1
|
Zeng X, Liu T, Tang S, Dong X, Li Y, Liao L, Chen S, Chen L, Kong J, Dai Z, Feng K, Wong YH, Xie Q. Exosomal miR-7-25207 Increases Subgroup J Avian Leukosis Virus Titers by Targeting the Akt-CyclinQ1 and PRC1-YAF2 Dual Pathways. Microorganisms 2024; 12:1495. [PMID: 39065263 PMCID: PMC11279298 DOI: 10.3390/microorganisms12071495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/05/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Subgroup J avian leukosis virus (ALV-J) is a major pathogen in poultry, causing substantial economic losses to the poultry industry worldwide. Exosomal small RNAs derived from virus-infected cells or biological fluids can serve as viral transmission vectors. However, the role and mechanism of exosomal miRNA in ALV-J infection are unclear. In this study, we demonstrated that exosomal microRNA-7-25207 (miR-7-25207) could increase the titers of ALV-J. Exosomes isolated from ALV-J-infected DF-1 cells (Exo-ALV-J) contained partial viral proteins from ALV-J and could transmit the infection to uninfected DF-1 cells, leading to productive infection. Additionally, the RNA expression profile of exosomes was altered following ALV-J infection. miRNA analysis revealed that the expression of exosomal miR-7-25207 increased. Overexpression of miR-7-25207 significantly increased the titers of ALV-J in transfected cells. Furthermore, miR-7-25207 directly suppressed the expression of Akt and PRC1. Akt, in turn, directly inhibited CyclinQ1 expression, while PRC1 directly interfered with YAF2 expression. In conclusion, ALV-J infection activates the expression of miR-7-25207, which is subsequently delivered via exosomes to uninfected cells, increasing ALV-J titers by targeting Akt-CyclinQ1 and PRC1-YAF2 dual pathways. These findings suggest that exosomal miR-7-25207 may serve as a potential biomarker for clinical parameters in ALV-J infection.
Collapse
Affiliation(s)
- Xiaona Zeng
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (T.L.); (Y.L.); (L.L.); (S.C.); (L.C.); (J.K.); (Z.D.); (K.F.)
- Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan 512005, China; (S.T.); (X.D.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Tongfei Liu
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (T.L.); (Y.L.); (L.L.); (S.C.); (L.C.); (J.K.); (Z.D.); (K.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Shengqiu Tang
- Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan 512005, China; (S.T.); (X.D.)
| | - Xiaoying Dong
- Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan 512005, China; (S.T.); (X.D.)
| | - Yajuan Li
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (T.L.); (Y.L.); (L.L.); (S.C.); (L.C.); (J.K.); (Z.D.); (K.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Liqin Liao
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (T.L.); (Y.L.); (L.L.); (S.C.); (L.C.); (J.K.); (Z.D.); (K.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Sheng Chen
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (T.L.); (Y.L.); (L.L.); (S.C.); (L.C.); (J.K.); (Z.D.); (K.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Liyi Chen
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (T.L.); (Y.L.); (L.L.); (S.C.); (L.C.); (J.K.); (Z.D.); (K.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Jie Kong
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (T.L.); (Y.L.); (L.L.); (S.C.); (L.C.); (J.K.); (Z.D.); (K.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Zhenkai Dai
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (T.L.); (Y.L.); (L.L.); (S.C.); (L.C.); (J.K.); (Z.D.); (K.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Keyu Feng
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (T.L.); (Y.L.); (L.L.); (S.C.); (L.C.); (J.K.); (Z.D.); (K.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Yung-Hou Wong
- Division of Life Sciences, Biotechnology Research Institute, Hong Kong University of Science and Technology, Hong Kong, China;
| | - Qingmei Xie
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (T.L.); (Y.L.); (L.L.); (S.C.); (L.C.); (J.K.); (Z.D.); (K.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| |
Collapse
|
2
|
Rajpurohit YS, Sharma DK, Lal M, Soni I. A perspective on tumor radiation resistance following high-LET radiation treatment. J Cancer Res Clin Oncol 2024; 150:226. [PMID: 38696003 PMCID: PMC11065934 DOI: 10.1007/s00432-024-05757-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 04/22/2024] [Indexed: 05/05/2024]
Abstract
High-linear energy transfer (LET) radiation is a promising alternative to conventional low-LET radiation for therapeutic gain against cancer owing to its ability to induce complex and clustered DNA lesions. However, the development of radiation resistance poses a significant barrier. The potential molecular mechanisms that could confer resistance development are translesion synthesis (TLS), replication gap suppression (RGS) mechanisms, autophagy, epithelial-mesenchymal transition (EMT) activation, release of exosomes, and epigenetic changes. This article will discuss various types of complex clustered DNA damage, their repair mechanisms, mutagenic potential, and the development of radiation resistance strategies. Furthermore, it highlights the importance of careful consideration and patient selection when employing high-LET radiotherapy in clinical settings.
Collapse
Affiliation(s)
- Yogendra Singh Rajpurohit
- Molecular Biology Division, Bhabha Atomic Research Centre, 2-46-S, Modular Lab, A-Block, Mumbai, 400085, India.
- Homi Bhabha National Institute, DAE- Deemed University, Mumbai, 400094, India.
| | - Dhirendra Kumar Sharma
- Molecular Biology Division, Bhabha Atomic Research Centre, 2-46-S, Modular Lab, A-Block, Mumbai, 400085, India
| | - Mitu Lal
- Molecular Biology Division, Bhabha Atomic Research Centre, 2-46-S, Modular Lab, A-Block, Mumbai, 400085, India
| | - Ishu Soni
- Homi Bhabha National Institute, DAE- Deemed University, Mumbai, 400094, India
| |
Collapse
|
3
|
Shimura T, Sunaga K, Yamazaki M, Honoka N, Sasatani M, Kamiya K, Ushiyama A. Nuclear DNA damage-triggered ATM-dependent AMPK activation regulates the mitochondrial radiation response. Int J Radiat Biol 2024; 100:584-594. [PMID: 38166485 DOI: 10.1080/09553002.2023.2295297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/11/2023] [Indexed: 01/04/2024]
Abstract
PURPOSE AMP-activated protein kinase (AMPK) acts as a cellular energy sensor and is essential for controlling mitochondrial homeostasis. Here, we investigated the regulatory mechanisms involved in AMPK activation to elucidate how networks of intracellular signaling pathways respond to stress conditions. MATERIALS AND METHODS Inhibitors of ATM, DNA-PK, and AKT were tested in normal TIG-3 and MRC-5 human fibroblasts to determine which upstream kinases are responsible for AMPK activation. SV40 transformed-human ATM-deficient fibroblasts (AT5BIVA) and their ATM-complemented cells (i.e., AT5BIVA/ATMwt) were also used. Protein expression associated with AMPK signaling was examined by immunostaining and/or Western blotting. RESULTS Radiation-induced nuclear DNA damage activates ATM-dependent AMPK signaling pathways that regulate mitochondrial quality control. In contrast, hypoxia and glucose starvation caused ATP depletion and activated AMPK via a pathway independent of ATM. DNA-PK and AKT are not involved in AMPK-mediated mitochondrial signaling pathways. CONCLUSION Activation of the AMPK signaling pathway differs depending on the stimulus. Radiation activates AMPK through two pathways: depletion of ATP-mediated LKB1 signaling and nuclear DNA damage-induced ATM signaling. Nuclear DNA damage signaling to mitochondria therefore plays a pivotal role in determining the cell fates of irradiated cells.
Collapse
Affiliation(s)
- Tsutomu Shimura
- Department of Environmental Health, National Institute of Public Health Wako, Saitama, Japan
| | - Kenta Sunaga
- Faculty of Pharmaceutical Sciences Student, Meiji Pharmaceutical University, Kiyose, Japan
| | - Mayu Yamazaki
- Faculty of Pharmaceutical Sciences Student, Meiji Pharmaceutical University, Kiyose, Japan
| | - Nara Honoka
- Faculty of Pharmaceutical Sciences Student, Meiji Pharmaceutical University, Kiyose, Japan
| | - Megumi Sasatani
- Department of Experimental Oncology; Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan
| | - Kenji Kamiya
- Department of Experimental Oncology; Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan
| | - Akira Ushiyama
- Department of Environmental Health, National Institute of Public Health Wako, Saitama, Japan
| |
Collapse
|
4
|
Kempska J, Oliveira-Ferrer L, Grottke A, Qi M, Alawi M, Meyer F, Borgmann K, Hamester F, Eylmann K, Rossberg M, Smit DJ, Jücker M, Laakmann E, Witzel I, Schmalfeldt B, Müller V, Legler K. Impact of AKT1 on cell invasion and radiosensitivity in a triple negative breast cancer cell line developing brain metastasis. Front Oncol 2023; 13:1129682. [PMID: 37483521 PMCID: PMC10358765 DOI: 10.3389/fonc.2023.1129682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/30/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction The PI3K/AKT pathway is activated in 43-70% of breast cancer (BC)-patients and promotes the metastatic potential of BC cells by increasing cell proliferation, invasion and radioresistance. Therefore, AKT1-inhibition in combination with radiotherapy might be an effective treatment option for triple-negative breast cancer (TNBC)-patients with brain metastases. Methods The impact of AKT1-knockout (AKT1_KO) and AKT-inhibition using Ipatasertib on MDA-MB-231 BR cells was assessed using in vitro cell proliferation and migration assays. AKT1-knockout in MDA-MB-231BR cells was performed using CRISPR/Cas9. The effect of AKT1-knockout on radiosensitivity of MDA-MB-231BR cell lines was determined via colony formation assays after cell irradiation. To detect genomic variants in AKT1_KO MDA-MB-231BR cells, whole-genome sequencing (WGS) was performed. Results Pharmacological inhibition of AKT with the pan-AKT inhibitor Ipatasertib led to a significant reduction of cell viability but did not impact cell migration. Moreover, only MDA-MB-231BR cells were sensitized following Ipatasertib-treatment. Furthermore, specific AKT1-knockout in MDA-MB-231BR showed reduced cell viability in comparison to control cells, with significant effect in one of two analyzed clones. Unexpectedly, AKT1 knockout led to increased cell migration and clonogenic potential in both AKT1_KO clones. RNAseq-analysis revealed the deregulation of CTSO, CYBB, GPR68, CEBPA, ID1, ID4, METTL15, PBX1 and PTGFRN leading to the increased cell migration, higher clonogenic survival and decreased radiosensitivity as a consequence of the AKT1 knockout in MDA-MB-231BR. Discussion Collectively, our results demonstrate that Ipatasertib leads to radiosensitization and reduced cell proliferation of MDA-MB-231BR. AKT1-inhibition showed altered gene expression profile leading to modified cell migration, clonogenic survival and radioresistance in MDA-MB-231BR. We conclude, that AKT1-inhibition in combination with radiotherapy contribute to novel treatment strategies for breast cancer brain metastases.
