Inhibitors pharmacology, Angiogenesis Inhibitors therapeutic use, Animals, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carcinoma, Squamous Cell blood supply, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell pathology, Cell Line, Tumor, Cells, Cultured, Female, HSP90 Heat-Shock Proteins metabolism, Head and Neck Neoplasms blood supply, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms metabolism, Head and Neck Neoplasms pathology, Human Umbilical Vein Endothelial Cells cytology, Human Umbilical Vein Endothelial Cells drug effects, Human Umbilical Vein Endothelial Cells metabolism, Humans, Isoxazoles administration & dosage, Isoxazoles pharmacology, Mice, Mice, Nude, Molecular Targeted Therapy, Mouth Neoplasms blood supply, Mouth Neoplasms metabolism, Mouth Neoplasms pathology, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins metabolism, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Random Allocation, Resorcinols administration & dosage, Resorcinols pharmacology, Sirolimus administration & dosage, Sirolimus pharmacology, Sirolimus therapeutic use, Squamous Cell Carcinoma of Head and Neck, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Squamous Cell drug therapy, HSP90 Heat-Shock Proteins antagonists & inhibitors, Isoxazoles therapeutic use, Mouth Neoplasms drug therapy, Resorcinols therapeutic use, Sirolimus analogs & derivatives, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Background and Aim: Heat shock protein 90 (HSP90) and mammalian target of rapamycin (mTOR) are involved in the molecular pathogenesis of advanced oral squamous cell carcinoma. HSP90 inhibitors are capable of effectively interfering with multiple signaling pathways, including the mTOR signaling pathway. However, the combined effects of HSP90 and mTOR inhibitors on oral squamous cell carcinoma are still unknown. In this study, we investigated the dual treatment of the novel HSP90 inhibitor NVP-AUY922 and temsirolimus against oral squamous cell carcinoma., Materials and Methods: The effect of the combination of NVP-AUY922 and temsirolimus on oral squamous cell carcinoma in vitro and in vivo was determined by MTS assay and mouse xenograft models. The effect of the combination on angiogenesis was determined by tube formation assay and angioreactor., Results: The combination treatment of NVP-AUY922 and temsirolimus significantly inhibited the proliferation of SAS oral squamous cell carcinoma cells in vitro and suppressed the growth of oral squamous cell carcinoma xenografts in vivo. We have clearly shown that the combination treatment of NVP-AUY922 and temsirolimus inhibited vascular formation both in vitro and in vivo. Moreover, the combination treatment of NVP-AUY922 and temsirolimus prolonged the survival rate in mice xenografted with oral squamous cell carcinoma., Conclusions: Here, we showed the activity of a combination of mTOR and HSP90 inhibitors for the treatment of advanced oral squamous carcinoma.
- Published
- 2013
- Full Text
- View/download PDF
125. Hypoxia-induced up-regulation of angiogenin, besides VEGF, is related to progression of oral cancer.
- Author
-
Kishimoto K, Yoshida S, Ibaragi S, Yoshioka N, Okui T, Hu GF, and Sasaki A
- Subjects
- Animals, Blotting, Western, Cell Hypoxia, Enzyme-Linked Immunosorbent Assay, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Male, Mice, Mice, Nude, Real-Time Polymerase Chain Reaction, Carcinoma, Squamous Cell metabolism, Mouth Neoplasms metabolism, Ribonuclease, Pancreatic metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
Objectives: Angiogenin (ANG) is a prominent angiogenic factor that has been shown to have a dual effect on tumor progression by inducing both angiogenesis and cancer cell proliferation through stimulating ribosomal RNA transcription in both endothelial cells and cancer cells. In the present study, we investigated the expression profiles of ANG and vascular endothelial growth factor (VEGF) in oral cancer and their correlation with hypoxia and evaluated the possible value of ANG as a therapeutic target for oral cancer., Materials and Methods: Immunohistochemistry (IHC), ELISA, real-time RT-PCR and Western blotting were used to examine the expression of ANG, VEGF, and hypoxia-inducible factor 1α (HIF-1α) in oral squamous cell carcinoma (OSCC) specimens and human OSCC cell lines. In order to examine the role of ANG, we knocked down ANG expression in HSC-2 cells by means of plasmid-mediated RNA interference., Results: IHC showed that the expression of ANG was significantly correlated with that of HIF-1α in 50 OSCC specimens (P = 0.031). However, no significant correlation between VEGF and HIF-1α expression was found (P = 0.243). Consistently, ANG secretion increased under hypoxia in all of the 10 OSCC cell lines tested; and a significant increase was observed in 6 of them. In contrast, there was no noticeable increase in VEGF secretion under hypoxia in any of these cell lines. In HSC-2 and SAS OSCC cells, the increase in ANG mRNA expression correlated very well with that of HIF-1α protein expression after hypoxia onset. However, no noticeable increase in VEGF mRNA expression was observed even after 12 h of hypoxia. Down-regulation of ANG expression in HSC-2 cells highly expressing and secreting VEGF inhibited ribosome biogenesis, cell proliferation, tumor angiogenesis, and xenograft growth in athymic mice., Conclusion: These results suggest that ANG is up-regulated in the hypoxic environment of oral cancers and that its inhibition can have a therapeutic implication., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
126. Angiogenin as a molecular target for the treatment of prostate cancer.
- Author
-
Li S, Ibaragi S, and Hu GF
- Abstract
Angiogenin (ANG), a 14 kDa angiogenic ribonuclease, is upregulated in human prostate cancers, especially in hormone refractory diseases, and is the highest upregulated gene in Akt-driven prostate intraepithelial neoplasia (PIN) in mice. ANG has been shown to undergo nuclear translocation in both prostate cancer cells and cancer-associated endothelial cells where it binds to the promoter region of ribosomal DNA (rDNA) and stimulates ribosomal RNA (rRNA) transcription. ANG thus plays an essential role in prostate cancer progression by stimulating both cancer cell proliferation and tumor angiogenesis. A variety of ANG antagonists, including its antisense oligonucleotide, siRNA, soluble binding proteins, monoclonal antibody, enzymatic inhibitors, and nuclear translocation blockers, have all been shown to inhibit prostate cancer in various animal models. Accumulating evidence indicates that ANG is a molecular target for prostate cancer drug development.
