Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Oncol Rep ; 49(3)2023 03.
Article in English | MEDLINE | ID: mdl-36799199

ABSTRACT

Colon cancer is one of the most frequent malignant neoplasms worldwide. Epidemiological studies suggested that the development of colon cancer can be prevented by plant­derived ingredients. In the present study, the chemopreventive activity of buddlejasaponin IV (BS­IV), isolated from the aerial part of Pleurospermum kamtschaticum, was investigated using cell viability, DNA fragmentation, caspase­3 activity, anoikis, cell adhesion, and flow cytometry assays and a murine lung metastasis model. Protein expression levels were detected by western blotting. Treatment with BS­IV significantly reduced cell viability and caused DNA fragmentation in HT­29 human colorectal cancer cells. BS­IV increased the ratio of Bax to Bcl­2 by significantly inhibiting Bcl­2 expression levels. BS­IV reduced expression levels of procaspase­9, procaspase­3, and full­length poly (ADP­ribose) polymerase (PARP) and increased cleaved PARP and nonsteroidal anti­inflammatory drug activated gene­1 expression levels and caspase­3 activity. In addition, BS­IV decreased the attachment of HT­29 cells to the extracellular matrix proteins collagen type I and IV and downregulated cell surface expression of α2ß1 integrin by inhibiting its glycosylation. BS­IV also reduced the expression and phosphorylation levels of focal adhesion kinase (FAK) and Akt, and the reduced FAK and Akt levels were rescued by treatment with a caspase­3 inhibitor Z­VAD­FMK. Furthermore, orally administered BS­IV inhibited the formation of tumor nodules in Balb/C mice intravenously injected with CT­26 murine colorectal cancer cells. Collectively, these findings indicated that BS­IV induces apoptosis via the mitochondrial­dependent pathway by increasing the ratio of Bax to Bcl­2 and activating caspases. BS­IV also induces anoikis by inhibiting α2ß1 integrin­mediated cell adhesion and signaling and inhibits the lung metastasis of colon cancer cells. Therefore, BS­IV may serve as a promising cancer chemopreventive agent.


Subject(s)
Colonic Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Animals , Mice , HT29 Cells , Caspase 3 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , bcl-2-Associated X Protein , Apoptosis , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Cell Adhesion , Anoikis , Integrins/metabolism
2.
Am J Chin Med ; 50(6): 1645-1661, 2022.
Article in English | MEDLINE | ID: mdl-35848124

ABSTRACT

Platycodin D is a major constituent in the root of Platycodon grandiflorum and has diverse pharmacologic activities, including anti-inflammatory, anti-allergic, and antitumor activities. Vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8) are potent angiogenic factors and contribute to tumor angiogenesis by directly and indirectly promoting angiogenic processes, including the proliferation, adhesion, migration, and tube formation of endothelial cells. Here, we found that platycodin D at noncytotoxic concentrations inhibited VEGF-induced proliferation, adhesion to the extracellular matrix proteins fibronectin and vitronectin, chemotactic motility, and tube formation of human umbilical vein endothelial cells (HUVECs). Platycodin D reduced the phosphorylation of extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK) and the secretion of IL-8 in VEGF-stimulated HUVECs. Moreover, platycodin D inhibited tube formation and the phosphorylation of ERK and p38 in IL-8-stimulated HUVECs. The in vitro anti-angiogenic activity of platycodin D was confirmed by in vivo experimental models. Platycodin D inhibited the formation of new blood vessels into mouse Matrigel plugs with VEGF or IL-8. In mice injected with MDA-MB-231 human breast cancer cells, orally administered platycodin D inhibited tumor growth, the number of CD34 [Formula: see text]vessels, and the expression of VEGF and IL-8. Taken together, platycodin D directly and indirectly prevents VEGF-induced and IL-8-induced angiogenesis by blocking the activation of mitogen-activated protein kinases (MAPKs). Platycodin D may be beneficial for the prevention or treatment of tumor angiogenesis and angiogenesis-related human diseases.


Subject(s)
Interleukin-8 , Vascular Endothelial Growth Factor A , Angiogenesis Inhibitors/pharmacology , Animals , Cell Movement , Extracellular Signal-Regulated MAP Kinases/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-8/metabolism , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/prevention & control , Saponins , Triterpenes , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factors/metabolism , Vascular Endothelial Growth Factors/pharmacology
3.
Biomol Ther (Seoul) ; 30(3): 284-290, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35110423

