51
|
Liu J, Liu L, Yagüe E, Yang Q, Pan T, Zhao H, Hu Y, Zhang J. GGNBP2 suppresses triple-negative breast cancer aggressiveness through inhibition of IL-6/STAT3 signaling activation. Breast Cancer Res Treat 2018; 174:65-78. [PMID: 30450530 DOI: 10.1007/s10549-018-5052-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/13/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, lacking effective targeted therapies, and whose underlying mechanisms are still unclear. The gene coding for Gametogenetin-binding protein (GGNBP2), also known as Zinc Finger Protein 403 (ZNF403), is located on chromosome 17q12-q23, a region known as a breast cancer susceptibility locus. We have previously reported that GGNBP2 functions as a tumor suppressor in estrogen receptor-positive breast cancer. The aim of this study was to evaluate the role and mechanisms of GGNBP2 in TNBC. METHODS The effect of GGNBP2 on TNBC aggressiveness was investigated both in vitro and in vivo. The protein and mRNA expression levels were analyzed by western blotting and reverse transcription quantitative polymerase chain reaction, respectively. Fluorescence-activated cell sorting analysis was used to evaluate the cell cycle distribution and cell apoptosis. Immunohistochemistry was used to determine the expression of GGNBP2 in breast cancer tissues. RESULTS We find that GGNBP2 expression decreases in TNBC tissues and is associated with the outcome of breast cancer patients. Furthermore, experimental overexpression of GGNBP2 in MDA-MB-231 and Cal51 cells suppresses cell proliferation, migration and invasion, reduces the cancer stem cell subpopulation, and promotes cell apoptosis in vitro as well as inhibits tumor growth in vivo. In these cell models, overexpression of GGNBP2 decreases the activation of IL-6/STAT3 signaling. CONCLUSION Our data demonstrate that GGNBP2 suppresses cancer aggressiveness by inhibition of IL-6/STAT3 activation in TNBC.
Collapse
Affiliation(s)
- Jingjing Liu
- The 3rd Department of Breast Cancer, Treatment and Research Center, China Tianjin Breast Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan Hu Xi Road, Ti Yuan Bei, He Xi District, Tianjin, 300060, People's Republic of China
| | - Lei Liu
- The 3rd Department of Breast Cancer, Treatment and Research Center, China Tianjin Breast Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan Hu Xi Road, Ti Yuan Bei, He Xi District, Tianjin, 300060, People's Republic of China
| | - Ernesto Yagüe
- Division of Cancer, Faculty of Medicine, Cancer Research Center, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Qianxi Yang
- The 3rd Department of Breast Cancer, Treatment and Research Center, China Tianjin Breast Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan Hu Xi Road, Ti Yuan Bei, He Xi District, Tianjin, 300060, People's Republic of China
| | - Teng Pan
- The 3rd Department of Breast Cancer, Treatment and Research Center, China Tianjin Breast Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan Hu Xi Road, Ti Yuan Bei, He Xi District, Tianjin, 300060, People's Republic of China
| | - Hui Zhao
- The 3rd Department of Breast Cancer, Treatment and Research Center, China Tianjin Breast Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan Hu Xi Road, Ti Yuan Bei, He Xi District, Tianjin, 300060, People's Republic of China
| | - Yunhui Hu
- The 3rd Department of Breast Cancer, Treatment and Research Center, China Tianjin Breast Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan Hu Xi Road, Ti Yuan Bei, He Xi District, Tianjin, 300060, People's Republic of China.
| | - Jin Zhang
- The 3rd Department of Breast Cancer, Treatment and Research Center, China Tianjin Breast Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan Hu Xi Road, Ti Yuan Bei, He Xi District, Tianjin, 300060, People's Republic of China.
| |
Collapse
|
52
|
Hermawan A, Putri H. Current report of natural product development against breast cancer stem cells. Int J Biochem Cell Biol 2018; 104:114-132. [DOI: 10.1016/j.biocel.2018.09.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 02/08/2023]
|
53
|
Resveratrol eliminates cancer stem cells of osteosarcoma by STAT3 pathway inhibition. PLoS One 2018; 13:e0205918. [PMID: 30356255 PMCID: PMC6200233 DOI: 10.1371/journal.pone.0205918] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022] Open
Abstract
Resveratrol shows potent anti-tumor therapeutic properties in various tumors. However, the exact effect of resveratrol on osteosarcoma cells, especially cancer stem cells, remains unclear. In this study, we examined the effect of resveratrol on osteosarcoma stem cells and explored the underlying molecular mechanisms. Resveratrol inhibited cell viability, self-renewal ability and tumorigenesis of osteosarcoma cells, whereas showed no significant inhibition effects to normal osteoblast cells. Mechanically, resveratrol treatment decreased cytokines synthesis and inhibited JAK2/STAT3 signaling, which was consistent with the decline of cancer stem cells marker, CD133. Exogenous STAT3 activation attenuated the cancer stem cell elimination effects of resveratrol treatment. Our results demonstrated that resveratrol inhibited osteosarcoma cell proliferation and tumorigenesis ability, which was correlated with cytokines inhibition related JAK2/STAT3 signaling blockage. Resveratrol may be a promising therapeutic agent for osteosarcoma management.
Collapse
|
54
|
Antitumor effects and mechanisms of olaparib in combination with carboplatin and BKM120 on human triple‑negative breast cancer cells. Oncol Rep 2018; 40:3223-3234. [PMID: 30272286 PMCID: PMC6196642 DOI: 10.3892/or.2018.6716] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) refers to a heterogeneous group of tumors, for which there is currently a lack of targeted therapies. Poly(ADP-ribose) polymerase (PARP) inhibitors, phosphatidylinositol 3-kinase (PI3K) inhibitors and carboplatin (CBP) have demonstrated sufficient efficacy and safety for their use as individual drugs for the treatment of TNBC; however, their effects on TNBC when used as a combination have not been investigated. The primary objectives of the present study were to determine the effects of a combination of CBP, olaparib and NVP-BKM120 (BKM120), and to investigate the mechanism underlying their effects on TNBC cells. The drug combination was cytotoxic to TNBC cells, both with regards to short-term and long-term sensitivity, as determined using colony forming assays, and they exerted strong synergistic effects on MDA-MB-231 and CAL51 cell lines. All drugs affected cell cycle progression, and western blotting and immunofluorescence indicated that the the drug combination exerted its cytotoxicity via DNA damage, enhancing non-homologous end joining repair and inhibiting homologous recombination repair. These data provide a strong rationale to explore the therapeutic use of olaparib in combination with CBP and BKM120 in animal models, and later in clinical trials on patients with TNBC.
