1
|
Owaki R, Hosoya K, Deguchi T, Konnai S, Maekawa N, Okagawa T, Yasui H, Kim S, Sunaga T, Okumura M. Enhancement of radio-sensitivity by inhibition of Janus kinase signaling with oclacitinib in canine tumor cell lines. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200946. [PMID: 40051504 PMCID: PMC11883359 DOI: 10.1016/j.omton.2025.200946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/14/2025] [Accepted: 01/30/2025] [Indexed: 03/09/2025]
Abstract
A combination of irradiation and oclacitinib, a Janus kinase (JAK) inhibitor used in dogs, could lead to synergistic anticancer effects in canine tumors. However, the anti-tumor effects of oclacitinib remain unclear. This study investigated the radio-sensitizing effect of oclacitinib in canine tumors and determined its underlying mechanisms using osteosarcoma (HMPOS), malignant melanoma (CMeC), and thyroid adenocarcinoma (CTAC) cell lines. A clonogenic assay and a tumor growth assessment in a xenograft mouse model (BALB/cAJcl-nu/nu) were performed to evaluate the radio-sensitizing effects of oclacitinib. Oclacitinib enhanced the radio-sensitivity of tumor cells both in vitro and in vivo. The signal transducer and activator of transcription (STAT)3 expression was activated and suppressed by oclacitinib in X-irradiation-exposed cells. Oclacitinib enhanced radiation-induced apoptosis only in HMPOS cells by inhibiting anti-apoptotic genes. In addition, oclacitinib inhibited the transcription of cell-cycle-regulating genes and arrested cell cycle progression from the G1 phase to subsequent phases. In conclusion, oclacitinib enhanced radio-sensitivity both in vitro and in vivo by triggering apoptosis and impeding cell cycle progression via STAT3 inhibition in canine tumor cell lines. This study suggested the clinical therapeutic potential of oclacitinib and radiation therapy in enhancing treatment efficacy and outcomes in canine tumors.
Collapse
Affiliation(s)
- Ryo Owaki
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kenji Hosoya
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuya Deguchi
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Satoru Konnai
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Naoya Maekawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Tomohiro Okagawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Department of Applied Veterinary Science, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Sangho Kim
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takafumi Sunaga
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Masahiro Okumura
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
2
|
Kwon YS, Nguyen PA, Dao HY, Jang H, Kim S. Advantages of single high-dose radiation therapy compared with conventional fractionated radiation therapy in overcoming radioresistance. Int J Radiat Biol 2024:1-12. [PMID: 39446049 DOI: 10.1080/09553002.2024.2418493] [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: 01/10/2024] [Revised: 09/25/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Radioresistance is a major clinical challenge in cancer treatment, as it reduces the effectiveness of radiation therapy (RT). While advances in radiation delivery have enabled the clinical use of high-dose hypofractionated RT, its impact on radioresistant tumors remains unclear. This study aimed to compare the effects of single high-dose RT with conventional fractionated RT on radioresistant breast cancer cells and explore the underlying mechanisms. METHODS Radioresistant cell lines were previously established by exposing SK-BR-3 and MCF-7 cells to 48 Gy and 70 Gy of radiation, respectively, in multiple fractions. We compared the effects of 2 Gy × 5 and 7 Gy × 1 fractions on these cells using clonogenic survival assays and western blot analysis. In vivo antitumor effects were assessed in SR tumor-bearing BALB/c mice irradiated with either 2 Gy × 5 or 7 Gy × 1 fractions. RESULTS 7 Gy x1 was more efficient at killing radioresistant breast cancer cells than 2 Gy x5. Furthermore, the 7 Gy x1 fraction produced higher levels of reactive oxygen species (ROS) and decreased the expression of radioresistance factors such as p-STAT3, ACSL4, FOXM1, RAD51, Bcl-xL, and survivin. Consistent with the in vitro studies, the 7 Gy × 1 fraction also showed superior antitumor effects in SR tumor-bearing BALB/c mice. CONCLUSIONS Single high-dose RT offers superior advantages over conventional fractionated RT in regard to overcoming radioresistance, supporting its potential as a promising treatment for recurrent tumors.
Collapse
Affiliation(s)
- Yun-Suk Kwon
- Research Institute of Climate Change and Agriculture, National Institute of Horticultural and Herbal Science, Jeju, Jeju-do, Republic of Korea
- Department of Pharmacology, College of Medicine, Dongguk University, Gyeongju, Gyeongsangbuk-do, Republic of Korea
| | - Phuong Anh Nguyen
- Department of Pharmacology, College of Medicine, Dongguk University, Gyeongju, Gyeongsangbuk-do, Republic of Korea
| | - Hai Yen Dao
- Department of Pharmacology, College of Medicine, Dongguk University, Gyeongju, Gyeongsangbuk-do, Republic of Korea
| | - Hyunsoo Jang
- Department of Radiation Oncology, Pohang St. Mary's Hospital, Pohang, Gyeongsangbuk-do, Republic of Korea
| | - Soyoung Kim
- Department of Pharmacology, College of Medicine, Dongguk University, Gyeongju, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
3
|
Yang X, Liu Z, Wang X, Tian W, Zhao T, Yang Q, Li W, Yang L, Yang H, Jia Y. Anti-cancer effects of nitazoxanide in epithelial ovarian cancer in-vitro and in-vivo. Chem Biol Interact 2024; 400:111176. [PMID: 39084502 DOI: 10.1016/j.cbi.2024.111176] [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: 06/06/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 08/02/2024]
Abstract
Epithelial ovarian cancer is one of the most lethal gynecologic malignancies and poses a considerable threat to women's health. Although the progression-free survival of patients has been prolonged with the application of anti-angiogenesis drugs and Poly (ADP-ribose) polymerases (PARP) inhibitors, overall survival has not substantially improved. Thus, new therapeutic strategies are essential for the treatment of ovarian cancer. Nitazoxanide (NTZ), an FDA-approved anti-parasitic drug, has garnered attention for its potential anti-cancer activity. However, the anti-tumor effects and possible underlying mechanisms of NTZ on ovarian cancer remain unclear. In this study, we investigated the anti-tumor effects and the mechanism of NTZ on ovarian cancer in vitro and in vivo. We found that NTZ inhibited the proliferation of A2780 and SKOV3 epithelial ovarian cancer cells in a time- and concentration-dependent manner; Furthermore, NTZ suppressed the metastasis and invasion of A2780 and SKOV3 cells in vitro, correlating with the inhibition of epithelial-mesenchymal transition; Additionally, NTZ suppressed the Hippo/YAP/TAZ signaling pathway both in vitro and in vivo and demonstrated a good binding activity with core genes of Hippo pathway, including Hippo, YAP, TAZ, LATS1, and LATS2. Oral administration of NTZ inhibited tumor growth in xenograft ovarian cancer mice models without causing considerable damage to major organs. Overall, these data suggest that NTZ has therapeutic potential for treating epithelial ovarian cancer.