Collapse
Affiliation(s)
- Joanna Kempska
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Astrid Grottke
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Minyue Qi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Meyer
- Laboratory of Radiobiology & Experimental Radio Oncology, Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Borgmann
- Laboratory of Radiobiology & Experimental Radio Oncology, Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabienne Hamester
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kathrin Eylmann
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maila Rossberg
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel J. Smit
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Laakmann
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabell Witzel
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karen Legler
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
5
|
Li X, Lu J, Liu L, Li F, Xu T, Chen L, Yan Z, Li Y, Guo W. FOXK1 regulates malignant progression and radiosensitivity through direct transcriptional activation of CDC25A and CDK4 in esophageal squamous cell carcinoma. Sci Rep 2023; 13:7737. [PMID: 37173384 PMCID: PMC10182098 DOI: 10.1038/s41598-023-34979-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/10/2023] [Indexed: 05/15/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a serious malignancy with poor prognosis, necessitating identification of oncogenic mechanisms for novel therapeutic strategies. Recent studies have highlighted the significance of the transcription factor forkhead box K1 (FOXK1) in diverse biological processes and carcinogenesis of multiple malignancies, including ESCC. However, the molecular pathways underlying FOXK1's role in ESCC progression are not fully understood, and its potential role in radiosensitivity remains unclear. Here, we aimed to elucidate the function of FOXK1 in ESCC and explore the underlying mechanisms. Elevated FOXK1 expression levels were found in ESCC cells and tissues, positively correlated with TNM stage, invasion depth, and lymph node metastasis. FOXK1 markedly enhanced the proliferative, migratory and invasive capacities of ESCC cells. Furthermore, silencing FOXK1 resulted in heightened radiosensitivity by impeding DNA damage repair, inducing G1 arrest, and promoting apoptosis. Subsequent studies demonstrated that FOXK1 directly bound to the promoter regions of CDC25A and CDK4, thereby activating their transcription in ESCC cells. Moreover, the biological effects mediated by FOXK1 overexpression could be reversed by knockdown of either CDC25A or CDK4. Collectively, FOXK1, along with its downstream target genes CDC25A and CDK4, may serve as a promising set of therapeutic and radiosensitizing targets for ESCC.
Collapse
Affiliation(s)
- Xiaoxu Li
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, Hebei, China
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Juntao Lu
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, Hebei, China
| | - Lei Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fei Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tongxin Xu
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, Hebei, China
| | - Liying Chen
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, Hebei, China
| | - Zhaoyang Yan
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, Hebei, China
| | - Yan Li
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, Hebei, China
| | - Wei Guo
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, Hebei, China.
| |
Collapse
|
6
|
Ouellette MM, Zhou S, Yan Y. Cell Signaling Pathways That Promote Radioresistance of Cancer Cells. Diagnostics (Basel) 2022; 12:diagnostics12030656. [PMID: 35328212 PMCID: PMC8947583 DOI: 10.3390/diagnostics12030656] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/26/2022] [Accepted: 03/02/2022] [Indexed: 12/20/2022] Open
Abstract
Radiation therapy (RT) is a standard treatment for solid tumors and about 50% of patients with cancer, including pediatric cancer, receive RT. While RT has significantly improved the overall survival and quality of life of cancer patients, its efficacy has still been markedly limited by radioresistance in a significant number of cancer patients (intrinsic or acquired), resulting in failure of the RT control of the disease. Radiation eradicates cancer cells mainly by causing DNA damage. However, radiation also concomitantly activates multiple prosurvival signaling pathways, which include those mediated by ATM, ATR, AKT, ERK, and NF-κB that promote DNA damage checkpoint activation/DNA repair, autophagy induction, and/or inhibition of apoptosis. Furthermore, emerging data support the role of YAP signaling in promoting the intrinsic radioresistance of cancer cells, which occurs through its activation of the transcription of many essential genes that support cell survival, DNA repair, proliferation, and the stemness of cancer stem cells. Together, these signaling pathways protect cancer cells by reducing the magnitude of radiation-induced cytotoxicity and promoting radioresistance. Thus, targeting these prosurvival signaling pathways could potentially improve the radiosensitivity of cancer cells. In this review, we summarize the contribution of these pathways to the radioresistance of cancer cells.
Collapse
Affiliation(s)
- Michel M. Ouellette
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Sumin Zhou
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Ying Yan
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Correspondence:
| |
Collapse
|
7
|
Dong D, Fu Y, Chen F, Zhang J, Jia H, Li J, Wang H, Wen J. Hyperoxia sensitizes hypoxic HeLa cells to ionizing radiation by downregulating HIF‑1α and VEGF expression. Mol Med Rep 2021; 23:62. [PMID: 33215223 PMCID: PMC7706008 DOI: 10.3892/mmr.2020.11700] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/02/2020] [Indexed: 12/16/2022] Open
Abstract
The current study investigated whether hyperoxia may reverse hypoxia‑induced radioresistance (RR) in cervical cancer. Human HeLa cells exposed to hypoxic, normoxic or hyperoxic conditions were irradiated using X‑rays. Cell proliferation and apoptosis were analyzed using MTT assays and flow cytometry. The expression levels of hypoxia‑inducible factor‑1α (HIF‑1α), VEGF165, VEGFRs, Akt and ERK were measured via western blotting and/or ELISA. The results demonstrated that hypoxia stimulated HIF‑1α and VEGF expression, and induced RR in HeLa cells. The administration of recombinant VEGF or the forced expression of VEGF promoted RR, whereas inactivating HIF‑1α or blocking the VEGF‑VEGFR interaction abrogated hypoxia‑induced RR. Notably, hyperoxia decreased the level of hypoxia‑stimulated HIF‑1α and VEGF, and enhanced radiosensitivity in hypoxic HeLa cells. The results demonstrated that hyperoxia suppressed the hypoxia‑activated Akt and ERK signaling pathways in HeLa cells. Therefore, a high O2 concentration may be considered as a radiotherapeutic sensitizer for hypoxic HeLa cells.
Collapse
Affiliation(s)
- Dan Dong
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan Fu
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Feng Chen
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jing Zhang
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Haiyan Jia
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jia Li
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Huailin Wang
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jihong Wen
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
8
|
Kim KH, Kim HS, Kim SC, Kim D, Kim YB, Chung HC, Rha SY. Gene Expression Profiling Identifies Akt as a Target for Radiosensitization in Gastric Cancer Cells. Front Oncol 2020; 10:562284. [PMID: 33042843 PMCID: PMC7517358 DOI: 10.3389/fonc.2020.562284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/18/2020] [Indexed: 12/24/2022] Open
Abstract
Background Despite the important role of radiotherapy in cancer treatment, a subset of patients responds poorly to treatment majorly due to radioresistance. Particularly the role of radiotherapy has not been established in gastric cancer (GC). Herein, we aimed to identify a radiosensitivity gene signature and to discover relevant targets to enhance radiosensitivity in GC cells. Methods An oligonucleotide microarray (containing 22,740 probes) was performed in 12 GC cell lines prior to radiation. A clonogenic assay was performed to evaluate the survival fraction at 2 Gy (SF2) as a surrogate marker for radiosensitivity. Genes differentially expressed (fold change > 6, q-value < 0.025) were identified between radiosensitive and radioresistant cell lines, and quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) was performed for validation. Gene set and pathway analyses were performed using Ingenuity Pathway Analysis (IPA). Results Radiosensitive (SF2 < 0.4) and radioresistant cell lines (SF2 ≥ 0.6) exhibited a marked difference in gene expression. We identified 68 genes that are differentially expressed between radiosensitive and radioresistant cell lines. The identified genes showed interactions via AKT, HIF1A, TGFB1, and TP53, and their functions were associated with the genetic networks associated with cellular growth and proliferation, cellular movement, and cell cycle. The Akt signaling pathway exhibited the highest association with radiosensitivity. Combinatorial treatment with MK-2206, an allosteric Akt inhibitor, and radiotherapy significantly increased cell death compared with radiotherapy alone in two radioresistant cell lines (YCC-2 and YCC-16). Conclusion We identified a GC-specific radiosensitivity gene signature and suggest that the Akt signaling pathway could serve as a therapeutic target for GC radiosensitization.
Collapse
Affiliation(s)
- Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Han Sang Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Cheol Kim
- Division of Biomedical Informatics, Center for Genome Science, National Institute of Health, KCDC, Cheongju, South Korea
| | - DooA Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Yong Bae Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Cheol Chung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Sun Young Rha
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
9
|
Joseph N, Kirkby NF, Hoskin PJ, West CML, Choudhury A, Dale RG. Radiobiologically derived biphasic fractionation schemes to overcome the effects of tumour hypoxia. Br J Radiol 2020; 93:20190250. [PMID: 32462907 DOI: 10.1259/bjr.20190250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE As a fractionated course of radiotherapy proceeds tumour shrinkage leads to resolution of hypoxia and the initiation of accelerated proliferation of radioresistant cancer cells with better repair capacity. We hypothesise that, in tumours with significant hypoxia, improved tumour control could be achieved with biphasic fractionation schedules that either use acceleration after 3-4 weeks of conventional radiotherapy or deliver a higher proportional dose towards the end of a course of treatment. We conducted a modelling study based on the concept of biological effective dose (BED) comparing such novel regimens with conventional fractionation. METHODS The comparator conventional fractionation schedule 70 Gy in 35 fractions delivered over 7 weeks was tested against the following novel regimens, both of which were designed to be isoeffective in terms of late normal tissue toxicity.40 Gy in 20 fractions over 4 weeks followed by 22.32 Gy in 6 consecutive daily fractions (delayed acceleration)30.4 Gy in 27 fractions over 4 weeks followed by 40 Gy in 15 fractions over 3 weeks (temporal dose redistribution)The delayed acceleration regimen is exactly identical to that of the comparator schedule over the first 28 days and the BED gains with the novel schedule are achieved during the second phase of treatment when reoxygenation is complete. For the temporal redistribution regimen, it was assumed that the reoxygenation fraction progressively increases during the first 4 weeks of treatment and an iterative approach was used to calculate the final tumour BED for varying hypoxic fractions. RESULTS Novel fractionation with delayed acceleration or temporal fractionation results in tumour BED gains equivalent to 3.5-8 Gy when delivered in 2 Gy fractions. CONCLUSION In hypoxic tumours, novel fractionation strategies result in significantly higher tumour BED in comparison to conventional fractionation. ADVANCES IN KNOWLEDGE We demonstrate that novel biphasic fractionation regimens could overcome the effects of tumour hypoxia resulting in biological dose escalation.