Collapse
|
55
|
Hu Y, Yagüe E, Zhao J, Wang L, Bai J, Yang Q, Pan T, Zhao H, Liu J, Zhang J. Sabutoclax, pan-active BCL-2 protein family antagonist, overcomes drug resistance and eliminates cancer stem cells in breast cancer. Cancer Lett 2018; 423:47-59. [DOI: 10.1016/j.canlet.2018.02.036] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/18/2018] [Accepted: 02/22/2018] [Indexed: 02/07/2023]
|
56
|
Padthaisong S, Dokduang H, Yothaisong S, Techasen A, Namwat N, Yongvanit P, Khuntikeo N, Titapun A, Sangkhamanon S, Loilome W. Inhibitory effect of NVP-BKM120 on cholangiocarcinoma cell growth. Oncol Lett 2018; 16:1627-1633. [PMID: 30008846 PMCID: PMC6036373 DOI: 10.3892/ol.2018.8848] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 01/25/2018] [Indexed: 01/17/2023] Open
Abstract
Abnormal activation of the phosphatidylinositol 3-kinase (PI3K) pathway has been demonstrated in certain types of cancer, including cholangiocarcinoma (CCA). This pathway may therefore be a promising target for CCA treatment. The present study assessed the inhibitory effect of NVP-BKM120, a pan-class I PI3K inhibitor, on CCA cell growth. This inhibitory effect was determined using CCA cell lines and in CCA-inoculated mice. The result from sulforhodamine B (SRB) assay demonstrated that NVP-BKM120 treatment inhibited CCA cell growth in a dose-dependent manner, even at the lowest tested concentration. The in vivo study revealed that oral administration of NVP-BKM120 (10 or 30 mg/kg) to CCA-inoculated nude mice led to a reduction in tumor growth when compared with controls, which was indicated by an immunohistochemical assay for Ki67 expression. In addition, the result from TUNEL assay demonstrated that NVP-BKM120 induced cancer cell death without any signs of toxicity, which indicated by the body weight of mice (data not shown). Western blot analysis demonstrated that NVP-BKM120 inhibited CCA cell growth by suppressing RAC serine/threonine protein kinase/mechanistic target of rapamycin activation and inhibiting the phosphorylation of phosphatase and tensin homolog, which is the inactivation form of the negative regulator of this pathway. Therefore, the results of the present study indicated that NVP-BKM120 should be considered as a therapeutic agent against CCA that could be used to improve treatment.
Collapse
Affiliation(s)
- Sureerat Padthaisong
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Research Institute (CARI), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Hasaya Dokduang
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Research Institute (CARI), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen 40002, Thailand
| | - Supak Yothaisong
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Research Institute (CARI), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Anchalee Techasen
- Cholangiocarcinoma Research Institute (CARI), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Nisana Namwat
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Research Institute (CARI), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen 40002, Thailand
| | - Puangrat Yongvanit
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Research Institute (CARI), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen 40002, Thailand
| | - Narong Khuntikeo
- Cholangiocarcinoma Research Institute (CARI), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen 40002, Thailand.,Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Attapol Titapun
- Cholangiocarcinoma Research Institute (CARI), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sakkarn Sangkhamanon
- Cholangiocarcinoma Research Institute (CARI), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen 40002, Thailand.,Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Watcharin Loilome
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
57
|
Zhou Q, Chen Y, Zhang L, Zhong Y, Zhang Z, Wang R, Jin M, Gong M, Qiu Y, Kong D. Antiproliferative effect of ZSTK474 alone or in combination with chemotherapeutic drugs on HL60 and HL60/ADR cells. Oncotarget 2018; 8:39064-39076. [PMID: 28388564 PMCID: PMC5503595 DOI: 10.18632/oncotarget.16589] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/28/2017] [Indexed: 02/06/2023] Open
Abstract
While chemotherapy remains to be one of the main approaches in the clinical treatment of acute myeloid leukemia (AML), multidrug resistance (MDR) has become a serious problem which limits the therapeutic efficacy. The important roles of the PI3K/Akt pathway in modulating cell proliferation and MDR suggest that PI3K inhibitor might be effective for treatment of AML. In the present study, the antiproliferative effects of PI3K inhibitor ZSTK474 on AML cell HL60 and the adriamycin (ADR)-resistant HL60/ADR cells were investigated. Our data indicated that ZSTK474 exhibited potent antiproliferative activity, induced G1 cell cycle arrest, but no obvious apoptosis in both cell lines. Moreover, ZSTK474 affected the protein levels of cell-cycle-related molecules including increased p27, decreased cyclin D1 and phosphorylated Rb in dose-dependent manner. The proteins downstream of PI3K including phosphorylated PDK1, Akt and GSK-3β were reduced in a dose-dependent manner after ZSTK474 treatment. ZSTK474 reversed ADR resistance, increased the intracellular accumulation of ADR, and reduced the expression and function of multidrug resistance (MDR) proteins including both P-gp and MRP1 in HL60/ADR cells. The combination of ZSTK474 and chemotherapeutic drugs cytarabine or vincristine led to a synergistic effect in HL60 and HL60/ADR cells. In conclusion, ZSTK474 showed potent antiproliferative effect on HL60 and HL60/ADR cells; combination with cytarabine or vincristine resulted in synergistic effect. Our results suggest ZSTK474 has the potential to be applied in the treatment of AML patients, while further evidences particularly those about in vivo efficacy are needed.