- Published
- 2011
- Full Text
- View/download PDF
127. Induction of MMP-13 expression in bone-metastasizing cancer cells by type I collagen through integrin α1β1 and α2β1-p38 MAPK signaling.
- Author
-
Ibaragi S, Shimo T, Hassan NM, Isowa S, Kurio N, Mandai H, Kodama S, and Sasaki A
- Subjects
- Animals, Blotting, Western, Bone Neoplasms genetics, Bone Neoplasms secondary, Breast Neoplasms genetics, Breast Neoplasms pathology, Collagen Type I genetics, Female, Focal Adhesion Protein-Tyrosine Kinases genetics, Focal Adhesion Protein-Tyrosine Kinases metabolism, Humans, Immunoenzyme Techniques, Integrin alpha1beta1 genetics, Integrin alpha2beta1 genetics, Matrix Metalloproteinase 13 genetics, Mice, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Tumor Cells, Cultured, p38 Mitogen-Activated Protein Kinases genetics, Bone Neoplasms metabolism, Breast Neoplasms metabolism, Collagen Type I metabolism, Integrin alpha1beta1 metabolism, Integrin alpha2beta1 metabolism, Matrix Metalloproteinase 13 metabolism, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
Background: Breast cancer cells frequently metastasize to the skeleton and produce and secrete proteinases, such as matrix metalloproteinase-13 (MMP-13), which promote destruction of the bone matrix. However, the mechanism of MMP-13 expression induced in areas of bone metastasis is unknown. Here, the interaction between tumors and type I collagen in bone metastasis was investigated., Materials and Methods: A mouse model of bone metastasis was prepared by inoculating mice with suspensions of cells of the human metastatic breast cancer cell line MDA-MB-231 via the left cardiac ventricle. MMP-13 expression was examined by immunohistochemical, Western blot, and real-time RT-PCR analyses., Results: MMP-13 expression was highly up-regulated in MDA-MB-231 cells, and attachment of these cells to type I collagen and the induction of MMP-13 were down-regulated by treatment with integrin α1, α2 or β1 neutralizing antibodies. The attachment of MDA-MB-231 cells to type I collagen induced the activation of focal adhesion kinase (FAK) and p38 mitogen-activated protein kinase (MAPK). Inhibition of FAK and p38 MAPK down-regulated type I collagen-induced MMP-13 expression., Conclusion: Our study indicates that metastatic breast cancer cells in the bone microenvironment attached to type I collagen, which stimulated integrins α1β1 and α2β1, via FAK and p38 MAPK pathways, to induce MMP13 expression and further osteolysis.
- Published
- 2011
128. Parathyroid hormone-related peptide regulates matrix metalloproteinase-13 gene expression in bone metastatic breast cancer cells.
- Author
-
Ibaragi S, Shimo T, Iwamoto M, Hassan NM, Kodama S, Isowa S, and Sasaki A
- Subjects
- Animals, Bone Neoplasms enzymology, Bone Neoplasms genetics, Bone Neoplasms secondary, Breast Neoplasms genetics, Cell Line, Tumor, Cyclic AMP-Dependent Protein Kinases metabolism, Disease Models, Animal, Female, Flavonoids pharmacology, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Humans, Matrix Metalloproteinase 13 genetics, Mice, Mice, Inbred BALB C, Mice, Nude, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Parathyroid Hormone-Related Protein biosynthesis, Parathyroid Hormone-Related Protein pharmacology, Phosphorylation, Protein Kinase C metabolism, Breast Neoplasms enzymology, Breast Neoplasms pathology, Matrix Metalloproteinase 13 biosynthesis, Parathyroid Hormone-Related Protein metabolism
- Abstract
Background: Breast cancer (BC) cells often metastasize to bone where they express large amounts of parathyroid hormone-related protein (PTHrP). In this study, we investigated the possibility that PTHrP may have roles in breast cancer bone metastasis independently of, or in addition to, its roles in osteoclastic function., Materials and Methods: A mouse model of bone metastasis was prepared by inoculating mice with suspensions of the human BC cell line MDA-MB-231 tumor cells via the left cardiac ventricle. Matrix metalloproteinase-13 (MMP-13) expression in the bone microenvironment was examined by Western blot and Real-time RT-PCR (RT-PCR) analysis, as well as by confocal microscopy., Results: The invading MDA-MB-231 cells contained conspicuous amounts of both PTHrP and MMP-13, an important matrix-degrading enzyme; and treatment of the cells in culture with exogenous PTHrP markedly stimulated MMP13 gene expression. Analysis of signaling mechanisms showed that PTHrP treatment led to rapid increases in the levels of phosphorylated protein kinase C (PKCα) and extracellular signal-regulated kinase (ERK1/2). Pharmacologic inhibition of ERK1/2 and PKC as well as of PKA activities counteracted the PTHrP-dependent stimulation of MMP13 expression. Indeed, pharmacologic activation of PKA or PKC was sufficient for stimulation of MMP13 expression., Conclusion: Consistent with these findings, the inhibition of PKC prevented PTHrP-induced activation of ERK1/2, whereas 12-O-tetradecanoylphorbol-13-acetate (TPA), a stimulator of PKC, up-regulated the PTHrP-induced activation of ERK1/2. Taken together, our data indicate that the MDA-MB-231 breast cancer cells may carry out bone destruction and favor their own metastatic behavior by producing MMP-13. Given that the cells expressed PTHrP and that this factor stimulated MMP-13 expression, metastatic bone destruction may result from a PTHrP autocrine loop involving a PKC-ERK1/2 signaling pathway.
