1
|
Zhao Y, Hu F, Wang Q. Cortactin contributes to the tumorigenesis of gastric cancer by activating ERK/MMP pathway. Heliyon 2023; 9:e18289. [PMID: 37539204 PMCID: PMC10395536 DOI: 10.1016/j.heliyon.2023.e18289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 08/05/2023] Open
Abstract
Gastric cancer is a malignant tumor with high mortality and high incidence. This study aims to explore the function and molecular mechanism of Cortactin on gastric cancer progression in vitro and in vivo. A bioinformatics analysis from TCGA displayed that Cortactin was highly expressed in gastric cancer samples, and patients with a high Cortactin level had a worse survival rate. Subsequently, we investigated the specific mechanism of action of A in gastric cancer by collecting patient samples for immunohistochemistry, WB, qRT-PCR, cell transfection, cell invasion and metastasis, and constructing tumor xenografts in nude mice. Overexpression of Cortactin inhibited apoptosis and enhanced cellular proliferation and mobility in AGS cells, while those activities were reversed by the knockdown of MMP2 or MMP9. Conversely, the deletion of Cortactin induced apoptosis and suppressed cell growth and metastasis in SGC7901 cells, whereas those behaviors were inhibited by overexpression of MMP2 or MMP9. Additionally, the ERK pathway was activated by Cortactin upregulation. In vivo studies presented that overexpression of Cortactin promoted tumor growth, increased Ki67 expression, and reduced caspase 3 expression, which was reversed by ERK inhibitor treatment. In conclusion, Cortactin acted as an oncogene in gastric cancer and exerted its function by ERK/MMP2/MMP9 signaling pathway.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui 233004, China
| | - Fang Hu
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui 233004, China
| | - Qizhi Wang
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui 233004, China
| |
Collapse
|
2
|
Suppression of osteoclastogenesis signalling pathways and attenuation of ameloblastic osteolysis induced by local administration of CaP-bisphosphonate and CaP-doxycycline cements: Review of the literature and therapeutic hypothesis. ADVANCES IN ORAL AND MAXILLOFACIAL SURGERY 2022. [DOI: 10.1016/j.adoms.2021.100241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
3
|
Peng Y, Liu Q, Xu D, Li K, Li H, Qiu L, Lin J. Inhibition of zoledronic acid derivatives with extended methylene linkers on osteoclastogenesis involve downregulation of JNK and Akt pathways. Cell Biol Int 2021; 45:1015-1029. [PMID: 33404170 DOI: 10.1002/cbin.11546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/09/2020] [Accepted: 01/03/2021] [Indexed: 11/11/2022]
Abstract
Bisphosphonates (BPs), especially zoledronic acid (ZOL), are clinically used to treat osteolytic bone lesions. However, serious side-effects may be also induced during the therapeutic process. To improve the BPs drugs, here, we investigated the effects of a series of ZOL derivatives with increasing number of methylene linker between the imidazole ring and the P-C-P backbone named IPrDP, IBDP, IPeDP, and IHDP on cell viability and receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation, function and apoptosis induction in mouse bone marrow-derived macrophages (BMMs). Our results suggested that IPeDP and IHDP, which contains 4 and 5 methylene linkers, respectively, exerted lower toxicity on BMMs compared with ZOL, IPrDP, and IBDP, which contains 1, 2, and 3 methylene linkers respectively. At concentrations below cytotoxicity threshold, IPeDP and IHDP possessed strong abilities of antiosteoclast formation, antibone absorption, and inducing osteoclast apoptosis, which were similar to ZOL and more powerful than IPrDP and IBDP. The mechanism behind these effects of IPeDP and IHDP might involve the interference of small GTPases prenylation through suppression of mevalonate pathway. The downregulation of JNK and Akt phosphorylation and subsequent inhibition of the expression of c-Fos and NFATc1 might also be involved. Our results supported the potential usage of IPeDP and IHDP to treat bone-related disorders involving increased osteoclastogenesis. Our attempt to extend the methylene linker between the imidazole ring and the P-C-P backbone of ZOL also reveals some regularities between the structure and properties of the BPs drugs.
Collapse
Affiliation(s)
- Ying Peng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China.,Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qingzhu Liu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Dong Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Ke Li
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Hang Li
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China.,School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| | - Ling Qiu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China.,School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| | - Jianguo Lin
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China.,Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Li YY, Gao LJ, Zhang YX, Liu SJ, Cheng S, Liu YP, Jia CX. Bisphosphonates and risk of cancers: a systematic review and meta-analysis. Br J Cancer 2020; 123:1570-1581. [PMID: 32901134 PMCID: PMC7652831 DOI: 10.1038/s41416-020-01043-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 07/28/2020] [Accepted: 08/14/2020] [Indexed: 01/02/2023] Open
Abstract
Background It is unclear whether bisphosphonates are associated with risk of cancers. Therefore, this meta-analysis aimed to evaluate the effect of bisphosphonates on overall cancers. Methods A search in Pubmed, Embase, Cochrane Library and Web of Science databases was conducted, from the inception date of each resource to September 26, 2019. The summarised effect estimates with 95% CIs were calculated using a random-effect model. Heterogeneity and publication bias were explored. Results Thirty-four articles were included in this study (4,508,261 participants; 403,196 cases). The results revealed that bisphosphonates significantly decreased the risk of colorectal cancer (RR = 0.89, 95% CI: 0.81–0.98), breast cancer (RR = 0.87, 95% CI: 0.82–0.93) and endometrial cancer (RR = 0.75, 95% CI: 0.61–0.94), but no significant association was observed in all-cause cancer. Furthermore, nitrogen-containing bisphosphonates only had protective effects both on breast cancer (RR = 0.94, 95% CI: 0.90–0.99) and endometrial cancer (RR = 0.70, 95% CI: 0.54–0.92). Non-nitrogen-containing bisphosphonates tended to increase the risk of liver cancer (RR = 2.14, 95% CI: 1.23–3.72) and pancreas cancer (RR = 1.75, 95% CI: 1.32–2.33). Conclusion Bisphosphonates are significantly associated with risk reduction of colorectal, breast and endometrial cancer, especially nitrogen-containing bisphosphonates. It should be noted that non-nitrogen-containing bisphosphonates might increase the risk of liver and pancreas cancer. Large prospective cohort studies are needed to find the causal association between bisphosphonates and risk of cancers.