Collapse
Affiliation(s)
- Qianxiang Zhou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yali Chen
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Lei Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yuxu Zhong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Zhe Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Ran Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Meihua Jin
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Min Gong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Yuling Qiu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
58
|
Yndestad S, Austreid E, Svanberg IR, Knappskog S, Lønning PE, Eikesdal HP. Activation of Akt characterizes estrogen receptor positive human breast cancers which respond to anthracyclines. Oncotarget 2018; 8:41227-41241. [PMID: 28476032 PMCID: PMC5522318 DOI: 10.18632/oncotarget.17167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 03/24/2017] [Indexed: 12/18/2022] Open
Abstract
Anthracyclines are key components of human breast cancer chemotherapy. Here, we explored the role of Akt signaling in anthracycline resistance. The antitumor activity of doxorubicin and Akt inhibitor A-443654 alone or combined was examined in estrogen receptor (ER) positive and negative human breast cancer cell lines. Further, we examined mRNA changes induced by anthracyclines in locally advanced breast cancers biopsied before and after treatment in two clinical trials. Doxorubicin increased Akt phosphorylation in ER positive MCF7 and T47D cell lines, with no effect in ER negative MDA-MB231 breast cancer cells. A-443654 was significantly more cytotoxic in doxorubicin-resistant compared to doxorubicin-naïve MCF7. This difference was not observed in MDA-MB231. Among 24 patients, AKT1 gene expression increased 24 hrs after the initial epirubicin exposure in ER positive tumors responding to therapy (n=6), as compared to ER positive non-responders (n=7) or ER negative tumors (n=11). In contrast, AKT1 mRNA changes after 16 weeks of doxorubicin were unrelated to clinical response and ER status (n=30). In conclusion, rapid Akt activation was observed in ER positive breast cancers which responded to anthracyclines. Increased cytotoxicity of A-443654 in doxorubicin-resistant MCF7 cells indicates a possible role for Akt inhibitors in ER positive breast cancers where chemoresistance evolves.
Collapse
Affiliation(s)
- Synnøve Yndestad
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Eilin Austreid
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ida R Svanberg
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Stian Knappskog
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Per E Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Hans P Eikesdal
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
59
|
Di D, Chen L, Guo Y, Wang L, Zhao C, Ju J. BCSC-1 suppresses human breast cancer metastasis by inhibiting NF-κB signaling. Int J Oncol 2018; 52:1674-1684. [PMID: 29512758 DOI: 10.3892/ijo.2018.4309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/07/2018] [Indexed: 11/06/2022] Open
Abstract
Breast cancer suppressor candidate-1 (BCSC-1; also termed von Willebrand factor A domain containing 5A and LOH11CR2A) is a newly identified candidate tumor suppressor gene that has been implicated in several types of cancer in previous studies. However, there have been few reports about the association between BCSC-1 and human breast cancer in recent years. In the present study, the expression of BCSC-1 in breast cancer was determined by immunohistochemistry (IHC) staining of tissue microarrays and clinical tissue specimens. Subsequently, BCSC-1 gene expression was evaluated in different breast cancer cell lines by quantitative polymerase chain reaction and the MDA-MB-231 cell line was selected for further use in subsequent experiments, due to its low BCSC-1 expression. An MDA-MB-231 cell line with stable overexpression of BCSC-1 was established through transfection with plasmid containing the BCSC-1 gene, and then screening for G418 resistance. Wound-healing, migration and invasion assays were conducted to detect the effect of BCSC-1 on MDA-MB-231 cells. Furthermore, changes in matrix metalloproteinases (MMPs), osteopontin (OPN) and the nuclear factor-κB (NF-κB) pathway were detected in the current study. Additionally, stable silencing of BCSC-1 expression in MCF-7 cells was performed using a lentivirus. The results of IHC indicated that BCSC-1 is expressed at low levels in breast cancer tissues compared with in normal breast tissue. Results of the wound healing, migration and invasion assays demonstrated that BCSC-1 overexpression reduced the metastasis ability of MDA-MB-231 cells in vitro. Further research confirmed that the BCSC-1 overexpression reduced the expression levels of MMP7, MMP9 and OPN, and the phosphorylation of NF-κB p65. Furthermore, inhibition of BCSC-1 via lentivirus-mediated RNA interference revealed that the downregulation of BCSC-1 increased the invasive ability of MCF-7 cells. In summary, the results demonstrated that BCSC-1 is expressed at low levels in breast cancer tissues, and that it can suppress human breast cancer cell migration and invasion, potentially altering the expression of MMP7, MMP9, OPN, and the activity of the NF-κB pathway. Therefore, BCSC-1 may be useful as a biomarker for the treatment of breast cancer in the future.
Collapse
Affiliation(s)
- Dalin Di
- Department of Immunology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Lei Chen
- Department of Hematology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Yingying Guo
- Department of Immunology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Lina Wang
- Department of Immunology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Chunling Zhao
- School of Biological Science, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Jiyu Ju
- Department of Immunology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
60
|
Ai Y, Zhou Q, Li L, Pan Z, Guo M, Han J. Interference of P-REX2a may inhibit proliferation and reverse the resistance of SGC7901 cells to doxorubicin. Oncol Lett 2018; 15:3185-3191. [PMID: 29435055 DOI: 10.3892/ol.2017.7693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/15/2017] [Indexed: 12/23/2022] Open
Abstract
Drug resistance inhibits the efficacy of doxorubicin in gastric cancer. Phosphatidylinositol 3,4,5-trisphosphate RAC exchanger 2a (P-REX2a) activates the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) signaling pathway by binding to and inactivating phosphatase and tensin homolog (PTEN), which functions as a tumor promoter in a number of types of cancer. However, there is no research concerning the association between P-REX2a expression and drug resistance in gastric cancer. In the present study, the expression of P-REX2a in clinical gastric cancer tissues was detected, and the mechanism of doxorubicin resistance in the gastric cancer cell line SGC7901 was investigated. Using reverse transcription-quantitative polymerase chain reaction and western blotting, it was demonstrated that the mRNA and protein expression of P-REX2a was increased in gastric cancer tissues. MTT assays were also used to determine proliferation, and proliferation was revealed to be reduced following transfection of P-REX2a small interfering (si)RNA. When the cells were treated with 0.3 µM doxorubicin for 24 h, the rate of apoptosis in the siRNA-transfected groups significantly increased and no marked changes in of PTEN and Akt expression were observed. By contrast, the activity of PTEN increased, and the expression of p-Akt (S473) decreased in the P-REX2a siRNA-transfected group compared with the control. The detection of PTEN enzymatic activity in the present study was based on phosphatidylinositol-3,4,5-trisphosphate. Therefore, it was concluded that P-REX2a may participate in the generation of resistance to doxorubicin in gastric cancer, and this may be associated with the upregulation of the PI3K/Akt signaling pathway via inactivation of PTEN.