Collapse
Affiliation(s)
- Nuradh Joseph
- Ministry of Health, Colombo, Sri Lanka.,Sri Lanka Cancer Research Group, Maharagama, Sri Lanka
| | - Norman F Kirkby
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Peter J Hoskin
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Cancer Centre, Mount Vernon Hospital, Northwood, Middlesex, UK
| | - Catharine M L West
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ananya Choudhury
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Roger G Dale
- Faculty of Medicine, Imperial College, London, UK
| |
Collapse
|
10
|
Assani G, Segbo J, Yu X, Yessoufou A, Xiong Y, Zhou F, Zhou Y. Downregulation of TMPRSS4 Enhances Triple-Negative Breast Cancer Cell Radiosensitivity Through Cell Cycle and Cell Apoptosis Process Impairment. Asian Pac J Cancer Prev 2019; 20:3679-3687. [PMID: 31870109 PMCID: PMC7173382 DOI: 10.31557/apjcp.2019.20.12.3679] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Indexed: 12/09/2022] Open
Abstract
Background: Radioresistance remains a challenge for cancer radiotherapy. The present study aims to investigate the role of TMPRSS4 in triple negative breast cancer (TNBC) cell radiosensitivity. Materials and Methods: After transfection of MDA-MD-468 triple negative breast cancer cells line by using the lentivirus vector, the effect of TMPRSS4 down-regulation on TNBC radiosensitivity was evaluated by using cloning assay and CCK-8 assay. The CCK-8 assay was also used for performing cell proliferation analysis. Western blot was carried out to detect the expression of certain proteins related to cell cycle pathways (cyclin D1), cell apoptosis pathways (Bax, Bcl2, and Caspase3), DNA damage and DNA damage repair (TRF2, Ku80 , ˠH2AX) . The cell cycle and cell apoptosis were also investigated using flow cytometer analysis. Results: TMPRSS4 expression was down-regulated in MDA-MB-468 cells which enhanced MDA-MB-468 cells radiosensitivity. TMPRSS4 silencing also improved IR induced cell proliferation ability reduction and promoted cell arrested at G2/M phase mediated by 6 Gy IR associated with cyclin D1 expression inhibition. Moreover, TMPRSS4 inhibition enhanced TNBC apoptosis induced by 6 Gy IR following by over-expression of (Bax, Caspase3) and down-regulation of Bcl2 as the pro-apoptotic and anti-apoptotic proteins, respectively. Otherwise, TMPRSS4 down-regulation increases DNA damage induced by 6 Gy IR and delays DNA damage repair respectively illustrated by downregulation of TRF2 and permanent increase of Ku80 and ˠH2AX expression at 1 h and 10 h post-IR. Conclusion: Down-regulation of TMPRSS4 increases triple negative breast cancer cell radiosensitivity and the use of TMPRSS4 inhibitor can be encouraged for improving radiotherapy effectiveness in TNBC radioresistant patients.
Collapse
Affiliation(s)
- Ganiou Assani
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors; Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China.,Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Julien Segbo
- University of Abomey Calavi, BP 526, Cotonou, Benin
| | - Xiaoyan Yu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors; Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China.,Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | | | - Yudi Xiong
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors; Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China.,Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Fuxiang Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors; Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China.,Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Yunfeng Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors; Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China.,Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
11
|
Rouhani M, Ramshini S, Omidi M. The Psychiatric Drug Lithium Increases DNA Damage and Decreases Cell Survival in MCF-7 and MDA-MB-231 Breast Cancer Cell Lines Expos ed to Ionizing Radiation. Curr Mol Pharmacol 2019; 12:301-310. [DOI: 10.2174/1874467212666190503151753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/25/2019] [Accepted: 04/04/2019] [Indexed: 01/24/2023]
Abstract
Background:
Breast cancer is the most common cancer among women. Radiation therapy
is used for treating almost every stage of breast cancer. A strategy to reduce irradiation side effects and
to decrease the recurrence of cancer is concurrent use of radiation and radiosensitizers. We studied the
effect of the antimanic drug lithium on radiosensitivity of estrogen-receptor (ER)-positive MCF-7 and
ER-negative, invasive, and radioresistant MDA-MB-231 breast cancer cell lines.
Methods:
MCF-7 and MDA-MB-231 breast cancer cell lines were treated with 30 mM and 20 mM
concentrations of lithium chloride (LiCl), respectively. These concentrations were determined by
MTT viability assay. Growth curves were depicted and comet assay was performed for control and
LiCl-treated cells after exposure to X-ray. Total and phosphorylated inactive levels of glycogen
synthase kinase-3beta (GSK-3β) protein were determined by ELISA assay for control and treated
cells.
Results:
Treatment with LiCl decreased cell proliferation after exposure to X-ray as indicated by
growth curves of MCF-7 and MDA-MB-231 cell lines within six days following radiation. Such
treatment increased the amount of DNA damages represented by percent DNA in Tails of comets at
0, 1, 4, and even 24 hours after radiation in both studied cell lines. The amount of active GSK-3β
was increased in LiCl-treated cells in ER-positive and ER-negative breast cancer cell lines.
Conclusion:
Treatment with LiCl that increased the active GSK-3β protein, increased DNA damages
and decreased survival independent of estrogen receptor status in breast cancer cells exposed to
ionizing radiation.
Collapse
Affiliation(s)
- Maryam Rouhani
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Samira Ramshini
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Maryam Omidi
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| |
Collapse
|
12
|
Ouellette MM, Yan Y. Radiation‐activated prosurvival signaling pathways in cancer cells. PRECISION RADIATION ONCOLOGY 2019. [DOI: 10.1002/pro6.1076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Michel M. Ouellette
- Department of Internal MedicineUniversity of Nebraska Medical Center Omaha Nebraska USA
| | - Ying Yan
- Department of Radiation OncologyUniversity of Nebraska Medical Center Omaha Nebraska USA
| |
Collapse
|
13
|
Wang M, Wang X, Li Y, Xiao Q, Cui XH, Xiao GD, Wang JC, Xu CW, Ren H, Liu D. Nutlin-3-Induced Sensitization of Non-Small Cell Lung Cancer Stem Cells to Axitinib-Induced Apoptosis Through Repression of Akt1/Wnt Signaling. Oncol Res 2019; 27:987-995. [PMID: 30832755 PMCID: PMC7848271 DOI: 10.3727/096504018x15424918479652] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to investigate the potential biological activities of nutlin-3 in the regulation of growth and proliferation of non-small cell lung cancer (NSCLC) stem cells (CSCs), which may help in sensitizing to axitinib-induced apoptosis. Nutlin-3 induction of p53 expression was used to test its role in controlling the cell division pattern and apoptosis of NSCLC cells. A549 cells and H460 cells were pretreated with nutlin-3 and then treated with either an Akt1 activator or shRNA-GSK3β, to investigate the potential role of p53 sensitization in the biological effects of axitinib. We also determined the expression levels of GSK3β and p-Akt1 in patients with NSCLC and determined their potential association with survival data using Kaplan-Meier plots and CBIOTAL. Increased p53 expression stimulated the induction of apoptosis by axitinib and promoted asymmetric cell division (ACD) of NSCLC CSCs. The repression of Akt phosphorylation induced by nutlin-3 promoted the ACD of lung CSCs, decreasing the proportion of the stem cell population. In addition to the induction of apoptosis by axitinib through inhibition of Wnt signaling, nutlin-3 treatment further enhanced axitinib-induced apoptosis by inhibiting Akt1/GSK3β/Wnt signaling. The low expression of GSK3β and increased expression of p-Akt in patients with NSCLC were closely associated with the development of NSCLC. TP53 stimulates the induction of apoptosis in NSCLC by axitinib and the ACD of lung CSCs through its regulatory effects on the p53/Akt/GSK3β pathways.
Collapse
Affiliation(s)
- Meng Wang
- Department of Thoracic Surgery and Oncology, the Second Department of Thoracic Surgery, Cancer Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Xin Wang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Yuan Li
- School of Humanities and Social Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Qiang Xiao
- Department of Medical Oncology, Cancer Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Xiao-Hai Cui
- Department of Thoracic Surgery and Oncology, the Second Department of Thoracic Surgery, Cancer Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Guo-Dong Xiao
- Department of Thoracic Surgery and Oncology, the Second Department of Thoracic Surgery, Cancer Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Ji-Chang Wang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Chong-Wen Xu
- Department of Otorhinolaryngology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Hong Ren
- Department of Thoracic Surgery and Oncology, the Second Department of Thoracic Surgery, Cancer Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Dapeng Liu
- Department of Thoracic Surgery and Oncology, the Second Department of Thoracic Surgery, Cancer Center, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| |
Collapse
|
14
|
Dong X, Luo Z, Liu T, Chai J, Ke Q, Shen L. Identification of Integrin β1 as a Novel PAG1-Interacting Protein Involved in the Inherent Radioresistance of Human Laryngeal Carcinoma. J Cancer 2018; 9:4128-4138. [PMID: 30519312 PMCID: PMC6277618 DOI: 10.7150/jca.26885] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/19/2018] [Indexed: 12/22/2022] Open
Abstract
Inherent radioresistance plays a crucial role in the failure of radiotherapy. Using the inherent radioresistant (Hep-2max) and radiosensitive (Hep-2min) cell lines established from the parental cell line Hep-2, we previously reported that phosphoprotein associated with glycosphingolipid-enriched microdomains 1(PAG1) overexpression in laryngeal carcinoma cells was correlated with inherent radioresistant phenotypes. However, the underlying mechanisms of this effect remain unknown. In the present study, we performed a proteomic screen to investigate the interactome of PAG1 in Hep-2max cells resulting in the identification of several interaction partners. Bioinformatic analysis and immunofluorescence experiments indicated the integrin β1 to be a crucial interaction partner of PAG1. PAG1 was also highly expressed in laryngeal carcinoma radioresistant tissues and showed co-localization with integrin β1. In addition, we demonstrated that integrin β1's binding to PAG1 could be interrupted by MβCD, an inhibitor of lipid rafts formation. Moreover, knockdown of integrin β1 by RNA interference sensitized radioresistant cells to irradiation. Importantly, we identified 2 potential interaction sites (Pro216-Arg232 and Asn356-Gly377) in the cytoplasmic domain of PAG1 using high throughput peptide arrays. Taken together, these results suggest that the binding of PAG1 to integrin β1 in lipid rafts is essential for inherent radioresistance of human laryngeal carcinoma.