Collapse
Affiliation(s)
- Xiangqun Yang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, 650118, PR China
| | - Zhenyan Liu
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, 650118, PR China
| | - Xin Wang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, 650118, PR China
| | - Wenda Tian
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, 650118, PR China
| | - Taoyu Zhao
- Department of Obstetrics and Gynecology, Dehong Affiliated Hospital of Kunming Medical University, Dehong People's Hospital of Yunnan Province, Yunnan, 678400, PR China
| | - Qiaoling Yang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, 650118, PR China
| | - Wenliang Li
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, 650118, PR China
| | - Linlin Yang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, 650118, PR China.
| | - Hongying Yang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, 650118, PR China.
| | - Yue Jia
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, 650118, PR China.
| |
Collapse
|
4
|
Boothe PF, Kumar VP, Kong Y, Wang K, Levinson H, Mu D, Brown ML. Radiation Induced Skin Fibrosis (RISF): Opportunity for Angiotensin II-Dependent Intervention. Int J Mol Sci 2024; 25:8261. [PMID: 39125831 PMCID: PMC11312688 DOI: 10.3390/ijms25158261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Medical procedures, such as radiation therapy, are a vital element in treating many cancers, significantly contributing to improved survival rates. However, a common long-term complication of such exposure is radiation-induced skin fibrosis (RISF), a complex condition that poses substantial physical and psychological challenges. Notably, about 50% of patients undergoing radiation therapy may achieve long-term remission, resulting in a significant number of survivors managing the aftereffects of their treatment. This article delves into the intricate relationship between RISF, reactive oxygen species (ROS), and angiotensin II (Ang II) signaling. It proposes the underlying mechanisms and examines potential treatments for mitigating skin fibrosis. The primary goal is to offer essential insights in order to better care for and improve the quality of life of cancer survivors who face the risk of developing RISF.
Collapse
Affiliation(s)
- Patricia F. Boothe
- Department of Internal Medicine, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Vidya P. Kumar
- Armed Forces Radiobiology Research Institute, The Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Yali Kong
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (Y.K.); (D.M.)
| | - Kan Wang
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (Y.K.); (D.M.)
| | - Howard Levinson
- The Center for Plastic Surgery at Sentara, 301 Riverview Ave. #400, Norfolk, VA 23510, USA;
| | - David Mu
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (Y.K.); (D.M.)
- Leroy T. Canoles Jr. Cancer Research Center, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Milton L. Brown
- Department of Internal Medicine, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| |
Collapse
|
5
|
Pennel K, Dutton L, Melissourgou-Syka L, Roxburgh C, Birch J, Edwards J. Novel radiation and targeted therapy combinations for improving rectal cancer outcomes. Expert Rev Mol Med 2024; 26:e14. [PMID: 38623751 PMCID: PMC11140547 DOI: 10.1017/erm.2024.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 01/29/2024] [Accepted: 03/22/2024] [Indexed: 04/17/2024]
Abstract
Neoadjuvant radiotherapy (RT) is commonly used as standard treatment for rectal cancer. However, response rates are variable and survival outcomes remain poor, highlighting the need to develop new therapeutic strategies. Research is focused on identifying novel methods for sensitising rectal tumours to RT to enhance responses and improve patient outcomes. This can be achieved through harnessing tumour promoting effects of radiation or preventing development of radio-resistance in cancer cells. Many of the approaches being investigated involve targeting the recently published new dimensions of cancer hallmarks. This review article will discuss key radiation and targeted therapy combination strategies being investigated in the rectal cancer setting, with a focus on exploitation of mechanisms which target the hallmarks of cancer.
Collapse
Affiliation(s)
- Kathryn Pennel
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Louise Dutton
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Lydia Melissourgou-Syka
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
- CRUK Scotland Institute, Glasgow, G611BD, UK
| | - Campbell Roxburgh
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
- Academic Unit of Surgery, Glasgow Royal Infirmary, University of Glasgow, Glasgow, G4 0SF, UK
| | - Joanna Birch
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Joanne Edwards
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| |
Collapse
|
6
|
Qin F, Chen G, Yu KN, Yang M, Cao W, Kong P, Peng S, Sun M, Nie L, Han W. Golgi Phosphoprotein 3 Mediates Radiation-Induced Bystander Effect via ERK/EGR1/TNF-α Signal Axis. Antioxidants (Basel) 2022; 11:2172. [PMID: 36358544 PMCID: PMC9686538 DOI: 10.3390/antiox11112172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 07/30/2023] Open
Abstract
The radiation-induced bystander effect (RIBE), an important non-targeted effect of radiation, has been proposed to be associated with irradiation-caused secondary cancers and reproductive damage beyond the irradiation-treated area after radiotherapy. However, the mechanisms for RIBE signal(s) regulation and transduction are not well understood. In the present work, we found that a Golgi protein, GOLPH3, was involved in RIBE transduction. Knocking down GOLPH3 in irradiated cells blocked the generation of the RIBE, whereas re-expression of GOLPH3 in knockdown cells rescued the RIBE. Furthermore, TNF-α was identified as an important intercellular signal molecule in the GOLPH3-mediated RIBE. A novel signal axis, GOLPH3/ERK/EGR1, was discovered to modulate the transcription of TNF-α and determine the level of released TNF-α. Our findings provide new insights into the molecular mechanism of the RIBE and a potential target for RIBE modulation.