Collapse
Affiliation(s)
- Yaowei Ai
- Department of Gastroenterology, The First People's Hospital of Yichang, China Three Georges University, Yichang, Hubei 443000, P.R. China
| | - Qiaohui Zhou
- Department of Gastroenterology, The First People's Hospital of Yichang, China Three Georges University, Yichang, Hubei 443000, P.R. China
| | - Ling Li
- Department of Gastroenterology, The First People's Hospital of Yichang, China Three Georges University, Yichang, Hubei 443000, P.R. China
| | - Zhihong Pan
- Department of Gastroenterology, The First People's Hospital of Yichang, China Three Georges University, Yichang, Hubei 443000, P.R. China
| | - Mingwen Guo
- Department of Gastroenterology, The First People's Hospital of Yichang, China Three Georges University, Yichang, Hubei 443000, P.R. China
| | - Jingbo Han
- Department of Anesthesiology, Ren He Hospital of Three Gorges University, Yichang, Hubei 443000, P.R. China
| |
Collapse
|
61
|
Rychahou P, Bae Y, Reichel D, Zaytseva YY, Lee EY, Napier D, Weiss HL, Roller N, Frohman H, Le AT, Mark Evers B. Colorectal cancer lung metastasis treatment with polymer-drug nanoparticles. J Control Release 2018; 275:85-91. [PMID: 29421609 PMCID: PMC5908241 DOI: 10.1016/j.jconrel.2018.02.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 01/27/2018] [Accepted: 02/04/2018] [Indexed: 12/18/2022]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer deaths in the United States; the predominant cause for mortality is metastasis to distant organs (e.g., lung). A major problem limiting the success of chemotherapy in metastatic CRC is the inability to target tumor tissues selectively and avoid severe side effects to normal tissues and organs. Here, we demonstrate polymeric nanoparticles (PNPs) entrapping chemotherapeutic agents provide a new therapeutic option for treating CRC that has metastasized to the lung. PNPs assembled from FDA approved biocompatible block copolymer accumulated predominantly in lung tissue. PNPs showed negligible accumulation in liver, spleen and kidneys, which was confirmed by fluorescent nanoparticle imaging and analysis of PI3K inhibition in the organs. PNPs entrapping PI3K inhibitors (i.e., wortmannin and PX866) suppressed CRC lung metastasis growth, and SN-38-loaded PNPs completely eliminated CRC lung metastasis. Our results demonstrate that polymer-drug nanoparticles offer a new approach to reduce toxicity of cancer therapy and has the potential to improve outcomes for patients with lung metastasis.
Collapse
Affiliation(s)
- Piotr Rychahou
- Markey Cancer Center, The University of Kentucky, Lexington, KY 40536, United States; Department of Surgery, The University of Kentucky, Lexington, KY 40536, United States
| | - Younsoo Bae
- Department of Pharmaceutical Sciences, The University of Kentucky, Lexington, KY 40536, United States
| | - Derek Reichel
- Department of Pharmaceutical Sciences, The University of Kentucky, Lexington, KY 40536, United States
| | - Yekaterina Y Zaytseva
- Markey Cancer Center, The University of Kentucky, Lexington, KY 40536, United States
| | - Eun Y Lee
- Markey Cancer Center, The University of Kentucky, Lexington, KY 40536, United States; Pathology and Laboratory Medicine, The University of Kentucky, Lexington, KY 40536, United States
| | - Dana Napier
- Pathology and Laboratory Medicine, The University of Kentucky, Lexington, KY 40536, United States
| | - Heidi L Weiss
- Markey Cancer Center, The University of Kentucky, Lexington, KY 40536, United States
| | - Nick Roller
- Markey Cancer Center, The University of Kentucky, Lexington, KY 40536, United States
| | - Heather Frohman
- Markey Cancer Center, The University of Kentucky, Lexington, KY 40536, United States; Department of Surgery, The University of Kentucky, Lexington, KY 40536, United States
| | - Anh-Thu Le
- Markey Cancer Center, The University of Kentucky, Lexington, KY 40536, United States; Department of Surgery, The University of Kentucky, Lexington, KY 40536, United States
| | - B Mark Evers
- Markey Cancer Center, The University of Kentucky, Lexington, KY 40536, United States; Department of Surgery, The University of Kentucky, Lexington, KY 40536, United States.
| |
Collapse
|
62
|
Dong C, Yang R, Li H, Ke K, Luo C, Yang F, Shi XN, Zhu Y, Liu X, Wong MH, Lin G, Wang X, Leung KS, Kung HF, Chen C, Lin MCM. Econazole nitrate inhibits PI3K activity and promotes apoptosis in lung cancer cells. Sci Rep 2017; 7:17987. [PMID: 29269744 PMCID: PMC5740072 DOI: 10.1038/s41598-017-18178-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 12/06/2017] [Indexed: 11/09/2022] Open
Abstract
The phosphatidylinositol-3-kinase (PI3K)/AKT signaling pathway plays a pivotal role in many cellular processes, including the proliferation, survival and differentiation of lung cancer cells. Thus, PI3K is a promising therapeutic target for lung cancer treatment. In this study, we applied free and open-source protein-ligand docking software, screened 3167 FDA-approved small molecules, and identified putative PI3Kα inhibitors. Among them, econazole nitrate, an antifungal agent, exhibited the highest activity in decreasing cell viability in pathological types of NSCLC cell lines, including H661 (large cell lung cancer) and A549 (adenocarcinoma). Econazole decreased the protein levels of p-AKT and Bcl-2, but had no effect on the phosphorylation level of ERK. It inhibited cell growth and promote apoptosis in a dose-dependent manner. Furthermore, the combination of econazole and cisplatin exhibited additive and synergistic effects in the H661 and A549 lung cancer cell lines, respectively. Finally, we demonstrated that econazole significantly suppressed A549 tumor growth in nude mice. Our findings suggest that econazole is a new PI3K inhibitor and a potential drug that can be used in lung cancer treatment alone or in combination with cisplatin.