Collapse
Affiliation(s)
- Xiaoxia Dong
- Department of pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Zhiguo Luo
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Tiantian Liu
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Jingjing Chai
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Qing Ke
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Li Shen
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
15
|
Plant-Derived Products for Treatment of Vascular Intima Hyperplasia Selectively Inhibit Vascular Smooth Muscle Cell Functions. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:3549312. [PMID: 30405738 PMCID: PMC6201497 DOI: 10.1155/2018/3549312] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/01/2018] [Accepted: 09/20/2018] [Indexed: 12/11/2022]
Abstract
Natural products are used widely for preventing intimal hyperplasia (IH), a common cardiovascular disease. Four different cells initiate and progress IH, namely, vascular smooth muscle, adventitial and endothelial cells, and circulation or bone marrow-derived cells. Vascular smooth muscle cells (VSMCs) play a critical role in initiation and development of intimal thickening and formation of neointimal hyperplasia. In this review, we describe the different originating cells involved in vascular IH and emphasize the effect of different natural products on inhibiting abnormal cellular functions, such as VSMC proliferation and migration. We further present a classification for the different natural products like phenols, flavonoids, terpenes, and alkaloids that suppress VSMC growth. Abnormal VSMC physiology involves disturbance in MAPKs, PI3K/AKT, JAK-STAT, FAK, and NF-κB signal pathways. Most of the natural isolate studies have revealed G1/S phase of cell cycle arrest, decreased ROS production, induced cell apoptosis, restrained migration, and downregulated collagen deposition. It is necessary to screen optimal drugs from natural sources that preferentially inhibit VSMC rather than vascular endothelial cell growth to prevent early IH, restenosis following graft implantation, and atherosclerotic diseases.
Collapse
|
16
|
Chan KKL, Siu MKY, Jiang YX, Wang JJ, Leung THY, Ngan HYS. Estrogen receptor modulators genistein, daidzein and ERB-041 inhibit cell migration, invasion, proliferation and sphere formation via modulation of FAK and PI3K/AKT signaling in ovarian cancer. Cancer Cell Int 2018; 18:65. [PMID: 29743815 PMCID: PMC5930957 DOI: 10.1186/s12935-018-0559-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 04/19/2018] [Indexed: 01/09/2023] Open
Abstract
Background Ovarian cancer is the most lethal gynaecological malignancy. Chemotherapy is the main stay of treatment for metastatic disease, with modest response rates but significant side effects. Therefore, there is a need for alternative therapies that can control the disease while offering good quality of life. Ovarian cancer cells express both estrogen receptor subtypes (ERα and ERβ). There is growing evidence that ERβ is anti-oncogenic. Genistein and daidzein are phytoestrogens found in soybeans and they display higher affinity to bind ERβ. ERB-041 is a potent selective ERβ agonist. In this study, we aimed to investigate the effects of genistein, daidzein and ERB-041 on ovarian cancer. Methods Ovarian cancer cell lines were treated with genistein, daidzein and ERB-041 in pharmacological doses. Cell migration, invasion, proliferation, cell cycle arrest, apoptosis and sphere formation were assessed by Transwell migration and invasion assays, XTT assay, focus formation, flow cytometry and sphere formation assay, respectively. Immunoblotting analysis was performed to determine the downstream signaling pathways. Results We found that genistein, daidzein and ERB-041 significantly inhibited ovarian cancer cell migration, invasion, proliferation, as well as induced cell cycle arrest and apoptosis. Significantly inhibitory effect on the size and number of sphere formed in genistein, daidzein and ERB-041 treated cells was also demonstrated. Moreover, genistein, daidzein and ERB-041 treatment reduced p-FAK, p-PI3K, p-AKT, p-GSK3β, p21 or cyclin D1 expression in ovarian cancer cells. Conclusion Genistein, daidzein and ERB-041 decreased ovarian cancer cell migration, invasion, proliferation and sphere formation, and induced cell cycle arrest and apoptosis with altered FAK and PI3K/AKT/GSK signaling and p21/cyclin D1 expression, suggesting their roles on ovarian cancer cell metastasis, tumorigenesis and stem-like properties and their potential as alternative therapies for ovarian cancer patients. Electronic supplementary material The online version of this article (10.1186/s12935-018-0559-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karen K L Chan
- Department of Obstetrics and Gynaecology, Queen Mary Hospital, University of Hong Kong, 6/F Professorial Block, Pokfulam, Hong Kong, SAR China
| | - Michelle K Y Siu
- Department of Obstetrics and Gynaecology, Queen Mary Hospital, University of Hong Kong, 6/F Professorial Block, Pokfulam, Hong Kong, SAR China
| | - Yu-Xin Jiang
- Department of Obstetrics and Gynaecology, Queen Mary Hospital, University of Hong Kong, 6/F Professorial Block, Pokfulam, Hong Kong, SAR China
| | - Jing-Jing Wang
- Department of Obstetrics and Gynaecology, Queen Mary Hospital, University of Hong Kong, 6/F Professorial Block, Pokfulam, Hong Kong, SAR China
| | - Thomas H Y Leung
- Department of Obstetrics and Gynaecology, Queen Mary Hospital, University of Hong Kong, 6/F Professorial Block, Pokfulam, Hong Kong, SAR China
| | - Hextan Y S Ngan
- Department of Obstetrics and Gynaecology, Queen Mary Hospital, University of Hong Kong, 6/F Professorial Block, Pokfulam, Hong Kong, SAR China
| |
Collapse
|
17
|
Clinically relevant radioresistant cell line: a simple model to understand cancer radioresistance. Med Mol Morphol 2017; 50:195-204. [PMID: 29067564 DOI: 10.1007/s00795-017-0171-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022]
Abstract
Radiotherapy (RT) is one of the major modalities for the treatment of human cancers and has been established as an excellent local treatment for malignant tumors. Conventional fractionated RT consists of 2-Gy X-rays, fractionated once a day, 5 days a week for 5-7 weeks in total 60 Gy. The efficacy of RT depends on the existence of radioresistant cells, which remains one of the most critical obstacles in RT and radio-chemotherapy. To improve the efficacy of RT, understanding the characteristics of radioresistant cells is one of the important subjects in radiation biology. Several studies have been reported to find out molecules implicated in radioresistance. However, it is noteworthy that cellular radioresistance has been mainly studied among cells with different genetic backgrounds and different origins. Therefore, making a system to compare between radioresistant and sensitive cells with the isogenic background is required. In this review, some aspects of cellular radioresistance mainly focusing on clinically relevant radioresistant (CRR) cell lines that can continue to proliferate even under exposure to 2-Gy X-rays, once a day, for more than 30 days, which is consistent with the conventional fractionated RT are discussed.
Collapse
|
18
|
Zang C, Liu X, Li B, He Y, Jing S, He Y, Wu W, Zhang B, Ma S, Dai W, Li S, Peng Z. IL-6/STAT3/TWIST inhibition reverses ionizing radiation-induced EMT and radioresistance in esophageal squamous carcinoma. Oncotarget 2017; 8:11228-11238. [PMID: 28061440 PMCID: PMC5355260 DOI: 10.18632/oncotarget.14495] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/27/2016] [Indexed: 01/06/2023] Open
Abstract
The acquisition of radioresistance by esophageal squamous carcinoma (ESC) cells during radiotherapy may lead to cancer recurrence and poor survival. Previous studies have demonstrated that ionizing radiation (IR) induces epithelial–mesenchymal transition (EMT) of ESC cells accompanied by increased migration, invasion, and radioresistance. However, the underlying molecular mechanisms of IR-induced EMT and radioresistance are not well established, hampering the development of potential solutions. To address this issue, we investigated the role of the IL-6/STAT3/TWIST signaling pathway in IR-induced EMT. We found not only the pathway was activated during IR-induced EMT but also STAT3 inhibition or Twist depletion reversed the EMT process and attenuated radioresistance. These results improve our understanding of the underlying mechanisms involved in IR-induced EMT and suggest potential interventions to prevent EMT-induced acquisition of radioresistance.
Collapse
Affiliation(s)
- Chunbao Zang
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Xujie Liu
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Bing Li
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chonqqing Medical University, Chongqing, China
| | - Yanqiong He
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Shen Jing
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Yujia He
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Wenli Wu
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Bingqian Zhang
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Shuhong Ma
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Weiwei Dai
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Shaolin Li
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| | - Zhiping Peng
- Department of Radiological Medicine, Chongqing Medical University, Chonging, China
| |
Collapse
|
19
|
Liu B, Wang C, Chen P, Wang L, Cheng Y. RACK1 promotes radiation resistance in esophageal cancer via regulating AKT pathway and Bcl-2 expression. Biochem Biophys Res Commun 2017; 491:622-628. [PMID: 28760343 DOI: 10.1016/j.bbrc.2017.07.153] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 07/27/2017] [Indexed: 01/18/2023]
Abstract
RACK1 is a seven Trp-Asp 40 repeat protein, which interacts with a wide range of kinases and proteins. RACK1 plays an important role in the proliferation and progression of various cancers. The aim of this study is to detect the role of RACK1 in the radioresistance in esophageal cancer. The results indicated that downregulation of RACK1 reduced the colony formation ability, proliferation ability and resistance of cells to radiation effection through regulating the radiation-related proteins including pAKT, Bcl-2 and Bim; whereas upregulation of RACK1 promoted the ability and radioresistance of ESCC cells. Our findings suggest that RACK1 promotes proliferation and radioresistance in ESCC cells by activating the AKT pathway, upregulating Bcl-2 expression and downregulating protein levels of Bim. Our study fills in gaps in the field of RACK1 and radiation resistance and may provide new possibilities for improving strategies of radiotherapy in esophageal cancer.
Collapse
Affiliation(s)
- Bowen Liu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China.
| | - Cong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China.
| | - Pengxiang Chen
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China.
| | - Lu Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China.
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
20
|
Chang R, He H, Mao G, Kong Z. Upregulating DAB2IP expression via EGR-1 inhibition, a new approach for overcoming fractionated-irradiation-induced cross-tolerance to ionizing radiation and mitomycin C in tumor cells. Int J Radiat Biol 2016; 93:386-393. [PMID: 27834104 DOI: 10.1080/09553002.2016.1257831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE To evaluate the effect of fractionated irradiation (FI) on tumor cells' sensitivity to ionizing radiation (IR) and antineoplastic drugs, and examine the potential of early growth response-1 (EGR-1) inhibition to sensitize tumor cells to IR. MATERIALS AND METHODS PC3 and HepG2 cells were subjected 10 times to γ-rays at 2 Gy. The surviving cells were named PC3/R and HepG2/R, respectively. The cells' sensitivity to irradiation and chemotherapeutic drugs, including cisplatin (PT), doxorubicin (DOX), mitomycin C (MMC) and 5-fluorouracil (5-FU), were identified by colony formation assay and MMT method, respectively. Quantitative real-time polymerase chain reaction (RT-qPCR) analysis was utilized to compare the difference of gene expression between radioresistant cells and parental cells. The small interfering RNA system was implemented to inhibit endogenous EGR-1 expression in radiation-resistant cells. Western blot was employed to identify the possible mechanism by which EGR-1 regulates cells' radiosensitivity. RESULTS FI induced cross-resistant to IR and MMC in tumor cells. Along with the reduction of ovarian cancer-2/disabled homolog 2 (DOC-2/DAB2) interactive protein (DAB2IP) expression, EGR-1 gene was upregulated in FI-treated cells. On the other hand, downregulation of EGR-1 gene expression sensitized radioresistant cells to IR accompanied by DAB2IP overexpression and STAT3 inactivation. In addition, NF-κB inhibitor, BAY11-7082 enhanced resistant cells' radiosensitivity and chemosensitivity. CONCLUSIONS Conventionally FI has a higher risk of forming acquired radioresistance (ARR) in vitro. EGR-1 gene-targeted drug design could be an effective strategy to overcome DAB2IP-dysregulation-induced ARR in tumor patients.