ABSTRACT

Oral squamous cell carcinoma (OSCC) is mostly diagnosed at an advanced stage, with local and/or distal metastasis. Thus, locoregional and/or local control of the primary tumor is crucial for a better prognosis in patients with OSCC. Platelets have long been considered major players in cancer metastasis. Traditional antiplatelet agents, such as aspirin, are thought to be potential chemotherapeutics, but they need to be used with caution because of the increased bleeding risk. Podoplanin (PDPN)-expressing cancer cells can activate platelets and promote OSCC metastasis. However, the reciprocal effect of platelets on PDPN expression in OSCC has not been investigated. In this study, we found that direct contact with platelets upregulated PDPN and integrin ß1 at the protein level and promoted invasiveness of human OSCC Ca9.22 cells that express low levels of PDPN. In another human OSCC HSC3 cell line that express PDPN at an abundant level, silencing of the PDPN gene reduced cell invasiveness. Analysis of the public database further supported the co-expression of PDPN and integrin ß1 and their increased expression in metastatic tissues compared to normal and tumor tissues of the oral cavity. Taken together, these data suggest that PDPN is a potential target to regulate platelet-tumor interaction and metastasis for OSCC treatment, which can overcome the limitations of traditional antiplatelet drugs.

4.
Cancers (Basel) ; 13(9)2021 Apr 28.
Article in English | MEDLINE | ID: mdl-33924899

ABSTRACT

It is well-known that microbiota dysbiosis is closely associated with numerous diseases in the human body. The oral cavity and gut are the two largest microbial habitats, playing a major role in microbiome-associated diseases. Even though the oral cavity and gut are continuous regions connected through the gastrointestinal tract, the oral and gut microbiome profiles are well-segregated due to the oral-gut barrier. However, the oral microbiota can translocate to the intestinal mucosa in conditions of the oral-gut barrier dysfunction. Inversely, the gut-to-oral microbial transmission occurs as well in inter- and intrapersonal manners. Recently, it has been reported that oral and gut microbiomes interdependently regulate physiological functions and pathological processes. Oral-to-gut and gut-to-oral microbial transmissions can shape and/or reshape the microbial ecosystem in both habitats, eventually modulating pathogenesis of disease. However, the oral-gut microbial interaction in pathogenesis has been underappreciated to date. Here, we will highlight the oral-gut microbiome crosstalk and its implications in the pathogenesis of the gastrointestinal disease and cancer. Better understanding the role of the oral-gut microbiome axis in pathogenesis will be advantageous for precise diagnosis/prognosis and effective treatment.

5.
Am J Chin Med ; 49(3): 737-751, 2021.
Article in English | MEDLINE | ID: mdl-33683188

ABSTRACT

Angiogenesis plays a crucial role in tumor growth and metastasis. Vascular endothelial growth factor (VEGF)-stimulated endothelial cell proliferation and migration are critical steps in tumor angiogenesis. Here, we investigated the anti-angiogenic activity of xanthorrhizol, a sesquiterpenoid isolated from the Indonesian medicinal plant Curcuma xanthorrhiza. Xanthorrhizol at noncytotoxic concentrations inhibited the proliferation, migration, and formation of capillary-like tubes in VEGF-treated human umbilical vein endothelial cells (HUVECs). Xanthorrhizol inhibited the phosphorylation of Akt and endothelial nitric oxide synthase (eNOS) and the expression of vascular cell adhesion molecule (VCAM)-1 and E-selectin in VEGF-treated HUVECs. The expression and transcriptional activity of NF-[Formula: see text]B were downregulated by xanthorrhizol in VEGF-treated HUVECs. Furthermore, xanthorrhizol significantly inhibited VEGF-induced angiogenesis in the chorioallantoic membrane of fertilized eggs and Matrigel plugs subcutaneously injected into mice. Xanthorrhizol inhibited tumor volume and tumor-derived angiogenesis in mice inoculated with breast cancer cells. The in vitro and in vivo anti-angiogenic activities of xanthorrhizol were as potent as those of curcumin, a well-known anticancer agent derived from C. longa. Taken together, xanthorrhizol inhibits VEGF-induced angiogenesis of endothelial cells by blocking the activation of the PI3K/Akt/eNOS axis and subsequent upregulation of adhesion molecules induced by the transcriptional activation of NF-[Formula: see text]B. Xanthorrhizol is a promising anti-angiogenic agent and can serve as a beneficial agent to enhance anticancer treatments.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Gene Expression/drug effects , Gene Expression/genetics , NF-kappa B/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/prevention & control , Nitric Oxide Synthase Type III/metabolism , Phenols/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/adverse effects , Animals , Curcuma/chemistry , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neovascularization, Pathologic/chemically induced , Phenols/isolation & purification , Phenols/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Phytotherapy , Tumor Cells, Cultured
6.
Eur J Oral Sci ; 129(1): e12750, 2021 02.
Article in English | MEDLINE | ID: mdl-33503283