Collapse
Affiliation(s)
- Yuan-Yuan Li
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Li-Jie Gao
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu-Xue Zhang
- Department of Preventive Medicine, Public Health School, Harbin Medical University, Harbin, China
| | - Shu-Juan Liu
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuo Cheng
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu-Peng Liu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.
| | - Cun-Xian Jia
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
5
|
Tsubaki M, Seki S, Takeda T, Chihara A, Arai Y, Morii Y, Imano M, Satou T, Shimomura K, Nishida S. The HGF/Met/NF-κB Pathway Regulates RANKL Expression in Osteoblasts and Bone Marrow Stromal Cells. Int J Mol Sci 2020; 21:ijms21217905. [PMID: 33114380 PMCID: PMC7663721 DOI: 10.3390/ijms21217905] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/16/2020] [Accepted: 10/22/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM)-induced bone disease occurs through hyperactivation of osteoclasts by several factors secreted by MM cells. MM cell-secreted factors induce osteoclast differentiation and activation via direct and indirect actions including enhanced expression of receptor activator of nuclear factor κB ligand (RANKL) in osteoblasts and bone marrow stromal cells (BMSCs). Hepatocyte growth factor (HGF) is elevated in MM patients and is associated with MM-induced bone disease, although the mechanism by which HGF promotes bone disease remains unclear. In the present study, we demonstrated that HGF induces RANKL expression in osteoblasts and BMSCs, and investigated the mechanism of induction. We found that HGF and MM cell supernatants induced RANKL expression in ST2 cells, MC3T3-E1 cells, and mouse BMSCs. In addition, HGF increased phosphorylation of Met and nuclear factor κB (NF-κB) in ST2 cells, MC3T3-E1 cells, or mouse BMSCs. Moreover, Met and NF-κB inhibitors suppressed HGF-induced RANKL expression in ST2 cells, MC3T3-E1 cells, and mouse BMSCs. These results indicated that HGF promotes RANKL expression in osteoblasts and BMSCs via the Met/NF-κB signaling pathway, and Met and NF-κB inhibitors suppressed HGF-induced RANKL expression. Our findings suggest that Met and NF-κB inhibitors are potentially useful in mitigating MM-induced bone disease in patients expressing high levels of HGF.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
| | - Shiori Seki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
| | - Akiko Chihara
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
| | - Yuuko Arai
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
| | - Yuusuke Morii
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
- Department of Pharmacy, Municipal Ikeda Hospital, Ikeda 563-0025, Japan;
| | - Motohiro Imano
- Department of Surgery, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-0014, Japan;
| | - Takao Satou
- Department of Pathology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-0014, Japan;
| | - Kazunori Shimomura
- Department of Pharmacy, Municipal Ikeda Hospital, Ikeda 563-0025, Japan;
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
- Correspondence: ; Tel.: +81-6-6721-2332
| |
Collapse
|
6
|
Tsubaki M, Takeda T, Noguchi M, Jinushi M, Seki S, Morii Y, Shimomura K, Imano M, Satou T, Nishida S. Overactivation of Akt Contributes to MEK Inhibitor Primary and Acquired Resistance in Colorectal Cancer Cells. Cancers (Basel) 2019; 11:cancers11121866. [PMID: 31769426 PMCID: PMC6966459 DOI: 10.3390/cancers11121866] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/21/2022] Open
Abstract
RAS and BRAF-mutated colorectal cancers are associated with resistance to chemotherapy and poor prognosis, highlighting the need for new therapeutic strategies. Although these cancers sometimes respond to mitogen activated protein kinase kinase (MEK) inhibitor treatment, they often acquire resistance via mechanisms, which are poorly understood. Here, we investigated the mechanism of MEK inhibitor resistance in primary- and acquired-resistant cells. Cell viability was examined using the trypan blue dye exclusion assay. Protein expression was analyzed by western blotting. Somatic mutations in colorectal cancer cells were investigated using the polymerase chain reaction array. PD0325901 and trametinib induced cell death in LoVo and Colo-205 cells but not in DLD-1 and HT-29 cells, which have a PIK3CA mutation constitutively activating Akt and NF-κB. Treatment with PD0325901 and trametinib suppressed ERK1/2 activation in all four cell lines but only induced Akt and NF-κB activation in DLD-1 and HT-29 cells. Inhibition of Akt but not NF-κB, overcame MEK inhibitor resistance in DLD-1 and HT-29 cells. Acquired-resistant LoVo/PR, Colo-205/PR and LoVo/TR cells have constitutively active Akt due to a M1043V mutation in the kinase activation loop of PIK3CA and Akt inhibitor resensitized these cells to MEK inhibitor. These results demonstrate that the overactivation of Akt plays a critical role in MEK inhibitor primary and acquired resistance and implicate combined Akt/MEK inhibition as a potentially useful treatment for RAS/BRAF-mutated colorectal cancer.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Masaki Noguchi
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Minami Jinushi
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Shiori Seki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Yuusuke Morii
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
- Department of Phamacy, Municipal Ikeda Hospital, Ikeda, Osaka 563-8510, Japan;
| | - Kazunori Shimomura
- Department of Phamacy, Municipal Ikeda Hospital, Ikeda, Osaka 563-8510, Japan;
| | - Motohiro Imano
- Department of Surgery, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-0014, Japan;
| | - Takao Satou
- Department of Pathology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-0014, Japan.;
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
- Correspondence:
| |
Collapse
|
7
|
Rittler D, Baranyi M, Molnár E, Garay T, Jalsovszky I, Varga IK, Hegedűs L, Aigner C, Tóvári J, Tímár J, Hegedűs B. The Antitumor Effect of Lipophilic Bisphosphonate BPH1222 in Melanoma Models: The Role of the PI3K/Akt Pathway and the Small G Protein Rheb. Int J Mol Sci 2019; 20:ijms20194917. [PMID: 31623406 PMCID: PMC6801414 DOI: 10.3390/ijms20194917] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/24/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
Malignant melanoma is one of the most metastatic cancer types, and despite recent success with novel treatment strategies, there is still a group of patients who do not respond to any therapies. Earlier, the prenylation inhibitor hydrophilic bisphosphonate zoledronic acid (ZA) was found to inhibit melanoma growth in vitro, but only a weaker effect was observed in vivo due to its hydrophilic properties. Recently, lipophilic bisphosphonates (such as BPH1222) were developed. Accordingly, for the first time, we compared the effect of BPH1222 to ZA in eight melanoma lines using viability, cell-cycle, clonogenic and spheroid assays, videomicroscopy, immunoblot, and xenograft experiments. Based on 2D and spheroid assays, the majority of cell lines were more sensitive to BPH. The activation of Akt and S6 proteins, but not Erk, was inhibited by BPH. Additionally, BPH had a stronger apoptotic effect than ZA, and the changes of Rheb showed a correlation with apoptosis. In vitro, only M24met cells were more sensitive to ZA than to BPH; however, in vivo growth of M24met was inhibited more strongly by BPH. Here, we present that lipophilic BPH is more effective on melanoma cells than ZA and identify the PI3K pathway, particularly Rheb as an important mediator of growth inhibition.
Collapse
Affiliation(s)
- Dominika Rittler
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
| | - Marcell Baranyi
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
| | - Eszter Molnár
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
| | - Tamás Garay
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, H-1083 Budapest, Hungary.