Collapse
Affiliation(s)
- Feng Qin
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Scinece Island Branch, Graduate School of USTC, Hefei 230026, China
- Institute of Sericultural, Anhui Academy of Agricultural Sciences, Hefei 230061, China
| | - Guodong Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| | - Kwan Ngok Yu
- Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong 999077, Hong Kong
- State Key Laboratory in Marine Pollution, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong 999077, Hong Kong
| | - Miaomiao Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Scinece Island Branch, Graduate School of USTC, Hefei 230026, China
| | - Wei Cao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Scinece Island Branch, Graduate School of USTC, Hefei 230026, China
| | - Peizhong Kong
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Shengjie Peng
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Scinece Island Branch, Graduate School of USTC, Hefei 230026, China
| | - Mingyu Sun
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Scinece Island Branch, Graduate School of USTC, Hefei 230026, China
| | - Lili Nie
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Institute of Sericultural, Anhui Academy of Agricultural Sciences, Hefei 230061, China
| | - Wei Han
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Institute of Sericultural, Anhui Academy of Agricultural Sciences, Hefei 230061, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215006, China
| |
Collapse
|
7
|
Tian YT, Ma LP, Ding CY, Liu MM, Wang SN, Tian M, Gao L, Liu QJ. Autophagy regulates X-ray radiation-induced premature senescence through STAT3-Beclin1-p62 pathway in lung adenocarcinoma cells. Int J Radiat Biol 2022; 98:1432-1441. [DOI: 10.1080/09553002.2022.2055799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Yu-Ting Tian
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, China Centers for Disease Control, Beijing, P.R. China
| | - Li-Ping Ma
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, China Centers for Disease Control, Beijing, P.R. China
| | - Chun-Yan Ding
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, China Centers for Disease Control, Beijing, P.R. China
- Qingdao Central Medical Group, Qingdao, P.R. China
| | - Meng-Meng Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, China Centers for Disease Control, Beijing, P.R. China
| | - Si-Nian Wang
- PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Mei Tian
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, China Centers for Disease Control, Beijing, P.R. China
| | - Ling Gao
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, China Centers for Disease Control, Beijing, P.R. China
| | - Qing-Jie Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, China Centers for Disease Control, Beijing, P.R. China
| |
Collapse
|
8
|
Sweha SR, Chung C, Natarajan SK, Panwalkar P, Pun M, Ghali A, Bayliss J, Pratt D, Shankar A, Ravikumar V, Rao A, Cieslik M, Wilder-Romans K, Scott AJ, Wahl DR, Jessa S, Kleinman CL, Jabado N, Mackay A, Jones C, Martinez D, Santi M, Judkins AR, Yadav VN, Qin T, Phoenix TN, Koschmann CJ, Baker SJ, Chinnaiyan AM, Venneti S. Epigenetically defined therapeutic targeting in H3.3G34R/V high-grade gliomas. Sci Transl Med 2021; 13:eabf7860. [PMID: 34644147 DOI: 10.1126/scitranslmed.abf7860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
High-grade gliomas with arginine or valine substitutions of the histone H3.3 glycine-34 residue (H3.3G34R/V) carry a dismal prognosis, and current treatments, including radiotherapy and chemotherapy, are not curative. Because H3.3G34R/V mutations reprogram epigenetic modifications, we undertook a comprehensive epigenetic approach using ChIP sequencing and ChromHMM computational analysis to define therapeutic dependencies in H3.3G34R/V gliomas. Our analyses revealed a convergence of epigenetic alterations, including (i) activating epigenetic modifications on histone H3 lysine (K) residues such as H3K36 trimethylation (H3K36me3), H3K27 acetylation (H3K27ac), and H3K4 trimethylation (H3K4me3); (ii) DNA promoter hypomethylation; and (iii) redistribution of repressive histone H3K27 trimethylation (H3K27me3) to intergenic regions at the leukemia inhibitory factor (LIF) locus to drive increased LIF abundance and secretion by H3.3G34R/V cells. LIF activated signal transducer and activator of transcription 3 (STAT3) signaling in an autocrine/paracrine manner to promote survival of H3.3G34R/V glioma cells. Moreover, immunohistochemistry and single-cell RNA sequencing from H3.3G34R/V patient tumors revealed high STAT3 protein and RNA expression, respectively, in tumor cells with both inter- and intratumor heterogeneity. We targeted STAT3 using a blood-brain barrier–penetrable small-molecule inhibitor, WP1066, currently in clinical trials for adult gliomas. WP1066 treatment resulted in H3.3G34R/V tumor cell toxicity in vitro and tumor suppression in preclinical mouse models established with KNS42 cells, SJ-HGGx42-c cells, or in utero electroporation techniques. Our studies identify the LIF/STAT3 pathway as a key epigenetically driven and druggable vulnerability in H3.3G34R/V gliomas. This finding could inform development of targeted, combination therapies for these lethal brain tumors.
Collapse
Affiliation(s)
- Stefan R Sweha
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chan Chung
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Siva Kumar Natarajan
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Molecular and Cellular Pathology Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Pooja Panwalkar
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Matthew Pun
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Amer Ghali
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jill Bayliss
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Drew Pratt
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anand Shankar
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Visweswaran Ravikumar
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Marcin Cieslik
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Andrew J Scott
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Selin Jessa
- Quantitative Life Sciences, McGill University, Montreal, Quebec H3A 2A7, Canada.,Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec H3T 1E2, Canada
| | - Claudia L Kleinman
- Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec H3T 1E2, Canada.,Department of Human Genetics, McGill University, Montreal, Quebec H3A 0C7, Canada
| | - Nada Jabado
- Department of Human Genetics, McGill University, Montreal, Quebec H3A 0C7, Canada.,Department of Pediatrics, McGill University, and Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Alan Mackay
- Division of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London SM2 5NG, UK
| | - Chris Jones
- Division of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London SM2 5NG, UK
| | - Daniel Martinez
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Mariarita Santi
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexander R Judkins
- Department of Pathology, Children's Hospital of Los Angeles, Los Angeles, CA 90027, USA
| | - Viveka Nand Yadav
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Timothy N Phoenix
- Division of Pharmaceutical Sciences, College of Pharmacy, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Carl J Koschmann
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Suzanne J Baker
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Michigan Center for Translational Pathology, Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sriram Venneti
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
9
|
Wang CI, Chang YF, Sie ZL, Ho AS, Chang JS, Peng CL, Cheng CC. Irradiation Suppresses IFNγ-Mediated PD-L1 and MCL1 Expression in EGFR-Positive Lung Cancer to Augment CD8 + T Cells Cytotoxicity. Cells 2021; 10:cells10102515. [PMID: 34685495 PMCID: PMC8533912 DOI: 10.3390/cells10102515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022] Open
Abstract
Tumor cells express immune checkpoints to exhaust CD8+ T cells. Irradiation damages tumor cells and augments tumor immunotherapy in clinical applications. However, the radiotherapy-mediated molecular mechanism affecting CD8+ T cell activity remains elusive. We aimed to uncover the mechanism of radiotherapy augmenting cytotoxic CD8+ T cells in non-small-cell lung cancer (NSCLC). EGFR-positive NSCLC cell lines were co-cultured with CD8+ T cells from healthy volunteers. Tumor cell viability and apoptosis were consequently measured. IFNγ was identified secreted by CD8+ T cells and PBMCs. Therefore, RNAseq was used to screen the IFNγ-mediated gene expression in A549 cells. The irradiation effect to IFNγ-mediated gene expression was investigated using qPCR and western blots. We found that the co-culture of tumor cells stimulated the increase of granzyme B and IFNγ in CD8+ T, but A549 exhibited resistance against CD8+ T cytotoxicity compared to HCC827. Irradiation inhibited A549 proliferation and enhanced apoptosis, augmenting PBMCs-mediated cytotoxicity against A549. We found that IFNγ simultaneously increased phosphorylation on STAT1 and STAT3 in EGFR-positive lung cancer, resulting in overexpression of PD-L1 (p < 0.05). In RNAseq analysis, MCL1 was identified and increased by the IFNγ-STAT3 axis (p < 0.05). We demonstrated that irradiation specifically inhibited phosphorylation on STAT1 and STAT3 in IFNγ-treated A549, resulting in reductions of PD-L1 and MCL1 (both p < 0.05). Moreover, knockdowns of STAT3 and MCL1 increased the PBMCs-mediated anti-A549 effect. This study demonstrated that A549 expressed MCL1 to resist CD8+ T cell-mediated tumor apoptosis. In addition, we found that irradiation suppressed IFNγ-mediated STAT3 phosphorylation and PD-L1 and MCL1 expression, revealing a potential mechanism of radiotherapy augmenting immune surveillance.