Collapse
Affiliation(s)
- Chao Dong
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan, China.,Department of the second medical oncology, The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, China
| | - Runxiang Yang
- Department of the second medical oncology, The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, China
| | - Hongjian Li
- Institute of Future Cities, Chinese University of Hong Kong, Hong Kong, China.,Department of Computer Science and Engineering, Chinese University of Hong Kong, Hong Kong, China
| | - Kunbin Ke
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan, China.,Department of Urology, the 1st Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chunxiang Luo
- Department of the second medical oncology, The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, China
| | - Fang Yang
- Department of the second medical oncology, The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, China
| | - Xi-Nan Shi
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan, China
| | - Ying Zhu
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan, China
| | - Xu Liu
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan, China
| | - Man-Hon Wong
- Department of Computer Science and Engineering, Chinese University of Hong Kong, Hong Kong, China
| | - Guimiao Lin
- School of Basic Medical Sciences, Shenzhen University Health Sciences Center, Shenzhen, China
| | - Xiaomei Wang
- School of Basic Medical Sciences, Shenzhen University Health Sciences Center, Shenzhen, China
| | - Kwong-Sak Leung
- Institute of Future Cities, Chinese University of Hong Kong, Hong Kong, China.,Department of Computer Science and Engineering, Chinese University of Hong Kong, Hong Kong, China
| | - Hsiang-Fu Kung
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Marie Chia-Mi Lin
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan Province, China.
| |
Collapse
|
63
|
García-Aranda M, Redondo M. Protein Kinase Targets in Breast Cancer. Int J Mol Sci 2017; 18:ijms18122543. [PMID: 29186886 PMCID: PMC5751146 DOI: 10.3390/ijms18122543] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 01/10/2023] Open
Abstract
With 1.67 million new cases and 522,000 deaths in the year 2012, breast cancer is the most common type of diagnosed malignancy and the second leading cause of cancer death in women around the world. Despite the success of screening programs and the development of adjuvant therapies, a significant percentage of breast cancer patients will suffer a metastatic disease that, to this day, remains incurable and justifies the research of new therapies to improve their life expectancy. Among the new therapies that have been developed in recent years, the emergence of targeted therapies has been a milestone in the fight against cancer. Over the past decade, many studies have shown a causal role of protein kinase dysregulations or mutations in different human diseases, including cancer. Along these lines, cancer research has demonstrated a key role of many protein kinases during human tumorigenesis and cancer progression, turning these molecules into valid candidates for new targeted therapies. The subsequent discovery and introduction in 2001 of the kinase inhibitor imatinib, as a targeted treatment for chronic myelogenous leukemia, revolutionized cancer genetic pathways research, and lead to the development of multiple small-molecule kinase inhibitors against various malignancies, including breast cancer. In this review, we analyze studies published to date about novel small-molecule kinase inhibitors and evaluate if they would be useful to develop new treatment strategies for breast cancer patients.
Collapse
Affiliation(s)
- Marilina García-Aranda
- Biochemistry Department, Hospital Costa del Sol, Carretera de Cádiz km, 187, 29600 Marbella, Málaga, Spain.
| | - Maximino Redondo
- Biochemistry Department, Hospital Costa del Sol, Carretera de Cádiz km, 187, 29600 Marbella, Málaga, Spain.
- Biochemistry Department, Facultad de Medicina de la Universidad de Málaga, Bulevar Louis Pasteur 32, 29010 Málaga, Spain.
| |
Collapse
|
64
|
Valdés-Rives SA, Casique-Aguirre D, Germán-Castelán L, Velasco-Velázquez MA, González-Arenas A. Apoptotic Signaling Pathways in Glioblastoma and Therapeutic Implications. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7403747. [PMID: 29259986 PMCID: PMC5702396 DOI: 10.1155/2017/7403747] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/22/2017] [Accepted: 09/28/2017] [Indexed: 12/18/2022]
Abstract
Glioblastoma multiforme (GBM) is the most hostile type of brain cancer. Its aggressiveness is due to increased invasion, migration, proliferation, angiogenesis, and a decreased apoptosis. In this review, we discuss the role of key regulators of apoptosis in GBM and glioblastoma stem cells. Given their importance in the etiology and pathogenesis of GBM, these signaling molecules may represent potential therapeutic targets.