- Oncology Center, Semmelweis University, H-1091 Budapest, Hungary.
| | - István Jalsovszky
- Eötvös Loránd University, Faculty of Science, Institute of Chemistry, Department of Organic Chemistry; H-1117 Budapest, Hungary.
| | - Imre Károly Varga
- Eötvös Loránd University, Faculty of Science, Institute of Chemistry, Department of Organic Chemistry; H-1117 Budapest, Hungary.
| | - Luca Hegedűs
- Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, D-45239 Essen, Germany.
| | - Clemens Aigner
- Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, D-45239 Essen, Germany.
| | - József Tóvári
- Department of Experimental Pharmacology, National Institute of Oncology, H-1122 Budapest, Hungary.
| | - József Tímár
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
| | - Balázs Hegedűs
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
- Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, D-45239 Essen, Germany.
| |
Collapse
|
8
|
Nasulewicz-Goldeman A, Goldeman W, Mrówczyńska E, Wietrzyk J. Biological effects of aromatic bis[aminomethylidenebis(phosphonic)] acids in osteoclast precursors in vitro. Chem Biol Drug Des 2019; 94:1835-1848. [PMID: 31356729 DOI: 10.1111/cbdd.13597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/02/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022]
Abstract
Nitrogen-containing bisphosphonates (N-BPs) inhibit bone resorption by preventing osteoclast activity. Most clinically used BPs are hydroxybisphosphonates with the exception of incadronate, which belongs to the class of aminomethylidenebisphosphonic acids. The aim of this study was to evaluate the antiproliferative activity of two previously reported aminobisphosphonates (WG8185B2 and WG9001B) in combination with doxorubicin and cisplatin toward J774E cells (a model of osteoclast precursors in vitro). WG8185B2 and WG9001B BPs enhanced the cytotoxic activity of doxorubicin and cisplatin, especially when applied 24 hr prior to cytostatics. The antiproliferative effect of studied BPs was related to the changes in cell cycle progression. WG8185B2 leads to significant accumulation of J774E cells in S phase, whereas WG9001B causes transient arrest in G2 /M phase, followed by an increase in the percentage of cells in S phase. Moreover, WG8185B2 and WG9001B BPs showed enhanced proapoptotic activity in osteoclast precursors, which was manifested by an increase in caspase-3 activity and percentage of apoptotic cells. In addition, both compounds influenced the motility of J774E cells. The exact molecular mechanism of action of examined BPs remains to be determined; however, results show an interesting biological activity of these compounds, which may be of interest in the context of antiresorptive therapy.
Collapse
Affiliation(s)
- Anna Nasulewicz-Goldeman
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wrocław, Poland
| | - Waldemar Goldeman
- Department of Organic Chemistry, Wrocław University of Technology, Wrocław, Poland
| | - Ewa Mrówczyńska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wrocław, Poland
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wrocław, Poland
| |
Collapse
|
9
|
Tamai R, Kiyoura Y. Alendronate augments lipid A-induced IL-1β release and Smad3/NLRP3/ASC-dependent cell death. Life Sci 2018; 198:8-17. [DOI: 10.1016/j.lfs.2018.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 02/01/2018] [Accepted: 02/09/2018] [Indexed: 12/13/2022]
|
10
|
Takeda T, Tsubaki M, Tomonari Y, Kawashima K, Itoh T, Imano M, Satou T, Nishida S. Bavachin induces the apoptosis of multiple myeloma cell lines by inhibiting the activation of nuclear factor kappa B and signal transducer and activator of transcription 3. Biomed Pharmacother 2018; 100:486-494. [PMID: 29477912 DOI: 10.1016/j.biopha.2018.02.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/26/2018] [Accepted: 02/08/2018] [Indexed: 11/26/2022] Open
Abstract
Bavachin is a phytoestrogen purified from natural herbal plants such as Psoralea corylifolia. In this study, we examined the effect of bavachin in multiple myeloma (MM) cell lines. We found that bavachin decreased the viability of MM cell lines, but was not cytotoxic towards normal cells. It inhibited the activation of nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3). Furthermore, bavachin increased the expression of p53 and NOXA, and decreased the expression of X-linked inhibitor of apoptosis protein (XIAP), survivin, B cell lymphoma-extra large (Bcl-xL), and Bcl-2. Additionally, bavachin induced apoptosis by the activation of caspase-3 and caspase-9, implicating the involvement of the mitochondrial pathway. Our results suggest that bavachin induces apoptosis through the inhibition of NF-κB and STAT3 activation in MM cell lines. Most importantly, few NF-κB and STAT3 inhibitors with high efficiency, specificity, and safety are currently available for clinical cancer therapy. Hence, bavachin, which targets NF-κB and STAT3, is a potential anticancer agent for the treatment of MM.
Collapse
Affiliation(s)
- Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Osaka, Japan
| | - Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Osaka, Japan
| | - Yoshika Tomonari
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Osaka, Japan
| | - Keishi Kawashima
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Osaka, Japan
| | - Tatsuki Itoh
- Department of Food Science and Nutrition, Kindai University School of Agriculture, Nara, Nara, Japan
| | - Motohiro Imano
- Department of Surgery, Kindai University School of Medicine, Osakasayama, Osaka, Japan
| | - Takao Satou
- Department of Pathology, Kindai University School of Medicine, Osakasayama, Osaka, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Osaka, Japan.
| |
Collapse
|
11
|
Tsubaki M, Takeda T, Asano RT, Matsuda T, Fujimoto SI, Itoh T, Imano M, Satou T, Nishida S. Rebamipide suppresses 5-fluorouracil-induced cell death via the activation of Akt/mTOR pathway and regulates the expression of Bcl-2 family proteins. Toxicol In Vitro 2018; 46:284-293. [PMID: 29054700 DOI: 10.1016/j.tiv.2017.10.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 09/12/2017] [Accepted: 10/16/2017] [Indexed: 02/08/2023]
|
12
|
Tsubaki M, Takeda T, Tomonari Y, Mashimo K, Koumoto YI, Hoshida S, Itoh T, Imano M, Satou T, Sakaguchi K, Nishida S. The MIP-1α autocrine loop contributes to decreased sensitivity to anticancer drugs. J Cell Physiol 2017; 233:4258-4271. [PMID: 29057477 DOI: 10.1002/jcp.26245] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 10/18/2017] [Indexed: 01/04/2023]
Abstract