- Published
- 2010
129. Bone destruction by invading oral squamous carcinoma cells mediated by the transforming growth factor-beta signalling pathway.
- Author
-
Goda T, Shimo T, Yoshihama Y, Hassan NM, Ibaragi S, Kurio N, Okui T, Honami T, Kishimoto K, and Sasaki A
- Subjects
- Aged, Aged, 80 and over, Animals, Carcinoma, Squamous Cell genetics, Cell Line, Tumor, Connective Tissue Growth Factor biosynthesis, Connective Tissue Growth Factor genetics, Female, Gene Expression, Gingival Neoplasms genetics, Humans, Male, Mice, Mice, Nude, Middle Aged, Osteolysis genetics, Osteolysis metabolism, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases biosynthesis, RANK Ligand biosynthesis, RANK Ligand genetics, Receptor, Transforming Growth Factor-beta Type I, Receptors, Transforming Growth Factor beta antagonists & inhibitors, Receptors, Transforming Growth Factor beta biosynthesis, Signal Transduction, Transforming Growth Factor beta antagonists & inhibitors, Transplantation, Heterologous, Tumor Necrosis Factor-alpha biosynthesis, Tumor Necrosis Factor-alpha genetics, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell pathology, Gingival Neoplasms metabolism, Gingival Neoplasms pathology, Osteolysis pathology, Transforming Growth Factor beta metabolism
- Abstract
Background: Gingival squamous cell carcinoma (SCC) cells frequently invade mandibular bone, and this destruction is associated with a worse prognosis. However, the relationship between bone destruction and associated factors is unclear. In this study, the role and diagnostic utility of transforming growth factor-beta (TGF-beta) type I receptor (TbetaRI) in bone destruction of the mandible was investigated., Patients and Methods: The expression of TbetaRI was explored by using an immunohistochemical method on paraffin-embedded tissues from 21 cases of mandibular SCC. An inhibitor of the kinase activity of the TbetaRI (TbetaRI-I) was used to assess the role of TbetaRI in bone destruction by a human oral SCC cell line (HSC-2) that highly expresses TbetaRI., Results: TbetaRI-positive signals were closely associated with destructive invasion of the mandible by oral SCC cells. Consistent with these results, TbetaRI-I greatly reduced HSC-2 cell-induced bone destruction and osteoclast formation in vivo and in vitro. TbetaRI-I treatment reduced the expression of TNF-alpha, RANKL and connective tissue growth factor (CTGF/CCN2), all of which were up-regulated by TGF-beta in HSC-2 cells., Conclusion: These data demonstrated an important role for TGF-beta signalling in bone invasion by oral SCC cells, and suggest that the bone destruction is mediated by RANKL, TNF-alpha and CCN2.
- Published
- 2010
130. PTHrP regulates angiogenesis and bone resorption via VEGF expression.
- Author
-
Isowa S, Shimo T, Ibaragi S, Kurio N, Okui T, Matsubara K, Hassan NM, Kishimoto K, and Sasaki A
- Subjects
- Animals, Bone Neoplasms genetics, Bone Neoplasms metabolism, Bone Resorption genetics, Bone Resorption pathology, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, Culture Media, Cyclic AMP-Dependent Protein Kinases metabolism, Disease Models, Animal, Endothelial Cells metabolism, Endothelial Cells pathology, Enzyme Activation, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, Mitogen-Activated Protein Kinases metabolism, Neovascularization, Pathologic genetics, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Osteoclasts pathology, Parathyroid Hormone-Related Protein pharmacology, Promoter Regions, Genetic, Protein Kinase C metabolism, Rats, Receptor, Parathyroid Hormone, Type 1 metabolism, Transcriptional Activation, Vascular Endothelial Growth Factor A genetics, Vascular Endothelial Growth Factor A metabolism, Bone Neoplasms secondary, Bone Resorption metabolism, Breast Neoplasms blood supply, Parathyroid Hormone-Related Protein metabolism, Vascular Endothelial Growth Factor A biosynthesis
- Abstract
Background: Parathyroid hormone-related protein (PTHrP) is a key regulator of osteolytic metastasis of breast cancer (BC) cells, but its targets and mechanisms of action are not fully understood. This study investigated whether/how PTHrP (1-34) signaling regulates expression of vascular endothelial growth factor (VEGF) produced by BC cells., Materials and Methods: A mouse model of bone metastasis was prepared by inoculating mice with tumour cell suspensions of the human BC cell line MDA-MB-231 via the left cardiac ventricle. VEGF expression was examined by Western blot and real-time RT-PCR analysis, as well as by confocal microscopy in the bone microenvironment., Results: PTHrP was expressed in cancer cells producing PTH/PTHrP receptor and VEGF that had invaded the bone marrow, and PTHrP was up-regulated VEGF in MDA-MB-231 in vitro. The culture medium conditioned by PTHrP-treated MDA-MB-231 cells stimulated angiogenesis and osteoclastogenesis compared with control medium, giving a response that was inhibited by VEGF-neutralizing antibody treatment. Inhibition of protein kinase C (PKC) prevented PTHrP-induced extracellular signal-regulated kinase (ERK1/2) and p38 activation, and PTHrP-induced VEGF expression., Conclusion: PTHrP plays an important role in modulating the angiogenic and bone osteolytic actions of VEGF through PKC-dependent activation of an ERK1/2 and p38 signaling pathway during bone metastasis by breast cancer cells.