ABSTRACT

Patients with oral squamous cell carcinoma (OSCC) bone invasion are surgically treated with bone resection, which results in severe physical and psychological damage. Here, we investigated the potential of fractalkine (CX3CL1), which is regulated by transforming growth factor (TGF-ß), as a novel biomarker for correct prediction and early detection of OSCC-associated bone invasion. TGF-ß knockdown and treatment with a TGF-ß-neutralizing antibody decreased the level of fractalkine in the culture media of HSC-2 and YD10B OSCC cells. Treatment with a fractalkine-neutralizing antibody reduced TGF-ß-stimulated invasion by HSC-2 and YD10B cells. Fractalkine treatment increased the viability, invasion, and uPA secretion of both OSCC cell lines. Furthermore, OSCC cell bone invasion was assessed following subcutaneous inoculation of wild-type or TGF-ß knockdown OSCC cells in mouse calvaria. TGF-ß knockdown prevented erosive bone invasion, reduced the number of osteoclasts at the tumor-bone interface, and downregulated fractalkine expression in mouse tumor tissues. Our results indicate that the production of fractalkine is stimulated by TGF-ß and mediates TGF-ß-induced cell invasion in several OSCC cell lines showing an erosive pattern of bone invasion. Fractalkine may be a useful predictive marker and therapeutic target for OSCC-induced bone destruction.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Animals , Biomarkers , Cell Line, Tumor , Chemokine CX3CL1 , Humans , Mice , Neoplasm Invasiveness , Squamous Cell Carcinoma of Head and Neck , Transforming Growth Factor beta , Transforming Growth Factor beta1
7.
Front Immunol ; 12: 807600, 2021.
Article in English | MEDLINE | ID: mdl-34987523

ABSTRACT

Cancer tissues are not just simple masses of malignant cells, but rather complex and heterogeneous collections of cellular and even non-cellular components, such as endothelial cells, stromal cells, immune cells, and collagens, referred to as tumor microenvironment (TME). These multiple players in the TME develop dynamic interactions with each other, which determines the characteristics of the tumor. Platelets are the smallest cells in the bloodstream and primarily regulate blood coagulation and hemostasis. Notably, cancer patients often show thrombocytosis, a status of an increased platelet number in the bloodstream, as well as the platelet infiltration into the tumor stroma, which contributes to cancer promotion and progression. Thus, platelets function as one of the important stromal components in the TME, emerging as a promising chemotherapeutic target. However, the use of traditional antiplatelet agents, such as aspirin, has limitations mainly due to increased bleeding complications. This requires to implement new strategies to target platelets for anti-cancer effects. In oral squamous cell carcinoma (OSCC) patients, both high platelet counts and low tumor-stromal ratio (high stroma) are strongly correlated with increased metastasis and poor prognosis. OSCC tends to invade adjacent tissues and bones and spread to the lymph nodes for distant metastasis, which is a huge hurdle for OSCC treatment in spite of relatively easy access for visual examination of precancerous lesions in the oral cavity. Therefore, locoregional control of the primary tumor is crucial for OSCC treatment. Similar to thrombocytosis, higher expression of podoplanin (PDPN) has been suggested as a predictive marker for higher frequency of lymph node metastasis of OSCC. Cumulative evidence supports that platelets can directly interact with PDPN-expressing cancer cells via C-type lectin-like receptor 2 (CLEC2), contributing to cancer cell invasion and metastasis. Thus, the platelet CLEC2-PDPN axis could be a pinpoint target to inhibit interaction between platelets and OSCC, avoiding undesirable side effects. Here, we will review the role of platelets in cancer, particularly focusing on CLEC2-PDPN interaction, and will assess their potentials as therapeutic targets for OSCC treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Blood Platelets/drug effects , Cell Movement/drug effects , Lectins, C-Type/antagonists & inhibitors , Membrane Glycoproteins/antagonists & inhibitors , Mouth Neoplasms/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Antineoplastic Agents/adverse effects , Blood Platelets/metabolism , Humans , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Molecular Targeted Therapy , Mouth Neoplasms/blood , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Platelet Aggregation Inhibitors/adverse effects , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/blood , Squamous Cell Carcinoma of Head and Neck/secondary , Tumor Microenvironment
8.
Biochem Biophys Res Commun ; 534: 381-386, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33256984