Several autocrine soluble factors, including macrophage inflammatory protein-1α (MIP-1α), tumor necrosis factor-α, and hepatocyte growth factor, promote cell survival and growth in multiple myeloma (MM) cells. We hypothesized that inhibition of the MIP-1α autocrine loop may enhance the cytotoxic effect of anticancer drugs in MM cell lines. In the present study, an MIP-1α neutralizing antibody suppressed cell proliferation and enhanced the cytotoxic effect of melphalan or bortezomib on MM cells. In addition, melphalan resistance cells (RPMI8226/L-PAM and HS-sultan/L-PAM cells) secreted MIP-1α and neutralizing antibody of MIP-1α partially overcame melphalan resistance. Moreover, combination treatment with MIP-1α neutralizing antibody and melphalan or bortezomib inhibited extracellular signal regulated kinase 1/2 (ERK1/2), Akt, and mammalian target of rapamycin (mTOR) activation, Bcl-2, Bcl-xL, and Survivin expression, and upregulated the expression of Bim and cleaved Poly (ADP-ribose) polymerase (PARP). Treatment of IM9 cells with MIP-1α siRNA suppressed the activation of ERK1/2, Akt, and mTOR, and enhanced the cytotoxic effect of melphalan and bortezomib. These results indicate that MIP-1α neutralizing antibodies or MIP-1α siRNA enhance the cytotoxic effect of melphalan and bortezomib by suppressing the chemokine receptor/ERK and chemokine receptor/Akt/mTOR pathways. The inhibition of MIP-1α may thus provide a new therapeutic approach to control tumor progression and bone destruction in patients with MM.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Tomoya Takeda
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Yoshika Tomonari
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Kenji Mashimo
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan.,Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Yu-Ichi Koumoto
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Sachi Hoshida
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Tatsuki Itoh
- Faculty of Agriculture, Department of Food Science and Nutrition, Kindai University, Nara, Nara, Japan
| | - Motohiro Imano
- Faculty of Medicine, Department of Surgery, Kindai University, Osakasayama, Osaka, Japan
| | - Takao Satou
- Faculty of Medicine, Department of Pathology, Kindai University, Osakasayama, Osaka, Japan
| | - Katsuhiko Sakaguchi
- Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Shozo Nishida
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| |
Collapse
|
13
|
Tsubaki M, Takeda T, Tomonari Y, Kawashima K, Itoh T, Imano M, Satou T, Nishida S. Pioglitazone inhibits cancer cell growth through STAT3 inhibition and enhanced AIF expression via a PPARγ-independent pathway. J Cell Physiol 2017; 233:3638-3647. [PMID: 29030979 DOI: 10.1002/jcp.26225] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 10/05/2017] [Indexed: 12/14/2022]
Abstract
Pioglitazone is an anti-diabetic agent that belongs to the thiazolidinedione class, which target peroxisome proliferator-activated receptor γ (PPARγ), a transcription factor in the nuclear receptor family. Different cancer cells expressing high levels of PPARγ and PPARγ ligands induce cell cycle arrest, cell differentiation, and apoptosis. However, the mechanisms underlying these processes remain unknown. Here, we investigated the mechanism underlying pioglitazone-induced apoptosis in human cancer cells. We showed that at similar concentrations, pioglitazone induced death in cancer cells expressing high or low levels of PPARγ. Combined treatment of pioglitazone and GW9662, a PPARγ antagonist, did not rescue this cell death phenotype. Z-VAD-fmk, a pan-caspase inhibitor, did not reverse pioglitazone-induced apoptosis in cancer cells expressing PPARγ at high or low levels. Pioglitazone suppressed the activation of signal transducers and activator of transcription 3 (STAT3) and Survivin expression, and enhanced the apoptosis-inducing factor (AIF) levels in these cells. Furthermore, pioglitazone enhanced the cytotoxic effect of cisplatin and oxaliplatin by suppressing Survivin and increasing AIF expression. These results indicated that pioglitazone induced apoptosis via a PPARγ-independent pathway, thus describing pioglitazone as a potential therapeutic agent for controlling the progression of different cancers.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Faculty of Pharmacy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Tomoya Takeda
- Division of Pharmacotherapy, Faculty of Pharmacy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Yoshika Tomonari
- Division of Pharmacotherapy, Faculty of Pharmacy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Keishi Kawashima
- Division of Pharmacotherapy, Faculty of Pharmacy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Tatsuki Itoh
- Department of Food Science and Nutrition, Faculty of Agriculture, Kindai University, Nara, Nara, Japan
| | - Motohiro Imano
- Department of Surgery, Faculty of Medicine, Kindai University, Osakasayama, Osaka, Japan
| | - Takao Satou
- Department of Pathology, Faculty of Medicine, Kindai University, Osakasayama, Osaka, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Faculty of Pharmacy, Kindai University, Kowakae, Higashi-Osaka, Japan
| |
Collapse
|
14
|
Fujiwara D, Tsubaki M, Takeda T, Tomonari Y, Koumoto YI, Sakaguchi K, Nishida S. Statins induce apoptosis through inhibition of Ras signaling pathways and enhancement of Bim and p27 expression in human hematopoietic tumor cells. Tumour Biol 2017; 39:1010428317734947. [PMID: 28990465 DOI: 10.1177/1010428317734947] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recently, statins have been demonstrated to improve cancer-related mortality or prognosis in patients of various cancers. However, the details of the apoptosis-inducing mechanisms remain unknown. This study showed that the induction of apoptosis by statins in hematopoietic tumor cells is mediated by mitochondrial apoptotic signaling pathways, which are activated by the suppression of mevalonate or geranylgeranyl pyrophosphate biosynthesis. In addition, statins decreased the levels of phosphorylated extracellular signal-regulated kinase 1/2 and mammalian target of rapamycin through suppressing Ras prenylation. Furthermore, inhibition of extracellular signal-regulated kinase 1/2 and mammalian target of rapamycin by statins induced Bim expression via inhibition of Bim phosphorylation and ubiquitination and cell-cycle arrest at G1 phase via enhancement of p27 expression. Moreover, combined treatment of U0126, a mitogen-activated protein kinase kinase 1/2 inhibitor, and rapamycin, a mammalian target of rapamycin inhibitor, induced Bim and p27 expressions. The present results suggested that statins induce apoptosis by decreasing the mitochondrial transmembrane potential, increasing the activation of caspase-9 and caspase-3, enhancing Bim expression, and inducing cell-cycle arrest at G1 phase through inhibition of Ras/extracellular signal-regulated kinase and Ras/mammalian target of rapamycin pathways. Therefore, our findings support the use of statins as potential anticancer agents or concomitant drugs of adjuvant therapy.