- Published
- 2010
131. Epithelial-mesenchymal transition and cell cooperativity in metastasis.
- Author
-
Tsuji T, Ibaragi S, and Hu GF
- Subjects
- Animals, Epithelial Cells pathology, Humans, Mesoderm pathology, Neoplasm Metastasis, Cell Communication physiology, Neoplasms pathology
- Abstract
The role of epithelial-mesenchymal transition (EMT) in metastasis remains controversial. EMT has been postulated as an absolute requirement for tumor invasion and metastasis. Three different models including incomplete EMT, mesenchymal-epithelial transition (MET), and collective migration have been proposed for the role of EMT in cancer invasion and metastasis. However, skepticism remains about whether EMT truly occurs during cancer progression, and if it does, whether it plays an indispensible role in metastasis. Our recent findings suggest that EMT cells are responsible for degrading the surrounding matrix to enable invasion and intravasation of both EMT and non-EMT cells. Only non-EMT cells that have entered the blood stream are able to re-establish colonies in the secondary sites. Here, we discuss an alternative model for the role of EMT in cancer metastasis in which EMT and non-EMT cells cooperate to complete the entire process of spontaneous metastasis.
- Published
- 2009
- Full Text
- View/download PDF
132. Neamine inhibits prostate cancer growth by suppressing angiogenin-mediated rRNA transcription.
- Author
-
Ibaragi S, Yoshioka N, Li S, Hu MG, Hirukawa S, Sadow PM, and Hu GF
- Subjects
- Angiogenesis Inhibitors pharmacology, Animals, Cell Line, Tumor, Cell Proliferation drug effects, Humans, Male, Mice, Prostatic Neoplasms pathology, Proto-Oncogene Proteins c-akt physiology, Ribonuclease, Pancreatic physiology, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Framycetin pharmacology, Prostatic Neoplasms drug therapy, RNA, Ribosomal genetics, Ribonuclease, Pancreatic antagonists & inhibitors, Transcription, Genetic drug effects
- Abstract
Purpose: Angiogenin (ANG) undergoes nuclear translocation and stimulates rRNA transcription in both prostate cancer cells and endothelial cells. The purpose of this study is to assess the antitumor activity of neamine, a nontoxic degradation product of neomycin that blocks nuclear translocation of ANG., Experimental Design: The anti-prostate cancer activity of neamine was first evaluated in a xenograft animal model. It was then examined in the murine prostate-restricted AKT transgenic mice that develop prostate intraepithelial neoplasia (PIN) owing to AKT transgene overexpression., Results: Neamine inhibits xenograft growth of PC-3 human prostate cancer cells in athymic mice. It blocks nuclear translocation of ANG and inhibits rRNA transcription, cell proliferation, and angiogenesis. Neamine also prevents AKT-induced PIN formation as well as reverses fully developed PIN in murine prostate-restricted AKT mice, accompanied by a decrease in rRNA synthesis, cell proliferation, and angiogenesis and an increase in prostate epithelial cell apoptosis., Conclusion: We confirmed that ANG is a molecular target for cancer drug development and that blocking nuclear translocation of ANG is an effective means to inhibit its activity. Our results also suggested that neamine is a lead compound for further preclinical evaluation.
- Published
- 2009
- Full Text
- View/download PDF
133. Angiogenin-stimulated rRNA transcription is essential for initiation and survival of AKT-induced prostate intraepithelial neoplasia.
- Author
-
Ibaragi S, Yoshioka N, Kishikawa H, Hu JK, Sadow PM, Li M, and Hu GF
- Subjects
- Animals, Antibiotics, Antineoplastic pharmacology, Cell Proliferation drug effects, Dactinomycin pharmacology, Male, Mice, Neomycin pharmacology, Prostatic Intraepithelial Neoplasia drug therapy, Prostatic Intraepithelial Neoplasia metabolism, Prostatic Intraepithelial Neoplasia pathology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms enzymology, Prostatic Neoplasms pathology, Protein Synthesis Inhibitors pharmacology, RNA, Small Interfering genetics, Ribonuclease, Pancreatic antagonists & inhibitors, Ribonuclease, Pancreatic genetics, Ribonuclease, Pancreatic metabolism, Transcription, Genetic drug effects, Transcriptional Activation, Up-Regulation drug effects, Oncogene Protein v-akt metabolism, Prostatic Intraepithelial Neoplasia genetics, Prostatic Neoplasms genetics, RNA, Ribosomal genetics, Ribonuclease, Pancreatic biosynthesis
- Abstract
Angiogenin (ANG), originally identified as an angiogenic ribonuclease, has recently been shown to play a direct role in prostate cancer cell proliferation by mediating rRNA transcription. ANG is up-regulated in human prostate cancer and is the most significantly up-regulated gene in AKT-driven prostate intraepithelial neoplasia (PIN) in mice. Enhanced cell proliferation in the PIN lesions requires increased ribosome biogenesis, a multistep process involving an orchestrated production of ribosomal proteins and rRNA. AKT is known to enhance ribosomal protein production through the mammalian target of rapamycin pathway. However, it was unknown how rRNA is proportionally increased. Here, we report that ANG is essential for AKT-driven PIN formation and survival. We showed that up-regulation of ANG in the AKT-overexpressing mouse prostates is an early and lasting event. It occurs before PIN initiation and lasts beyond PIN is fully developed. Knocking down ANG expression by intraprostate injection of lentivirus-mediated ANG-specific small interfering RNA prevents AKT-induced PIN formation without affecting AKT expression and its signaling through the mammalian target of rapamycin pathway. Neomycin, an aminoglycoside that blocks nuclear translocation of ANG, and N65828, a small-molecule enzymatic inhibitor of the ribonucleolytic activity of ANG, both prevent AKT-induced PIN formation and reverse established PIN. They also decrease nucleolar organizer region, restore cell size, and normalize luminal architectures of the prostate despite continuous activation of AKT. All three types of the ANG inhibitor suppress rRNA transcription of the prostate luminal epithelial cells and inhibit AKT-induced PIN, indicating an essential role of ANG in AKT-mediated cell proliferation and survival.