ABSTRACT

Oral squamous cell carcinoma (OSCC) frequently invades nearby bone and bone involvement determines the prognosis of patients. Growth factors, stored in the bone matrix and released during bone destruction, are known as key components in the bone-tumor interaction. However, the coordination of growth factor signals and the precise mechanism of bone destruction in oral cancer are still unclear. In the study, we investigated the differential cytokine expression profile of oral cancer cells by TGF-ß treatment and the function of altered expression of cytokines on the osteoclast differentiation. We established TGFBR2-knockdown cells using small hairpin RNA. TGF-ß was treated to both TGFBR2 expressing and knockdown cells and the culture supernatants were analyzed using a cytokine array kit. We found that the TGF-ß inhibited IGFBP3 level and enhanced MMP9 level. We confirmed this regulation of IGFBP3 and MMP9 by TGF-ß using ELISA and zymography, respectively. IGFBP3 is known as to modulate the bioavailability of IGF1, which is abundant in the bone microenvironment and regulates osteoclast differentiation. Therefore, we further analyzed the function of IGFBP3 on osteoclastogenesis. Although IGFBP3 increased the viability of murine bone marrow macrophages, the osteoclast differentiation of these cells was blocked by IGFBP3 in a dose-dependent manner. These results revealed a novel pathway for the regulation of osteoclastogenesis by oral cancer cells, which may be a new therapeutic target for osteolysis induced by oral cancer infiltrating into the bone.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/metabolism , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line, Tumor , Down-Regulation , Gene Knockdown Techniques , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred ICR , Mouth Neoplasms/genetics , Osteogenesis/genetics , Osteogenesis/physiology , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Receptor, Transforming Growth Factor-beta Type II/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type II/genetics , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/genetics
9.
Int J Mol Sci ; 21(8)2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32325994

ABSTRACT

Chemerin is secreted as prochemerin from various cell types and then cleaved into the bioactive isoform by specific proteases. In various cancer types, chemerin exhibits pro- or antitumor effects. In the present study, chemerin treatment significantly inhibited the viability and invasion of breast cancer cells in the absence or presence of transforming growth factor (TGF)-ß and insulin-like growth factor (IGF)-1. The expression levels of E-cadherin and vimentin were reduced in chemerin-treated breast cancer cells. However, chemerin treatment recovered the reduced E-cadherin expression level in breast cancer cells treated with TGF-ß or IGF-1. Chemerin treatment inhibited nuclear ß-catenin levels in breast cancer cells stimulated with or without TGF-ß or IGF-1. In addition, chemerin treatment blocked the increase in the receptor activator of nuclear factor kappa-Β ligand (RANKL)/osteoprotegerin (OPG) ratio in osteoblastic cells exposed to metastatic breast cancer cell-derived conditioned medium. Chemerin treatment inhibited RANKL-induced osteoclast formation and bone resorption by reducing the secretion of matrix metalloproteinase (MMP)-2, MMP-9, and cathepsin K. Intraperitoneal administration of chemerin inhibited tumor growth in MCF-7 breast cancer cell-injected mice and reduced the development of osteolytic lesions resulting from intratibial inoculation of MDA-MB-231 cells. Taken together, chemerin inhibits the growth and invasion of breast cancer cells and prevents bone loss resulting from breast cancer cells by inhibiting finally osteoclast formation and activity.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/secondary , Chemokines/pharmacology , Animals , Biomarkers , Bone Neoplasms/diagnosis , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Disease Models, Animal , Female , Gene Expression , Humans , Immunophenotyping , Mice , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Xenograft Model Antitumor Assays
10.
J Clin Invest ; 129(12): 5381-5399, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31487270

ABSTRACT

Oral squamous cell carcinoma (OSCC) frequently invades the maxillary or mandibular bone, and this bone invasion is closely associated with poor prognosis and survival. Here, we show that CCL28 functions as a negative regulator of OSCC bone invasion. CCL28 inhibited invasion and epithelial-mesenchymal transition (EMT), and its inhibition of EMT was characterized by induced E-cadherin expression and reduced nuclear localization of ß-catenin in OSCC cells with detectable RUNX3 expression levels. CCL28 signaling via CCR10 increased retinoic acid receptor-ß (RARß) expression by reducing the interaction between RARα and HDAC1. In addition, CCL28 reduced RANKL production in OSCC and osteoblastic cells and blocked RANKL-induced osteoclastogenesis in osteoclast precursors. Intraperitoneally administered CCL28 inhibited tumor growth and osteolysis in mouse calvaria and tibia inoculated with OSCC cells. RARß expression was also increased in tumor tissues. In patients with OSCC, low CCL28, CCR10, and RARß expression levels were highly correlated with bone invasion. Patients with OSCC who had higher expression of CCL28, CCR10, or RARß had significantly better overall survival. These findings suggest that CCL28, CCR10, and RARß are useful markers for the prediction and treatment of OSCC bone invasion. Furthermore, CCL28 upregulation in OSCC cells or CCL28 treatment can be a therapeutic strategy for OSCC bone invasion.