Collapse
Affiliation(s)
- Daichiro Fujiwara
- 1 Division of Pharmacotherapy, School of Pharmacy, Kindai University, Higashi-Osaka, Japan.,2 Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Masanobu Tsubaki
- 1 Division of Pharmacotherapy, School of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | - Tomoya Takeda
- 1 Division of Pharmacotherapy, School of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | - Yoshika Tomonari
- 1 Division of Pharmacotherapy, School of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | - Yu-Ichi Koumoto
- 1 Division of Pharmacotherapy, School of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | - Katsuhiko Sakaguchi
- 2 Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Shozo Nishida
- 1 Division of Pharmacotherapy, School of Pharmacy, Kindai University, Higashi-Osaka, Japan
| |
Collapse
|
15
|
Tsubaki M, Fujiwara D, Takeda T, Kino T, Tomonari Y, Itoh T, Imano M, Satou T, Sakaguchi K, Nishida S. The sensitivity of head and neck carcinoma cells to statins is related to the expression of their Ras expression status, and statin-induced apoptosis is mediated via suppression of the Ras/ERK and Ras/mTOR pathways. Clin Exp Pharmacol Physiol 2017; 44:222-234. [PMID: 27805296 DOI: 10.1111/1440-1681.12690] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 12/30/2022]
Abstract
Statins induce apoptosis of tumour cells by inhibiting the prenylation of small G-proteins. However, the details of the apoptosis-inducing mechanisms remain poorly understood. The present study showed that the induction of apoptosis by statins in four different human head and neck squamous cell carcinoma (HNSCC) cell lines, HSC-3, HEp-2, Ca9-22, and SAS cells was mediated by increased caspase-3 activity. Statins induced apoptosis by the suppression of geranylgeranyl pyrophosphate biosynthesis. Furthermore, statins decreased the levels of phosphorylated ERK and mTOR by inhibiting the membrane localization of Ras and enhancing Bim expression in HSC-3 and HEp-2 cells. We also found that in all the cell types analyzed, the IC50 values for fluvastatin and simvastatin were highest in HEp-2 cells. In addition, HSC-3, Ca9-22, and SAS cells had higher Ras expression and membrane localization, higher activation of ERK1/2 and mTOR, and lower levels of Bim expression than HEp-2 cells. Our results indicate that statins induce apoptosis by increasing the activation of caspase-3 and by enhancing Bim expression through inhibition of the Ras/ERK and Ras/mTOR pathways. Furthermore, the sensitivity of HNSCC cells to statin treatment was closely related to Ras expression and prenylation levels, indicating that statins may act more effectively against tumours with high Ras expression and Ras-variability. Therefore, our findings support the use of statins as potential anticancer agents.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Japan
| | - Daichiro Fujiwara
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Japan.,Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Japan
| | - Toshiki Kino
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Japan
| | - Yoshika Tomonari
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Japan
| | - Tatsuki Itoh
- Department of Food Science and Nutrition, Kindai University School of Agriculture, Nara, Nara, Japan
| | - Motohiro Imano
- Department of Surgery, Kindai University School of Medicine, Osakasayama, Osaka, Japan
| | - Takao Satou
- Department of Pathology, Kindai University School of Medicine, Osakasayama, Osaka, Japan
| | - Katsuhiko Sakaguchi
- Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Kowakae, Higashi-Osaka, Japan
| |
Collapse
|
16
|
Nikfar Z, Shariatinia Z. Phosphate functionalized (4,4)-armchair CNTs as novel drug delivery systems for alendronate and etidronate anti-osteoporosis drugs. J Mol Graph Model 2017; 76:86-105. [DOI: 10.1016/j.jmgm.2017.06.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 06/17/2017] [Accepted: 06/19/2017] [Indexed: 11/26/2022]
|
17
|
Agabiti SS, Li J, Wiemer AJ. Geranylgeranyl diphosphate synthase inhibition induces apoptosis that is dependent upon GGPP depletion, ERK phosphorylation and caspase activation. Cell Death Dis 2017; 8:e2678. [PMID: 28300835 PMCID: PMC5386513 DOI: 10.1038/cddis.2017.101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/26/2017] [Accepted: 02/15/2017] [Indexed: 02/08/2023]
Abstract
Bisphosphonates are diphosphate analogs that inhibit the intermediate enzymes of the mevalonate pathway. Here, we compared the effects of a farnesyl diphosphate synthase inhibitor, zoledronate, and a geranylgeranyl diphosphate synthase (GGDPS) inhibitor, digeranyl bisphosphonate (DGBP), on lymphocytic leukemia cell proliferation and apoptosis. Both zoledronate and DGBP inhibited proliferation with DGBP doing so more potently. DGBP was markedly less toxic than zoledronate toward the viability of healthy human peripheral blood mononuclear cells. Addition of GGPP, but not farnesyl diphosphate (FPP), prevented the anti-proliferative effects of DGBP. Both GGPP and FPP partially rescued the effects of zoledronate. Co-treatment with DGBP and zoledronate was antagonistic. To further assess the effects of the bisphosphonates, we analyzed annexin V and propidium iodide staining via flow cytometry and found that DGBP induced apoptosis more potently than zoledronate. Western blots show that DGBP treatment altered expression and membrane affinity of some but not all geranylgeranylated small GTPases, activated caspases and increased ERK phosphorylation. Importantly, the anti-proliferative effects of DGBP were blocked by treatment with a caspase inhibitor and by treatment with a MEK inhibitor. Together, our findings indicate that DGBP is a more potent and selective compound than zoledronate in inducing apoptosis mediated through pathways that include caspases and MEK/ERK. These findings support the further development of GGDPS inhibitors as anticancer therapeutics.
Collapse
Affiliation(s)
- Sherry S Agabiti
- Department of Pharmaceutical Sciences, University of Connecticut, School of Pharmacy, Storrs, CT, USA
| | - Jin Li
- Department of Pharmaceutical Sciences, University of Connecticut, School of Pharmacy, Storrs, CT, USA
| | - Andrew J Wiemer
- Department of Pharmaceutical Sciences, University of Connecticut, School of Pharmacy, Storrs, CT, USA.,Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
18
|
Song XF, Tian H, Zhang P, Zhang ZX. Expression of Cyt-c-Mediated Mitochondrial Apoptosis-Related Proteins in Rat Renal Proximal Tubules during Development. Nephron Clin Pract 2016; 135:77-86. [PMID: 27665619 DOI: 10.1159/000450585] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 09/01/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Apoptosis regulates embryogenesis, organ metamorphosis and tissue homeostasis. Mitochondrial signaling is an apoptotic pathway, in which Cyt-c and Apaf-1 are transformed into an apoptosome, which activates procaspase-9 and triggers apoptosis. This study evaluated Cyt-c, Apaf-1 and caspase-9 expression during renal development. METHODS Kidneys from embryonic (E) 16-, 18-, and 20-day-old fetuses and postnatal (P) 1-, 3-, 5-, 7-, 14-, and 21-day-old pups were obtained. Immunohistochemical analysis, dual-labeled immunofluorescence, terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) technique assay and Western blot were performed in addition to histological analysis. RESULTS Immunohistochemistry showed that Cyt-c was strongly expressed in proximal and distal tubules (DTs) at all time points. Caspase-9 and Apaf-1 were strongly expressed in proximal tubules (PTs) but only weakly expressed in DTs. Dual-labeled immunofluorescence showed that most tubules expressed both Cyt-c and Apaf-1, except for some tubules that only expressed Cyt-c. The TUNEL assay showed a greater percentage of apoptotic cells in PTs compared to DTs. Apaf-1 and cleaved caspase-9 protein expression gradually increased during the embryonic period and peaked during the early postnatal period but apparently declined from P7. Cyt-c protein expression was weak during the embryonic period but obviously increased after P1. CONCLUSION This study showed that PTs are more sensitive to apoptosis than DTs during rat renal development, even though both tubule segments contain a large number of mitochondria. Furthermore, Cyt-c-mediated mitochondrial apoptosis-related proteins play an important role in PTs during the early postnatal kidney development.