- Published
- 2009
- Full Text
- View/download PDF
134. Epithelial-mesenchymal transition induced by growth suppressor p12CDK2-AP1 promotes tumor cell local invasion but suppresses distant colony growth.
- Author
-
Tsuji T, Ibaragi S, Shima K, Hu MG, Katsurano M, Sasaki A, and Hu GF
- Subjects
- Animals, Cadherins biosynthesis, Cadherins genetics, Cricetinae, Epithelial Cells pathology, Humans, Keratinocytes pathology, Keratinocytes physiology, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Mesoderm pathology, Mice, Mice, Inbred BALB C, Mice, Nude, Mouth Neoplasms genetics, Mouth Neoplasms metabolism, Neoplasm Invasiveness, Neoplasm Metastasis, Protein Kinases biosynthesis, Protein Kinases genetics, Transfection, Transforming Growth Factor beta pharmacology, Tumor Suppressor Proteins biosynthesis, Tumor Suppressor Proteins genetics, Lung Neoplasms secondary, Mouth Neoplasms pathology, Protein Kinases physiology, Tumor Suppressor Proteins physiology
- Abstract
Epithelial-mesenchymal transition (EMT) has been considered essential for metastasis, a multistep process including local invasion, intravasation, extravasation, and proliferation at distant sites. However, controversy remains as to whether EMT truly happens and how important it is to metastasis. We studied the involvement of EMT in individual steps of metastasis and found that p12(CDK2-AP1), a down-stream effector of transforming growth factor beta, induced EMT of hamster cheek pouch carcinoma-1 cells by promoting the expression of Twist2. EMT cells have an increased invasive but decreased metastatic phenotype. When s.c. inoculated, both EMT and non-EMT cells established primary tumors, but only EMT cells invaded into the adjacent tissues and blood vessels; however, neither cells formed lung metastases. When i.v. inoculated, only non-EMT cells established lung metastases. Moreover, s.c. inoculation of a mixture of the two cell types resulted in intravasation of both cell types and formation of lung metastasis from non-EMT cells. Our results allowed us to propose a novel model for the role of EMT in cancer metastasis. We showed that EMT and non-EMT cells cooperate to complete the spontaneous metastasis process. We thus hypothesize that EMT cells are responsible for degrading the surrounding matrix to lead the way of invasion and intravasation. Non-EMT cells then enter the blood stream and reestablish colonies in the secondary sites.
- Published
- 2008
- Full Text
- View/download PDF
135. Nicotine promotes mammary tumor migration via a signaling cascade involving protein kinase C and CDC42.
- Author
-
Guo J, Ibaragi S, Zhu T, Luo LY, Hu GF, Huppi PS, and Chen CY
- Subjects
- Animals, Breast drug effects, Breast enzymology, Cell Cycle drug effects, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Humans, Lung Neoplasms secondary, Mice, Breast Neoplasms pathology, Nicotine toxicity, Protein Kinase C-alpha physiology, Signal Transduction drug effects, cdc42 GTP-Binding Protein physiology
- Abstract
Nicotine, one of the major components in tobacco, is at high concentrations in the bloodstream of cigarette smokers. However, the mechanisms of how nicotine affects tumor development and whether nicotine is a potential carcinogen for malignancies induced by secondhand smoking are not fully understood yet. Here, we investigate the signaling pathways by which nicotine potentiates tumorigenesis in human mammary epithelial-like MCF10A or cancerous MCF7 cells. We show that human MCF10A and MCF7 cells both express four subunits of nicotine acetylcholine receptor (nAChR). The treatment of these cells with nicotine enhances the activity of protein kinase C (PKC) alpha without altering the expression level of this kinase. Nicotine also stimulates [(3)H]thymidine incorporation into the genome of these cells as well as forces serum-starved cells to enter S phase of the cell cycle, resulting in growth promotion. Importantly, on nicotine treatment, the mobility of MCF10A and MCF7 cells is enhanced, which can be blocked by the addition of nAChR or PKC inhibitor. Experiments using small interfering RNA knockdown or ectopic expression of cdc42 showed that cdc42 functions as a downstream effector of PKC and is crucial in the regulation of nicotine-mediated migratory activity in the cells. Together, our findings suggest that nicotine, through interacting with its receptor, initiates a signaling cascade that involves PKC and cdc42 and consequently promotes migration in mammary epithelial or tumor cells.