Collapse
Affiliation(s)
- Silvia Anahi Valdés-Rives
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Diana Casique-Aguirre
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Liliana Germán-Castelán
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Marco A. Velasco-Velázquez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Unidad Periférica de Investigación en Biomedicina Translacional, ISSSTE C.M.N. 20 de Noviembre, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Aliesha González-Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
65
|
Novel pan PI3K inhibitor-induced apoptosis in APL cells correlates with suppression of telomerase: An emerging mechanism of action of BKM120. Int J Biochem Cell Biol 2017; 91:1-8. [DOI: 10.1016/j.biocel.2017.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/30/2017] [Accepted: 08/14/2017] [Indexed: 12/21/2022]
|
66
|
Yang S, Li X, Guan W, Qian M, Yao Z, Yin X, Zhao H. NVP-BKM120 inhibits colon cancer growth via FoxO3a-dependent PUMA induction. Oncotarget 2017; 8:83052-83062. [PMID: 29137323 PMCID: PMC5669949 DOI: 10.18632/oncotarget.20943] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/26/2017] [Indexed: 01/01/2023] Open
Abstract
NVP-BKM120, a potent and highly selective PI3K inhibitor, is currently being investigated in phase I/II clinical trials. The mechanisms of action of NVP-BKM120 in colon cancer cells are unclear. In the present study, we investigated how NVP-BKM120 suppresses colon cancer cells growth and potentiates effects of other chemotherapeutic drugs. We found that NVP-BKM120 treatment enhance PUMA induction irrespective of p53 status through the FoxO3a pathway following AKT inhibition. Furthermore, PUMA is required for NVP-BKM120-induced apoptosis in colon cancer cells. In addition, NVP-BKM120 also synergized with 5-Fluorouracil or regorafenib to induce marked apoptosis via PUMA induction. Deficiency of PUMA suppressed apoptosis and antitumor effect of NVP-BKM120 in xenograft model. These results demonstrate a key role of PUMA in mediating the anticancer effects of NVP-BKM120 and suggest that PUMA could be used as an indicator of NVP-BKM120 sensitivity, and also have important implications for it clinical applications.
Collapse
Affiliation(s)
- Shida Yang
- Department of Laboratory Medicine, The People's Hospital of Liaoning Province, Shenyang, China
| | - Xin Li
- Department of Anesthesia, The People's Hospital of Liaoning Province, Shenyang, China
| | - Wenchang Guan
- Department of Gynaecology and Obstetrics, The People's Hospital of Liaoning Province, Shenyang, China
| | - Mingqin Qian
- Department of Ultrasound Diagnosis, The People's Hospital of Liaoning Province, Shenyang, China
| | - Zhicheng Yao
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Xiaoxue Yin
- Department of Laboratory Medicine, The People's Hospital of Liaoning Province, Shenyang, China
| | - Hongmei Zhao
- Department of Laboratory Medicine, The People's Hospital of Liaoning Province, Shenyang, China
| |
Collapse
|
67
|
Guan J, Zhao Q, Lv J, Zhang Z, Sun S, Mao W. Triptolide induces DNA breaks, activates caspase-3-dependent apoptosis and sensitizes B-cell lymphoma to poly(ADP-ribose) polymerase 1 and phosphoinositide 3-kinase inhibitors. Oncol Lett 2017; 14:4965-4970. [PMID: 29085508 DOI: 10.3892/ol.2017.6771] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 06/15/2017] [Indexed: 12/31/2022] Open
Abstract
Triptolide is the primary compound isolated from Tripterygium wilfordii, which has been reported to inhibit nucleotide excision repair as well as exhibit anti-inflammatory and antitumor activities. However, the action of triptolide in DNA breaks remains unknown. The present study investigated the effects of triptolide in the induction of DNA breaks and apoptosis in a murine B-cell lymphoma cell line, CH12F3. An MTT assay revealed that X-ray repair cross-complementing protein 1 (XRCC1)-/- CH12F3 cells were more sensitive to 6 nM triptolide compared with the wild-type CH12F3 cells, which suggests that low levels of triptolide induce DNA breaks in a manner that is dependent on the XRCC1-mediated repair pathway. Flow cytometric analysis identified that 50 nM triptolide increased the phospho-histone H2AX level, demonstrating that triptolide induces double-strand breaks. Western blot analysis revealed that triptolide up-regulated Rad51 and nuclear proliferating cell nuclear antigen. Annexin V/propidium iodide staining identified that triptolide promoted apoptosis and western blot analysis confirmed that triptolide activated caspase-3-dependent apoptosis. The results of the present study also demonstrated that 5 nM triptolide sensitized CH12F3 lymphoma cells to poly(ADP-ribose) polymerase 1 and phosphoinositide 3-kinase inhibitors, suggesting that triptolide may be a potent antitumor drug for sensitizing lymphoma cells to chemotherapeutic agents.
Collapse
Affiliation(s)
- Jiawei Guan
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Qian Zhao
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Jian Lv
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Zhiwei Zhang
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Shijie Sun
- Department of Immunology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Weifeng Mao
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
68
|
Shaker H, Harrison H, Clarke R, Landberg G, Bundred NJ, Versteeg HH, Kirwan CC. Tissue Factor promotes breast cancer stem cell activity in vitro. Oncotarget 2017; 8:25915-25927. [PMID: 28033108 PMCID: PMC5432226 DOI: 10.18632/oncotarget.13928] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/25/2016] [Indexed: 01/01/2023] Open
Abstract
Cancer stem cells (CSCs) are a subpopulation of cells that can self-renew and initiate tumours. The clotting-initiating protein Tissue Factor (TF) promotes metastasis and may be overexpressed in cancer cells with increased CSC activity. We sought to determine whether TF promotes breast CSC activity in vitro using human breast cancer cell lines. TF expression was compared in anoikis-resistant (CSC-enriched) and unselected cells. In cells sorted into of TF-expressing and TF-negative (FACS), and in cells transfected to knockdown TF (siRNA) and overexpress TF (cDNA), CSC activity was compared by (i) mammosphere forming efficiency (MFE) (ii) holoclone colony formation (Hc) and (iii) ALDH1 activity. TF expression was increased in anoikis-resistant and high ALDH1-activity T47D cells compared to unselected cells. FACS sorted TF-expressing T47Ds and TF-overexpressing MCF7s had increased CSC activity compared to TF-low cells. TF siRNA cells (MDAMB231,T47D) had reduced CSC activity compared to control cells. FVIIa increased MFE and ALDH1 in a dose-dependent manner (MDAMB231, T47D). The effects of FVIIa on MFE were abrogated by TF siRNA (T47D). Breast CSCs (in vitro) demonstrate increased activity when selected for high TF expression, when induced to overexpress TF, and when stimulated (with FVIIa). Targeting the TF pathway in vivo may abrogate CSC activity.