Collapse
Affiliation(s)
- Xiao-Feng Song
- Department of Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | | | | | | |
Collapse
|
19
|
Agabiti SS, Liang Y, Wiemer AJ. Molecular mechanisms linking geranylgeranyl diphosphate synthase to cell survival and proliferation. Mol Membr Biol 2016; 33:1-11. [PMID: 27537059 DOI: 10.1080/09687688.2016.1213432] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Geranylgeranyl diphosphate is a 20-carbon isoprenoid phospholipid whose lipid moiety can be post-translationally incorporated into proteins to promote membrane association. The process of geranylgeranylation has been implicated in anti-proliferative effects of clinical agents that inhibit enzymes of the mevalonate pathway (i.e. statins and nitrogenous bisphosphonates) as well as experimental agents that deplete geranylgeranyl diphosphate. Inhibitors of geranylgeranyl diphosphate synthase are an attractive way to block geranylgeranylation because they possess a calcium-chelating substructure to allow localization to bone and take advantage of a unique position of the enzyme within the biosynthetic pathway. Here, we describe recent advances in geranylgeranyl diphosphate synthase expression and inhibitor development with a particular focus on the molecular mechanisms that link geranylgeranyl diphosphate to cell proliferation via geranylgeranylated small GTPases.
Collapse
Affiliation(s)
- Sherry S Agabiti
- a Department of Pharmaceutical Sciences , University of Connecticut , Storrs , CT , USA
| | - Yilan Liang
- a Department of Pharmaceutical Sciences , University of Connecticut , Storrs , CT , USA
| | - Andrew J Wiemer
- a Department of Pharmaceutical Sciences , University of Connecticut , Storrs , CT , USA.,b Institute for Systems Genomics, University of Connecticut , Storrs , CT , USA
| |
Collapse
|
20
|
Minegaki T, Fukushima S, Morioka C, Takanashi H, Uno J, Tsuji S, Yamamoto S, Watanabe A, Tsujimoto M, Nishiguchi K. Effects of bisphosphonates on human esophageal squamous cell carcinoma cell survival. Dis Esophagus 2016; 29:656-62. [PMID: 25894100 DOI: 10.1111/dote.12370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most malignant cancers in Japan. Anticancer chemotherapy has been useful for ESCC treatment. However, therapeutic options are limited. Recently, bisphosphonates (BPs), which are osteoporosis drugs, have shown anticancer effects in several cancer cell lines, but the effects against ESCC cell lines are unknown. In this study, we examined the cytotoxic effects of BPs and their mechanisms of cytotoxicity in human ESCC cell lines. A first-generation BP (etidronate), two second-generation BPs (alendronate and pamidronate), and two third-generation BPs (risedronate and zoledronate) were used in this study. All BPs, except etidronate, were cytotoxic, as indicated by increased caspase-3/7 activity and numbers of Annexin-fluorescein isothiocyanate positive cells in ESCC cell lines. From cell cycle analysis, G0/G1-phase arrest was observed upon treatment with second- and third-generation BPs. In addition, Cyclin D1 protein expression levels were decreased by second- and third-generation BP treatment. Although squalene and trans, trans-farnesol minimally affected BP cytotoxicity, treatment with geranylgeraniol inhibited BP cytotoxicity almost completely. We concluded that second- and third-generation BPs are cytotoxic to ESCC cell lines as they induce apoptosis and inhibit the cell cycle through mevalonate pathway inhibition. Therefore, BP treatment may be a beneficial therapy in ESCC patients.
Collapse
Affiliation(s)
- T Minegaki
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - S Fukushima
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - C Morioka
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - H Takanashi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - J Uno
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - S Tsuji
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - S Yamamoto
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - A Watanabe
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - M Tsujimoto
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - K Nishiguchi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
21
|
Antitumor effects of minodronate, a third-generation nitrogen-containing bisphosphonate, in synergy with γδT cells in human glioblastoma in vitro and in vivo. J Neurooncol 2016; 129:231-41. [PMID: 27393349 DOI: 10.1007/s11060-016-2186-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/11/2016] [Indexed: 12/30/2022]
Abstract
Nitrogen-containing bisphosphonates (N-BPs), which prevent bone resorption, exert direct and γδT cell (GDT)-mediated antitumor effects against several tumor cell types, including glioblastoma (GBM). However, limited information is available regarding the antitumor effects of N-BPs in GBM. Specifically, the antitumor effects of minodronate (MDA), a third-generation N-BP, in GBM are yet unclear. This study aimed to investigate the antitumor effects of MDA in GBM in vitro and in vivo. We performed growth inhibition and apoptosis detection assays using the GBM cell lines U87MG and U138MG. Apoptosis inhibition assays were also conducted. In vivo xenograft assays were performed in highly immunodeficient NOD.Cg-Prkdc(scid) Il2rg(tm1Sug)/Jic mice subcutaneously implanted with U87MG and U138MG cells. Growth inhibition and apoptosis detection assays demonstrated that MDA inhibited GBM cell growth via apoptosis, which was markedly enhanced by ex vivo expanded GDT. A pan-caspase inhibitor, z-VAD-fmk, inhibited MDA-induced U138MG apoptosis and MDA/GDT-induced U87MG and U138MG apoptosis. But z-VAD-fmk increased MDA-induced U87MG apoptosis. MDA/GDT-mediated apoptosis was blocked by the anti-T cell receptor (TCR) Vγ9, mevalonate pathway inhibitor, granzyme B inhibitor, and antitumor necrosis factor (TNF)-α. In vivo xenograft assays showed that combined intraperitoneal administration of MDA/GDT induced antitumor effects on unestablished U87MG-derived subcutaneous tumors. MDA exerted direct and GDT-mediated anti-GBM apoptotic effects in a caspase-dependent manner. GDT recognized MDA-exposed GBM cells via TCRVγ9 and induced apoptosis via granzyme B and TNF-α release. Because MDA elicited anti-GBM effects in synergy with GDT in vivo, a combination of MDA and ex vivo-generated GDT could be an effective treatment in patients with GBM.
Collapse
|
22
|
Elsayed M, Kobayashi D, Kubota T, Matsunaga N, Murata R, Yoshizawa Y, Watanabe N, Matsuura T, Tsurudome Y, Ogino T, Ohdo S, Shimazoe T. Synergistic Antiproliferative Effects of Zoledronic Acid and Fluvastatin on Human Pancreatic Cancer Cell Lines: An in Vitro Study. Biol Pharm Bull 2016; 39:1238-46. [PMID: 27181081 DOI: 10.1248/bpb.b15-00746] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bisphosphonates and statins are known to have antitumor activities against different types of cancer cell lines. In the present study, we investigated the antiproliferative effects of the combination of zoledronic acid (ZOL), a bisphophosphonate, and fluvastatin (FLU), a statin, in vitro on two types of human pancreatic cancer cell lines, Mia PaCa-2 and Suit-2. The pancreatic cancer cell lines were treated with ZOL and FLU both individually and in combination to evaluate their antiproliferative effects using WST-8 cell proliferation assay. In this study, we demonstrated a potent synergistic antiproliferative effect of both drugs when used in combination in both cell lines. Moreover, we studied the molecular mechanism behind this synergistic effect, which was inhibited by the addition of the mevalonate pathway products, farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP). Furthermore, we aimed to determine the effect of ZOL and FLU combination on RhoA and Ras guanosine 5'-triphosphate (GTP)-proteins. The combination induced a marked accumulation in RhoA and unprenylated Ras. GGPP and FPP reversed the increase in the amount of both proteins. These results indicated that the combination treatment impaired RhoA and Ras signaling pathway by the inhibition of geranylgeranylation and/or farnesylation. This study provides a potentially effective approach for the treatment of pancreatic cancer using a combination treatment of ZOL and FLU.