- Published
- 2008
- Full Text
- View/download PDF
136. Novel transcription-factor-like function of human matrix metalloproteinase 3 regulating the CTGF/CCN2 gene.
- Author
-
Eguchi T, Kubota S, Kawata K, Mukudai Y, Uehara J, Ohgawara T, Ibaragi S, Sasaki A, Kuboki T, and Takigawa M
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cell Line, Tumor metabolism, Cell Nucleus metabolism, Connective Tissue Growth Factor, Consensus Sequence, Female, Humans, Immediate-Early Proteins biosynthesis, Intercellular Signaling Peptides and Proteins biosynthesis, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Osteoarthritis metabolism, Protease Inhibitors pharmacology, Rats, Rats, Wistar, Recombinant Fusion Proteins physiology, Sequence Alignment, Sequence Homology, Nucleic Acid, Chondrocytes metabolism, Enhancer Elements, Genetic genetics, Gene Expression Regulation genetics, Immediate-Early Proteins genetics, Intercellular Signaling Peptides and Proteins genetics, Matrix Metalloproteinase 3 physiology
- Abstract
Matrix metalloproteinase 3 (MMP3) is well known as a secretory endopeptidase that degrades extracellular matrices. Recent reports indicated the presence of MMPs in the nucleus (A. J. Kwon et al., FASEB J. 18:690-692, 2004); however, its function has not been well investigated. Here, we report a novel function of human nuclear MMP3 as a trans regulator of connective tissue growth factor (CCN2/CTGF). Initially, we cloned MMP3 cDNA as a DNA-binding factor for the CCN2/CTGF gene. An interaction between MMP3 and transcription enhancer dominant in chondrocytes (TRENDIC) in the CCN2/CTGF promoter was confirmed by a gel shift assay and chromatin immunoprecipitation. The CCN2/CTGF promoter was activated by overexpressed MMP3, whereas a TRENDIC mutant promoter lost the response. Also, the knocking down of MMP3 suppressed CCN2/CTGF expression. By cytochemical and histochemical analyses, MMP3 was detected in the nuclei of chondrocytic cells in culture and also in the nuclei of normal and osteoarthritic chondrocytes in vivo. The nuclear translocation of externally added recombinant MMP3 and six putative nuclear localization signals in MMP3 also were shown. Furthermore, we determined that heterochromatin protein gamma coordinately regulates CCN2/CTGF by interacting with MMP3. The involvement of this novel role of MMP3 in the development, tissue remodeling, and pathology of arthritic diseases through CCN2/CTGF regulation thus is suggested.
- Published
- 2008
- Full Text
- View/download PDF
137. SOX9 is expressed in human fetal prostate epithelium and enhances prostate cancer invasion.
- Author
-
Wang H, Leav I, Ibaragi S, Wegner M, Hu GF, Lu ML, Balk SP, and Yuan X
- Subjects
- Animals, Cell Growth Processes physiology, Cell Line, Tumor, Down-Regulation, Epithelial Cells metabolism, High Mobility Group Proteins genetics, Humans, Male, Mice, Mice, Inbred ICR, Mice, SCID, Neoplasm Invasiveness, Neovascularization, Pathologic metabolism, Prostatic Neoplasms blood supply, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, RNA, Small Interfering genetics, SOX9 Transcription Factor, Transcription Factors genetics, High Mobility Group Proteins biosynthesis, Prostate embryology, Prostate metabolism, Prostatic Neoplasms metabolism, Transcription Factors biosynthesis
- Abstract
SOX9 is a transcription factor that plays a critical role in the development of multiple tissues. We previously reported that SOX9 in normal human adult prostate was restricted to basal epithelium. SOX9 was also expressed in a subset of prostate cancer (PCa) cells and was increased in relapsed hormone-refractory PCa. Moreover, SOX9 expression in PCa cell lines enhanced tumor cell proliferation and was beta-catenin regulated. Here we report additional in vivo results showing that SOX9 is highly expressed during fetal prostate development by epithelial cells expanding into the mesenchyme, suggesting it may contribute to invasive growth in PCa. Indeed, SOX9 overexpression in LNCaP PCa xenografts enhanced growth, angiogenesis, and invasion. Conversely, short hairpin RNA-mediated SOX9 suppression inhibited the growth of CWR22Rv1 PCa xenografts. These results support important functions of SOX9 in both the development and maintenance of normal prostate, and indicate that these functions contribute to PCa tumor growth and invasion.
- Published
- 2008
- Full Text
- View/download PDF
138. Specific inhibitor of MEK-mediated cross-talk between ERK and p38 MAPK during differentiation of human osteosarcoma cells.
- Author
-
Shimo T, Matsumura S, Ibaragi S, Isowa S, Kishimoto K, Mese H, Nishiyama A, and Sasaki A
- Abstract
Osteosarcoma is the most common primary malignant bone tumor, accounting for approximately 20% of all primary sarcomas in bone. Although treatment modalities have been improved over the past decades, it is still a tumor with a high mortality rate in children and young adults. Based on histological considerations, osteosarcoma arises from impaired differentiation of these immature cells into more mature types and that correction of this impairment may reduce malignancy and increase the efficiency of chemotherapy. The purpose of this study was to determine the effect of specific inhibitors of MAPK extracellular signaling-regulated kinase (ERK) kinase (MEK) and p38 on the differentiation of human osteosarcoma cell line SaOS-2 cells. We found that PD98059, a specific inhibitor of MEK, inhibited the serum-stimulated proliferation of SaOS-2 cells; whereas SB203580, a specific inhibitor of p38 MAPK, had little effect on it. SB203580 suppressed ALPase activity, gene expression of type I collagen, and expression of ALP and BMP-2 mRNAs; whereas PD98059 upregulated them dose dependently. In addition, immunoblot and immunostaining analysis revealed that phosphorylation of ERK was increased by treatment with SB203580; whereas PD98059 increased the phosphorylation of p38, which implies a seesaw-like balance between ERK and p38 phosphorylation. We suggest that osteosarcoma cell differentiation is regulated by the balance between the activities of the ERK and p38 pathways and that the MEK/ERK pathway negatively regulates osteosarcoma cell differentiation, whereas the p38 pathway does so positively. MEK inhibitor may thus be a good candidate for altering the expression of the osteosarcoma malignant phenotype.
- Published
- 2007
- Full Text
- View/download PDF
139. Ornithine decarboxylase antizyme upregulates DNA-dependent protein kinase and enhances the nonhomologous end-joining repair of DNA double-strand breaks in human oral cancer cells.