Collapse
Affiliation(s)
- Rulve Chang
- a The Institute of Radiation Medicine , Fudan University , Shanghai , China
| | - Hui He
- a The Institute of Radiation Medicine , Fudan University , Shanghai , China
| | - Guangmin Mao
- a The Institute of Radiation Medicine , Fudan University , Shanghai , China
| | - Zhaolu Kong
- a The Institute of Radiation Medicine , Fudan University , Shanghai , China
| |
Collapse
|
21
|
Shimura T. Targeting the AKT/cyclin D1 pathway to overcome intrinsic and acquired radioresistance of tumors for effective radiotherapy. Int J Radiat Biol 2016; 93:381-385. [PMID: 27910734 DOI: 10.1080/09553002.2016.1257832] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Radiotherapy (RT) is a powerful tool in the treatment of cancer, having the advantage of preserving normal tissues. Clinical outcomes of RT are significantly improved by technological advances, enabling increased radiation doses directed very specifically to a tumor. However, tumor radioresistance remains a major impediment to effective RT. We have shown that human tumor cells surviving after repeated exposure to fractionated radiation (FR) of X-rays for 1 month have acquired radioresistance through constitutive activation of AKT and downstream cyclin D1 nuclear retention. Tumor radioresistance is also proposed to be an intrinsic characteristic of cancer stem cells (CSC), whose efficient DNA repair is thought to confer this phenotype. We have isolated radioresistant CD133-positive cells following exposure to long-term FR. These cells exhibited the CSC phenotype with activation of the AKT/cyclin D1 pathway. In this review, I summarize our current understanding of the molecular mechanisms underlying tumor radioresistance and propose a strategy for overcoming radioresistance by targeting the AKT/cyclin D1 pathway. CONCLUSION Two different mechanisms: acquired radioresistance of surviving tumor cells after RT and intrinsic radioresistance of CSC are associated with tumor radioresistance. Inhibition of the AKT pathway results in radiosensitization of both types of tumor radioresistance.
Collapse
Affiliation(s)
- Tsutomu Shimura
- a Department of Environmental Health , National Institute of Public Health , Minami, Wako , Saitama , Japan
| |
Collapse
|
22
|
Hein AL, Post CM, Sheinin YM, Lakshmanan I, Natarajan A, Enke CA, Batra SK, Ouellette MM, Yan Y. RAC1 GTPase promotes the survival of breast cancer cells in response to hyper-fractionated radiation treatment. Oncogene 2016; 35:6319-6329. [PMID: 27181206 PMCID: PMC5112160 DOI: 10.1038/onc.2016.163] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 03/08/2016] [Accepted: 03/29/2016] [Indexed: 01/22/2023]
Abstract
Radiation therapy is a staple approach for cancer treatment, whereas radioresistance of cancer cells remains a substantial clinical problem. In response to ionizing radiation (IR) induced DNA-damage, cancer cells can sustain/activate pro-survival signaling pathways, leading to apoptotic resistance and induction of cell cycle checkpoint/DNA repair. Previous studies show that Rac1 GTPase is overexpressed/hyperactivated in breast cancer cells and is associated with poor prognosis. Studies from our laboratory reveal that Rac1 activity is necessary for G2/M checkpoint activation and cell survival in response to IR exposure of breast and pancreatic cancer cells. In the present study, we investigated the effect of Rac1 on the survival of breast cancer cells treated with hyper-fractionated radiation (HFR), which is used clinically for cancer treatment. Results in this report indicate that Rac1 protein expression is increased in the breast cancer cells that survived HFR compared to parental cells. Furthermore, this increase of Rac1 is associated with enhanced activities of ERK1/2 and NF-κB signaling pathways and increased levels of anti-apoptotic protein Bcl-xL and Mcl-1, which are downstream targets of ERK1/2 and NF-κB signaling pathways. Using Rac1 specific inhibitor and dominant negative mutant N17Rac1, here we demonstrate that Rac1 inhibition decreases the phosphorylation of ERK1/2 and IκBα, as well as the levels of Bcl-xL and Mcl-1 protein in the HFR-selected breast cancer cells. Moreover, inhibition of Rac1 using either small molecule inhibitor or dominant negative N17Rac1 abrogates clonogenic survival of HFR-selected breast cancer cells and decreases the level of intact PARP, which is indicative of apoptosis induction. Collectively, results in this report suggest that Rac1 signaling is essential for the survival of breast cancer cells subjected to HFR and implicate Rac1 in radioresistance of breast cancer cells. These studies also provide the basis to explore Rac1 as a therapeutic target for radioresistant breast cancer cells.
Collapse
Affiliation(s)
- A L Hein
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - C M Post
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Y M Sheinin
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - I Lakshmanan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - A Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - C A Enke
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - M M Ouellette
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Y Yan
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
23
|
Paradoxical roles of cyclin D1 in DNA stability. DNA Repair (Amst) 2016; 42:56-62. [PMID: 27155130 DOI: 10.1016/j.dnarep.2016.04.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/18/2016] [Accepted: 04/27/2016] [Indexed: 12/20/2022]
Abstract
Maintenance of DNA integrity is vital for all of the living organisms. Consequence of DNA damaging ranges from, introducing harmless synonymous mutations, to causing disease-associated mutations, genome instability, and cell death. A cell cycle protein cyclin D1 is an established cancer-driving protein. However, contribution of cyclin D1 to cancer formation and cancer survival is not entirely known. In cancer tissues, overexpression of cyclin D1 is associated with both cancer genome instability, and resistance to DNA-damaging cancer drugs. Emerging evidence indicated that cyclin D1 may play novel direct roles in regulating DNA repair. Here we provide an insight how cyclin D1 expression may contribute to DNA repair and chromosome instability, and how these functions may facilitate cancer formation, and drug resistance.
Collapse
|
24
|
Gray JE, Infante JR, Brail LH, Simon GR, Cooksey JF, Jones SF, Farrington DL, Yeo A, Jackson KA, Chow KH, Zamek-Gliszczynski MJ, Burris HA. A first-in-human phase I dose-escalation, pharmacokinetic, and pharmacodynamic evaluation of intravenous LY2090314, a glycogen synthase kinase 3 inhibitor, administered in combination with pemetrexed and carboplatin. Invest New Drugs 2015; 33:1187-96. [PMID: 26403509 DOI: 10.1007/s10637-015-0278-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 08/11/2015] [Indexed: 11/25/2022]
Abstract
PURPOSE LY2090314 (LY) is a glycogen synthase kinase 3 inhibitor with preclinical efficacy in xenograft models when combined with platinum regimens. A first-in-human phase 1 dose-escalation study evaluated the combination of LY with pemetrexed/carboplatin. PATIENTS AND METHODS Forty-one patients with advanced solid tumors received single-dose LY monotherapy lead-in and 37 patients received LY (10-120 mg) plus pemetrexed/carboplatin (500 mg/m(2) and 5-6 AUC, respectively) across 8 dose levels every 21 days. Primary objective was maximum tolerated dose (MTD) determination; secondary endpoints included safety, antitumor activity, pharmacokinetics, and beta-catenin pharmacodynamics. RESULTS MTD of LY with pemetrexed/carboplatin was 40 mg. Eleven dose-limiting toxicities (DLTs) occurred in ten patients. DLTs during LY monotherapy occurred at ≥ 40 mg: grade 2 visual disturbance (n = 1) and grade 3/4 peri-infusional thoracic pain during or shortly post infusion (n = 4; chest, upper abdominal, and back pain). Ranitidine was added after de-escalation to 80 mg LY to minimize peri-infusional thoracic pain. Following LY with pemetrexed/carboplatin therapy, DLTs included grade 3/4 thrombocytopenia (n = 4) and grade 4 neutropenia (n = 1). Best overall response by RECIST included 5 confirmed partial responses (non-small cell lung cancer [n = 3], mesothelioma, and breast cancer) and 19 patients having stable disease. Systemic LY exposure was approximately linear over dose range studied. Transient upregulation of beta-catenin measured in peripheral blood mononuclear cells (PBMCs) occurred at 40 mg LY. CONCLUSIONS The initial safety profile of LY2090314 was established. MTD LY dose with pemetrexed/carboplatin is 40 mg IV every 3 weeks plus ranitidine. Efficacy of LY plus pemetrexed/carboplatin requires confirmation in randomized trials.
Collapse
Affiliation(s)
- Jhanelle E Gray
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jeffrey R Infante
- Sarah Cannon Research Institute and Tennessee Oncology, 250 25th Avenue North, Suite 200, Nashville, TN, 37203, USA
| | - Les H Brail
- Eli Lilly and Company, Indianapolis, IN, USA
- Infinity Pharmaceuticals, Cambridge, MA, USA
| | | | | | - Suzanne F Jones
- Sarah Cannon Research Institute and Tennessee Oncology, 250 25th Avenue North, Suite 200, Nashville, TN, 37203, USA
| | | | - Adeline Yeo
- DOCS, San Diego, CA, USA
- Stat4ward LLC, Pittsburgh, PA, USA
| | | | - Kay H Chow
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Howard A Burris
- Sarah Cannon Research Institute and Tennessee Oncology, 250 25th Avenue North, Suite 200, Nashville, TN, 37203, USA.
| |
Collapse
|
25
|
Inhibition of RAC1 GTPase sensitizes pancreatic cancer cells to γ-irradiation. Oncotarget 2015; 5:10251-70. [PMID: 25344910 PMCID: PMC4279370 DOI: 10.18632/oncotarget.2500] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/16/2014] [Indexed: 12/31/2022] Open
Abstract
Radiation therapy is a staple treatment for pancreatic cancer. However, owing to the intrinsic radioresistance of pancreatic cancer cells, radiation therapy often fails to increase survival of pancreatic cancer patients. Radiation impedes cancer cells by inducing DNA damage, which can activate cell cycle checkpoints. Normal cells possess both a G1 and G2 checkpoint. However, cancer cells are often defective in G1 checkpoint due to mutations/alterations in key regulators of this checkpoint. Accordingly, our results show that normal pancreatic ductal cells respond to ionizing radiation (IR) with activation of both checkpoints whereas pancreatic cancer cells respond to IR with G2/M arrest only. Overexpression/hyperactivation of Rac1 GTPase is detected in the majority of pancreatic cancers. Rac1 plays important roles in survival and Ras-mediated transformation. Here, we show that Rac1 also plays a critical role in the response of pancreatic cancer cells to IR. Inhibition of Rac1 using specific inhibitor and dominant negative Rac1 mutant not only abrogates IR-induced G2 checkpoint activation, but also increases radiosensitivity of pancreatic cancer cells through induction of apoptosis. These results implicate Rac1 signaling in the survival of pancreatic cancer cells following IR, raising the possibility that this pathway contributes to the intrinsic radioresistance of pancreatic cancer.