Subject(s)
Bone and Bones/pathology , Chemokines, CC/pharmacology , Mouth Neoplasms/pathology , Receptors, Retinoic Acid/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Histone Deacetylase 1/physiology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Neoplasm Invasiveness , Osteoclasts/cytology , RANK Ligand/physiology , Receptors, CCR10/physiology , Retinoic Acid Receptor alpha/physiology
11.
PLoS One ; 14(5): e0216661, 2019.
Article in English | MEDLINE | ID: mdl-31071178

ABSTRACT

OBJECTIVES: This study aimed at investigating the molecular mechanism underlying PKM2-mediated cancer invasion. MATERIALS & METHODS: To optimize the investigation of PKM2-specific effects, we used two immortalized oral cell lines. The two cell lines drastically differed in PKM2 expression level, particularly in the level of nuclear PKM2, and subsequently in glucose metabolism and tumorigenicity. RESULTS: Knockdown of PKM2 reduced not only the glucose metabolism but also the invasive activity by curtailing the expressions of matrix metalloproteinases (MMP): PKM2 could modulate MMP-9 expression by regulating ETS-1 inside the nucleus. These results were further confirmed in an oral squamous cell carcinoma (OSCC) cell line. In correspondence with in vitro findings, clinicopathological data from OSCC patients indicated strong association between PKM2 expression and poor survival rate. Additionally, upon analysis of public database, significant positive correlation was found between PKM2 and ETS-1 in OSCC. CONCLUSION: Collectively, this study unveiled the molecular mechanism underlying PKM2-mediated cancer invasion, thereby providing novel targets for therapeutics development against invasive OSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carrier Proteins/metabolism , Matrix Metalloproteinase 9/biosynthesis , Membrane Proteins/metabolism , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Proto-Oncogene Protein c-ets-1/metabolism , Thyroid Hormones/metabolism , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Squamous Cell/genetics , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , Enzyme Induction , ErbB Receptors/metabolism , Female , Gene Expression , Gene Knockdown Techniques , Glucose/metabolism , Heterografts , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/virology , Male , Matrix Metalloproteinase 2/biosynthesis , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Middle Aged , Mouth Neoplasms/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thyroid Hormones/genetics , Transfection , Thyroid Hormone-Binding Proteins
12.
Cell Physiol Biochem ; 49(4): 1460-1475, 2018.
Article in English | MEDLINE | ID: mdl-30205376

ABSTRACT

BACKGROUND/AIMS: Bone metastasis of cancer cells decreases patient survival and quality of life. Hybridization via the covalent coupling of two bioactive natural products is a useful strategy for developing more potent anticancer agents by enhancing their bioavailability and avoiding drug resistance. METHODS: The in vivo activities of artemisinin-daumone hybrid 15 (ARTD) were estimated in cancer cell-inoculated mice and ovariectomized mice. The viability, migration, and invasion of cancer cells were measured via MTT, wound-healing, and transwell invasion assays. ARTD-regulated transcription factors were detected with an RT2 profiler PCR array kit and Western blotting. Osteoclastogenesis and osteoclast activity were detected with tartrate-resistant acid phosphatase staining, a pit formation assay, gelatin zymography, and a cathepsin K ELISA assay. RESULTS: ARTD blocked cancer-associated osteolysis more potently than artemisinin in mice with intratibially inoculated breast cancer or lung cancer cells. ARTD inhibited the viability, migration, and invasion of breast and lung cancer cells in the absence or presence of transforming growth factor-ß1. ARTD treatment induced the expression of tumor suppressive activating transcription factor 3 and inhibited oncogenic E2F transcription factor 1 expression at the mRNA and protein levels. ARTD inhibited receptor activator of nuclear factor kappa-B ligand-induced osteoclast formation and bone resorbing activity by reducing the secreted levels of matrix metalloproteinase-9 and cathepsin K. Furthermore, ARTD prevented estrogen deficiency-induced bone loss in ovariectomized mice. CONCLUSION: ARTD may be a promising candidate for inhibiting cancer-induced bone destruction. The application of ARTD may be extended to patients with chemotherapy-induced ovarian failure or postmenopausal osteoporosis.


Subject(s)
Artemisinins/chemistry , Bone Density Conservation Agents/therapeutic use , Fatty Acids/chemistry , Osteolysis/prevention & control , Pheromones/chemistry , Animals , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/pharmacology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Cathepsin K/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Female , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteolysis/etiology , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta1/pharmacology
13.
Arch Oral Biol ; 96: 46-51, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30172945