Collapse
Affiliation(s)
- Chun-I. Wang
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Linkou 333, Taiwan; (C.-I.W.); (Z.-L.S.)
| | - Yi-Fang Chang
- Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 104, Taiwan;
- Laboratory of Good Clinical Research Center, Department of Medical Research, Mackay Memorial Hospital, Tamsui District, New Taipei City 251, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
| | - Zong-Lin Sie
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Linkou 333, Taiwan; (C.-I.W.); (Z.-L.S.)
| | - Ai-Sheng Ho
- Division of Gastroenterology, Cheng Hsin General Hospital, Taipei 112, Taiwan;
| | - Jung-Shan Chang
- Graduate Institute of Medical Sciences, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Cheng-Liang Peng
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan 325, Taiwan;
| | - Chun-Chia Cheng
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Linkou 333, Taiwan; (C.-I.W.); (Z.-L.S.)
- Correspondence:
| |
Collapse
|
10
|
Dahl H, Eide DM, Tengs T, Duale N, Kamstra JH, Oughton DH, Olsen AK. Perturbed transcriptional profiles after chronic low dose rate radiation in mice. PLoS One 2021; 16:e0256667. [PMID: 34428250 PMCID: PMC8384182 DOI: 10.1371/journal.pone.0256667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
Adverse health outcomes of ionizing radiation given chronically at low dose rates are highly debated, a controversy also relevant for other stressors. Increased knowledge is needed for a more comprehensive understanding of the damaging potential of ionizing radiation from all dose rates and doses. There is a lack of relevant low dose rate data that is partly ascribed to the rarity of exposure facilities allowing chronic low dose rate exposures. Using the FIGARO facility, we assessed early (one day post-radiation) and late (recovery time of 100-200 days) hepatic genome-wide transcriptional profiles in male mice of two strains (CBA/CaOlaHsd and C57BL/6NHsd) exposed chronically to a low dose rate (2.5 mGy/h; 1200h, LDR), a mid-dose rate (10 mGy/h; 300h, MDR) and acutely to a high dose rate (100 mGy/h; 30h, HDR) of gamma irradiation, given to an equivalent total dose of 3 Gy. Dose-rate and strain-specific transcriptional responses were identified. Differently modulated transcriptional responses across all dose rate exposure groups were evident by the representation of functional biological pathways. Evidence of changed epigenetic regulation (global DNA methylation) was not detected. A period of recovery markedly reduced the number of differentially expressed genes. Using enrichment analysis to identify the functional significance of the modulated genes, perturbed signaling pathways associated with both cancer and non-cancer effects were observed, such as lipid metabolism and inflammation. These pathways were seen after chronic low dose rate and were not restricted to the acute high dose rate exposure. The transcriptional response induced by chronic low dose rate ionizing radiation suggests contribution to conditions such as cardiovascular diseases. We contribute with novel genome wide transcriptional data highlighting dose-rate-specific radiation responses and emphasize the importance of considering both dose rate, duration of exposure, and variability in susceptibility when assessing risks from ionizing radiation.
Collapse
Affiliation(s)
- Hildegunn Dahl
- Department of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
- Centre for Environmental Radiation (CERAD), Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Dag M. Eide
- Department of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
- Centre for Environmental Radiation (CERAD), Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Torstein Tengs
- Department of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
- Centre for Environmental Radiation (CERAD), Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Nur Duale
- Department of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
- Centre for Environmental Radiation (CERAD), Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Jorke H. Kamstra
- Centre for Environmental Radiation (CERAD), Norwegian University of Life Sciences (NMBU), Ås, Norway
- Faculty of Veterinary Medicine, Department of Population Health Sciences, Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Deborah H. Oughton
- Centre for Environmental Radiation (CERAD), Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Ann-Karin Olsen
- Department of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
- Centre for Environmental Radiation (CERAD), Norwegian University of Life Sciences (NMBU), Ås, Norway
| |
Collapse
|
11
|
Wang F, Ma X, Mao G, Zhang X, Kong Z. STAT3 enhances radiation-induced tumor migration, invasion and stem-like properties of bladder cancer. Mol Med Rep 2020; 23:87. [PMID: 33236137 PMCID: PMC7716396 DOI: 10.3892/mmr.2020.11728] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 10/21/2020] [Indexed: 01/17/2023] Open
Abstract
Bladder cancer (BCa) is the most common cancer of the human urinary system, and is associated with poor patient prognosis and a high recurrence rate. Cancer stem cells (CSCs) are the primary cause of tumor recurrence and metastasis, possessing self-renewal properties and resistance to radiation therapy. Our previous studies indicated that phosphorylated signal transduction and transcription activator 3 (STAT3) may be a potential biomarker to predict radiation tolerance and tumor recurrence in patients with BCa, following conventional radiotherapy. The aim of the present study was to investigate the underlying mechanism of STAT3 in the radio-resistance of BCa cells. It was found that fractionated irradiation promoted the activation of two STAT3-associated CSCs signaling pathways in BCa cells, namely suppressor of variegation 3–9 homolog 1/GATA binding protein 3/STAT3 and Janus kinase 2/STAT3. Surviving cells exhibited elevated migratory and invasive abilities, enhanced CSC-like characteristics and radio-resistance. Furthermore, knockdown of STAT3 expression or inhibition of STAT3 activation markedly decreased the self-renewal ability and tumorigenicity of radiation-resistant BCa cells. Kaplan-Meier analysis revealed that decreased STAT3 mRNA levels were associated with increased overall survival times in patients with BCa. Taken together, these data indicated that STAT3 may be an effective therapeutic target for inhibiting the progression, metastasis and recurrence of BCa in patients receiving radiotherapy.
Collapse
Affiliation(s)
- Fang Wang
- Department of Radiobiology, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Xiangli Ma
- Department of Radiobiology, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Guangmin Mao
- Department of Radiobiology, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Xiangyan Zhang
- Department of Radiobiology, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Zhaolu Kong
- Department of Radiobiology, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
12
|
Yang PL, Liu LX, Li EM, Xu LY. STAT3, the Challenge for Chemotherapeutic and Radiotherapeutic Efficacy. Cancers (Basel) 2020; 12:cancers12092459. [PMID: 32872659 PMCID: PMC7564975 DOI: 10.3390/cancers12092459] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 02/05/2023] Open
Abstract
Chemoradiotherapy is one of the most effective and extensively used strategies for cancer treatment. Signal transducer and activator of transcription 3 (STAT3) regulates vital biological processes, such as cell proliferation and cell growth. It is constitutively activated in various cancers and limits the application of chemoradiotherapy. Accumulating evidence suggests that STAT3 regulates resistance to chemotherapy and radiotherapy and thereby impairs therapeutic efficacy by mediating its feedback loop and several target genes. The alternative splicing product STAT3β is often identified as a dominant-negative regulator, but it enhances sensitivity to chemotherapy and offers a new and challenging approach to reverse therapeutic resistance. We focus here on exploring the role of STAT3 in resistance to receptor tyrosine kinase (RTK) inhibitors and radiotherapy, outlining the potential of targeting STAT3 to overcome chemo(radio)resistance for improving clinical outcomes, and evaluating the importance of STAT3β as a potential therapeutic approach to overcomes chemo(radio)resistance. In this review, we discuss some new insights into the effect of STAT3 and its subtype STAT3β on chemoradiotherapy sensitivity, and we explore how these insights influence clinical treatment and drug development for cancer.