Collapse
Affiliation(s)
- Mahitab Elsayed
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Tsubaki M, Mashimo K, Takeda T, Kino T, Fujita A, Itoh T, Imano M, Sakaguchi K, Satou T, Nishida S. Statins inhibited the MIP-1α expression via inhibition of Ras/ERK and Ras/Akt pathways in myeloma cells. Biomed Pharmacother 2016; 78:23-29. [DOI: 10.1016/j.biopha.2015.12.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/15/2015] [Indexed: 11/28/2022] Open
|
24
|
Reel B, Korkmaz CG, Arun MZ, Yildirim G, Ogut D, Kaymak A, Micili SC, Ergur BU. The Regulation of Matrix Metalloproteinase Expression and the Role of Discoidin Domain Receptor 1/2 Signalling in Zoledronate-treated PC3 Cells. J Cancer 2015; 6:1020-9. [PMID: 26366216 PMCID: PMC4565852 DOI: 10.7150/jca.12733] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/27/2015] [Indexed: 11/05/2022] Open
Abstract
Discoidin Domain Receptors (DDR1/DDR2) are tyrosine kinase receptors which are activated by collagen. DDR signalling regulates cell migration, proliferation, apoptosis and matrix metalloproteinase (MMP) production. MMPs degrade extracellular matrix (ECM) and play essential role in tumor growth, invasion and metastasis. Nitrogen-containing bisphosphonates (N-BPs) which strongly inhibit osteoclastic activity are commonly used for osteoporosis treatment. They also have MMP inhibitory effect. In this study, we aimed to investigate the effects of zoledronate in PC3 cells and the possible role of DDR signalling and downstream pathways in these inhibitory effects. We studied messenger RNA (mRNA) and protein expressions of MMP-2,-9,-8, DDR1/DDR2 type I procollagen (TIP) and mRNA levels of PCA-1, MMP-13 and DDR-initiated signalling pathway players including K-Ras oncogene, ERK1, JNK1, p38, AKT-1 and BCLX in PC3 cells in the presence or absence of zoledronate (10-100 μM) for 2-3 days. Zoledronate (100 μM) down-regulated DDR1/ DDR2, TIP mRNAs but did not change MMP-13 (collagenase-3) mRNA. However, zoledronate up-regulated MMP-8 (collagenase-2) mRNA. Zoledronate also inhibited mRNA expressions of K-Ras, ERK1, AKT-1, BCLX and PCA-1; but did not change JNK1, p38 mRNA levels. Zoledronate (100 μM) supressed DDR1/DDR2, TIP expressions; and gelatinase (MMP-2/MMP-9) expressions/activities. Conversely, zoledronate up-regulated MMP-8 expression in PC3 cells. Zoledronate down-regulates MMP-2/-9 expressions in PC3 prostate cancer cells. DDR1/DDR2 signalling and DDR-initiated downstream Ras/Raf/ERK and PI3K/AKT pathways may at least partially responsible for MMP inhibitory effect of zoledronate.
Collapse
Affiliation(s)
- Buket Reel
- 1. Department of Pharmacology, Faculty of Pharmacy, Ege University, Bornova, Izmir, Turkey
| | - Ceren Gonen Korkmaz
- 1. Department of Pharmacology, Faculty of Pharmacy, Ege University, Bornova, Izmir, Turkey
| | - Mehmet Zuhuri Arun
- 1. Department of Pharmacology, Faculty of Pharmacy, Ege University, Bornova, Izmir, Turkey
| | - Gokce Yildirim
- 1. Department of Pharmacology, Faculty of Pharmacy, Ege University, Bornova, Izmir, Turkey
| | - Deniz Ogut
- 1. Department of Pharmacology, Faculty of Pharmacy, Ege University, Bornova, Izmir, Turkey
| | - Aysegul Kaymak
- 1. Department of Pharmacology, Faculty of Pharmacy, Ege University, Bornova, Izmir, Turkey
| | - Serap Cilaker Micili
- 2. Department of Histology and Embriology, School of Medicine, Dokuz Eylul University, Inciralti, Izmir, Turkey
| | - Bekir Ugur Ergur
- 2. Department of Histology and Embriology, School of Medicine, Dokuz Eylul University, Inciralti, Izmir, Turkey
| |
Collapse
|
25
|
Tanaka M, Mori H, Kayasuga R, Kawabata K. Induction of creatine kinase release from cultured osteoclasts via the pharmacological action of aminobisphosphonates. SPRINGERPLUS 2015; 4:59. [PMID: 25664231 PMCID: PMC4315803 DOI: 10.1186/s40064-015-0848-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/22/2015] [Indexed: 11/25/2022]
Abstract
An increase of serum creatine kinase (CK) has been observed in clinical studies of nitrogen-containing aminobisphosphonates (N-BPs). Osteoclasts are thought to be the source of the CK, but there is no clear evidence for the hypothesis. In this study, CK release from rabbit osteoclasts induced by N-BPs was examined in an in vitro culture system. Rabbit bone-derived cells were cultured for 3 days on the N-BPs pretreated cortical bone slices. CK activity in the culture medium was measured at 3 days of culture. The CK activity was increased with all N-BPs at concentrations at which showed antiresorptive activity over 60% inhibition of C-terminal cross-linking telopeptide of type I collagen (CTX-1) release. The maximum induction of CK activity was 2.6 times the control level. The lowest N-BP concentration inducing CK release was 3 times lower than that required to decrease the osteoclast number. Bafilomycin A1, an inhibitor of vacuolar H+-ATPase, abrogated all N-BP actions, including CK release. Bone-derived cells except osteoclasts were insensitive to bafilomycin A1, suggesting that osteoclasts were the source of CK. Regarding the time course, CK release occurred after a 1 day lag time and increased steadily until day 3 of culture. These results show that CK release is induced by N-BPs from osteoclasts at concentrations at which N-BPs show antiresorptive activity over 60% inhibition of CTX-1 release in vitro. These findings explain the mechanism of the CK increase induced by clinical use of N-BPs.
Collapse
Affiliation(s)
- Makoto Tanaka
- Research Promotion, Ono Pharmaceutical Co., Ltd, 3-1-1, Sakurai, Shimamoto-cho, Mishima-gun Osaka, 618-8585 Japan
| | - Hiroshi Mori
- Discovery Research Laboratories, Ono Pharmaceutical Co., Ltd, Shimamoto-cho, Mishima-gun Osaka, 618-8585 Japan
| | - Ryoji Kayasuga
- Discovery Research Laboratories, Ono Pharmaceutical Co., Ltd, Shimamoto-cho, Mishima-gun Osaka, 618-8585 Japan
| | - Kazuhito Kawabata
- Discovery Research Laboratories, Ono Pharmaceutical Co., Ltd, Shimamoto-cho, Mishima-gun Osaka, 618-8585 Japan
| |
Collapse
|
26
|
Chandrasekaran V, Kalyan S, Biel V, Lettau M, Nerdal PT, Oberg HH, Wesch D, Lindhorst TK, Kabelitz D. Novel synthesis of fluorochrome-coupled zoledronate with preserved functional activity on gamma/delta T cells and tumor cells. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00063g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel fluorescent derivative of bisphosphonate zoledronate was synthesized and shown to have comparable functional activity as native zoledronate.