ABSTRACT

OBJECTIVE: Remodeling of alveolar bone is controlled by osteoclast-mediated bone resorption and osteoblast-induced bone formation. LPS of Porphyromonas gingivalis, a major causative agent of periodontitis, produces proinflammatory cytokines in host immune cells, which thereby triggers osteoclastogenesis and leads to alveolar bone resorption. We investigated the anti-periodontitis potential of Platycarya strobilacea leaf extract (PLE), which is used as a traditional medicine in Asian countries. DESIGN: TNF-α levels in cell culture media were measured using a commercially available enzyme-linked immunosorbent assay kit. Osteoclast differentiation was observed by tartrate-resistant acid phosphatase staining, and the expression levels of osteoclastogenic genes were measured by quantitative real-time PCR. Bone-resorbing activity was confirmed by the resorption pit formation, gelatin zymographic, and the cathepsin K activity assays. Osteogenic differentiation was confirmed with an ALP activity assay and alizarin red S staining. RESULTS: PLE treatment inhibited the production of TNF-α in P. gingivalis LPS-stimulated RAW264.7 macrophages. In bone marrow-derived macrophages serving as osteoclast precursors, PLE treatment blocked RANKL-induced osteoclastogenesis and gene expression levels of the osteoclastogenic transcription factor NFATc1, DC-STAMP for osteoclast fusion, and cathepsin K for osteoclast activity. In addition, PLE treatment reduced the formation of resorption pits and the secretion of MMP 9 and cathepsin K from the differentiated osteoclasts. Furthermore, PLE treatment induced osteogenesis by increasing ALP activity and calcium content in preosteoblastic cells. CONCLUSION: PLE inhibits P. gingivalis LPS-induced TNF-α production and bone resorption and induces bone formation. PLE may be a beneficial agent to promote oral health by inhibiting periodontitis-induced alveolar bone loss.


Subject(s)
Alveolar Bone Loss/prevention & control , Juglandaceae/classification , Plant Extracts/pharmacology , Plant Leaves , Porphyromonas gingivalis/metabolism , Tumor Necrosis Factor-alpha/metabolism , Alveolar Bone Loss/microbiology , Animals , Cell Differentiation/drug effects , Enzyme-Linked Immunosorbent Assay , Lipopolysaccharides/pharmacology , Male , Mice , Osteoclasts/drug effects , Osteogenesis/drug effects , Real-Time Polymerase Chain Reaction
14.
Sci Rep ; 7(1): 17332, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29230013

ABSTRACT

The activities of osteoclasts and osteoblasts are balanced to maintain normal bone density. Many pathological conditions cause osteoclastic bone resorption in excess of osteoblastic bone formation, resulting in osteoporosis. We found that oral administration of Artemisia annua ethanol extract (AaE) or major components, artemisinin and arteannuin B, to ovariectomized (OVX) mice prevented bone loss, as verified by examining three-dimensional images and bone morphometric parameters derived from microcomputed tomography analysis, as well as serum levels of bone turnover markers and proinflammatory cytokines. The administered doses were not toxic to the liver or kidney and showed promising effects that were comparable to those of 17ß-estradiol treatment. At non-cytotoxic concentrations, AaE and active components, artemisinin, artemisinic acid, and arteannuin B, potently inhibited receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastogenesis and the formation of osteoclast-mediated resorption pits. Furthermore, AaE, artemisinin, and arteannuin B remarkably reduced the expression of the c-Fos and NFATc1 transcription factors, which play critical roles in RANKL-induced osteoclast differentiation. Taken together, the in vivo anti-osteoporotic activity of AaE may be derived from the anti-osteoclastic and anti-bone resorptive activities of its active components. AaE has beneficial applications for the prevention and inhibition of osteoporosis and osteoclast-mediated bone diseases.


Subject(s)
Artemisia annua/chemistry , Bone Resorption/prevention & control , Cell Differentiation , Osteoclasts/drug effects , Ovariectomy/adverse effects , Plant Extracts/pharmacology , RANK Ligand/antagonists & inhibitors , Animals , Bone Resorption/etiology , Bone Resorption/metabolism , Bone Resorption/pathology , Female , Mice , Mice, Inbred ICR , Osteoclasts/metabolism , Osteoclasts/pathology
15.
Sci Rep ; 7(1): 9610, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851919

ABSTRACT

Breast cancer mainly spreads to bone, causing decreased survival of patient. Human antigen R (HuR) and chemokines are important molecules associated with mRNA stability and cell-cell interaction in cancer biology. Here, HuR knockdown inhibited bone metastasis and osteolysis of metastatic breast cancer cells in mice and HuR expression promoted the metastatic ability of cancer cells via CCL20 and GM-CSF. In contrast with the findings for GM-CSF, ELAVL1 and CCL20 expressions were markedly increased in breast tumor tissues and ELAVL1 expression showed a strong positive correlation with CCL20 expression in breast cancer subtypes, particularly the basal-like subtype. Metastasis-free survival and overall survival were decreased in the breast cancer patients with high CCL20 expression. We further confirmed the role of CCL20 in breast cancer bone metastasis. Intraperitoneal administration of anti-CCL20 antibodies inhibited osteolytic breast cancer bone metastasis in mice. Treatment with CCL20 noticeably promoted cell invasion and the secretion of MMP-2/9 in the basal-like triple-negative breast cancer cell lines, not the luminal. Moreover, CCL20 elevated the receptor activator of nuclear factors kappa-B ligand/osteoprotegerin ratio in breast cancer and osteoblastic cells and mediated the crosstalk between these cells. Collectively, HuR-regulated CCL20 may be an attractive therapeutic target for breast cancer bone metastasis.