Collapse
Affiliation(s)
- Ping-Lian Yang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China; (P.-L.Y.); (L.-X.L.)
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Lu-Xin Liu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China; (P.-L.Y.); (L.-X.L.)
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China; (P.-L.Y.); (L.-X.L.)
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
- Correspondence: (E.-M.L.); (L.-Y.X.); Tel.: +86-754-88900460 (L.-Y.X.); Fax: +86-754-88900847 (L.-Y.X.)
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China; (P.-L.Y.); (L.-X.L.)
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China
- Correspondence: (E.-M.L.); (L.-Y.X.); Tel.: +86-754-88900460 (L.-Y.X.); Fax: +86-754-88900847 (L.-Y.X.)
| |
Collapse
|
13
|
Ott M, Kassab C, Marisetty A, Hashimoto Y, Wei J, Zamler D, Leu JS, Tomaszowski KH, Sabbagh A, Fang D, Gupta P, Priebe W, Zielinski RJ, Burks JK, Long JP, Kong LY, Fuller GN, DeGroot J, Sulman EP, Heimberger AB. Radiation with STAT3 Blockade Triggers Dendritic Cell-T cell Interactions in the Glioma Microenvironment and Therapeutic Efficacy. Clin Cancer Res 2020; 26:4983-4994. [PMID: 32605912 DOI: 10.1158/1078-0432.ccr-19-4092] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/14/2020] [Accepted: 06/24/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Patients with central nervous system (CNS) tumors are typically treated with radiotherapy, but this is not curative and results in the upregulation of phosphorylated STAT3 (p-STAT3), which drives invasion, angiogenesis, and immune suppression. Therefore, we investigated the combined effect of an inhibitor of STAT3 and whole-brain radiotherapy (WBRT) in a murine model of glioma. EXPERIMENTAL DESIGN C57BL/6 mice underwent intracerebral implantation of GL261 glioma cells, WBRT, and treatment with WP1066, a blood-brain barrier-penetrant inhibitor of the STAT3 pathway, or the two in combination. The role of the immune system was evaluated using tumor rechallenge strategies, immune-incompetent backgrounds, immunofluorescence, immune phenotyping of tumor-infiltrating immune cells (via flow cytometry), and NanoString gene expression analysis of 770 immune-related genes from immune cells, including those directly isolated from the tumor microenvironment. RESULTS The combination of WP1066 and WBRT resulted in long-term survivors and enhanced median survival time relative to monotherapy in the GL261 glioma model (combination vs. control P < 0.0001). Immunologic memory appeared to be induced, because mice were protected during subsequent tumor rechallenge. The therapeutic effect of the combination was completely lost in immune-incompetent animals. NanoString analysis and immunofluorescence revealed immunologic reprograming in the CNS tumor microenvironment specifically affecting dendritic cell antigen presentation and T-cell effector functions. CONCLUSIONS This study indicates that the combination of STAT3 inhibition and WBRT enhances the therapeutic effect against gliomas in the CNS by inducing dendritic cell and T-cell interactions in the CNS tumor.
Collapse
Affiliation(s)
- Martina Ott
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cynthia Kassab
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anantha Marisetty
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuuri Hashimoto
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jun Wei
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Zamler
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jia-Shiun Leu
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Karl-Heinz Tomaszowski
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aria Sabbagh
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dexing Fang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pravesh Gupta
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Waldemar Priebe
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rafal J Zielinski
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James P Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ling-Yuan Kong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregory N Fuller
- Department of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John DeGroot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Erik P Sulman
- Department of Radiation Oncology, NYU Langone Health Perlmutter Cancer Center, New York, New York
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
14
|
Bharadwaj U, Kasembeli MM, Robinson P, Tweardy DJ. Targeting Janus Kinases and Signal Transducer and Activator of Transcription 3 to Treat Inflammation, Fibrosis, and Cancer: Rationale, Progress, and Caution. Pharmacol Rev 2020; 72:486-526. [PMID: 32198236 PMCID: PMC7300325 DOI: 10.1124/pr.119.018440] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Before it was molecularly cloned in 1994, acute-phase response factor or signal transducer and activator of transcription (STAT)3 was the focus of intense research into understanding the mammalian response to injury, particularly the acute-phase response. Although known to be essential for liver production of acute-phase reactant proteins, many of which augment innate immune responses, molecular cloning of acute-phase response factor or STAT3 and the research this enabled helped establish the central function of Janus kinase (JAK) family members in cytokine signaling and identified a multitude of cytokines and peptide hormones, beyond interleukin-6 and its family members, that activate JAKs and STAT3, as well as numerous new programs that their activation drives. Many, like the acute-phase response, are adaptive, whereas several are maladaptive and lead to chronic inflammation and adverse consequences, such as cachexia, fibrosis, organ dysfunction, and cancer. Molecular cloning of STAT3 also enabled the identification of other noncanonical roles for STAT3 in normal physiology, including its contribution to the function of the electron transport chain and oxidative phosphorylation, its basal and stress-related adaptive functions in mitochondria, its function as a scaffold in inflammation-enhanced platelet activation, and its contributions to endothelial permeability and calcium efflux from endoplasmic reticulum. In this review, we will summarize the molecular and cellular biology of JAK/STAT3 signaling and its functions under basal and stress conditions, which are adaptive, and then review maladaptive JAK/STAT3 signaling in animals and humans that lead to disease, as well as recent attempts to modulate them to treat these diseases. In addition, we will discuss how consideration of the noncanonical and stress-related functions of STAT3 cannot be ignored in efforts to target the canonical functions of STAT3, if the goal is to develop drugs that are not only effective but safe. SIGNIFICANCE STATEMENT: Key biological functions of Janus kinase (JAK)/signal transducer and activator of transcription (STAT)3 signaling can be delineated into two broad categories: those essential for normal cell and organ development and those activated in response to stress that are adaptive. Persistent or dysregulated JAK/STAT3 signaling, however, is maladaptive and contributes to many diseases, including diseases characterized by chronic inflammation and fibrosis, and cancer. A comprehensive understanding of JAK/STAT3 signaling in normal development, and in adaptive and maladaptive responses to stress, is essential for the continued development of safe and effective therapies that target this signaling pathway.