Collapse
Affiliation(s)
- Hudhaifah Shaker
- The University of Manchester, Manchester Academic Health Science Centre, Department of Academic Surgery, University Hospital of South Manchester, Manchester, UK
| | - Hannah Harrison
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Robert Clarke
- Breast Biology Group, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Goran Landberg
- Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Nigel J. Bundred
- The University of Manchester, Manchester Academic Health Science Centre, Department of Academic Surgery, University Hospital of South Manchester, Manchester, UK
| | - Henri H. Versteeg
- Department of Thrombosis and Hemostasis, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Cliona C. Kirwan
- The University of Manchester, Manchester Academic Health Science Centre, Department of Academic Surgery, University Hospital of South Manchester, Manchester, UK
| |
Collapse
|
69
|
Zhao X, Zhao J, Hu R, Yao Q, Zhang G, Shen H, Yagüe E, Hu Y. Ruanjian Sanjie decoction exhibits antitumor activity by inducing cell apoptosis in breast cancer. Oncol Lett 2017; 13:3071-3079. [PMID: 28529560 PMCID: PMC5431657 DOI: 10.3892/ol.2017.5832] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/06/2017] [Indexed: 12/28/2022] Open
Abstract
Traditional Chinese medicine, based on theories developed and practiced for >2,000 years, is one of the most common complementary and alternative types of medicine currently used in the treatment of patients with breast cancer. Ruanjian Sanjie (RJSJ) decoction, is composed of four herbs, including Ban xia (Pinellia ternata), Xia ku cao (Prunella vulgaris), Shan ci gu (Cremastra appendiculata) and Hai zao (Sargassum pallidum), and has traditionally been used for softening hard lumps and resolving hard tissue masses. However, the active compounds and mechanisms of action of RJSJ remain unknown. The present study demonstrated the antitumor activity of RJSJ against Ehrlich ascites carcinoma in Swiss albino mice and breast cancer xenografts in nude mice. Notably, RJSJ does not induce body weight loss, immune function toxicity or myelosuppression in mice, indicating that it is safe and well tolerated. In addition, RJSJ shows potent cytotoxicity against breast cancer cells in vitro by the suppression of the anti-apoptotic proteins B-cell lymphoma 2 and survivin, leading to the activation of caspase-3/7 and caspase-9, and the apoptotic cascade. These findings provide a clear rationale to explore the therapeutic strategy of using RJSJ alone or in combination with chemotherapeutic agents for breast cancer patients and the characterization of its active principles.
Collapse
Affiliation(s)
- Xiumei Zhao
- Centre for Research and Development of Anti Tumor Drugs, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin 300020, P.R. China
| | - Jing Zhao
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Renjie Hu
- Centre for Research and Development of Anti Tumor Drugs, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin 300020, P.R. China
| | - Qiang Yao
- Tianjin People's Hospital, Tianjin 300121, P.R. China
| | - Guixian Zhang
- Centre for Research and Development of Anti Tumor Drugs, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin 300020, P.R. China
| | - Hongsheng Shen
- Centre for Research and Development of Anti Tumor Drugs, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin 300020, P.R. China
| | - Ernesto Yagüe
- Cancer Research Center, Division of Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Yunhui Hu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| |
Collapse
|
70
|
Reversal effects of local anesthetics on P-glycoprotein-mediated cancer multidrug resistance. Anticancer Drugs 2017; 28:243-249. [DOI: 10.1097/cad.0000000000000455] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
71
|
Activating Akt1 mutations alter DNA double strand break repair and radiosensitivity. Sci Rep 2017; 7:42700. [PMID: 28209968 PMCID: PMC5314324 DOI: 10.1038/srep42700] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022] Open
Abstract
The survival kinase Akt has clinical relevance to radioresistance. However, its contributions to the DNA damage response, DNA double strand break (DSB) repair and apoptosis remain poorly defined and often contradictory. We used a genetic approach to explore the consequences of genetic alterations of Akt1 for the cellular radiation response. While two activation-associated mutants with prominent nuclear access, the phospho-mimicking Akt1-TDSD and the clinically relevant PH-domain mutation Akt1-E17K, accelerated DSB repair and improved survival of irradiated Tramp-C1 murine prostate cancer cells and Akt1-knockout murine embryonic fibroblasts in vitro, the classical constitutively active membrane-targeted myrAkt1 mutant had the opposite effects. Interestingly, DNA-PKcs directly phosphorylated Akt1 at S473 in an in vitro kinase assay but not vice-versa. Pharmacological inhibition of DNA-PKcs or Akt restored radiosensitivity in tumour cells expressing Akt1-E17K or Akt1-TDSD. In conclusion, Akt1-mediated radioresistance depends on its activation state and nuclear localization and is accessible to pharmacologic inhibition.
Collapse
|
72
|
Bashash D, Safaroghli-Azar A, Delshad M, Bayati S, Nooshinfar E, Ghaffari SH. Inhibitor of pan class-I PI3K induces differentially apoptotic pathways in acute leukemia cells: Shedding new light on NVP-BKM120 mechanism of action. Int J Biochem Cell Biol 2016; 79:308-317. [DOI: 10.1016/j.biocel.2016.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 09/02/2016] [Indexed: 10/21/2022]
|
73
|
miRNA-205 targets VEGFA and FGF2 and regulates resistance to chemotherapeutics in breast cancer. Cell Death Dis 2016; 7:e2291. [PMID: 27362808 PMCID: PMC5108343 DOI: 10.1038/cddis.2016.194] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/19/2016] [Accepted: 06/06/2016] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) have critical roles in regulating cancer cell survival, proliferation and sensitivity to chemotherapy. The potential application of using miRNAs to predict chemotherapeutic response to cancer treatment is highly promising. However, the underlying mechanisms of chemotherapy response control by miRNAs remain to be fully identified and their prognostic value has not been fully evaluated. Here we show a strong correlation between miR-205 expression and chemosensitivtiy to TAC (docetaxol, doxorubicin plus cyclophosphamide), a widely-used neoadjuvant chemotherapy (NAC) regimen, for breast cancer patients. High level of miR-205 predicted better response to TAC regimen NAC in breast cancer patients. We found miR-205 downregulated in both MCF-7/A02 and CALDOX cells, two drug-resistant derivatives of MCF-7 and Cal51 cells, and its ectopic expression led to an increase in apoptosis resensitization of both drug-resistant cell lines to doxorubicin and taxol. We further show that miR-205 directly binds VEGFA and FGF2 mRNA 3′-UTRs and confirm that miR-205 levels are negatively correlated with VEGFA and FGF2 mRNA expression in breast cancer patients. Adding VEGFA and FGF2 exogenously to chemosensitive breast cancer cells and chemoresistant cells with miR-205 overexpression led to drug resistance. Consistently, low VEGFA and FGF2 expression correlated with better response to NAC in breast cancer patients. In addition, inhibition of tumor growth and resensitization to doxorubicin were also observed in mouse tumor xenografts from cells overexpressing miR-205. Taken together, our data suggest that miR-205 enhances chemosensitivity of breast cancer cells to TAC chemotherapy by suppressing both VEGFA and FGF2, leading to evasion of apoptosis. MiR-205 may serve as a predictive biomarker and a potential therapeutic target in breast cancer treatment.