Subject(s)
Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Chemokine CCL20/metabolism , ELAV-Like Protein 1/metabolism , Osteolysis , Animals , Biomarkers , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/mortality , Cell Line, Tumor , Chemokine CCL20/antagonists & inhibitors , Disease Models, Animal , ELAV-Like Protein 1/genetics , Female , Gene Knockdown Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Osteoblasts/metabolism , Prognosis , RANK Ligand/metabolism , Xenograft Model Antitumor Assays
16.
J Endod ; 43(8): 1317-1322, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28578892

ABSTRACT

INTRODUCTION: Previous occasional successes after delayed replantation suggest that the presence of viable cells may not be the only factor for successful periodontal regeneration in delayed replantation. Various other factors such as proteins or the extracellular matrix (ECM) may play a role in this process. The purpose of this study was to characterize changes in the proteome components of periodontal ligament (PDL) tissue after hypothermic preservation of the tooth. METHODS: Extracted teeth were divided into 4 groups: immediate sampling, sampling after 1 week of preservation at 4°C, sampling after 2 weeks of preservation at 4°C, and sampling after 1 week of dry storage at room temperature as a negative control. PDL tissues were collected from the root and stored immediately in liquid nitrogen. The tissues were subjected to 2-dimensional gel electrophoresis, and spot selection was executed. Selected spots that maintained the protein volume were then processed with matrix-assisted laser desorption and ionization time-of-flight mass spectrometry to identify the nature of the proteins. RESULTS: Thirty-five selected spots were analyzed. Sixteen spots were identified as vimentin, and 3 spots were type VI collagen. The size of the 16 vimentin spots decreased gradually with increasing storage time, from 0 to 2 weeks, and decreased rapidly after dry storage. However, only the dry storage group differed significantly from the immediately sampled group. CONCLUSIONS: Vimentin was the most prominent protein followed by type VI collagen in volumetrically maintained protein spots. Although these proteins are known to be closely related with ECM integrity, the role of these proteins in delayed replantation is beyond the scope of this study. Further studies are needed to elucidate the possible role of these proteins for periodontal healing of delayed replantation.


Subject(s)
Collagen/metabolism , Cryopreservation/methods , Periodontal Ligament/metabolism , Proteome/metabolism , Vimentin/metabolism , Electrophoresis, Gel, Two-Dimensional , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Survival , Tooth Replantation
17.
J Cancer Prev ; 22(1): 22-32, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28382283

ABSTRACT

BACKGROUND: Nicotine is a major toxic component of tobacco smoke and has been recognized as a risk factor to induce oxidative tissue damage, which is a precursor to cardiovascular diseases, lung-related diseases, and cancers. Peaches (Prunus persica) have been used for the treatment of degenerative disorders, such as hypermenorrhea, dysmenorrhea, and infertility in Asian countries. In this study, we investigated the effects of white-fleshed peach on the excretion of nicotine metabolites and 1-hydroxypyrene in smokers and chronic nicotine-induced tissue damages in mice. METHODS: The concentrations of cotinine and 1-hydroxypyrene were measured in urine of smokers before or after intake of white-fleshed peaches. In addition, ICR mice were injected with nicotine (5 mg/kg body weight) and then orally administered with white-fleshed peach extracts (WFPE) (250 or 500 mg/kg body weight) for 36 days. The oxidative stress parameters and the activities of antioxidant enzymes were measured in liver and kidney tissues. Also, histological changes and nitrotyrosine expression were assessed. RESULTS: Intake of white-fleshed peaches increased the urinary concentration of nicotine metabolites and 1-hydroxypyrene in 91.67% and 83.33% of smokers, respectively. WFPE decreased the malondialdehyde levels and recovered the activities of antioxidant enzymes in nicotine-injected mice. In addition, WFPE inhibited nitrotyrosine expression and inflammatory responses in the liver, kidney, and lung tissues of nicotine-treated mice. CONCLUSIONS: White-fleshed peaches may increase the metabolism of toxic components in tobacco smoke in smokers and protect normal tissues against nicotine toxicity in mice. Therefore, supplementation of white-fleshed peaches might be beneficial to smokers.