Collapse
|
26
|
Miyasaka A, Oda K, Ikeda Y, Sone K, Fukuda T, Inaba K, Makii C, Enomoto A, Hosoya N, Tanikawa M, Uehara Y, Arimoto T, Kuramoto H, Wada-Hiraike O, Miyagawa K, Yano T, Kawana K, Osuga Y, Fujii T. PI3K/mTOR pathway inhibition overcomes radioresistance via suppression of the HIF1-α/VEGF pathway in endometrial cancer. Gynecol Oncol 2015; 138:174-80. [PMID: 25913131 DOI: 10.1016/j.ygyno.2015.04.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/10/2015] [Indexed: 12/11/2022]
Abstract
Radiation therapy is a key therapeutic strategy for endometrial carcinomas. However, biomarkers that predict radiosensitivity and drugs to enhance this sensitivity have not yet been established. We aimed to investigate the roles of TP53 and MAPK/PI3K pathways in endometrial carcinomas and to identify appropriate radiosensitizing therapeutics. D10 values (the irradiating dose required to reduce a cell population by 90%) were determined in eight endometrial cancer cell lines with known mutational statuses for TP53, PIK3CA, and KRAS. Cells were exposed to ionizing radiation (2-6Gy) and either a dual PI3K/mTOR inhibitor (NVP-BEZ235) or a MEK inhibitor (UO126), and their radiosensitizing effects were evaluated using clonogenic assays. The effects of silencing hypoxia-inducible factor-1 α (HIF-1α) expression with small interfering RNAs (siRNAs) were evaluated following exposure to ionizing radiation (2-3Gy). D10 values ranged from 2.0 to 3.1Gy in three cell lines expressing wild-type TP53 or from 3.3 to more than 6.0Gy in five cell lines expressing mutant TP53. NVP-BEZ235, but not UO126, significantly improved radiosensitivity through the suppression of HIF-1α/vascular endothelial growth factor-A expression. HIF-1α silencing significantly increased the induction of the sub-G1 population by ionizing radiation. Our study data suggest that TP53 mutation and PI3K pathway activation enhances radioresistance in endometrial carcinomas and that targeting the PI3K/mTOR or HIF-1α pathways could improve radiosensitivity.
Collapse
Affiliation(s)
- Aki Miyasaka
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Katsutoshi Oda
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Yuji Ikeda
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kenbun Sone
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tomohiko Fukuda
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kanako Inaba
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Chinami Makii
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Atsushi Enomoto
- Laboratory of Molecular Radiology, Centre for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noriko Hosoya
- Laboratory of Molecular Radiology, Centre for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Michihiro Tanikawa
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yuriko Uehara
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Takahide Arimoto
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | | | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kiyoshi Miyagawa
- Laboratory of Molecular Radiology, Centre for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsu Yano
- Department of Obstetrics and Gynaecology, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Kei Kawana
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
27
|
Improving chemoradiation efficacy by PI3-K/AKT inhibition. Cancer Treat Rev 2014; 40:1182-91. [DOI: 10.1016/j.ctrv.2014.09.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 09/23/2014] [Accepted: 09/25/2014] [Indexed: 12/28/2022]
|
28
|
HEIN ASHLEYL, OUELLETTE MICHELM, YAN YING. Radiation-induced signaling pathways that promote cancer cell survival (review). Int J Oncol 2014; 45:1813-9. [PMID: 25174607 PMCID: PMC4203326 DOI: 10.3892/ijo.2014.2614] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 08/01/2014] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy is a staple cancer treatment approach that has significantly improved local disease control and the overall survival of cancer patients. However, its efficacy is still limited by the development of radiation resistance and the presence of residual disease after therapy that leads to cancer recurrence. Radiation impedes cancer cell growth by inducing cytotoxicity, mainly caused by DNA damage. However, radiation can also simultaneously induce multiple pro-survival signaling pathways, such as those mediated by AKT, ERK and ATM/ATR, which can lead to suppression of apoptosis, induction of cell cycle arrest and/or initiation of DNA repair. These signaling pathways act conjointly to reduce the magnitude of radiation-induced cytotoxicity and promote the development of radioresistance in cancer cells. Thus, targeting these pro-survival pathways has great potential for the radiosensitization of cancer cells. In the present review, we summarize the current literature on how these radiation‑activated signaling pathways promote cancer cell survival.
Collapse
Affiliation(s)
- ASHLEY L. HEIN
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - MICHEL M. OUELLETTE
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - YING YAN
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
29
|
Wong P, Houghton P, Kirsch DG, Finkelstein SE, Monjazeb AM, Xu-Welliver M, Dicker AP, Ahmed M, Vikram B, Teicher BA, Coleman CN, Machtay M, Curran WJ, Wang D. Combining targeted agents with modern radiotherapy in soft tissue sarcomas. J Natl Cancer Inst 2014; 106:dju329. [PMID: 25326640 DOI: 10.1093/jnci/dju329] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Improved understanding of soft-tissue sarcoma (STS) biology has led to better distinction and subtyping of these diseases with the hope of exploiting the molecular characteristics of each subtype to develop appropriately targeted treatment regimens. In the care of patients with extremity STS, adjunctive radiation therapy (RT) is used to facilitate limb and function, preserving surgeries while maintaining five-year local control above 85%. In contrast, for STS originating from nonextremity anatomical sites, the rate of local recurrence is much higher (five-year local control is approximately 50%) and a major cause of death and morbidity in these patients. Incorporating novel technological advancements to administer accurate RT in combination with novel radiosensitizing agents could potentially improve local control and overall survival. RT efficacy in STS can be increased by modulating biological pathways such as angiogenesis, cell cycle regulation, cell survival signaling, and cancer-host immune interactions. Previous experiences, advancements, ongoing research, and current clinical trials combining RT with agents modulating one or more of the above pathways are reviewed. The standard clinical management of patients with STS with pretreatment biopsy, neoadjuvant treatment, and primary surgery provides an opportune disease model for interrogating translational hypotheses. The purpose of this review is to outline a strategic vision for clinical translation of preclinical findings and to identify appropriate targeted agents to combine with radiotherapy in the treatment of STS from different sites and/or different histology subtypes.
Collapse
Affiliation(s)
- Philip Wong
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Peter Houghton
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - David G Kirsch
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Steven E Finkelstein
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Arta M Monjazeb
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Meng Xu-Welliver
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Adam P Dicker
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Mansoor Ahmed
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Bhadrasain Vikram
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Beverly A Teicher
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - C Norman Coleman
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Mitchell Machtay
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Walter J Curran
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Dian Wang
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW).
| |
Collapse
|
30
|
Fukumoto M, Amanuma T, Kuwahara Y, Shimura T, Suzuki M, Mori S, Kumamoto H, Saito Y, Ohkubo Y, Duan Z, Sano K, Oguchi T, Kainuma K, Usami S, Kinoshita K, Lee I, Fukumoto M. Guanine nucleotide-binding protein 1 is one of the key molecules contributing to cancer cell radioresistance. Cancer Sci 2014; 105:1351-9. [PMID: 25098609 PMCID: PMC4462352 DOI: 10.1111/cas.12489] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 07/18/2014] [Accepted: 07/22/2014] [Indexed: 01/07/2023] Open
Abstract
Standard fractionated radiotherapy for the treatment of cancer consists of daily irradiation of 2-Gy X-rays, 5 days a week for 5-8 weeks. To understand the characteristics of radioresistant cancer cells and to develop more effective radiotherapy, we established a series of novel, clinically relevant radioresistant (CRR) cells that continue to proliferate with 2-Gy X-ray exposure every 24 h for more than 30 days in vitro. We studied three human and one murine cell line, and their CRR derivatives. Guanine nucleotide-binding protein 1 (GBP1) gene expression was higher in all CRR cells than their corresponding parental cells. GBP1 knockdown by siRNA cancelled radioresistance of CRR cells in vitro and in xenotransplanted tumor tissues in nude mice. The clinical relevance of GBP1 was immunohistochemically assessed in 45 cases of head and neck cancer tissues. Patients with GBP1-positive cancer tended to show poorer response to radiotherapy. We recently reported that low dose long-term fractionated radiation concentrates cancer stem cells (CSCs). Immunofluorescence staining of GBP1 was stronger in CRR cells than in corresponding parental cells. The frequency of Oct4-positive CSCs was higher in CRR cells than in parental cells, however, was not as common as GBP1-positive cells. GBP1-positive cells were radioresistant, but radioresistant cells were not necessarily CSCs. We concluded that GBP1 overexpression is necessary for the radioresistant phenotype in CRR cells, and that targeting GBP1-positive cancer cells is a more efficient method in conquering cancer than targeting CSCs.
Collapse
Affiliation(s)
- Motoi Fukumoto
- Department of Pathology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
AKT-mediated enhanced aerobic glycolysis causes acquired radioresistance by human tumor cells. Radiother Oncol 2014; 112:302-7. [PMID: 25150637 DOI: 10.1016/j.radonc.2014.07.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/07/2014] [Accepted: 07/29/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND PURPOSE Cellular radioresistance is a major impediment to effective radiotherapy. Here, we demonstrated that long-term exposure to fractionated radiation conferred acquired radioresistance to tumor cells due to AKT-mediated enhanced aerobic glycolysis. MATERIAL AND METHODS Two human tumor cell lines with acquired radioresistance were established by long-term exposure to fractionated radiation with 0.5 Gy of X-rays. Glucose uptake was inhibited using 2-deoxy-D-glucose, a non-metabolizable glucose analog. Aerobic glycolysis was assessed by measuring lactate concentrations. Cells were then used for assays of ROS generation, survival, and cell death as assessed by annexin V staining. RESULTS Enhanced aerobic glycolysis was shown by increased glucose transporter Glut1 expression and a high lactate production rate in acquired radioresistant cells compared with parental cells. Inhibiting the AKT pathway using the AKT inhibitor API-2 abrogated these phenomena. Moreover, we found that inhibiting glycolysis with 2-deoxy-D-glucose suppressed acquired tumor cell radioresistance. CONCLUSIONS Long-term fractionated radiation confers acquired radioresistance to tumor cells by AKT-mediated alterations in their glucose metabolic pathway. Thus, tumor cell metabolic pathway is an attractive target to eliminate radioresistant cells and improve radiotherapy efficacy.