Collapse
|
74
|
Dirican E, Akkiprik M, Özer A. Mutation distributions and clinical correlations of PIK3CA gene mutations in breast cancer. Tumour Biol 2016; 37:7033-45. [PMID: 26921096 DOI: 10.1007/s13277-016-4924-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/28/2016] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BCa) is the most common cancer and the second cause of death among women. Phosphoinositide 3-kinase (PI3K) signaling pathway has a crucial role in the cellular processes such as cell survival, growth, division, and motility. Moreover, oncogenic mutations in the PI3K pathway generally involve the activation phosphatidylinositol-4,5-bisphosphate 3-kinase-catalytic subunit alpha (PIK3CA) mutation which has been identified in numerous BCa subtypes. In this review, correlations between PIK3CA mutations and their clinicopathological parameters on BCa will be described. It is reported that PIK3CA mutations which have been localized mostly on exon 9 and 20 hot spots are detected 25-40 % in BCa. This relatively high frequency can offer an advantage for choosing the best treatment options for BCa. PIK3CA mutations may be used as biomarkers and have been major focus of drug development in cancer with the first clinical trials of PI3K pathway inhibitors currently in progress. Screening of PIK3CA gene mutations might be useful genetic tests for targeted therapeutics or diagnosis. Increasing data about PIK3CA mutations and its clinical correlations with BCa will help to introduce new clinical applications in the near future.
Collapse
Affiliation(s)
- Ebubekir Dirican
- Department of Medical Biology, School of Medicine, Marmara University, Başıbüyük Mah., Maltepe Başıbüyük Yolu Sok., No: 9/1, 34854, Maltepe, Istanbul, Turkey
| | - Mustafa Akkiprik
- Department of Medical Biology, School of Medicine, Marmara University, Başıbüyük Mah., Maltepe Başıbüyük Yolu Sok., No: 9/1, 34854, Maltepe, Istanbul, Turkey.
| | - Ayşe Özer
- Department of Medical Biology, School of Medicine, Marmara University, Başıbüyük Mah., Maltepe Başıbüyük Yolu Sok., No: 9/1, 34854, Maltepe, Istanbul, Turkey
| |
Collapse
|
75
|
Podolski-Renić A, Milošević Z, Dinić J, Stanković T, Banković J, Pešić M. Mutual regulation and targeting of multidrug resistance and cancer stem phenotype. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00391e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Targeting stemness mechanisms leads to the suppression of ABC transporter activity and elimination of CSCs.
Collapse
Affiliation(s)
- Ana Podolski-Renić
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| | - Zorica Milošević
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| | - Jelena Dinić
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| | - Tijana Stanković
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| | - Jasna Banković
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| | - Milica Pešić
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| |
Collapse
|
76
|
Yousefnia S, Seyed Forootan F, Seyed Forootan S, Nasr Esfahani MH, Gure AO, Ghaedi K. Activated coagulation time in monitoring heparinized dogs. Am J Vet Res 1981; 10:452. [PMID: 32426267 PMCID: PMC7212408 DOI: 10.3389/fonc.2020.00452] [Citation(s) in RCA: 33] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/13/2020] [Indexed: 12/13/2022]
Abstract
Breast cancer stem cells (BCSCs) are the minor population of breast cancer (BC) cells that exhibit several phenotypes such as migration, invasion, self-renewal, and chemotherapy as well as radiotherapy resistance. Recently, BCSCs have been more considerable due to their capacity for recurrence of tumors after treatment. Recognition of signaling pathways and molecular mechanisms involved in stemness phenotypes of BCSCs could be effective for discovering novel treatment strategies to target BCSCs. This review introduces BCSC markers, their roles in stemness phenotypes, and the dysregulated signaling pathways involved in BCSCs such as mitogen-activated protein (MAP) kinase, PI3K/Akt/nuclear factor kappa B (NFκB), TGF-β, hedgehog (Hh), Notch, Wnt/β-catenin, and Hippo pathway. In addition, this review presents recently discovered molecular mechanisms implicated in chemotherapy and radiotherapy resistance, migration, metastasis, and angiogenesis of BCSCs. Finally, we reviewed the role of microRNAs (miRNAs) in BCSCs as well as several other therapeutic strategies such as herbal medicine, biological agents, anti-inflammatory drugs, monoclonal antibodies, nanoparticles, and microRNAs, which have been more considerable in the last decades.
Collapse
Affiliation(s)
- Saghar Yousefnia
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Farzad Seyed Forootan
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute of Biotechnology, ACECR, Isfahan, Iran
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
- *Correspondence: Farzad Seyed Forootan ;
| | - Shiva Seyed Forootan
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Mohammad Hossein Nasr Esfahani
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute of Biotechnology, ACECR, Isfahan, Iran
| | - Ali Osmay Gure
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
- Ali Osmay Gure
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute of Biotechnology, ACECR, Isfahan, Iran
- Kamran Ghaedi ; ;
| |
Collapse
|