Collapse
Affiliation(s)
| | - Shirin Kalyan
- Institute of Immunology
- Christian-Albrechts-University of Kiel
- D-24105 Kiel
- Germany
| | - Valentina Biel
- Institute of Immunology
- Christian-Albrechts-University of Kiel
- D-24105 Kiel
- Germany
| | - Marcus Lettau
- Institute of Immunology
- Christian-Albrechts-University of Kiel
- D-24105 Kiel
- Germany
| | | | - Hans-Heinrich Oberg
- Institute of Immunology
- Christian-Albrechts-University of Kiel
- D-24105 Kiel
- Germany
| | - Daniela Wesch
- Institute of Immunology
- Christian-Albrechts-University of Kiel
- D-24105 Kiel
- Germany
| | - Thisbe K. Lindhorst
- Otto Diels Institute of Organic Chemistry
- Christian-Albrechts-University of Kiel
- D-24118 Kiel
- Germany
| | - Dieter Kabelitz
- Institute of Immunology
- Christian-Albrechts-University of Kiel
- D-24105 Kiel
- Germany
| |
Collapse
|
27
|
Barszczyk A, Sun HS, Quan Y, Zheng W, Charlton MP, Feng ZP. Differential roles of the mevalonate pathway in the development and survival of mouse Purkinje cells in culture. Mol Neurobiol 2014; 51:1116-29. [PMID: 24973985 DOI: 10.1007/s12035-014-8778-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 06/01/2014] [Indexed: 11/29/2022]
Abstract
The cerebellum is an important locus for motor learning and higher cognitive functions, and Purkinje cells constitute a key component of its circuit. Biochemically, significant turnover of cholesterol occurs in Purkinje cells, causing the activation of the mevalonate pathway. The mevalonate pathway has important roles in cell survival and development. In this study, we investigated the outcomes of mevalonate inhibition in immature and mature mouse cerebellar Purkinje cells in culture. Specifically, we found that the inhibition of the mevalonate pathway by mevastatin resulted in cell death, and geranylgeranylpyrophosphate (GGPP) supplementation significantly enhanced neuronal survival. The surviving immature Purkinje cells, however, exhibited dendritic developmental deficits. The morphology of mature cells was not affected. The inhibition of squalene synthase by zaragozic acid caused impaired dendritic development, similar to that seen in the GGPP-rescued Purkinje cells. Our results indicate GGPP is required for cell survival and squalene synthase for the cell development of Purkinje cells. Abnormalities in Purkinje cells are linked to motor-behavioral learning disorders such as cerebellar ataxia. Thus, serious caution should be taken when using drugs that inhibit geranylgeranylation or the squalene-cholesterol branch of the pathway in the developing stage.
Collapse
Affiliation(s)
- Andrew Barszczyk
- Department of Physiology, University of Toronto, Medical Sciences Building, Rm. 3306, 1 King's College, Toronto, ON, M5S 1A8, Canada
| | | | | | | | | | | |
Collapse
|
28
|
The effect of alendronate on proteome of hepatocellular carcinoma cell lines. INTERNATIONAL JOURNAL OF PROTEOMICS 2014; 2014:532953. [PMID: 24653834 PMCID: PMC3932719 DOI: 10.1155/2014/532953] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 12/13/2013] [Accepted: 12/14/2013] [Indexed: 12/12/2022]
Abstract
Cancer is a life threatening disorder effecting 11 million people worldwide annually. Among various types of cancers, Hepatocellular carcinoma (HCC) has a higher rate of mortality and is the fifth leading cause of cancer related deaths around the world. Many chemotherapeutic drugs have been used for the treatment of HCC with many side effects. These drugs are inhibitors of different cell regulatory pathways. Mevalonate (MVA) pathway is an important cellular cascade vital for cell growth. A variety of inhibitors of MVA pathway have been reported for their anticancerous activity. Bisphosphonates (BPs) are members of a family involved in the treatment of skeletal complications. In recent years, their anticancer potential has been highlighted. Current study focuses on exploring the effects of alendronate (ALN), a nitrogen containing BP, on hepatocellular carcinoma cell line using genomic and proteomics approach. Our results identified ten differentially expressed proteins, of which five were up regulated and five were down regulated in ALN treated cells. Furthermore, we also performed gene expression analysis in treated and control cell lines. The study may help in understanding the molecular mechanism involved in antitumor activity of ALN, identification of possible novel drug targets, and designing new therapeutic strategies for HCC.
Collapse
|
29
|
Tsubaki M, Komai M, Itoh T, Imano M, Sakamoto K, Shimaoka H, Takeda T, Ogawa N, Mashimo K, Fujiwara D, Mukai J, Sakaguchi K, Satou T, Nishida S. Nitrogen-containing bisphosphonates inhibit RANKL- and M-CSF-induced osteoclast formation through the inhibition of ERK1/2 and Akt activation. J Biomed Sci 2014; 21:10. [PMID: 24490900 PMCID: PMC3996180 DOI: 10.1186/1423-0127-21-10] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 01/20/2014] [Indexed: 11/12/2022] Open
Abstract
Background Bisphosphonates are an important class of antiresorptive drugs used in the treatment of metabolic bone diseases. Recent studies have shown that nitrogen-containing bisphosphonates induced apoptosis in rabbit osteoclasts and prevented prenylated small GTPase. However, whether bisphosphonates inhibit osteoclast formation has not been determined. In the present study, we investigated the inhibitory effect of minodronate and alendronate on the osteoclast formation and clarified the mechanism involved in a mouse macrophage-like cell lines C7 and RAW264.7. Results It was found that minodronate and alendronate inhibited the osteoclast formation of C7 cells induced by receptor activator of NF-κB ligand and macrophage colony stimulating factor, which are inhibited by the suppression of geranylgeranyl pyrophosphate (GGPP) biosynthesis. It was also found that minodronate and alendronate inhibited the osteoclast formation of RAW264.7 cells induced by receptor activator of NF-κB ligand. Furthermore, minodronate and alendornate decreased phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt; similarly, U0126, a mitogen protein kinase kinase 1/2 (MEK1/2) inhibitor, and LY294002, a phosphatidylinositol 3-kinase (PI3K) inhibitor, inhibited osteoclast formation. Conclusions This indicates that minodronate and alendronate inhibit GGPP biosynthesis in the mevalonate pathway and then signal transduction in the MEK/ERK and PI3K/Akt pathways, thereby inhibiting osteoclast formation. These results suggest a novel effect of bisphosphonates that could be effective in the treatment of bone metabolic diseases, such as osteoporosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Shozo Nishida
- Division of Pharmacotherapy, Kinki University School of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan.
| |
Collapse
|