18.
Article in English | MEDLINE | ID: mdl-29333179

ABSTRACT

Once breast cancer cells grow aggressively and become lodged in the skeleton through migration and invasion, they interact with bone microenvironment and accelerate much more tumor growth and bone destruction. We investigated whether liensinine and nuciferine, major active components in Nelumbo nucifera (lotus), could prevent breast cancer cell-mediated bone destruction. Liensinine and nuciferine inhibited the growth of MDA-MB-231 and MCF-7 human breast cancer cells by inducing apoptosis and inhibiting proliferation via cell cycle arrest. Liensinine treatment led to the increased Bax/Bcl-2 ratio, activation of caspase-3, and subsequent cleavage of PARP. Liensinine also displayed significant inhibition on the migration and invasion of both MDA-MB-231 and MCF-7 human breast cancer cells compared with nuciferine. In addition, liensinine and nuciferine inhibited the receptor activator of nuclear factor kappa-B ligand- (RANKL-) induced osteoclast differentiation in mouse bone marrow macrophage cells and mature osteoclast-mediated bone resorption. Furthermore, oral administration of liensinine reduced the osteolysis in nude mice with intratibial injection of MDA-MB-231 cells. Collectively, liensinine and nuciferine may be promising candidates for preventing and treating breast cancer bone metastasis and the resulting osteolytic bone loss by targeting both cancer cells and osteoclasts. Liensinine has more potent anticancer and antibone resorptive activities than nuciferine.

19.
Article in English | MEDLINE | ID: mdl-29358968

ABSTRACT

Oral squamous cell carcinoma (OSCC) frequently invades mandibular bone, and outcomes for treatment with surgical resection are typically poor, ultimately resulting in death. Holarrhena antidysenterica L. (Apocynaceae), distributed throughout Sri Lanka and India, has been used as a folk remedy to treat various diseases. Treatment with methanol extract of H. antidysenterica bark (HABE) inhibited cell viability and BrdU incorporation and induced apoptotic cell death in Ca9-22 gingival and HSC-3 tongue SCC cells. Flow cytometric analysis indicated that HABE treatment preferentially induces apoptotic cell death via increasing the sub-G1 peak in Ca9-22 cells and cell cycle arrest at the G1 phase in HSC-3 cells. HABE treatment in the presence of zVAD-fmk, a pan-caspase inhibitor, rescued cell viabilities in both OSCC cell lines. The ratio of Bax to Bcl-2 increased with reductions in the Bcl-2 protein expression, and the activation of caspase 3 and subsequent cleavage of PARP was detected in HABE-treated Ca9-22 and HSC-3 cells. Furthermore, HABE treatment at noncytotoxic concentrations inhibited osteoclast formation in RANKL-stimulated bone marrow macrophages. Taken together, HABE possesses the inhibitory activity on the growth of OSCC cells and antiosteoclastogenic activity. Therefore, HABE may be a promising alternative and complementary agent for preventing and treating OSCC.

20.
Oncotarget ; 8(6): 9079-9092, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28030842

ABSTRACT

High recurrence and lower survival rates in patients with oral squamous cell carcinoma (OSCC) are associated with its bone invasion. We identified the oncogenic role of RUNX3 during bone invasion by OSCC. Tumor growth and the generation of osteolytic lesions were significantly inhibited in mice that were subcutaneously inoculated with RUNX3-knockdown human OSCC cells. RUNX3 knockdown enhanced TGF-ß-induced growth arrest and inhibited OSCC cell migration and invasion in the absence or presence of transforming growth factor-ß (TGF-ß), a major growth factor abundant in the bone microenvironment. RUNX3 knockdown induced cell cycle arrest at the G1 and G2 phases and promoted G2 arrest by TGF-ß in Ca9.22 OSCC cells. RUNX3 knockdown also inhibited both the basal and TGF-ß-induced epithelial-to-mesenchymal transition by increasing E-cadherin expression and suppressing the nuclear translocation of ß-catenin. In addition, the expression and TGF-ß-mediated induction of parathyroid hormone-related protein (PTHrP), one of key osteolytic factors, was blocked in RUNX3-knockdown OSCC cells. Furthermore, treating human osteoblastic cells with conditioned medium derived from RUNX3-knockdown OSCC cells reduced the receptor activator of nuclear factor-kappaB ligand (RANKL)/osteoprotegerin ratio compared with treatment with conditioned medium from RUNX3-expressing cells. These findings indicate that RUNX3 expression in OSCC cells contributes to their bone invasion and the resulting osteolysis by inducing their malignant behaviors and production of osteolytic factors. RUNX3 alone or in combination with TGF-ß and PTHrP may be a useful predictive biomarker and therapeutic target for bone invasion by oral cancer.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Movement , Core Binding Factor Alpha 3 Subunit/metabolism , Head and Neck Neoplasms/metabolism , Mouth Neoplasms/metabolism , Osteolysis/metabolism , Skull/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Core Binding Factor Alpha 3 Subunit/genetics , Epithelial-Mesenchymal Transition , G1 Phase Cell Cycle Checkpoints , G2 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Kaplan-Meier Estimate , Male , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/genetics , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Osteoblasts/metabolism , Osteoblasts/pathology , Osteolysis/genetics , Osteolysis/pathology , Paracrine Communication , Parathyroid Hormone-Related Protein/metabolism , RANK Ligand/metabolism , RNA Interference , Signal Transduction , Skull/pathology , Squamous Cell Carcinoma of Head and Neck , Time Factors , Transfection , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...