Collapse
|
32
|
Chen TJ, Lee SW, Lin LC, Lin CY, Chang KY, Li CF. Cyclin-dependent kinase 4 overexpression is mostly independent of gene amplification and constitutes an independent prognosticator for nasopharyngeal carcinoma. Tumour Biol 2014; 35:7209-7216. [PMID: 24771220 DOI: 10.1007/s13277-014-1884-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 03/24/2014] [Indexed: 12/26/2022] Open
Abstract
Data mining in the public domain demonstrates that cyclin-dependent kinase 4 (CDK4) is highly expressed in nasopharyngeal carcinomas (NPC). Associated with cyclin-D, CDK4 phosphorylates and inactivates retinoblastoma (Rb) protein family members and mediates progression through the G1- to the S-phase of the cell cycle. Amplification and overexpression of CDK4 has been identified in various human malignancies. However, its expression and amplification has never been systemically evaluated in NPC. This study aimed to evaluate the amplification and expression status, correlation with clinicopathological features, and prognostic implications of CDK4 based on public domain dataset and in our well-defined cohort of NPC patients. The association between CDK4 transcript level and gene dosage was explored by analysis of an independent public domain dataset. We retrospectively assessed CDK4 immunoexpression in biopsies of 124 consecutive NPC patients devoid of initial distant metastasis and treated according to consistent guidelines. The results were correlated with clinicopathological features, local recurrence-free survival (LRFS), distant metastasis-free survival (DMeFS), and disease-specific survival (DSS). High levels of CDK4 protein were positively correlated with the T 3, 4 status (p = 0.024); N 2, 3 status (p < 0.001); and the American Joint Committee on Cancer stage 3, 4 (p < 0.001). Multivariate analysis suggested high CDK4 expression was an independent prognostic indicator of worse DMeFS (p = 0.001, hazard ratio (HR) = 3.226) and DSS (p = 0.037, HR = 1.838). Although CDK4 is frequently upregulated, its gene locus is very uncommonly amplified in NPC. CDK4 overexpression is mostly independent with gene amplification and represents a potential prognostic biomarker in NPC and may indicate tumor aggressiveness through cell cycle dysregulation.
Collapse
Affiliation(s)
- Tzu-Ju Chen
- Department of Pathology, Chi-Mei Foundation Medical Center, Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
33
|
Fukumoto M. Radiation pathology: From thorotrast to the future beyond radioresistance. Pathol Int 2014; 64:251-62. [DOI: 10.1111/pin.12170] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 04/13/2014] [Indexed: 12/30/2022]
Affiliation(s)
- Manabu Fukumoto
- Department of Pathology; Institute of Development, Aging and Cancer; Tohoku University; Sendai Japan
| |
Collapse
|
34
|
Shimura T, Hamada N, Sasatani M, Kamiya K, Kunugita N. Nuclear accumulation of cyclin D1 following long-term fractionated exposures to low-dose ionizing radiation in normal human diploid cells. Cell Cycle 2014; 13:1248-55. [PMID: 24583467 DOI: 10.4161/cc.28139] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Cyclin D1 is a mitogenic sensor that responds to growth signals from the extracellular environment and regulates the G 1-to-S cell cycle transition. When cells are acutely irradiated with a single dose of 10 Gy, cyclin D1 is degraded, causing cell cycle arrest at the G 1/S checkpoint. In contrast, cyclin D1 accumulates in human tumor cells that are exposed to long-term fractionated radiation (0.5 Gy/fraction of X-rays). In this study we investigated the effect of fractionated low-dose radiation exposure on cyclin D1 localization in 3 strains of normal human fibroblasts. To specifically examine the nuclear accumulation of cyclin D1, cells were treated with a hypotonic buffer containing detergent to remove cytoplasmic cyclin D1. Proliferating cell nuclear antigen (PCNA) immunofluorescence was used to identify cells in S phase. With this approach, we observed S-phase nuclear retention of cyclin D1 following low-dose fractionated exposures, and found that cyclin D1 nuclear retention increased with exposure time. Cells that retained nuclear cyclin D1 were more likely to have micronuclei than non-retaining cells, indicating that the accumulation of nuclear cyclin D1 was associated with genomic instability. Moreover, inhibition of the v-akt murine thymoma viral oncogene homolog (AKT) pathway facilitated cyclin D1 degradation and eliminated cyclin D1 nuclear retention in cells exposed to fractionated radiation. Thus, cyclin D1 may represent a useful marker for monitoring long-term effects associated with exposure to low levels of radiation.
Collapse
Affiliation(s)
- Tsutomu Shimura
- Department of Environmental Health; National Institute of Public Health; Wako, Saitama, Japan
| | - Nobuyuki Hamada
- Radiation Safety Research Center; Nuclear Technology Research Laboratory; Central Research Institute of Electric Power Industry (CRIEPI); Tokyo, Japan
| | - Megumi Sasatani
- Department of Experimental Oncology; Research Center for Radiation Genome Medicine; Research Institute for Radiation Biology and Medicine (RIRBM); Hiroshima University; Hiroshima, Japan
| | - Kenji Kamiya
- Department of Experimental Oncology; Research Center for Radiation Genome Medicine; Research Institute for Radiation Biology and Medicine (RIRBM); Hiroshima University; Hiroshima, Japan
| | - Naoki Kunugita
- Department of Environmental Health; National Institute of Public Health; Wako, Saitama, Japan
| |
Collapse
|
35
|
Qiao Q, Jiang Y, Li G. Inhibition of the PI3K/AKT-NF-κB pathway with curcumin enhanced radiation-induced apoptosis in human Burkitt's lymphoma. J Pharmacol Sci 2013; 121:247-56. [PMID: 23603894 DOI: 10.1254/jphs.12149fp] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The phosphatidylinositol-3-kinase (PI3K) / protein kinase B (AKT) signal transduction pathway is commonly misregulated in lymphoma and associated with tumorigenesis and enhanced resistance to radiotherapy. Curcumin has been shown to inhibit the PI3K/AKT signal transduction pathway in several tumor models. In this study, we found that curcumin inhibits constitutive and radiation-induced expression of the PI3K/AKT pathway and its downstream regulator nuclear factor kappaB (NF-κB) in human Burkitt's lymphoma, a high-grade non-Hodgkin's lymphoma (NHL). We further demonstrated that the blockage of radiation-induced activation of the PI3K/AKT pathway and its downstream regulator NF-κB by either curcumin or specific PI3/AKT inhibitors (LY294002 for PI3K or SH-5 for AKT) enhance apoptosis in three human Burkitt's lymphoma cell lines (Namalwa, Ramos, and Raji) that were treated with ionizing radiation. However, no synergic effect on radiation-induced apoptosis was found in the cells co-pretreated with curcumin combined with LY294002 or curcumin combined with SH-5. The results from this study suggest that curcumin might play an important role in radiotherapy of high-grade NHL through inhibition of the PI3K/AKT-dependent NF-κB pathway.
Collapse
Affiliation(s)
- Qiao Qiao
- Department of Radiotherapy, the First Hospital of China Medical University, China
| | | | | |
Collapse
|
36
|
Shimura T, Fukumoto M, Kunugita N. The role of cyclin D1 in response to long-term exposure to ionizing radiation. Cell Cycle 2013; 12:2738-43. [PMID: 23974042 DOI: 10.4161/cc.25746] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The health-related hazards resulting from long-term exposure to radiation remain unknown. Thus, an appropriate molecular marker is needed to clarify these effects. Cyclin D1 regulates the cell cycle transition from the G 1 phase to the S phase. Cyclin D1 is degraded as a G1/S checkpoint after 10 Gy of single acute radiation exposure, whereas conversely, cyclin D1 is stabilized when human tumor cells are exposed to fractionated radiation (FR) with 0.5 Gy of x-rays for 31 d. In this article, we review new findings regarding cyclin D1 overexpression in response to long-term exposure to FR. Cyclin D1 overexpression is associated with induction of genomic instability in irradiated cells. Therefore, repression of cyclin D1 expression is likely to cancel the harmful effects of long-term exposure to FR. Thus cyclin D1 may be a marker of long-term exposure to radiation and is a putative molecular radioprotection target for radiation safety.
Collapse
Affiliation(s)
- Tsutomu Shimura
- Department of Environmental Health; National Institute of Public Health 2-3-6 Minami; Wako, Saitama, Japan
| | | | | |
Collapse
|
37
|
Role of fucosyltransferase IV in epithelial-mesenchymal transition in breast cancer cells. Cell Death Dis 2013; 4:e735. [PMID: 23887626 PMCID: PMC3730415 DOI: 10.1038/cddis.2013.241] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/13/2013] [Accepted: 05/30/2013] [Indexed: 11/09/2022]
Abstract
Epithelial–mesenchymal transition (EMT) is a crucial step in tumor progression and has an important role during cancer invasion and metastasis. Although fucosyltransferase IV (FUT4) has been implicated in the modulation of cell migration, invasion and cancer metastasis, its role during EMT is unclear. This study explores the molecular mechanisms of the involvement of FUT4 in EMT in breast cancer cells. Breast cancer cell lines display increased expression of FUT4, which is accompanied by enhanced appearance of the mesenchymal phenotype and which can be reversed by knockdown of endogenous FUT4. Moreover, FUT4 induced activation of phosphatidylinositol 3-kinase (PI3K)/Akt, and inactivation of GSK3β and nuclear translocation of NF-κB, resulting in increased Snail and MMP-9 expression and greater cell motility. Taken together, these findings indicate that FUT4 has a role in EMT through activation of the PI3K/Akt and NF-κB signaling systems, which induce the key mediators Snail and MMP-9 and facilitate the acquisition of a mesenchymal phenotype. Our findings support the possibility that FUT4 is a novel regulator of EMT in breast cancer cells and a promising target for cancer therapy.
Collapse
|
38
|
Deng X, Ma L, Wu M, Zhang G, Jin C, Guo Y, Liu R. miR-124 radiosensitizes human glioma cells by targeting CDK4. J Neurooncol 2013; 114:263-74. [PMID: 23761023 DOI: 10.1007/s11060-013-1179-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 06/02/2013] [Indexed: 12/15/2022]
Abstract
The aberrant expression of cyclin-dependent kinase-4 (CDK4) has previously been observed in human brain glioma. Furthermore, it is observed that up-regulation of CDK4 is associated with therapy resistance and relapse. However, the mechanisms behind these phenomena remain unclear. Here, we demonstrated that elevated CDK4 expression is correlated with poor prognosis in glioma after radiotherapy and that CDK4 knockdown conferred radiosensitivity in glioma cell lines. CDK4 was identified as potential downstream target of miR-124 through bioinformatics analysis and dual-firefly luciferase reporter assay. Furthermore, restoration of miR-124 could confer radiosensitivity. Cell differentiation agent-2 (CDA-2) mimicked the effect of miR-124 restoration and CDK4 knockdown, and sensitized xenografts to radiation in an animal model. Our findings demonstrated for the first time that CDK4 was a downstream target of miR-124 and that CDA-2 could radiosensitize Glioblastoma multiforme cells through the MiR-124-CDK4 axis.