- Author
-
Tsuji T, Katsurano M, Ibaragi S, Shima K, Sasaki A, and Hu GF
- Subjects
- Antigens, Nuclear genetics, Antigens, Nuclear metabolism, Azacitidine analogs & derivatives, Azacitidine pharmacology, Cadherins genetics, Cadherins metabolism, Carrier Proteins genetics, Carrier Proteins metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Cell Line, Tumor, Cell Nucleus genetics, Cell Nucleus metabolism, DNA-Activated Protein Kinase genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Decitabine, G1 Phase genetics, Gamma Rays, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Neoplastic radiation effects, Histones genetics, Histones metabolism, Humans, Keratinocytes metabolism, Ku Autoantigen, Mouth Neoplasms genetics, Mouth Neoplasms metabolism, Mouth Neoplasms pathology, Proteins genetics, Transfection, Up-Regulation drug effects, Up-Regulation radiation effects, DNA Breaks, Double-Stranded, DNA Repair, DNA-Activated Protein Kinase metabolism, Proteins metabolism
- Abstract
Ornithine decarboxylase (ODC) antizyme targets ODC for ubiquitin-independent proteosome degradation, thereby inhibiting polyamine synthesis. It has been shown to regulate DNA methylation and has tumor suppressor activity. Increasing evidence suggested that antizyme may also have ODC-independent functions. Here, we report that antizyme plays a role in DNA double-strand break repairs. A zinc-inducible human antizyme gene expression vector was transfected into UM1 human oral squamous cancer cells that do not express endogenous antizyme. The resultant upregulated genes were screened by cDNA arrays and confirmed by quantitative real-time polymerase chain reaction. DNA-dependent protein kinase including its catalytic subunit DNA-PKcs and regulatory subunit Ku70, two key proteins of the DNA damage repair machinery, was significantly upregulated after ectopic expression of antizyme. Consistently, we found that UM1 cells are sensitive to gamma irradiation and deficient in DNA damage repairs, as shown by radio-sensitivity and Comet assays. Ectopic expression of antizyme increased radio-resistance of UM1 cells and restored their capacity of DNA damage repairs to the level of UM2 cells that have an identical genetic background but express endogenous antizyme. Plasmid end-joining assays confirmed that antizyme enhances the ability of UM1 cells to repair DNA double-strand breaks by the nonhomologous end-joining pathway.
- Published
- 2007
- Full Text
- View/download PDF
140. [A case of acute respiratory distress syndrome (ARDS) induced by docetaxel administration for lung metastases from oral cancer].
- Author
-
Mese H, Yoshihama Y, Nakayama S, Ibaragi S, and Sasaki A
- Subjects
- Aged, 80 and over, Carcinoma, Squamous Cell secondary, Dexamethasone administration & dosage, Docetaxel, Gingival Neoplasms surgery, Humans, Lung Neoplasms secondary, Male, Mandible surgery, Neck Dissection, Antineoplastic Agents adverse effects, Carcinoma, Squamous Cell drug therapy, Gingival Neoplasms pathology, Lung Neoplasms drug therapy, Respiratory Distress Syndrome chemically induced, Taxoids adverse effects
- Abstract
We reported a case of acute respiratory distress syndrome (ARDS) induced by docetaxel in treating lung metastases from oral cancer. The patient was an 84-year-old man who had undergone partial mandibulectomy and radical neck dissection for lower gingival carcinoma. The patient developed ARDS after docetaxel administration (40 mg/body) for multiple lung metastases.
- Published
- 2007
141. [A successful case of lower gingival cancer with pulmonary metastases by adjuvant chemotherapy including paclitaxel, cisplatin and 5-fluorouracil following a surgical procedure].
- Author
-
Tsukamoto G, Ibaragi S, Shimo T, Oyama K, Kishimoto K, Mese H, Aoe M, Kiura K, and Sasaki A
- Subjects
- Carcinoma, Squamous Cell secondary, Carcinoma, Squamous Cell surgery, Chemotherapy, Adjuvant, Cisplatin administration & dosage, Combined Modality Therapy, Drug Administration Schedule, Female, Fluorouracil administration & dosage, Gingival Neoplasms pathology, Gingival Neoplasms surgery, Humans, Lung Neoplasms surgery, Lymphatic Metastasis, Middle Aged, Paclitaxel administration & dosage, Remission Induction, Thoracic Surgery, Video-Assisted, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Squamous Cell drug therapy, Gingival Neoplasms drug therapy, Lung Neoplasms secondary, Lymph Nodes pathology, Pneumonectomy methods
- Abstract
We report a successful case with pulmonary metastases from lower gingival cancer by a surgical procedure and four cycles of adjuvant chemotherapy including paclitaxel (PTX), cisplatin (CDDP) and 5-fluorouracil (5-FU). A 47-year-old woman underwent chemotherapy with CDDP and 5-FU after an operation for lower gingival squamous cell carcinoma and its neck lymph node metastases. At 4 months from the initial treatment, pulmonary metastatic lesion was resected by video-assisted thoracoscopic surgery (VATS). Fourteen months later, pulmonary metastatic lesion was found and dissected again using VATS. Furthermore, the patient was treated by adjuvant chemotherapy with PTX 135 mg/m(2) over 3 hours on day 1, CDDP 75 mg/m(2)on day 2 and 5-FU 350 mg/m(2)/day by continuous intravenous infusion on day 2 through 5. After that, there is no evidence of pulmonary recurrence for more than six years.