Collapse
Affiliation(s)
- Uddalak Bharadwaj
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Moses M Kasembeli
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Prema Robinson
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - David J Tweardy
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
15
|
Li Q, Zong Y, Li K, Jie X, Hong J, Zhou X, Wu B, Li Z, Zhang S, Wu G, Meng R. Involvement of endothelial CK2 in the radiation induced perivascular resistant niche (PVRN) and the induction of radioresistance for non-small cell lung cancer (NSCLC) cells. Biol Res 2019; 52:22. [PMID: 30992075 PMCID: PMC6466699 DOI: 10.1186/s40659-019-0231-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 04/06/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Tumor microenvironment (TME) plays a vital role in determining the outcomes of radiotherapy. As an important component of TME, vascular endothelial cells are involved in the perivascular resistance niche (PVRN), which is formed by inflammation or cytokine production induced by ionizing radiation (IR). Protein kinase CK2 is a constitutively active serine/threonine kinase which plays a vital role in cell proliferation and inflammation. In this study, we investigated the potential role of CK2 in PVRN after IR exposure. RESULT Specific CK2 inhibitors, Quinalizarin and CX-4945, were employed to effectively suppressed the kinase activity of CK2 in human umbilical vein endothelial cells (HUVECs) without affecting their viability. Results showing that conditioned medium from IR-exposed HUVECs increased cell viability of A549 and H460 cells, and the pretreatment of CK2 inhibitors slowed down such increment. The secretion of IL-8 and IL-6 in HUVECs was induced after exposure with IR, but significantly inhibited by the addition of CK2 inhibitors. Furthermore, IR exposure elevated the nuclear phosphorylated factor-κB (NF-κB) p65 expression in HUVECs, which was a master factor regulating cytokine production. But when pretreated with CK2 inhibitors, such elevation was significantly suppressed. CONCLUSION This study indicated that protein kinase CK2 is involved in the key process of the IR induced perivascular resistant niche, namely cytokine production, by endothelial cells, which finally led to radioresistance of non-small cell lung cancer cells. Thus, the inhibition of CK2 may be a promising way to improve the outcomes of radiation in non-small cell lung cancer cells.
Collapse
Affiliation(s)
- Qianwen Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yan Zong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ke Li
- Pharmacy Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaohua Jie
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiaxin Hong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoshu Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bian Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhenyu Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
16
|
Zhu H, Ruan S, Jia F, Chu J, Zhu Y, Huang Y, Liu G. In vitro and in vivo superior radiosensitizing effect of berbamine for head and neck squamous cell carcinoma. Onco Targets Ther 2018; 11:8117-8125. [PMID: 30532553 PMCID: PMC6241700 DOI: 10.2147/ott.s171212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Berbamine (BBM), one of the bis-benzylisoquinoline products isolated from Berberis amurensis, has been demonstrated for its anticancer effect against leukemia, breast cancer, liver cancer, etc. There are some studies focusing on the chemosensitization effect of BBM. However, there is no report about whether BBM could enhance the anticancer effect of radiation, which made us to explore the possible radiosensitization effect of BBM. Materials and methods Here, in vitro cytotoxicity of BBM was evaluated on two kinds of head and neck squamous cancer cell lines. Clonogenic assay was performed to study the radiosensitization effect of BBM. Western blot was utilized to elucidate the possible mechanism underlying the radiosensitization effect. Results BBM effectively inhibited the growth of two kinds of cancer cells in a time- and dose-dependent manner. Radiation plus BBM led to significantly more reduction of the colony-forming ability of cancer cells when compared with radiation alone. BBM plus radiation led to the most reduction of STAT3 phosphorylation, followed by the significant decrease of the ratio of Bax/Bcl-2. In vivo study demonstrated that the combinational administration of BBM and radiation generated the most significant tumor-delaying effect among all of the treatment regimens. Conclusion We reported, in the current study, the potential role of BBM in not only treating cancer by itself but also offering a promising way to improve the efficacy of radiotherapy by inhibiting the activation of STAT3 and subsequently inducing the apoptosis of cancer.
Collapse
Affiliation(s)
- Hongmei Zhu
- Department of Otolaryngology, Jiangsu Taizhou People's Hospital, Taizhou 225300, Jiangsu Province, China,
| | - Shu Ruan
- Department of Endocrinology, Yancheng Third Hospital, The Affiliated Hospital of Southeast University Medical College, Yancheng 224001, Jiangsu Province, China
| | - Feng Jia
- Department of Neurosurgery, Yancheng City No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong Medical College, Yancheng 224005, Jiangsu Province, China
| | - Jiusheng Chu
- Department of Otolaryngology, Jiangsu Taizhou People's Hospital, Taizhou 225300, Jiangsu Province, China,
| | - Yong Zhu
- Department of Otolaryngology, Jiangsu Taizhou People's Hospital, Taizhou 225300, Jiangsu Province, China,
| | - Yongjiu Huang
- Department of Otolaryngology, Jiangsu Taizhou People's Hospital, Taizhou 225300, Jiangsu Province, China,
| | - Guan Liu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, Zhejiang Province, China,
| |
Collapse
|
17
|
McKelvey KJ, Hudson AL, Back M, Eade T, Diakos CI. Radiation, inflammation and the immune response in cancer. Mamm Genome 2018; 29:843-865. [PMID: 30178305 PMCID: PMC6267675 DOI: 10.1007/s00335-018-9777-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/22/2018] [Indexed: 01/17/2023]
Abstract
Radiation is an important component of cancer treatment with more than half of all patients receive radiotherapy during their cancer experience. While the impact of radiation on tumour morphology is routinely examined in the pre-clinical and clinical setting, the impact of radiation on the tumour microenvironment and more specifically the inflammatory/immune response is less well characterised. Inflammation is a key contributor to short- and long-term cancer eradication, with significant tumour and normal tissue consequences. Therefore, the role of radiation in modulating the inflammatory response is highly topical given the current wave of targeted and immuno-therapeutic treatments for cancer. This review provides a general overview of how radiation modulates the inflammatory and immune response—(i) how radiation induces the inflammatory/immune system, (ii) the cellular changes that take place, (iii) how radiation dose delivery affects the immune response, and (iv) a discussion on research directions to improve patient survival, reduce side effects, improve quality of life, and reduce financial costs in the immediate future. Harnessing the benefits of radiation on the immune response will enhance its maximal therapeutic benefit and reduce radiation-induced toxicity.
Collapse
Affiliation(s)
- Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Northern Sydney Local Health District Research and the Northern Clinical School, University of Sydney, St Leonards, NSW, 2065, Australia. .,Sydney Neuro-Oncology Group, North Shore Private Hospital, St Leonards, NSW, 2065, Australia. .,Sydney Vital Translational Research Centre, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia.