Collapse
Affiliation(s)
- Xubin Deng
- Cancer Institute of Southern Medical University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
39
|
Jirawatnotai S, Hu Y, Livingston DM, Sicinski P. Proteomic identification of a direct role for cyclin d1 in DNA damage repair. Cancer Res 2012; 72:4289-93. [PMID: 22915759 DOI: 10.1158/0008-5472.can-11-3549] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The human CCND1 gene, which encodes the cell-cycle protein cyclin D1, is one of the most frequently amplified genes in human cancers. Cyclin D1 activates the cyclin-dependent kinases CDK4 and CDK6 and drives cell proliferation. Beyond the cell-cycle role, the full repertoire of cyclin D1 functions in cancer cells is still unclear. Emerging evidence indicates that cyclin D1 may play a role in DNA damage response. In this review, we discuss observations linking cyclin D1 to DNA damage repair and summarize our recent findings, which show a cyclin D1 function in homologous recombination-mediated DNA repair.
Collapse
Affiliation(s)
- Siwanon Jirawatnotai
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
40
|
Activation of the AKT/cyclin D1/Cdk4 survival signaling pathway in radioresistant cancer stem cells. Oncogenesis 2012; 1:e12. [PMID: 23552696 PMCID: PMC3412645 DOI: 10.1038/oncsis.2012.12] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Radioresistance, which is a major cause of failure of radiotherapy (RT), is proposed as one of the intrinsic characteristics of cancer stem cells (CSCs) whose unique DNA damage response (DDR), efficient DNA repair and resistance to apoptosis are thought to confer the phenotype. We have isolated surviving CSCs by exposure to long-term fractionated radiation for 82 days from HepG2 and A172 cells (82FR-31NR cells). 82FR-31NR cells exhibited CSC properties, such as high expression of CSC marker CD133 and the ABC transporters (MDR1 and BCRP1), and high tumorigenic potential after transplantation into nude mice. The advantage of our isolated CSCs is that they can proliferate in as the same growth medium as that of parental cells without loss of CSC properties. Therefore, we can analyze DDR of non-stem cells and CSCs without any influences caused by different culture conditions. 82FR-31NR cells showed efficient DNA repair of radiation-induced DNA damage and radioresistance with activation of the AKT/cyclin D1 survival signaling pathway. In contrast, DNA damage persisted for a long time after irradiation in parental cells compared with isolated CSCs. Persisted DNA damage induced apoptosis in parental cells without activation of the AKT/cyclin D1 pathway. Therefore, inhibition of the AKT/cyclin D1 pathway by an AKT inhibitor, API-2, or cyclin D1 siRNA resulted in a loss of efficient DNA repair and radiosensitization of 82FR-31NR cells. Furthermore, knockdown of Cdk4 by its siRNA or a Cdk4 inhibitor was sufficient to suppress radioresistance of CSCs. In this study, we present a newly discovered DDR regarding the AKT/cyclin D1/Cdk4 pathway in response to radiation in CSCs. Combination of fractionated RT and reagents targeting the AKT/cyclin D1/Cdk4 pathway to eradicate CSCs would be effective therapeutic modality.
Collapse
|
41
|
Williams TM, Flecha AR, Keller P, Ram A, Karnak D, Galbán S, Galbán CJ, Ross BD, Lawrence TS, Rehemtulla A, Sebolt-Leopold J. Cotargeting MAPK and PI3K signaling with concurrent radiotherapy as a strategy for the treatment of pancreatic cancer. Mol Cancer Ther 2012; 11:1193-202. [PMID: 22411900 DOI: 10.1158/1535-7163.mct-12-0098] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There is an urgent need for the development of novel therapies to treat pancreatic cancer, which is among the most lethal of all cancers. KRAS-activating mutations, which are found in more than 90% of pancreatic adenocarcinomas, drive tumor dependency on the Ras/MAPK and Akt signaling pathways. Radiation is currently being explored as a component of the standard treatment regimen for pancreatic cancer. This study's purpose was to test the hypothesis that MAP kinase kinase (MEK or MAP2K) inhibitors will offer clear therapeutic benefit when integrated into radiotherapy treatment regimens for treatment of this disease. We explored the activation of the mitogen-activated protein kinase (MAPK) and Akt pathways in response to radiation in multiple pancreatic tumor cell lines. Small molecule inhibitors of MEK (PD0325901) and Akt (API-2) were subsequently evaluated for their radiosensitizing potential alone and in combination. In vivo efficacy was tested in subcutaneous MIA-PaCa2 xenografts. Phosphorylated levels of extracellular signal-regulated kinase (ERK)-1/2 and Akt were found to increase in response to radiation treatment in our pancreatic tumor cell line panel. MEK inhibitor-induced radiosensitization was observed in vitro and in vivo. The further addition of an Akt inhibitor to the MEK inhibitor/radiation regimen resulted in enhanced therapeutic gain as determined by increased radiosensitization and tumor cell death. In conclusion, MEK inhibition results in growth arrest, apoptosis, and radiosensitization of multiple preclinical pancreatic tumor models, and the effects can be enhanced by combination with an Akt inhibitor. These results provide rationale for further testing of a treatment regimen in pancreatic cancer that combines MEK inhibition with radiation, optimally in conjunction with Akt inhibition.
Collapse
Affiliation(s)
- Terence M Williams
- Department of Radiation Oncology, University of Michigan Medical Center, Ann Arbor, Michigan, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Elumalai P, Gunadharini DN, Senthilkumar K, Banudevi S, Arunkumar R, Benson CS, Sharmila G, Arunakaran J. Ethanolic neem (Azadirachta indica A. Juss) leaf extract induces apoptosis and inhibits the IGF signaling pathway in breast cancer cell lines. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.bionut.2011.12.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
43
|
Shimura T. Acquired radioresistance of cancer and the AKT/GSK3β/cyclin D1 overexpression cycle. JOURNAL OF RADIATION RESEARCH 2011; 52:539-544. [PMID: 21881296 DOI: 10.1269/jrr.11098] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Fractionated radiotherapy (RT) is widely used in cancer therapy for its advantages in the preservation of normal tissues. However, repopulation of surviving tumor cells during fractionated RT limits the efficacy of RT. In fact, repopulating tumors often acquire radioresistance and this is the major cause of failure of RT. We have recently demonstrated that human tumor cells acquire radioresistance when exposed to fractionated radiation (FR) of X-rays every 12 hours for 1 month. The acquired radioresistance was associated with overexpression of cyclin D1, a result of a series of molecular changes; constitutive activation of DNA-PK and AKT with concomitant down-regulation of glycogen synthase kinase-3β (GSK3β) which results in suppression of cyclin D1 proteolysis. Aberrant cyclin D1 overexpression in S-phase induced DNA double strand breaks which activated DNA-PK and established the vicious cycle of cycling D1 overexpression. This overexpression of cyclin D1 is responsible for the radioresistance phenotype of long-term FR cells, since this phenotype was completely abrogated by treatment of FR cells by the API-2, an AKT inhibitor or by a Cdk4 inhibitor. Thus, targeting the AKT/GSK3β/cyclin D1/Cdk4 pathway can be an efficient modality to suppress acquired radioresistance of tumor cells. In this article, I overview the newly discovered molecular mechanisms underlying acquired radioresistance of tumor cells induced by FR, and propose a strategy for eradication of tumors using fractionated RT by overcoming tumor radioresistance.
Collapse
Affiliation(s)
- Tsutomu Shimura
- Department of Pathology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
| |
Collapse
|
44
|
Gupta K, Thakur VS, Bhaskaran N, Nawab A, Babcook MA, Jackson MW, Gupta S. Response of tertiary centres to pressure changes. Is there a mechano-electrical association? Cardiovasc Res 1990; 7:e52572. [PMID: 23285096 PMCID: PMC3527608 DOI: 10.1371/journal.pone.0052572] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 11/19/2012] [Indexed: 12/23/2022] Open
Abstract
Inactivation of the tumor suppressor gene p53 is commonly observed in human prostate cancer and is associated with therapeutic resistance. We have previously demonstrated that green tea polyphenols (GTP) induce apoptosis in prostate cancer cells irrespective of p53 status. However, the molecular mechanisms underlying these observations remain elusive. Here we investigated the mechanisms of GTP-induced apoptosis in human prostate cancer LNCaP cells stably-transfected with short hairpin-RNA against p53 (LNCaPshp53) and control vector (LNCaPshV). GTP treatment induced p53 stabilization and activation of downstream targets p21/waf1 and Bax in a dose-dependent manner specifically in LNCaPshV cells. However, GTP-induced FAS upregulation through activation of c-jun-N-terminal kinase resulted in FADD phosphorylation, caspase-8 activation and truncation of BID, leading to apoptosis in both LNCaPshV and LNCaPshp53 cells. In parallel, treatment of cells with GTP resulted in inhibition of survival pathway, mediated by Akt deactivation and loss of BAD phosphorylation more prominently in LNCaPshp53 cells. These distinct routes of cell death converged to a common pathway, leading to loss of mitochondrial transmembrane potential, cytochrome c release and activation of terminal caspases, resulting in PARP-cleavage. GTP-induced apoptosis was attenuated with JNK inhibitor, SP600125 in both cell lines; whereas PI3K-Akt inhibitor, LY294002 resulted in increased cell death prominently in LNCaPshp53 cells, establishing the role of two distinct pathways of GTP-mediated apoptosis. Furthermore, GTP exposure resulted in inhibition of class I HDAC protein, accumulation of acetylated histone-H3 in total cellular chromatin, resulting in increased accessibility of transcription factors to bind with the promoter sequences of p21/waf1 and Bax, regardless of the p53 status of cells, consistent with effects elicited by an HDAC inhibitor, trichostatin A. These results demonstrate that GTP induces prostate cancer cell death by two distinct mechanisms regardless of p53 status, thus identifying specific well-defined molecular mechanisms that may be targeted by chemopreventive and/or therapeutic strategies.
Collapse
Affiliation(s)
- Karishma Gupta
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Vijay S. Thakur
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, United States of America
- University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - Natarajan Bhaskaran
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, United States of America
- University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - Akbar Nawab
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, United States of America
- University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - Melissa A. Babcook
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, United States of America
- University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - Mark W. Jackson
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, United States of America
- University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|