- Published
- 2007
142. Pathogenic role of connective tissue growth factor (CTGF/CCN2) in osteolytic metastasis of breast cancer.
- Author
-
Shimo T, Kubota S, Yoshioka N, Ibaragi S, Isowa S, Eguchi T, Sasaki A, and Takigawa M
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibodies, Neoplasm pharmacology, Bone Neoplasms drug therapy, Bone Neoplasms pathology, Bone Neoplasms secondary, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Cell Line, Tumor, Connective Tissue Growth Factor, Drug Design, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Immediate-Early Proteins antagonists & inhibitors, Mice, Neoplasm Metastasis, Neoplasm Proteins antagonists & inhibitors, Neoplasm Transplantation, Neoplasms, Experimental drug therapy, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Osteolysis pathology, Parathyroid Hormone-Related Protein biosynthesis, Protein Kinases metabolism, Bone Neoplasms metabolism, Breast Neoplasms metabolism, Immediate-Early Proteins biosynthesis, Intercellular Signaling Peptides and Proteins biosynthesis, Neoplasm Proteins biosynthesis, Osteolysis metabolism, Signal Transduction
- Abstract
Unlabelled: The role of CTGF/CCN2 in osteolytic metastasis by breast cancer cells and its mechanism of action were studied. Osteolytic metastasis accompanied by CCN2 and PTHrP overproduction was efficiently inhibited by an anti-CCN2 antibody. Furthermore, we found that CCN2 was induced by PTHrP through PKA-, PKC-, and ERK-mediated pathways therein., Introduction: Connective tissue growth factor (CTGF/CCN2) is a mediator of local angiogenesis induced by breast cancer, but its role in osteolytic metastasis has not been evaluated. PTH-related peptide (PTHrP) is another critical factor in the development of the osteolytic metastasis. Using both in vivo and in vitro approaches, we studied whether/how neutralization of CCN2 prevented bone metastasis and how PTHrP signaling is related., Materials and Methods: A mouse model of bone metastasis by human breast cancer cell line MDA231 was treated with a CCN2-neutralizing antibody, and osteolytic bone metastases were assessed on radiographs and immunohistochemistry. Ccn2 gene expression and transcription were examined by Northern blot and luciferase analysis. Immunoblot analysis and kinase inhibitors were used to identify the signaling pathways implicated. Anti-angiogenic/osteoclastogenic effects of ccn2 downregulation were also evaluated., Results: Treatment of mice with a CCN2-neutralizing antibody greatly decreased osteolytic bone metastasis, microvasculature, and osteoclasts involved. The antibody also suppressed the growth of subcutaneous tumor in vivo and proliferation and migration of human umbilical vein endothelial cells (HUVECs) in vitro. Downregulation of ccn2 also repressed osteoclastogenesis. CCN2 expression was specifically observed in cancer cells producing PTHrP and type I PTH/PTHrP receptor (PTH1R) invaded the bone marrow, and PTHrP strongly upregulated ccn2 in MDA231 cells in vitro. Activation of protein kinase C (PKC) and protein kinase A (PKA) was necessary and sufficient for the stimulation of ccn2 by PTHrP. Indeed, inhibition of the extracellular signal-regulated kinase (ERK1/2), PKC, or PKA by specific inhibitors counteracted the stimulation of ccn2 expression. Incubation of MDA231 cells with PTHrP induced the activation of ERK1/2. Consistent with these findings, inhibition of PKC prevented PTHrP-induced ERK1/2 activation, whereas 12-O-tetradecanoylphorbol13-acetate (TPA), a stimulator of PKC, upregulated it., Conclusions: CCN2 was critically involved in osteolytic metastasis and was induced by PKA- and PKC-dependent activation of ERK1/2 signaling by PTHrP. Thus, CCN2 may be a new molecular target for anti-osteolytic therapy to shut off the PTHrP-CCN2 signaling pathway.
- Published
- 2006
- Full Text
- View/download PDF
143. Synthesis and secretion of an hEGF-like immunoreactive factor by human gastric cancer cells (MKN-45).
- Author
-
Mori K, Ibaragi S, Kurobe M, Furukawa S, and Hayashi K
- Subjects
- Chromatography, Gel, Culture Media, Epidermal Growth Factor biosynthesis, Epidermal Growth Factor immunology, Humans, Isoelectric Focusing, Molecular Weight, Peptides isolation & purification, Stomach Neoplasms analysis, Tumor Cells, Cultured, Epidermal Growth Factor metabolism, Stomach Neoplasms metabolism
- Abstract
A large amount of an immunoreactive factor was detected in the medium conditioned by human gastric cancer cells, strain MKN-45, by our enzyme immunoassay system for human epidermal growth factor (hEGF) based on hEGF isolated from urine. However, the dose-response curve of the immunoreactive factor (designated as MKN-45 EGF) was not parallel with the standard curve of hEGF. The molecular weight of MKN-45 EGF was slightly larger than that of hEGF and was estimated to be 7,000-8,000 by gel filtration on Sephadex G-50. On isoelectric focusing analysis, MKN-45 EGF gave a major peak at pH 5.0 and a minor one at pH 4.3. These results demonstrate that MKN-45 cells synthesize and secrete into the culture medium a polypeptide immunologically related to hEGF.
- Published
- 1987
144. Production of an hEGF-like immunoreactive factor by human gastric cancer cells depends on differentiational state of the cells.
- Author
-
Mori K, Ibaragi S, Kurobe M, Furukawa S, and Hayashi K
- Subjects
- Animals, Cell Differentiation, Cell Line, Cells, Cultured, Culture Media, DNA Replication drug effects, Epidermal Growth Factor pharmacology, Humans, Immunoenzyme Techniques, Mice, Mice, Inbred BALB C, Stomach Neoplasms pathology, Epidermal Growth Factor metabolism, Stomach Neoplasms metabolism
- Abstract
We have extended our recent observation that human gastric cancer cells (MKN-45) synthesize and secrete an hEGF-like immunoreactive factor (designated as EGF-LI) by characterization of EGF-LI produced by five human gastric cancer cell lines in culture. Two cell lines (MKN-45 and KATO-III) derived from poorly differentiated adenocarcinoma synthesized and secreted a much larger amount of EGF-LI than three cell lines (MKN-1, MKN-28, and MKN-74) derived from well-differentiated adenocarcinoma. Treatment of MKN-45 cells by retinoic acid reduced significantly synthesis and secretion of EGF-LI, suggesting that production of EGF-LI is dependent on differentiational state of gastric cancer cells.
- Published
- 1987
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.