| | - Amanda L Hudson
- Bill Walsh Translational Cancer Research Laboratory, Northern Sydney Local Health District Research and the Northern Clinical School, University of Sydney, St Leonards, NSW, 2065, Australia.,Sydney Neuro-Oncology Group, North Shore Private Hospital, St Leonards, NSW, 2065, Australia.,Sydney Vital Translational Research Centre, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Michael Back
- Sydney Neuro-Oncology Group, North Shore Private Hospital, St Leonards, NSW, 2065, Australia.,Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Tom Eade
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Connie I Diakos
- Sydney Vital Translational Research Centre, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia.,Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| |
Collapse
|
18
|
Najafi M, Motevaseli E, Shirazi A, Geraily G, Rezaeyan A, Norouzi F, Rezapoor S, Abdollahi H. Mechanisms of inflammatory responses to radiation and normal tissues toxicity: clinical implications. Int J Radiat Biol 2018; 94:335-356. [PMID: 29504497 DOI: 10.1080/09553002.2018.1440092] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 01/23/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE Cancer treatment is one of the most challenging diseases in the present era. Among a few modalities for cancer therapy, radiotherapy plays a pivotal role in more than half of all treatments alone or combined with other cancer treatment modalities. Management of normal tissue toxicity induced by radiation is one of the most important limiting factors for an appropriate radiation treatment course. The evaluation of mechanisms of normal tissue toxicity has shown that immune responses especially inflammatory responses play a key role in both early and late side effects of exposure to ionizing radiation (IR). DNA damage and cell death, as well as damage to some organelles such as mitochondria initiate several signaling pathways that result in the response of immune cells. Massive cell damage which is a common phenomenon following exposure to a high dose of IR cause secretion of a lot of inflammatory mediators including cytokines and chemokines. These mediators initiate different changes in normal tissues that may continue for a long time after irradiation. In this study, we reviewed the mechanisms of inflammatory responses to IR that are involved in normal tissue toxicity and considered as the most important limiting factors in radiotherapy. Also, we introduced some agents that have been proposed for management of these responses. CONCLUSIONS The early inflammation during the radiation treatment is often a limiting factor in radiotherapy. In addition to the limiting factors, chronic inflammatory responses may increase the risk of second primary cancers through continuous free radical production, attenuation of tumor suppressor genes, and activation of oncogenes. Moreover, these effects may influence non-irradiated tissues through a mechanism named bystander effect.
Collapse
Affiliation(s)
- Masoud Najafi
- a Radiology and Nuclear Medicine Department, School of Paramedical Sciences , Kermanshah University of Medical Science , Kermanshah , Iran
| | - Elahe Motevaseli
- b Department of Molecular Medicine, School of Advanced Technologies in Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Alireza Shirazi
- c Department of Medical Physics and Biomedical Engineering, Faculty of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Ghazale Geraily
- c Department of Medical Physics and Biomedical Engineering, Faculty of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Abolhasan Rezaeyan
- d Department of Medical Physics, School of Medicine , Iran University of Medical Sciences , Tehran , Iran
| | - Farzad Norouzi
- e Science and Research Branch , Azad University , Tehran , Iran
| | - Saeed Rezapoor
- f Department of Radiology, Faculty of Paramedical Sciences , Tehran University of Medical Sciences , Tehran , Iran
| | - Hamid Abdollahi
- d Department of Medical Physics, School of Medicine , Iran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
19
|
Galal SM, Abdel-Rafei MK, Hasan HF. Cholinergic and cytoprotective signaling cascades mediate the mitigative effect of erythropoietin on acute radiation syndrome. Can J Physiol Pharmacol 2017; 96:442-458. [PMID: 29220591 DOI: 10.1139/cjpp-2017-0578] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present investigation aimed to evaluate the radiomitigative efficacy of the recombinant human erythropoietin (EPO) against acute radiation syndrome (ARS) in a rat model. Rats were irradiated with a single sublethal dose of γ-radiation (7 Gy; total body irradiation; TBI) on the 1st day of experimental course, then received EPO (5000 IU/kg; i.p.) 24 h after irradiation, and rats were observed for 30 days of survival analysis. Administration of EPO improved 30-day survival, alleviated TBI-induced myelosuppression and pancytopenia, by augmenting lymphocytes and other white blood cells in the peripheral blood of rats, while bone marrow and spleen cellularity were restored. EPO post-exposure treatment alleviated hepatotoxicity biomarkers and restored splenic function. EPO abrogated radiation-induced oxidative stress through the upregulation of the cholinergic anti-inflammatory nicotinic acetylcholine receptor (α-7-nAChR) and the pro-survival Janus kinase-2 and signal transducers and activators of transcription JAK-2/STAT-3 signaling mediated via enhancing nuclear factor erythroid-2 related factor-2 (Nrf-2) cytoprotective machinery in liver and spleen of irradiated rats. Moreover, EPO treatment prevented hepatic and splenic apoptosis. The present study establishes the implication of α-7-nAChR-JAK-2/STAT-3-Nrf-2 signaling cascade in the radiomitigative potential of EPO against ARS.
Collapse
Affiliation(s)
- Shereen Mohamed Galal
- a Health Radiation Research Department, National Center for Radiation Research and Technology, Atomic Energy Authority, P.O. Box 29, Nasr City, Cairo, Egypt
| | - Mohamed Khairy Abdel-Rafei
- b Radiation Biology Department, National Center for Radiation Research and Technology, Atomic Energy Authority, P.O. Box 29, Nasr City, Cairo, Egypt
| | - Hesham Farouk Hasan
- b Radiation Biology Department, National Center for Radiation Research and Technology, Atomic Energy Authority, P.O. Box 29, Nasr City, Cairo, Egypt
| |
Collapse
|
20
|
Xu F, Li X, Yan L, Yuan N, Fang Y, Cao Y, Xu L, Zhang X, Xu L, Ge C, An N, Jiang G, Xie J, Zhang H, Jiang J, Li X, Yao L, Zhang S, Zhou D, Wang J. Autophagy Promotes the Repair of Radiation-Induced DNA Damage in Bone Marrow Hematopoietic Cells via Enhanced STAT3 Signaling. Radiat Res 2017; 187:382-396. [PMID: 28327001 DOI: 10.1667/rr14640.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Autophagy protects hematopoietic cells from radiation damage in part by promoting DNA damage repair. However, the molecular mechanisms by which autophagy regulates DNA damage repair remain largely elusive. Here, we report that this radioprotective effect of autophagy depends on STAT3 signaling in murine bone marrow mononuclear cells (BM-MNCs). Specifically, we found that STAT3 activation and nuclear translocation in BM-MNCs were increased by activation of autophagy with an mTOR inhibitor and decreased by knockout of the autophagy gene Atg7. The autophagic regulation of STAT3 activation is likely mediated by induction of KAP1 degradation, because we showed that KAP1 directly interacted with STAT3 in the cytoplasm and knockdown of KAP1 increased the phosphorylation and nuclear translocation of STAT3. Subsequently, activated STAT3 transcriptionally upregulated the expression of BRCA1, which increased the ability of BM-MNCs to repair radiation-induced DNA damage. This novel finding that activation of autophagy can promote DNA damage repair in BM-MNCs via the ATG-KAP1-STAT3-BRCA1 pathway suggests that autophagy plays an important role in maintaining genomic integrity of BM-MNCs and its activation may confer protection of BM-MNCs against radiation-induced genotoxic stress.
Collapse
Affiliation(s)
- Fei Xu
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Xin Li
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Lili Yan
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Na Yuan
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Yixuan Fang
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Yan Cao
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Li Xu
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Xiaoying Zhang
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Lan Xu
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Chaorong Ge
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Ni An
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Gaoyue Jiang
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Jialing Xie
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Han Zhang
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Jiayi Jiang
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Xiaotian Li
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Lei Yao
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| | - Suping Zhang
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China.,b Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas Medical Sciences, Little Rock, Arkansas 72205
| | - Daohong Zhou
- b Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas Medical Sciences, Little Rock, Arkansas 72205
| | - Jianrong Wang
- a Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University School of Medicine, Suzhou 215123, China
| |
Collapse
|
21
|
Lv Q, Hu JX, Li YJ, Xie N, Song DD, Zhao W, Yan YF, Li BS, Wang PY, Xie SY. MiR-320a effectively suppresses lung adenocarcinoma cell proliferation and metastasis by regulating STAT3 signals. Cancer Biol Ther 2017; 18:142-151. [PMID: 28106481 DOI: 10.1080/15384047.2017.1281497] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs play important roles in tumorigenesis of various types of cancers. MiR-320a can inhibits cell proliferation of some cancers, but the biologic roles of miR-320a in lung cancer need to be further studied. Here, we investigated the roles of miR-320a in suppressing the proliferation of lung adenocarcinoma cells. MiR-320a treatment was found to effectively suppress LTEP-a-2 and A549 cell proliferation, and induce more apoptotic cells with irradiation treatment compared with control treatment. Our results also showed that miR-320a, as a novel miRNA, directly regulated signal transducer and activator of transcription 3 (STAT3) and its signals, such as Bcl-2, Bax, and Caspase 3. The siRNA-inhibited STAT3 levels further proved its roles in regulating STAT3 signals. Moreover, miR-320a treatment effectively suppressed cancer cell growth in mice xenografts compared with controls, and significantly inhibited cell migration in vitro and in vivo. Our findings collectively demonstrated that miR-320a, by directly regulating STAT3 signals, not only suppressed cell proliferation and metastasis, but also enhanced irradiation-induced apoptosis of adenocarcinomia cells.
Collapse
Affiliation(s)
- Qing Lv
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Jin-Xia Hu
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - You-Jie Li
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Ning Xie
- b YanTaiShan Hospital , YanTai , Shandong , P.R. China
| | - Dan Dan Song
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Wei Zhao
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Yun-Fei Yan
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Bao-Sheng Li
- c Department of Radiation Oncology , Shandong Cancer Hospital, Shandong Academy of Medical Sciences , Jinan , P.R. China
| | - Ping-Yu Wang
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| | - Shu-Yang Xie
- a Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology , Binzhou Medical University , YanTai , Shandong , P.R. China
| |
Collapse
|
22
|
Hanson C, Cairns J, Wang L, Sinha S. Computational discovery of transcription factors associated with drug response. THE PHARMACOGENOMICS JOURNAL 2016; 16:573-582. [PMID: 26503816 PMCID: PMC4848185 DOI: 10.1038/tpj.2015.74] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 08/04/2015] [Accepted: 08/07/2015] [Indexed: 02/01/2023]
Abstract
This study integrates gene expression, genotype and drug response data in lymphoblastoid cell lines with transcription factor (TF)-binding sites from ENCODE (Encyclopedia of Genomic Elements) in a novel methodology that elucidates regulatory contexts associated with cytotoxicity. The method, GENMi (Gene Expression iN the Middle), postulates that single-nucleotide polymorphisms within TF-binding sites putatively modulate its regulatory activity, and the resulting variation in gene expression leads to variation in drug response. Analysis of 161 TFs and 24 treatments revealed 334 significantly associated TF-treatment pairs. Investigation of 20 selected pairs yielded literature support for 13 of these associations, often from studies where perturbation of the TF expression changes drug response. Experimental validation of significant GENMi associations in taxanes and anthracyclines across two triple-negative breast cancer cell lines corroborates our findings. The method is shown to be more sensitive than an alternative, genome-wide association study-based approach that does not use gene expression. These results demonstrate the utility of GENMi in identifying TFs that influence drug response and provide a number of candidates for further testing.
Collapse
Affiliation(s)
- C Hanson
- Department of Computer Science, University of Illinois at Urbana–Champaign, Urbana, IL, USA
| | - J Cairns
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - L Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - S Sinha
- Department of Computer Science and Institute of Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, IL, USA
| |
Collapse
|
23
|
Wu HH, Zhang S, Bian H, Li XX, Wang L, Pu YF, Wang YX, Guo CB. Bevacizumab Regulates Cancer Cell Migration by Activation of STAT3. Asian Pac J Cancer Prev 2016; 16:6501-6. [PMID: 26434865 DOI: 10.7314/apjcp.2015.16.15.6501] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
There are numerous clinical cases indicating that long-term use of bevacizumab may increase the invasiveness of tumors. However, to date, little is known about underlying molecular mechanisms. Therefore, the purpose of our study was to investigate effects of bevacizumab in four cancer cells lines (WSU-HN6, CAL27, Tca83, and HeLa). It was found to promote migration and invasion in the WSU-HN6 and Tca83 cases, while exerting inhibitory effects in CAL27 and HeLa cells. The signal transducer and activator of transcription (STAT) 3 inhibitors niclosamide and S3I-201 inhibited the STAT3 signal pathway, which is activated by bevacizumab. These inhibitors also substantially blocked bevacizumab-induced migration of WSU-HN6 and Tca83 cells. Bevacizumab upregulated interleukin (IL)-6 and phosphorylated (p)-STAT3 expression time-dependently. Therefore, we propose that bevacizumab has differential effects on the migration of different cancer cell lines and promotes migration via the IL-6/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Huan-Huan Wu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China E-mail : ;
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Persa E, Balogh A, Sáfrány G, Lumniczky K. The effect of ionizing radiation on regulatory T cells in health and disease. Cancer Lett 2015; 368:252-61. [PMID: 25754816 DOI: 10.1016/j.canlet.2015.03.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 02/07/2023]
Abstract
Treg cells are key elements of the immune system which are responsible for the immune suppressive phenotype of cancer patients. Interaction of Treg cells with conventional anticancer therapies might fundamentally influence cancer therapy response rates. Radiotherapy, apart from its direct tumor cell killing potential, has a contradictory effect on the antitumor immune response: it augments certain immune parameters, while it depresses others. Treg cells are intrinsically radioresistant due to reduced apoptosis and increased proliferation, which leads to their systemic and/or intratumoral enrichment. While physiologically Treg suppression is not enhanced by irradiation, this is not the case in a tumorous environment, where Tregs acquire a highly suppressive phenotype, which is further increased by radiotherapy. This is the reason why the interest for combined radiotherapy and immunotherapy approaches focusing on the abrogation of Treg suppression has increased in cancer therapy in the last few years. Here we summarize the basic mechanisms of Treg radiation response both in healthy and cancerous environments and discuss Treg-targeted pre-clinical and clinical immunotherapy approaches used in combination with radiotherapy. Finally, the discrepant findings regarding the predictive value of Tregs in therapy response are also reviewed.
Collapse
Affiliation(s)
- Eszter Persa
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Andrea Balogh
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Géza Sáfrány
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Katalin Lumniczky
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary.
| |
Collapse
|